Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28747501

RESUMEN

Latency-associated nuclear antigen (LANA) is a multifunctional protein encoded by members of the Rhadinovirus genus of gammaherpesviruses. Studies using murine gammaherpesvirus 68 (MHV68) demonstrated that LANA is important for acute replication, latency establishment, and reactivation in vivo Despite structural similarities in their DNA-binding domains (DBDs), LANA homologs from Kaposi sarcoma-associated herpesvirus (KSHV) and MHV68 exhibit considerable sequence divergence. We sought to determine if KSHV and MHV68 LANA homologs are functionally interchangeable. We generated an MHV68 virus that encodes KSHV LANA (kLANA) in place of MHV68 LANA (mLANA) and evaluated the virus's capacity to replicate, establish and maintain latency, and reactivate. kLANA knock-in (KLKI) MHV68 was replication competent in vitro and in vivo but exhibited slower growth kinetics and lower titers than wild-type (WT) MHV68. Following inoculation of mice, KLKI MHV68 established and maintained latency in splenocytes and peritoneal cells but did not reactivate efficiently ex vivo kLANA repressed the MHV68 promoter for ORF50, the gene that encodes the major lytic transactivator protein RTA, while mLANA did not, suggesting a likely mechanism for the KLKI MHV68 phenotypes. Bypassing this repression by providing MHV68 RTA in trans rescued KLKI MHV68 replication in tissue culture and enabled detection of KLKI MHV68 reactivation ex vivo These data demonstrate that kLANA and mLANA are functionally interchangeable for establishment and maintenance of latency and suggest that repression of lytic replication by kLANA, as previously shown with KSHV, is a kLANA-specific function that is transferable to MHV68.IMPORTANCE Kaposi sarcoma-associated herpesvirus (KSHV) and murine gammaherpesvirus 68 (MHV68) are members of the Rhadinovirus genus of gammaherpesviruses. These viruses establish lifelong infections that place their respective human and murine hosts at risk for cancer. Latency-associated nuclear antigen (LANA) is a conserved Rhadinovirus protein that is necessary for long-term chronic infection by these viruses. To better understand the conserved functions performed by LANA homologs, we generated a recombinant MHV68 virus that encodes the KSHV LANA protein in place of the MHV68 LANA homolog. We determined that the KSHV LANA protein is capable of supporting MHV68 latency in a mouse model of chronic infection but also functions to repress viral replication. This work describes an in vivo model system for defining evolutionarily conserved and divergent functions of LANA homologs in Rhadinovirus infection and disease.


Asunto(s)
Antígenos Virales/genética , Herpesvirus Humano 8/crecimiento & desarrollo , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/genética , Rhadinovirus/crecimiento & desarrollo , Transactivadores/genética , Latencia del Virus/genética , Células 3T3 , Animales , Antígenos Virales/biosíntesis , Línea Celular , Femenino , Técnicas de Sustitución del Gen , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas/genética , Rhadinovirus/genética , Rhadinovirus/metabolismo
2.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28701397

RESUMEN

Manipulation of host cellular pathways is a strategy employed by gammaherpesviruses, including mouse gammaherpesvirus 68 (MHV68), in order to negotiate a chronic infection. Ataxia-telangiectasia mutated (ATM) plays a unique yet incompletely understood role in gammaherpesvirus infection, as it has both proviral and antiviral effects. Chronic gammaherpesvirus infection is poorly controlled in a host with global ATM insufficiency, whether the host is a mouse or a human. In contrast, ATM facilitates replication, reactivation, and latency establishment of several gammaherpesviruses in vitro, suggesting that ATM is proviral in the context of infected cell cultures. The proviral role of ATM is also evident in vivo, as myeloid-specific ATM expression facilitates MHV68 reactivation during the establishment of viral latency. In order to better understand the complex relationship between host ATM and gammaherpesvirus infection, we depleted ATM specifically in B cells, a cell type critical for chronic gammaherpesvirus infection. B cell-specific ATM deficiency attenuated the establishment of viral latency due to compromised differentiation of ATM-deficient B cells. Further, we found that during long-term infection, peritoneal B-1b, but not related B-1a, B cells display the highest frequency of gammaherpesvirus infection. While ATM expression did not affect gammaherpesvirus tropism for B-1 B cells, B cell-specific ATM expression was necessary to support viral reactivation from peritoneal cells during long-term infection. Thus, our study reveals a role of ATM as a host factor that promotes chronic gammaherpesvirus infection of B cells.IMPORTANCE Gammaherpesviruses infect a majority of the human population and are associated with cancer, including B cell lymphomas. ATM is a unique host kinase that has both proviral and antiviral roles in the context of gammaherpesvirus infection. Further, there is insufficient understanding of the interplay of these roles in vivo during chronic infection. In this study, we show that ATM expression by splenic B cells is required for efficient establishment of gammaherpesvirus latency. We also show that ATM expression by peritoneal B cells is required to facilitate viral reactivation during long-term infection. Thus, our study defines a proviral role of B cell-specific ATM expression during chronic gammaherpesvirus infection.


