Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2060: 1-30, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31617170

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a prevalent and important human pathogen that has been studied in a wide variety of contexts. This book provides protocols currently in use in leading laboratories in many fields of HSV-1 research. This introductory chapter gives a brief overview of HSV-1 biology and life cycle, covering basic aspects of virus structure, the prevalence of and diseases caused by the virus, replication in cultured cells, viral latency, antiviral defenses, and the mechanisms that the virus uses to counteract these defenses.


Asunto(s)
Herpes Simple/metabolismo , Herpesvirus Humano 1/fisiología , Activación Viral/fisiología , Latencia del Virus/fisiología , Animales , Línea Celular , Humanos
2.
J Cell Sci ; 132(5)2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30745338

RESUMEN

Cancers that utilize the alternative lengthening of telomeres (ALT) mechanism for telomere maintenance are often difficult to treat and have a poor prognosis. They are also commonly deficient for expression of ATRX protein, a repressor of ALT activity, and a component of promyelocytic leukemia nuclear bodies (PML NBs) that are required for intrinsic immunity to various viruses. Here, we asked whether ATRX deficiency creates a vulnerability in ALT cancer cells that could be exploited for therapeutic purposes. We showed in a range of cell types that a mutant herpes simplex virus type 1 (HSV-1) lacking ICP0, a protein that degrades PML NB components including ATRX, was ten- to one thousand-fold more effective in infecting ATRX-deficient cells than wild-type ATRX-expressing cells. Infection of co-cultured primary and ATRX-deficient cancer cells revealed that mutant HSV-1 selectively killed ATRX-deficient cells. Sensitivity to mutant HSV-1 infection also correlated inversely with PML protein levels, and we showed that ATRX upregulates PML expression at both the transcriptional and post-transcriptional levels. These data provide a basis for predicting, based on ATRX or PML levels, which tumors will respond to a selective oncolytic herpesvirus.


Asunto(s)
Herpes Simple/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Riñón/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Nuclear Ligada al Cromosoma X/deficiencia , Animales , Muerte Celular , Línea Celular Tumoral , Cricetinae , Herpes Simple/patología , Humanos , Proteínas Inmediatas-Precoces/genética , Inmunidad Innata/genética , Riñón/patología , Mutación/genética , Viroterapia Oncolítica , Proteína de la Leucemia Promielocítica/genética , Homeostasis del Telómero , Ubiquitina-Proteína Ligasas/genética
3.
Nucleic Acids Res ; 45(20): 11673-11683, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-28981850

RESUMEN

The HIRA histone chaperone complex deposits histone H3.3 into nucleosomes in a DNA replication- and sequence-independent manner. As herpesvirus genomes enter the nucleus as naked DNA, we asked whether the HIRA chaperone complex affects herpesvirus infection. After infection of primary cells with HSV or CMV, or transient transfection with naked plasmid DNA, HIRA re-localizes to PML bodies, sites of cellular anti-viral activity. HIRA co-localizes with viral genomes, binds to incoming viral and plasmid DNAs and deposits histone H3.3 onto these. Anti-viral interferons (IFN) specifically induce HIRA/PML co-localization at PML nuclear bodies and HIRA recruitment to IFN target genes, although HIRA is not required for IFN-inducible expression of these genes. HIRA is, however, required for suppression of viral gene expression, virus replication and lytic infection and restricts murine CMV replication in vivo. We propose that the HIRA chaperone complex represses incoming naked viral DNAs through chromatinization as part of intrinsic cellular immunity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , ADN Viral/metabolismo , Herpesvirus Humano 1/metabolismo , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Cromatina/virología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , ADN Viral/genética , Células HEK293 , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Chaperonas de Histonas/genética , Chaperonas de Histonas/inmunología , Humanos , Cuerpos de Inclusión/inmunología , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/virología , Ratones Endogámicos C57BL , Muromegalovirus/genética , Muromegalovirus/fisiología , Proteína de la Leucemia Promielocítica/metabolismo , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/inmunología
4.
J Virol ; 90(21): 9664-9673, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535048