Asunto(s)
Linfocitos B/metabolismo , Infecciones por Herpesviridae/virología , Rhadinovirus/crecimiento & desarrollo , Activación Viral/fisiología , Latencia del Virus/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/biosíntesis , Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Proteínas de la Ataxia Telangiectasia Mutada/genética , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Infecciones por Herpesviridae/inmunología , Interacciones Huésped-Patógeno/inmunología , Ratones , Ratones Endogámicos C57BL , Peritoneo/citología , Peritoneo/inmunología , Rhadinovirus/inmunología , Bazo/citología , Bazo/inmunología , Activación Viral/genética
3.
Arch Virol ; 162(3): 657-667, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27837274

RESUMEN

γ-Herpesviruses (γHV) such as Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus are important human pathogens involved in lymphoproliferation and tumorigenesis. Murine gammaherpesvirus 68 (MHV-68, γHV-68) is an effective model for the study of γHV pathogenesis and host-virus interaction because it is closely related to human γHV. Similarly to human γHV, MHV-68 encodes 15 microRNAs (miRNAs). Although their functions remain unknown, they are thought to regulate the viral life cycle or host-virus interactions, similarly to other human γHV. Herein, we established stable cell lines expressing MHV-68 miRNAs and investigated the role of MHV-68 miRNAs in the regulation of viral life cycle. We found that mghv-miR-M1-1, -3, -5, -7, -8, -9, -10, -11, -13, and -15 repressed MHV-68 lytic replication by down-regulating expression of the replication and transcription activator (RTA) gene, whereas mghv-miR-M1-2, -4, -6, and -12 induced lytic replication by up-regulating RTA. We confirmed that the decrease in viral replication caused by mghv-miR-M1-1 was abolished by inhibition of miRNA expression via miRNA inhibitor treatment. In addition, we observed that mghv-miR-M1-1 down-regulated c-Jun indirectly and decreased cytokine production, suggesting that mghv-miR-M1-1 may inhibit MHV-68 lytic replication by inhibiting the activator protein 1 (AP-1) signaling pathway.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/veterinaria , MicroARNs/metabolismo , ARN Viral/metabolismo , Rhadinovirus/crecimiento & desarrollo , Enfermedades de los Roedores/virología , Animales , Infecciones por Herpesviridae/virología , Humanos , Ratones , MicroARNs/genética , ARN Viral/genética , Rhadinovirus/genética , Rhadinovirus/fisiología , Roedores , Latencia del Virus , Replicación Viral
4.
J Virol ; 90(5): 2455-72, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26676769

RESUMEN

UNLABELLED: Gammaherpesviruses are important human and animal pathogens. Despite the fact that they display the classical architecture of herpesviruses, the function of most of their structural proteins is still poorly defined. This is especially true for tegument proteins. Interestingly, a potential role in immune evasion has recently been proposed for the tegument protein encoded by Kaposi's sarcoma-associated herpesvirus open reading frame 63 (ORF63). To gain insight about the roles of ORF63 in the life cycle of a gammaherpesvirus, we generated null mutations in the ORF63 gene of murid herpesvirus 4 (MuHV-4). We showed that disruption of ORF63 was associated with a severe MuHV-4 growth deficit both in vitro and in vivo. The latter deficit was mainly associated with a defect of replication in the lung but did not affect the establishment of latency in the spleen. From a functional point of view, inhibition of caspase-1 or the inflammasome did not restore the growth of the ORF63-deficient mutant, suggesting that the observed deficit was not associated with the immune evasion mechanism identified previously. Moreover, this growth deficit was also not associated with a defect in virion egress from the infected cells. In contrast, it appeared that MuHV-4 ORF63-deficient mutants failed to address most of their capsids to the nucleus during entry into the host cell, suggesting that ORF63 plays a role in capsid movement. In the future, ORF63 could therefore be considered a target to block gammaherpesvirus infection at a very early stage of the infection. IMPORTANCE: The important diseases caused by gammaherpesviruses in human and animal populations justify a better understanding of their life cycle. In particular, the role of most of their tegument proteins is still largely unknown. In this study, we used murid herpesvirus 4, a gammaherpesvirus infecting mice, to decipher the role of the protein encoded by the viral ORF63 gene. We showed that the absence of this protein is associated with a severe growth deficit both in vitro and in vivo that was mainly due to impaired migration of viral capsids toward the nucleus during entry. Together, our results provide new insights about the life cycle of gammaherpesviruses and could allow the development of new antiviral strategies aimed at blocking gammaherpesvirus infection at the very early stages.