RESUMEN

It is now well established that several cellular proteins that are components of promyelocytic leukemia nuclear bodies (PML NBs, also known as ND10) have restrictive effects on herpesvirus infections that are countered by viral proteins that are either present in the virion particle or are expressed during the earliest stages of infection. For example, herpes simplex virus 1 (HSV-1) immediate early (IE) protein ICP0 overcomes the restrictive effects of PML-NB components PML, Sp100, hDaxx, and ATRX while human cytomegalovirus (HCMV) IE protein IE1 targets PML and Sp100, and its tegument protein pp71 targets hDaxx and ATRX. The functions of these viral regulatory proteins are in part interchangeable; thus, both IE1 and pp71 stimulate the replication of ICP0-null mutant HSV-1, while ICP0 increases plaque formation by pp71-deficient HCMV. Here, we extend these studies by examining proteins that are expressed by Epstein-Barr virus (EBV). We report that EBV tegument protein BNRF1, discovered by other investigators to target the hDaxx/ATRX complex, increases the replication of both ICP0-null mutant HSV-1 and pp71-deficient HCMV. In addition, EBV protein EBNA-LP, which targets Sp100, also augments ICP0-null mutant HSV-1 replication. The combination of these two EBV regulatory proteins had a greater effect than each one individually. These findings reinforce the concept that disruption of the functions of PML-NB proteins is important for efficient herpesvirus infections. IMPORTANCE: Whether a herpesvirus initiates a lytic infection in a host cell or establishes quiescence or latency is influenced by events that occur soon after the viral genome has entered the host cell nucleus. Certain cellular proteins respond in a restrictive manner to the invading pathogen's DNA, while viral functions are expressed that counteract the cell-mediated repression. One aspect of cellular restriction of herpesvirus infections is mediated by components of nuclear structures known as PML nuclear bodies (PML NBs), or ND10. Members of the alpha-, beta-, and gammaherpesvirus families all express proteins that interact with, degrade, or otherwise counteract the inhibitory effects of various PML NB components. Previous work has shown that there is the potential for a functional interchange between the viral proteins expressed by alpha- and betaherpesviruses, despite a lack of obvious sequence similarity. Here, this concept is extended to include a member of the gammaherpesviruses.


Asunto(s)
Citomegalovirus/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética , Replicación Viral/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos Nucleares/genética , Autoantígenos/genética , Línea Celular , Proteínas Co-Represoras , Infecciones por Citomegalovirus/genética , ADN Helicasas/genética , Replicación del ADN/genética , Regulación Viral de la Expresión Génica/genética , Herpes Simple/genética , Infecciones por Herpesviridae/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Chaperonas Moleculares , Mutación/genética , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica/genética , Factores de Transcripción/genética , Proteína Nuclear Ligada al Cromosoma X
5.
J Virol ; 90(19): 8621-33, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27440897