Asunto(s)
Transporte Biológico , Cápside/metabolismo , Rhadinovirus/fisiología , Proteínas Virales/metabolismo , Internalización del Virus , Animales , Línea Celular , Cricetinae , Femenino , Eliminación de Gen , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Histocitoquímica , Pulmón/patología , Pulmón/virología , Ratones Endogámicos BALB C , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Proteínas Virales/genética
5.
J Virol Methods ; 206: 105-14, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24928692

RESUMEN

Herpesvirus transmission is sporadic, and infection may be asymptomatic or present only with secondary lesions after dissemination. Consequently host entry remains ill-understood. Experimental infections can be informative, but depend on inoculations that are inherently artificial and so need validation. Mice are a widely used experimental host. Alert mice inhale readily small (5 µl) liquid volumes, and Indian ink, luciferase or radiolabel delivered thus distributed to the nasopharynx and oropharynx. Murid Herpesvirus-4 or Herpes simplex virus type 1 delivered thus infected only the nose, arguing that host entry is nasal rather than oral. Marker or virus delivery to the lung depended on general anesthesia and a large inoculum volume (30 µl), and so needs further validation of physiological relevance. While lungs could be infected at lower doses than the upper respiratory tract, tracking experiments showed that nasal inocula pass mostly into the oropharynx, even when restricted to 1 µl. Thus, the relative inefficiency of experimental upper respiratory tract infection was attributable to limited liquid retention in this site. Nonetheless low volume intranasal delivery to alert mice provides a convenient way to model experimentally an apparently natural mode of herpesvirus host entry.


Asunto(s)
Modelos Animales de Enfermedad , Herpesvirus Humano 1/crecimiento & desarrollo , Sistema Respiratorio/virología , Rhadinovirus/crecimiento & desarrollo , Administración Intranasal , Animales , Herpesvirus Humano 1/aislamiento & purificación , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Rhadinovirus/aislamiento & purificación
6.
J Virol ; 87(8): 4596-608, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23408606

RESUMEN

Human gammaherpesviruses cause morbidity and mortality associated with infection and transformation of lymphoid and endothelial cells. Knowledge of cell types involved in virus dissemination from primary virus entry to virus latency is fundamental for the understanding of gammaherpesvirus pathogenesis. However, the inability to directly trace cell types with respect to virus dissemination pathways has prevented definitive conclusions regarding the relative contribution of individual cell types. Here, we describe that the route of infection affects gammaherpesvirus dissemination pathways. We constructed a recombinant murine gammaherpesvirus 68 (MHV-68) variant harboring a cassette which switches fluorescent markers in a Cre-dependent manner. Since the recombinant virus which was constructed on the wild-type background was attenuated, in this study we used an M1-deleted version, which infected mice with normal kinetics. Infection of Cre-transgenic mice with this convertible virus was used to estimate the quantitative contribution of defined cell types to virus productivity and dissemination during the acute phase of MHV-68 infection. In systemic infection, we found splenic vascular endothelial cells (EC) among the first and main cells to produce virus. After local infection, the contribution of EC to splenic virus production did not represent such early kinetics. However, at later time points, B cell-derived viruses dominated splenic productivity independently of systemic or local infection. Systemic versus local infection also governed the cell types involved in loading peritoneal exudate cells, leading to latency in F4/80- and CD11b-positive target cells. Systemic infection supported EC-driven dissemination, whereas local infection supported B cell-driven dissemination.


Asunto(s)
Infecciones por Herpesviridae/virología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/virología , Tropismo Viral , Replicación Viral , Animales , Linfocitos B/virología , Línea Celular , Células Endoteliales/virología , Genes Reporteros , Infecciones por Herpesviridae/patología , Estudios Longitudinales , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Rhadinovirus/fisiología , Bazo/virología , Coloración y Etiquetado/métodos , Infecciones Tumorales por Virus/patología
7.
J Gen Virol ; 93(Pt 5): 1076-1080, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22258865

RESUMEN

Although ORF23 is conserved among gammaherpesviruses, its role during infection is unknown. Here, we studied the expression of ORF23 of murine gammaherpesvirus 68 (MHV-68) and its role during infection. ORF23 mRNA was detected in infected cells as a late transcript. The ORF23 protein product could be expressed and detected as an N-terminally FLAG-tagged protein by Western blot and indirect immunofluorescence. To investigate the role of ORF23 in the infection cycle of a gammaherpesvirus, we constructed an ORF23 deletion mutant of MHV-68. The analysis of the ORF23 deletion mutant suggested that ORF23 of MHV-68 is neither essential for replication in cell culture nor for lytic or latent infection in vivo. A phenotype of the ORF23 deletion mutant, reflected by a moderate reduction in lytic replication and latency amplification, was only detectable in the face of direct competition to the parental virus.


Asunto(s)
Sistemas de Lectura Abierta , Rhadinovirus/patogenicidad , Proteínas Virales/metabolismo , Replicación Viral , Animales , Western Blotting , Infecciones por Coronaviridae/patología , Infecciones por Coronaviridae/virología , Eliminación de Gen , Perfilación de la Expresión Génica , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Rhadinovirus/crecimiento & desarrollo , Bazo/virología , Transcripción Genética , Carga Viral , Proteínas Virales/genética
8.
J Gen Virol ; 91(Pt 4): 867-79, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19940063

RESUMEN

Two novel gammaherpesviruses were isolated, one from a field vole (Microtus agrestis) and the other from wood mice (Apodemus sylvaticus). The genome of the latter, designated wood mouse herpesvirus (WMHV), was completely sequenced. WMHV had the same genome structure and predicted gene content as murid herpesvirus 4 (MuHV4; murine gammaherpesvirus 68). Overall nucleotide sequence identity between WMHV and MuHV4 was 85 % and most of the 10 kb region at the left end of the unique region was particularly highly conserved, especially the viral tRNA-like sequences and the coding regions of genes M1 and M4. The partial sequence (71 913 bp) of another gammaherpesvirus, Brest herpesvirus (BRHV), which was isolated ostensibly from a white-toothed shrew (Crocidura russula), was also determined. The BRHV sequence was 99.2 % identical to the corresponding portion of the WMHV genome. Thus, WMHV and BRHV appeared to be strains of a new virus species. Biological characterization of WMHV indicated that it grew with similar kinetics to MuHV4 in cell culture. The pathogenesis of WMHV in wood mice was also extremely similar to that of MuHV4, except for the absence of inducible bronchus-associated lymphoid tissue at day 14 post-infection and a higher load of latently infected cells at 21 days post-infection.


Asunto(s)
Arvicolinae/virología , Gammaherpesvirinae/clasificación , Murinae/virología , Rhadinovirus/clasificación , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Viral/química , Gammaherpesvirinae/genética , Gammaherpesvirinae/crecimiento & desarrollo , Genoma Viral , Datos de Secuencia Molecular , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Proteínas de la Matriz Viral/análisis , Proteínas de la Matriz Viral/genética
9.
J Gen Virol ; 90(Pt 6): 1461-1470, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19264614

RESUMEN

Gammaherpesviruses infect at least 90 % of the world's population. Infection control is difficult, in part because some fundamental features of host colonization remain unknown, for example whether normal latency establishment requires viral lytic functions. Since human gammaherpesviruses have narrow species tropisms, answering such questions requires animal models. Murid herpesvirus-4 (MuHV-4) provides one of the most tractable. MuHV-4 genomes delivered to the lung or peritoneum persist without lytic replication. However, they fail to disseminate systemically, suggesting that the outcome is inoculation route-dependent. After upper respiratory tract inoculation, MuHV-4 infects mice without involving the lungs or peritoneum. We examined whether host entry by this less invasive route requires the viral thymidine kinase (TK), a gene classically essential for lytic replication in terminally differentiated cells. MuHV-4 TK knockouts delivered to the lung or peritoneum were attenuated but still reached lymphoid tissue. In contrast, TK knockouts delivered to the upper respiratory tract largely failed to establish a detectable infection. Therefore TK, and by implication lytic replication, is required for MuHV-4 to establish a significant infection by a non-invasive route.


Asunto(s)
Infecciones por Herpesviridae/virología , Rhadinovirus/crecimiento & desarrollo , Timidina Quinasa/fisiología , Infecciones Tumorales por Virus/virología , Proteínas Virales/fisiología , Animales , Femenino , Técnicas de Inactivación de Genes , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Peritoneo/virología , Sistema Respiratorio/virología , Timidina Quinasa/deficiencia
10.
J Virol ; 83(10): 4732-48, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19264770