RESUMEN

UNLABELLED: We previously reported that MORC3, a protein associated with promyelocytic leukemia nuclear bodies (PML NBs), is a target of herpes simplex virus 1 (HSV-1) ICP0-mediated degradation (E. Sloan, et al., PLoS Pathog 11:e1005059, 2015, http://dx.doi.org/10.1371/journal.ppat.1005059). Since it is well known that certain other components of the PML NB complex play an important role during an intrinsic immune response to HSV-1 and are also degraded or inactivated by ICP0, here we further investigate the role of MORC3 during HSV-1 infection. We demonstrate that MORC3 has antiviral activity during HSV-1 infection and that this antiviral role is counteracted by ICP0. In addition, MORC3's antiviral role extends to wild-type (wt) human cytomegalovirus (HCMV) infection, as its plaque-forming efficiency increased in MORC3-depleted cells. We found that MORC3 is recruited to sites associated with HSV-1 genomes after their entry into the nucleus of an infected cell, and in wt infections this is followed by its association with ICP0 foci prior to its degradation. The RING finger domain of ICP0 was required for degradation of MORC3, and we confirmed that no other HSV-1 protein is required for the loss of MORC3. We also found that MORC3 is required for fully efficient recruitment of PML, Sp100, hDaxx, and γH2AX to sites associated with HSV-1 genomes entering the host cell nucleus. This study further unravels the intricate ways in which HSV-1 has evolved to counteract the host immune response and reveals a novel function for MORC3 during the host intrinsic immune response. IMPORTANCE: Herpesviruses have devised ways to manipulate the host intrinsic immune response to promote their own survival and persistence within the human population. One way in which this is achieved is through degradation or functional inactivation of PML NB proteins, which are recruited to viral genomes in order to repress viral transcription. Because MORC3 associates with PML NBs in uninfected cells and is a target for HSV-1-mediated degradation, we investigated the role of MORC3 during HSV-1 infection. We found that MORC3 is also recruited to viral HSV-1 genomes, and importantly it contributes to the fully efficient recruitment of PML, hDaxx, Sp100, and γH2AX to these sites. Depletion of MORC3 resulted in an increase in ICP0-null HSV-1 and wt HCMV replication and plaque formation; therefore, this study reveals that MORC3 is an antiviral factor which plays an important role during HSV-1 and HCMV infection.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Citomegalovirus/inmunología , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 1/inmunología , Inmunidad Innata , Animales , Células Cultivadas , Cricetinae , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ensayo de Placa Viral , Replicación Viral
6.
J Virol ; 90(1): 167-79, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26468536

RESUMEN

UNLABELLED: Intrinsic immunity is an aspect of antiviral defense that operates through diverse mechanisms at the intracellular level through a wide range of constitutively expressed cellular proteins. In the case of herpesviruses, intrinsic resistance involves the repression of viral gene expression during the very early stages of infection, a process that is normally overcome by viral tegument and/or immediate-early proteins. Thus, the balance between cellular repressors and virus-counteracting proteins determines whether or not a cell becomes productively infected. One aspect of intrinsic resistance to herpes simplex virus 1 (HSV-1) is conferred by components of promyelocytic leukemia nuclear bodies (PML NBs), which respond to infection by accumulating at sites that are closely associated with the incoming parental HSV-1 genomes. Other cellular proteins, including IFI16, which has been implicated in sensing pathogen DNA and initiating signaling pathways that lead to an interferon response, also respond to viral genomes in this manner. Here, studies of the dynamics of the response of PML NB components and IFI16 to invading HSV-1 genomes demonstrated that this response is extremely rapid, occurring within the first hour after addition of the virus, and that human Daxx (hDaxx) and IFI16 respond more rapidly than PML. In the absence of HSV-1 regulatory protein ICP0, which counteracts the recruitment process, the newly formed, viral-genome-induced PML NB-like foci can fuse with existing PML NBs. These data are consistent with a model involving viral genome sequestration into such structures, thereby contributing to the low probability of initiation of lytic infection in the absence of ICP0. IMPORTANCE: Herpesviruses have intimate interactions with their hosts, with infection leading either to the productive lytic cycle or to a quiescent infection in which viral gene expression is suppressed while the viral genome is maintained in the host cell nucleus. Whether a cell becomes lytically or quiescently infected can be determined through the competing activities of cellular repressors and viral activators, some of which counteract cell-mediated repression. Therefore, the events that occur within the earliest stages of infection can be of crucial importance. This paper describes the extremely rapid response to herpes simplex virus 1 infection of cellular protein IFI16, a sensor of pathogen DNA, and also of the PML nuclear body proteins PML and hDaxx, as revealed by live-cell microscopy. The data imply that these proteins can accumulate on or close to the viral genomes in a sequential manner which may lead to their sequestration and repression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular , Proteínas Co-Represoras , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Chaperonas Moleculares , Proteína de la Leucemia Promielocítica , Factores de Tiempo , Ubiquitina-Proteína Ligasas/metabolismo
7.
PLoS Pathog ; 11(7): e1005059, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26200910