RESUMEN

NF-kappaB signaling is critical to the survival and transformation of cells infected by the human gammaherpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus. Here we have examined how elimination of the NF-kappaB transcription factor p50 from mice affects the life cycle of murine gammaherpesvirus 68 (MHV68). Notably, mice lacking p50 in every cell type were unable to establish a sufficiently robust immune response to control MHV68 infection, leading to high levels of latently infected B cells detected in the spleen and persistent virus replication in the lungs. The latter correlated with very low levels of virus-specific immunoglobulin G (IgG) in the infected p50(-/-) mice at day 48 postinfection. Because the confounding impact of the loss of p50 on the host response to MHV68 infection prevented a direct analysis of the role of this NF-kappaB family member on MHV68 latency in B cells, we generated and infected mixed p50(+/+)/p50(-/-) bone marrow chimeric mice. We show that the chimeric mice were able to control acute virus replication and exhibited normal levels of virus-specific IgG at 3 months postinfection, indicating the induction of a normal host immune response to MHV68 infection. However, in p50(+/+)/p50(-/-) chimeric mice the p50(-/-) B cells exhibited a significant defect compared to p50(+/+) B cells in supporting MHV68 latency. In addition to identifying a role for p50 in the establishment of latency, we determined that the absence of p50 in a subset of the hematopoietic compartment led to persistent virus replication in the lungs of the chimeric mice, providing evidence that p50 is required for controlling virus reactivation. Taken together, these data demonstrate that p50 is required for immune control by the host and has distinct tissue-dependent roles in the regulation of murine gammaherpesvirus latency during chronic infection.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Rhadinovirus/fisiología , Latencia del Virus , Animales , Linfocitos B/inmunología , Linfocitos B/virología , Línea Celular , Infecciones por Herpesviridae/virología , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p50 de NF-kappa B/inmunología , Rhadinovirus/crecimiento & desarrollo , Rhadinovirus/inmunología , Bazo/inmunología , Bazo/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Ensayo de Placa Viral
11.
Nucleic Acids Res ; 37(1): e2, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18988631

RESUMEN

Bacterial artificial chromosomes (BACs) derived from genomes of large DNA viruses are powerful tools for functional delineation of viral genes. Current methods for cloning the genomes of large DNA viruses as BACs require prior knowledge of the viral sequences or the cloning of viral DNA fragments, and are tedious because of the laborious process of multiple plaque purifications, which is not feasible for some fastidious viruses. Here, we describe a novel method for cloning the genomes of large DNA viruses as BACs, which entails direct in vitro transposition of viral genomes with a BAC cassette, and subsequent recovery in Escherichia coli. Determination of insertion sites and adjacent viral sequences identify the BAC clones for genetic manipulation and functional characterization. Compared to existing methods, this new approach is highly efficient, and does not require any information on viral sequences or cloning of viral DNA fragments, and plaque purifications. This method could potentially be used for discovering previously unidentified viruses.


Asunto(s)
Cromosomas Artificiales Bacterianos , Clonación Molecular/métodos , Elementos Transponibles de ADN , Virus ADN/genética , Genoma Viral , Animales , Secuencia de Bases , Línea Celular , ADN Viral/química , Genes Virales , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo
12.
J Gen Virol ; 90(Pt 1): 21-32, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19088269

RESUMEN

Luciferase-based imaging allows a global view of microbial pathogenesis. We applied this technique to gammaherpesvirus infection by inserting a luciferase expression cassette into the genome of murine herpesvirus-4 (MuHV-4). The recombinant virus strongly expressed luciferase in lytically infected cells without significant attenuation. We used it to compare different routes of virus inoculation. After intranasal infection of anaesthetized mice, luciferase was expressed in the nose and lungs for 7-10 days and in lymphoid tissue, most consistently the superficial cervical lymph nodes, for up to 30 days. Gastrointestinal infection was not observed. Intraperitoneal infection was very different to intranasal, with strong luciferase expression in the liver, kidneys, intestines, reproductive tract and spleen, but none in the nose or lungs. The nose has not previously been identified as a site of MuHV-4 infection. After intranasal infection of non-anaesthetized mice, it was the only site of non-lymphoid luciferase expression. Nevertheless, lymphoid colonization and persistence were still established, even at low inoculation doses. In contrast, virus delivered orally was very poorly infectious. Inoculation route therefore had a major impact on pathogenesis. Low dose intranasal infection without anaesthesia seems most likely to mimic natural transmission, and may therefore be particularly informative about normal viral gene functions.


Asunto(s)
Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Rhadinovirus/crecimiento & desarrollo , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Imagen de Cuerpo Entero , Estructuras Animales/virología , Animales , Femenino , Genes Reporteros , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos BALB C
13.
J Virol ; 83(4): 1811-22, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073723

RESUMEN

Gammaherpesviruses Kaposi's sarcoma-associated herpesvirus and Epstein-Barr virus are associated with multiple human cancers. Our goal was to develop a quantitative, high-throughput functional profiling system to identify viral cis-elements and protein subdomains critical for virus replication in the context of the herpesvirus genome. In gamma-2 herpesviruses, the transactivating factor RTA is essential for initiation of lytic gene expression and viral reactivation. We used the RTA locus as a model to develop the functional profiling approach. The mutant murine gammaherpesvirus 68 viral library, containing 15-bp random insertions in the RTA locus, was passaged in murine fibroblast cells for multiple rounds of selection. The effect of each 15-bp insertion was characterized using fluorescent-PCR profiling. We identified 1,229 insertions in the 3,845-bp RTA locus, of which 393, 282, and 554 were critically impaired, attenuated, and tolerated, respectively, for viral growth. The functional profiling phenotypes were verified by examining several individual RTA mutant clones for transactivating function of the RTA promoter and transcomplementing function of the RTA-null virus. Thus, the profiling approach enabled us to identify several novel functional domains in the RTA locus in the context of the herpesvirus genome. Importantly, our study has demonstrated a novel system to conduct high-density functional genetic mapping. The genome-scale expansion of the genetic profiling approach will expedite the functional genomics research on herpesvirus.


Asunto(s)
Mutagénesis Insercional , Rhadinovirus/fisiología , Transactivadores/fisiología , Proteínas no Estructurales Virales/fisiología , Replicación Viral , Animales , Línea Celular , Fibroblastos/virología , Prueba de Complementación Genética , Ratones , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Transactivadores/genética , Proteínas no Estructurales Virales/genética
14.
J Gen Virol ; 89(Pt 5): 1114-1121, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18420788

RESUMEN

We used the murine gammaherpesvirus 68 (gammaHV-68), which serves as a model for human gammaherpesvirus infection, to determine whether age at infection altered the pattern of gammaherpesvirus pathogenesis. We infected mice intranasally at 8 days old (pups) and 6 weeks old (adults) to investigate differences in gammaHV-68 pathogenesis. There was no difference between adults or pups in acute infection in the lungs at 6 days post-infection (p.i.). However, mice infected as pups exhibited a more disseminated viral infection with viral DNA detected in the spleen, liver and heart as measured by quantitative PCR (Q-PCR). In addition, viral DNA was detected in the lungs of mice infected as pups until 60 days p.i. Three viral transcripts (M2, M3 and M9) were expressed at both 30 and 60 days p.i. In contrast, no viral DNA or mRNA expression was detected in lungs of mice infected as adults at 30 or 60 days p.i. Mice infected as adults experienced a peak in latent infection in the spleen at 16 days p.i., corresponding with an increase in splenic weight and expansion of the Vbeta4+ CD8+ T-cell population, similar to infectious mononucleosis observed following infection of young adults with Epstein-Barr virus. However, the increase in splenic weight of infected pups was not as pronounced and no significant increase in Vbeta4+ CD8+ T-cell expansion was observed in infected pups. Together, these data suggest that the pathogenesis of murine gammaherpesvirus gammaHV-68 is age-dependent.


Asunto(s)
Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Mononucleosis Infecciosa/patología , Pulmón/virología , Rhadinovirus/crecimiento & desarrollo , Factores de Edad , Animales , Linfocitos T CD8-positivos/inmunología , ADN Viral/aislamiento & purificación , Genes Virales , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Viral/genética , Transcripción Genética
15.
Vet Pathol ; 43(4): 548-52, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16846998

RESUMEN

Granulomatous dermatitis in horses has been linked to many etiologies, including various parasites, fungi, and bacteria. Idiopathic forms of granulomatous inflammation-producing diseases, some of which are localized to the skin, also have been reported in horses. Herein we describe a case of recurrent equine granulomatous skin disease characterized by intranuclear viral inclusions within macrophages and giant cells. The histologic changes were primarily noted in the deep dermis and included multifocal to coalescing areas of necrosis marked by histiocytic cell infiltration and presence of giant cells. Electron microscopic examination revealed intranuclear and intracytoplasmic viral particles consistent with herpesvirus. Sequence results of the polymerase chain reaction product were consistent with equine herpesvirus 2, adding another possible etiology to the list of differentials in cases of equine granulomatous skin disease.


Asunto(s)
Dermatitis/veterinaria , Infecciones por Herpesviridae/veterinaria , Enfermedades de los Caballos/patología , Enfermedades de los Caballos/virología , Rhadinovirus/crecimiento & desarrollo , Infecciones Tumorales por Virus/veterinaria , Animales , Biopsia/veterinaria , ADN Viral/química , ADN Viral/genética , Dermatitis/patología , Dermatitis/virología , Resultado Fatal , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Histocitoquímica/veterinaria , Caballos , Masculino , Microscopía Electrónica de Transmisión/veterinaria , Reacción en Cadena de la Polimerasa/veterinaria , Análisis de Secuencia de ADN , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología
16.
J Vet Diagn Invest ; 17(2): 171-5, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15825499