RESUMEN

Covalent linkage to members of the small ubiquitin-like (SUMO) family of proteins is an important mechanism by which the functions of many cellular proteins are regulated. Sumoylation has roles in the control of protein stability, activity and localization, and is involved in the regulation of transcription, gene expression, chromatin structure, nuclear transport and RNA metabolism. Sumoylation is also linked, both positively and negatively, with the replication of many different viruses both in terms of modification of viral proteins and modulation of sumoylated cellular proteins that influence the efficiency of infection. One prominent example of the latter is the widespread reduction in the levels of cellular sumoylated species induced by herpes simplex virus type 1 (HSV-1) ubiquitin ligase ICP0. This activity correlates with relief from intrinsic immunity antiviral defence mechanisms. Previous work has shown that ICP0 is selective in substrate choice, with some sumoylated proteins such the promyelocytic leukemia protein PML being extremely sensitive, while RanGAP is completely resistant. Here we present a comprehensive proteomic analysis of changes in the cellular SUMO2 proteome during HSV-1 infection. Amongst the 877 potentially sumoylated species detected, we identified 124 whose abundance was decreased by a factor of 3 or more by the virus, several of which were validated by western blot and expression analysis. We found many previously undescribed substrates of ICP0 whose degradation occurs by a range of mechanisms, influenced or not by sumoylation and/or the SUMO2 interaction motif within ICP0. Many of these proteins are known or are predicted to be involved in the regulation of transcription, chromatin assembly or modification. These results present novel insights into mechanisms and host cell proteins that might influence the efficiency of HSV-1 infection.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 1 , Proteoma/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Proteoma/genética , Proteómica/métodos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/inmunología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Virales/metabolismo
8.
J Virol ; 89(6): 3062-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552717

RESUMEN

UNLABELLED: Human cytomegalovirus (HCMV) immediate early protein IE1 and the tegument protein pp71 are required for efficient infection. These proteins have some functional similarities with herpes simplex virus 1 (HSV-1) immediate early protein ICP0, which stimulates lytic HSV-1 infection and derepresses quiescent HSV-1 genomes. All three proteins counteract antiviral restriction mediated by one or more components of promyelocytic leukemia (PML) nuclear bodies, and IE1 and pp71, acting together, almost completely complement ICP0 null mutant HSV-1. Here, we investigated whether ICP0 might substitute for IE1 or pp71 during HCMV infection. Using human fibroblasts that express ICP0, IE1, or pp71 in an inducible manner, we found that ICP0 stimulated replication of both wild-type (wt) and pp71 mutant HCMV while IE1 increased wt HCMV plaque formation and completely complemented the IE1 mutant. Although ICP0 stimulated IE2 expression from IE1 mutant HCMV and increased the number of IE2-positive cells, it could not compensate for IE1 in full lytic replication. These results are consistent with previous evidence that both IE1 and IE2 are required for efficient HCMV gene expression, but they also imply that IE2 functionality is influenced specifically by IE1, either directly or indirectly, and that IE1 may include sequences that have HCMV-specific functions. We discovered a mutant form of IE1 (YL2) that fails to stimulate HCMV infection while retaining 30 to 80% of the activity of the wt protein in complementing ICP0 null mutant HSV-1. It is intriguing that the YL2 mutation is situated in the region of IE1 that is shared with IE2 and which is highly conserved among primate cytomegaloviruses. IMPORTANCE: Herpesvirus gene expression can be repressed by cellular restriction factors, one group of which is associated with structures known as ND10 or PML nuclear bodies (PML NBs). Regulatory proteins of several herpesviruses interfere with PML NB-mediated repression, and in some cases their activities are transferrable between different viruses. For example, the requirement for ICP0 during herpes simplex virus 1 (HSV-1) infection can be largely replaced by ICP0-related proteins expressed by other alphaherpesviruses and even by a combination of the unrelated IE1 and pp71 proteins of human cytomegalovirus (HCMV). Here, we report that ICP0 stimulates gene expression and replication of wt HCMV but cannot replace the need for IE1 during infection by IE1-defective HCMV mutants. Therefore, IE1 includes HCMV-specific functions that cannot be replaced by ICP0.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/enzimología , Proteínas Inmediatas-Precoces/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Citomegalovirus/genética , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Unión Proteica , Ubiquitina-Proteína Ligasas/genética , Proteínas Virales/genética , Replicación Viral
9.
J Virol ; 88(10): 5873-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24600001