RESUMEN

A malignant catarrhal fever (MCF)-like disease was induced experimentally in 3 sheep after aerosol inoculation with ovine herpesvirus-2 (OvHV-2). Each of 3 OvHV-2-negative sheep was nebulized with 2 ml of nasal secretions containing approximately 3.07 X 10(9) OvHV-2 DNA copies from a sheep experiencing an intensive viral-shedding episode. Ovine herpesvirus-2 DNA became detectable by polymerase chain reaction in the peripheral blood leukocytes of all 3 sheep within 3 days, and all 3 seroconverted between 6 and 8 days postinfection (PI). The sheep developed clinical signs, with copious mucopurulent nasal discharge and fever around 14 days PI. One of the 3 clinically affected sheep was euthanized at 18 days PI. Major lesions at necropsy were multifocal linear erosions and ulcers in mucosa of the cheeks, tongue, pharynx, and proximal esophagus and mild disseminated pneumonia. Microscopically, there was extensive moderate superficial histiocytic-lymphocytic rhinitis with epithelial dissociation and degeneration. Moderate multifocal histiocytic bronchointerstitial pneumonia was associated with loss of terminal bronchiolar epithelium. Lymphocytic vasculitis was present only in the lung. The remaining 2 sheep recovered clinically, approximately 25 days PI. The study revealed that clinical signs and lesions resembling MCF can develop when uninfected sheep are exposed to a high dose of aerosolized OvHV-2.


Asunto(s)
Leucocitos/virología , Fiebre Catarral Maligna/virología , Mucosa Nasal/virología , Rhadinovirus/crecimiento & desarrollo , Enfermedades de las Ovejas/virología , Animales , Bovinos , ADN Viral/análisis , ADN Viral/sangre , Pulmón/patología , Fiebre Catarral Maligna/diagnóstico , Fiebre Catarral Maligna/patología , Mucosa Bucal/patología , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Reacción en Cadena de la Polimerasa , Rhadinovirus/genética , Ovinos , Enfermedades de las Ovejas/diagnóstico , Enfermedades de las Ovejas/patología
17.
J Virol ; 78(16): 8878-84, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15280496

RESUMEN

Murid herpesvirus 4 (commonly called MHV-68) is closely related to Kaposi's sarcoma-associated herpesvirus (KSHV) and provides an excellent model system for investigating gammaherpesvirus-associated pathogenesis. MHV-76 is a naturally occurring deletion mutant of MHV-68 that lacks 9,538 bp of the left end of the unique portion of the genome encoding nonessential pathogenesis-related genes. The KSHV K1 protein has been shown to transform rodent fibroblasts in vitro and common marmoset T lymphocytes in vivo. Using homologous recombination techniques, we successfully generated recombinants of MHV-76 that encode green fluorescent protein (MHV76-GFP) and KSHV K1 (MHV76-K1). The replication of MHV76-GFP and MHV76-K1 in cell culture was identical to that of MHV-76. However, infection of BALB/c mice via the intranasal route revealed that MHV76-K1 replicated to a 10-fold higher titer than MHV76-GFP in the lungs at day 5 postinfection (p.i.). We observed type 2 pneumocyte proliferation in areas of consolidation and interstitial inflammation of mice infected with MHV76-K1 at day 10 p.i. MHV76-K1 established a 2- to 3-fold higher latent viral load than MHV76-GFP in the spleens of infected mice on days 10 and 14 p.i., although this was 10-fold lower than that established by wild-type MHV-76. A salivary gland tumor was present in one of four mice infected with MHV76-K1, as well as an increased inflammatory response in the lungs at day 120 p.i. compared with that of mice infected with MHV-76 and MHV76-GFP.


Asunto(s)
Transformación Celular Viral , Herpesvirus Humano 8/patogenicidad , Sistemas de Lectura Abierta/fisiología , Recombinación Genética , Rhadinovirus/patogenicidad , Proteínas Virales/metabolismo , Animales , Línea Celular , Cricetinae , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos BALB C , Sistemas de Lectura Abierta/genética , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Rhadinovirus/metabolismo , Proteínas Virales/genética
18.
Immunity ; 17(2): 143-55, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12196286

RESUMEN

Several gamma-herpesviruses encode homologs of host regulators of complement activation (RCA) proteins, suggesting that they have evolved immune evasion strategies targeting complement. We evaluated the role of complement factor C3 (C3) and the murine gamma-herpesvirus 68 (gammaHV68) RCA protein in viral pathogenesis. Deletion of the gammaHV68 RCA protein decreased virulence during acute CNS infection, and this attenuation was specifically reversed by deletion of host C3. The gammaHV68 RCA protein was also important for persistent viral replication and virulence in IFNgammaR(-/-) mice. In addition, C3 played a role in regulating latency, but this was not counteracted by the gammaHV68 RCA protein. We conclude that complement is a key host defense against gamma-herpesvirus infection and that gamma-herpesviruses have evolved an immune evasion strategy that is effective against complement-mediated antiviral responses during acute but not latent infection.