RESUMEN

Herpes simplex virus 1 (HSV-1) regulatory protein ICP0 stimulates efficient infection via its E3 ubiquitin ligase activity that causes degradation of several cellular proteins, some of which are sumoylated. Chicken adenovirus Gam1 protein also interferes with the sumoylation pathway, and both proteins disrupt promyelocytic leukemia protein (PML) nuclear bodies (NBs). We report that Gam1 increases the infection efficiency of ICP0-null mutant HSV-1 by approximately 100-fold, thus strengthening the hypothesis that PML NB- and sumoylation-related mechanisms are important factors in the control of HSV-1 infection.


Asunto(s)
Adenoviridae/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Proteínas Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Pollos , Prueba de Complementación Genética , Humanos , Sumoilación , Proteínas Virales/genética
10.
Methods Mol Biol ; 1144: 1-17, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24671673

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a common and important human pathogen that has been studied in a wide variety of contexts for several decades. This book presents chapters on protocols on many strands of HSV-1 research that are currently in use in leading laboratories. This chapter gives a brief overview of HSV-1 biology and life cycle, covering basic aspects of the virus and its replication in cultured cells, the diseases caused by the virus, viral latency, antiviral defenses, and the mechanisms that the virus uses to counteract these defenses.


Asunto(s)
Herpesvirus Humano 1/genética , Estadios del Ciclo de Vida , Biología Molecular/métodos , Replicación Viral/genética , Línea Celular , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/patogenicidad , Humanos , Latencia del Virus
11.
J Cell Sci ; 127(Pt 2): 365-75, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24190887

RESUMEN

Arsenic is a clinically effective treatment for acute promyelocytic leukaemia (APL) in which the promyelocytic leukaemia (PML) protein is fused to retinoic receptor alpha (RARα). PML-RARα is degraded by the proteasome by a SUMO-dependent, ubiquitin-mediated pathway in response to arsenic treatment, curing the disease. Six major PML isoforms are expressed as a result of alternative splicing, each of which encodes a unique C-terminal region. Using a system in which only a single EYFP-linked PML isoform is expressed, we demonstrate that PMLI, PMLII and PMLVI accumulate in the cytoplasm following arsenic treatment, whereas PMLIII, PMLIV and PMLV do not. 3D structured illumination was used to obtain super-resolution images of PML bodies, revealing spherical shells of PML along with associated SUMO. Arsenic treatment results in dramatic isoform-specific changes to PML body ultrastructure. After extended arsenic treatment most PML isoforms are degraded, leaving SUMO at the core of the nuclear bodies. A high-content imaging assay identifies PMLV as the isoform most readily degraded following arsenic treatment, and PMLIV as relatively resistant to degradation. Immunoprecipitation analysis demonstrates that all PML isoforms are modified by SUMO and ubiquitin after arsenic treatment, and by using siRNA, we demonstrate that arsenic-induced degradation of all PML isoforms is dependent on the ubiquitin E3 ligase RNF4. Intriguingly, depletion of RNF4 results in marked accumulation of PMLV, suggesting that this isoform is an optimal substrate for RNF4. Thus the variable C-terminal domain influences the rate and location of degradation of PML isoforms following arsenic treatment.


Asunto(s)
Arsénico/farmacología , Estructuras del Núcleo Celular/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteolisis/efectos de los fármacos , Western Blotting , Estructuras del Núcleo Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo
12.
J Virol ; 88(4): 2337-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24307587

RESUMEN

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.


Asunto(s)
Herpes Simple/metabolismo , Herpesvirus Humano 1/genética , MicroARNs/metabolismo , Latencia del Virus/genética , Animales , Técnicas de Cultivo de Célula , Fibroblastos/metabolismo , Herpes Simple/genética , Herpesvirus Humano 1/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Neuronas/metabolismo , Ratas
13.
J Virol ; 88(5): 2763-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24352468

RESUMEN

UNLABELLED: Herpes simplex virus type 1 immediate-early protein ICP0 is an E3 ubiquitin ligase of the RING finger class that degrades several cellular proteins during infection. This activity is essential for its functions in stimulating efficient lytic infection and productive reactivation from latency. ICP0 targets a number of proteins that are modified by the small ubiquitin-like SUMO family of proteins, and it includes a number of short sequences that are related to SUMO interaction motifs (SIMs). Therefore, ICP0 has characteristics that are related to those of cellular SUMO-targeted ubiquitin ligase enzymes. Here, we analyze the impact of mutation of a number of SIM-like sequences (SLSs) within ICP0 on HSV-1 replication and gene expression and their requirement for ICP0-mediated degradation of both sumoylated and unmodified promyelocytic leukemia (PML) and other sumoylated cellular proteins. One SLS in the central portion of the ICP0 sequence (SLS4) was found to be absolutely required for targeting cellular sumoylated species in general and sumoylated forms of PML other than those of PML isoform I. Mutation of a group of SLSs in the C-terminal quarter of ICP0 also reduced ICP0-mediated degradation of sumoylated PML in a cooperative manner. Although mutation of individual SLSs caused only modest decreases in viral replication, combined mutation of SLS4 with SLS sequences in the C-terminal quarter of the protein reduced plaque formation efficiency by up to two orders of magnitude. These results provide further evidence that the biological activities of ICP0 are connected with host cell sumoylation events. IMPORTANCE: Herpes simplex virus type 1 protein ICP0 plays important roles in regulating the initial stages of lytic infection and productive reactivation from latency. ICP0 mediates its effects through inducing the degradation of cellular proteins that have repressive effects on viral gene expression. An increasing number of cellular proteins are known to be sensitive to ICP0-mediated degradation; therefore, it is important to understand how ICP0 selects its substrates for degradation. This study identifies sequence motifs within ICP0 that are involved in targeting cellular proteins that are modified by the SUMO family of ubiquitin-like proteins and describes how mutation of combinations of these motifs causes a 100-fold defect in viral infectivity.


Asunto(s)
Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteína SUMO-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Regulación Viral de la Expresión Génica , Genoma Viral , Humanos , Proteínas Inmediatas-Precoces/química , Proteínas Inmediatas-Precoces/genética , Espacio Intracelular/metabolismo , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Unión Proteica , Transporte de Proteínas , Proteolisis , Dominios RING Finger , Alineación de Secuencia , Sumoilación , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ensayo de Placa Viral , Replicación Viral
14.
J Virol ; 87(24): 13422-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24089555

RESUMEN

The cellular protein IFI16 colocalizes with the herpes simplex virus 1 (HSV-1) ubiquitin ligase ICP0 at early times of infection and is degraded as infection progresses. Here, we report that the factors governing the degradation of IFI16 and its colocalization with ICP0 are distinct from those of promyelocytic leukemia protein (PML), a well-characterized ICP0 substrate. Unlike PML, IFI16 colocalization with ICP0 was dependent on the ICP0 RING finger and did not occur when proteasome activity was inhibited. Expression of ICP0 in the absence of infection did not destabilize IFI16, the degradation occurred efficiently in the absence of ICP0 if infection was progressing efficiently, and IFI16 was relatively stable in wild-type (wt) HSV-1-infected U2OS cells. Therefore, IFI16 stability appears to be regulated by cellular factors in response to active HSV-1 infection rather than directly by ICP0. Because IFI16 is a DNA sensor that becomes associated with viral genomes during the early stages of infection, we investigated its role in the recruitment of PML nuclear body (PML NB) components to viral genomes. Recruitment of PML and hDaxx was less efficient in a proportion of IFI16-depleted cells, and this correlated with improved replication efficiency of ICP0-null mutant HSV-1. Because the absence of interferon regulatory factor 3 (IRF3) does not increase the plaque formation efficiency of ICP0-null mutant HSV-1, we speculate that IFI16 contributes to cell-mediated restriction of HSV-1 in a manner that is separable from its roles in IRF3-mediated interferon induction, but that may be linked to the PML NB response to viral infection.