Asunto(s)
Complemento C3/inmunología , Factor B del Complemento/inmunología , Infecciones por Herpesviridae/inmunología , Meningoencefalitis/inmunología , Rhadinovirus/inmunología , Proteínas Virales/inmunología , Latencia del Virus/inmunología , Enfermedad Aguda , Animales , Encéfalo/patología , Encéfalo/virología , Línea Celular , Células Cultivadas , Enfermedad Crónica , Complemento C3/genética , Factor B del Complemento/genética , Femenino , Fibroblastos/citología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Huésped Inmunocomprometido , Pulmón/patología , Pulmón/virología , Masculino , Meningoencefalitis/patología , Meningoencefalitis/virología , Ratones , Ratones Endogámicos C57BL , Sistemas de Lectura Abierta , Receptores de Interferón/genética , Rhadinovirus/crecimiento & desarrollo , Rhadinovirus/patogenicidad , Rhadinovirus/fisiología , Proteínas Virales/genética , Proteínas Virales/fisiología , Virulencia , Replicación Viral , Receptor de Interferón gamma
19.
Nat Immunol ; 3(8): 733-40, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12101398

RESUMEN

The murine gamma-herpesvirus-68 (MHV-68) K3 protein, like that of the Kaposi's sarcoma associated herpesvirus, down-regulates major histocompatibility complex (MHC) class I expression. However, how this contributes to viral replication in vivo is unclear. After intranasal MHV-68 infection, K3 was transcribed both during acute lytic infection in the lung and during latency establishment in lymphoid tissue. K3-deficient viruses were not cleared more rapidly from the lung, but the number of latently infected spleen cells was reduced and the frequency of virus-specific CD8(+) cytotoxic T lymphocytes (CTLs) was increased. CTL depletion reversed the viral latency deficit. Thus, a major function of K3 appears to be CTL evasion during viral latency expansion.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Rhadinovirus/inmunología , Proteínas Virales/inmunología , Células 3T3 , Animales , Citometría de Flujo , Regulación de la Expresión Génica/inmunología , Genes MHC Clase I/inmunología , Hibridación in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutagénesis Insercional , Reacción en Cadena de la Polimerasa , ARN Viral/análisis , ARN Viral/genética , Rhadinovirus/genética , Rhadinovirus/crecimiento & desarrollo , Bazo/virología , Linfocitos T Citotóxicos/inmunología , Transcripción Genética/inmunología , Proteínas Virales/biosíntesis , Proteínas Virales/genética
20.
Arterioscler Thromb Vasc Biol ; 22(5): 793-8, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12006392

RESUMEN

Viral and bacterial infectious agents have been implicated in the etiology of atherosclerosis. We have previously shown that a gamma-herpesvirus can accelerate atherosclerosis in the apolipoprotein E-deficient (apoE-/-) mouse. To address whether a virally induced systemic immune response is sufficient to trigger enhanced atheroma formation, we infected apoE-/- mice with murine gamma-herpesvirus-68 (MHV-68) or herpes simplex virus-1 (HSV-1). In this study, we show that both viruses were able to induce a cell-mediated and humoral immune response in the apoE-/- mouse, which was sustained over a period of 24 weeks. Although intranasal or intraperitoneal infection with MHV-68 induced similar levels of virus-specific IgG1 and IgG2a antibodies in the serum of apoE-/- mice, those infected with HSV-1 showed higher anti-HSV-1 IgG2a compared with IgG1 antibody levels. In addition, viral message was not detected in the aortas of HSV-1-infected animals, whereas we have shown previously that MHV-68 mRNA can be detected in the aortas of infected mice as early as 5 days after infection. Compared with control mice, apoE-/- mice infected with MHV-68 showed accelerated atherosclerosis, whereas mice infected with HSV-1 did not. These data indicate that a systemic immune response to any particular infectious agent is insufficient to induce enhanced atherosclerosis in the apoE-/- mouse and point to specific infections or immune mechanisms that might be essential for virally enhanced atherogenesis.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/virología , Herpes Simple/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 1/patogenicidad , Rhadinovirus/patogenicidad , Animales , Anticuerpos Antivirales/biosíntesis , Aorta/virología , Arteriosclerosis/inmunología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Herpesvirus Humano 1/crecimiento & desarrollo , Herpesvirus Humano 1/aislamiento & purificación , Inmunidad Celular , Inmunoglobulina G/biosíntesis , Isotipos de Inmunoglobulinas/biosíntesis , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , ARN Viral/aislamiento & purificación , Rhadinovirus/crecimiento & desarrollo , Rhadinovirus/aislamiento & purificación , Bazo/virología , Infecciones Tumorales por Virus/inmunología , Células Vero/virología , Cultivo de Virus/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...