Asunto(s)
Herpes Simple/metabolismo , Herpesvirus Humano 1/enzimología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Co-Represoras , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Humanos , Proteínas Inmediatas-Precoces/genética , Chaperonas Moleculares , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteína de la Leucemia Promielocítica , Proteolisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética
16.
Nat Rev Microbiol ; 11(6): 400-11, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624814

RESUMEN

Post-translational modification by members of the small ubiquitin-like modifier (SUMO) family of proteins is important for the regulation of many cellular proteins and pathways. As obligate parasites, viruses must engage with the host cell throughout their replication cycles, and it is therefore unsurprising that there are many examples of interplay between viral proteins and the host sumoylation system. This article reviews recent advances in this field, summarizing information on sumoylated viral proteins, the varied ways in which viruses engage with SUMO-related pathways, and the consequences of these interactions for viral replication and engagement with innate and intrinsic immunity.


Asunto(s)
Sumoilación/fisiología , Virosis/virología , Virus , Animales , Regulación Viral de la Expresión Génica/fisiología , Humanos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virosis/metabolismo , Virus/metabolismo
17.
Nucleic Acids Res ; 41(6): 3532-50, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23396441

RESUMEN

Death domain-associated protein (Daxx) cooperates with X-linked α-thalassaemia retardation syndrome protein (ATRX), a putative member of the sucrose non-fermentable 2 family of ATP-dependent chromatin-remodelling proteins, acting as the core ATPase subunit in this complex, whereas Daxx is the targeting factor, leading to histone deacetylase recruitment, H3.3 deposition and transcriptional repression of cellular promoters. Despite recent findings on the fundamental importance of chromatin modification in host-cell gene regulation, it remains unclear whether adenovirus type 5 (Ad5) transcription is regulated by cellular chromatin remodelling to allow efficient virus gene expression. Here, we focus on the repressive role of the Daxx/ATRX complex during Ad5 replication, which depends on intact protein-protein interaction, as negative regulation could be relieved with a Daxx mutant that is unable to interact with ATRX. To ensure efficient viral replication, Ad5 E1B-55K protein inhibits Daxx and targets ATRX for proteasomal degradation in cooperation with early region 4 open reading frame protein 6 and cellular components of a cullin-dependent E3-ubiquitin ligase. Our studies illustrate the importance and diversity of viral factors antagonizing Daxx/ATRX-mediated repression of viral gene expression and shed new light on the modulation of cellular chromatin remodelling factors by Ad5. We show for the first time that cellular Daxx/ATRX chromatin remodelling complexes play essential roles in Ad gene expression and illustrate the importance of early viral proteins to counteract cellular chromatin remodelling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenovirus Humanos/genética , Cromatina/metabolismo , ADN Helicasas/metabolismo , Regulación Viral de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Adenovirus Humanos/metabolismo , Adenovirus Humanos/fisiología , Línea Celular , Cromatina/química , Proteínas Co-Represoras , Histonas/metabolismo , Humanos , Chaperonas Moleculares , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Proteína Nuclear Ligada al Cromosoma X
18.
J Virol ; 87(4): 2174-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221561

RESUMEN

Upon the entry of the viral genome into the nucleus, herpes simplex virus type 1 (HSV-1) gene expression is rapidly repressed by constitutively expressed cellular proteins. This intrinsic antiviral defense is normally counteracted by ICP0, which allows virus infection to proceed efficiently. Replication of ICP0-null mutant HSV-1, however, is severely repressed by mechanisms that are conferred, at least in part, by nuclear domain 10 (ND10) components, including hDaxx, the promyelocytic leukemia (PML) protein, and Sp100. To investigate if these ND10 components repress viral gene expression in a cooperative manner, we simultaneously depleted host cells for hDaxx, PML, and Sp100 by multiple short hairpin RNA (shRNA) knockdown from a single lentivirus vector. We found that replication and gene expression of ICP0-null mutant HSV-1 were cooperatively repressed by hDaxx, PML, and Sp100 immediately upon infection, and all stages of virus replication were inhibited. Plaque-forming efficiency was enhanced at least 50-fold in the triple-depleted cells, a much larger increase than achieved by depletion of any single ND10 protein. Similar effects were also observed during infection of triple-depleted cells with human cytomegalovirus (HCMV). Moreover, using a cell culture model of quiescent infection, we found that triple depletion resulted in a much larger number of viral genomes escaping repression. However, triple depletion was unable to fully overcome the ICP0-null phenotype, implying the presence of additional repressive host factors, possibly components of the SUMO modification or DNA repair pathways. We conclude that several ND10 components cooperate in an additive manner to regulate HSV-1 and HCMV infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Citomegalovirus/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 1/inmunología , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Replicación Viral , Línea Celular , Proteínas Co-Represoras , Citomegalovirus/fisiología , Eliminación de Gen , Herpesvirus Humano 1/fisiología , Humanos , Chaperonas Moleculares , Proteína de la Leucemia Promielocítica , Ensayo de Placa Viral , Proteínas Virales/genética , Proteínas Virales/metabolismo
19.
J Virol ; 87(2): 978-90, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23135716

RESUMEN

Herpes simplex virus 1 (HSV-1) immediate-early protein ICP0 is required for efficient lytic infection and productive reactivation from latency and induces derepression of quiescent viral genomes. Despite being unrelated at the sequence level, ICP0 and human cytomegalovirus proteins IE1 and pp71 share some functional similarities in their abilities to counteract antiviral restriction mediated by components of cellular nuclear structures known as ND10. To investigate the extent to which IE1 and pp71 might substitute for ICP0, cell lines were developed that express either IE1 or pp71, or both together, in an inducible manner. We found that pp71 dissociated the hDaxx-ATRX complex and inhibited accumulation of these proteins at sites juxtaposed to HSV-1 genomes but had no effect on the promyelocytic leukemia protein (PML) or Sp100. IE1 caused loss of the small ubiquitin-like modifier (SUMO)-conjugated forms of PML and Sp100 and inhibited the recruitment of these proteins to HSV-1 genome foci but had little effect on hDaxx or ATRX in these assays. Both IE1 and pp71 stimulated ICP0-null mutant plaque formation, but neither to the extent achieved by ICP0. The combination of IE1 and pp71, however, inhibited recruitment of all ND10 proteins to viral genome foci, stimulated ICP0-null mutant HSV-1 plaque formation to near wild-type levels, and efficiently induced derepression of quiescent HSV-1 genomes. These results suggest that ND10-related intrinsic resistance results from the additive effects of several ND10 components and that the effects of IE1 and pp71 on subsets of these components combine to mirror the overall activities of ICP0.


Asunto(s)
Citomegalovirus/genética , Prueba de Complementación Genética , Herpesvirus Humano 1/fisiología , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Proteínas Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Ensayo de Placa Viral , Proteínas Virales/genética
20.
J Gen Virol ; 94(Pt 3): 465-481, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23239572

RESUMEN

Immediate-early protein ICP0 of herpes simplex virus type 1 (HSV-1) is important for the regulation of lytic and latent viral infection. Like the related proteins expressed by other alphaherpesviruses, ICP0 has a zinc-stabilized RING finger domain that confers E3 ubiquitin ligase activity. This domain is essential for the core functions of ICP0 and its activity leads to the degradation of a number of cellular proteins, some of which are involved in cellular defences that restrict viral infection. The article reviews recent advances in ICP0-related research, with an emphasis on the mechanisms by which ICP0 and related proteins counteract antiviral restriction and the roles in this process of cellular nuclear substructures known as ND10 or PML nuclear bodies. We also summarize recent advances in the understanding of the biochemical aspects of ICP0 activity. These studies highlight the importance of the SUMO conjugation pathway in both intrinsic resistance to HSV-1 infection and in substrate targeting by ICP0. The topics discussed in this review are relevant not only to HSV-1 infection, but also to cellular intrinsic resistance against herpesviruses more generally and the mechanisms by which viruses can evade this restriction.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 1/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Herpesvirus Humano 1/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...