Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
PLoS Pathog ; 17(3): e1008979, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33657166

RESUMEN

The rhesus monkey rhadinovirus (RRV), a γ2-herpesvirus of rhesus macaques, shares many biological features with the human pathogenic Kaposi's sarcoma-associated herpesvirus (KSHV). Both viruses, as well as the more distantly related Epstein-Barr virus, engage cellular receptors from the Eph family of receptor tyrosine kinases (Ephs). However, the importance of the Eph interaction for RRV entry varies between cell types suggesting the existence of Eph-independent entry pathways. We therefore aimed to identify additional cellular receptors for RRV by affinity enrichment and mass spectrometry. We identified an additional receptor family, the Plexin domain containing proteins 1 and 2 (Plxdc1/2) that bind the RRV gH/gL glycoprotein complex. Preincubation of RRV with soluble Plxdc2 decoy receptor reduced infection by ~60%, while overexpression of Plxdc1 and 2 dramatically enhanced RRV susceptibility and cell-cell fusion of otherwise marginally permissive Raji cells. While the Plxdc2 interaction is conserved between two RRV strains, 26-95 and 17577, Plxdc1 specifically interacts with RRV 26-95 gH. The Plxdc interaction is mediated by a short motif at the N-terminus of RRV gH that is partially conserved between isolate 26-95 and isolate 17577, but absent in KSHV gH. Mutation of this motif abrogated the interaction with Plxdc1/2 and reduced RRV infection in a cell type-specific manner. Taken together, our findings characterize Plxdc1/2 as novel interaction partners and entry receptors for RRV and support the concept of the N-terminal domain of the gammaherpesviral gH/gL complex as a multifunctional receptor-binding domain. Further, Plxdc1/2 usage defines an important biological difference between KSHV and RRV.


Asunto(s)
Macaca mulatta/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Rhadinovirus/patogenicidad , Animales , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Humanos , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus
2.
JCI Insight ; 6(2)2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33491663

RESUMEN

The aryl-hydrocarbon receptor (AHR) is an intracellular sensor of aromatic hydrocarbons that sits at the top of various immunomodulatory pathways. Here, we present evidence that AHR plays a role in controlling IL-17 responses and the development of pulmonary fibrosis in response to respiratory pathogens following bone marrow transplant (BMT). Mice infected intranasally with gamma-herpesvirus 68 (γHV-68) following BMT displayed elevated levels of the AHR ligand, kynurenine (kyn), in comparison with control mice. Inhibition or genetic ablation of AHR signaling resulted in a significant decrease in IL-17 expression as well as a reduction in lung pathology. Lung CD103+ DCs expressed AHR following BMT, and treatment of induced CD103+ DCs with kyn resulted in altered cytokine production in response to γHV-68. Interestingly, mice deficient in the kyn-producing enzyme indolamine 2-3 dioxygenase showed no differences in cytokine responses to γHV-68 following BMT; however, isolated pulmonary fibroblasts infected with γHV-68 expressed the kyn-producing enzyme tryptophan dioxygenase (TDO2). Our data indicate that alterations in the production of AHR ligands in response to respiratory pathogens following BMT results in a pro-Th17 phenotype that drives lung pathology. We have further identified the TDO2/AHR axis as a potentially novel form of intercellular communication between fibroblasts and DCs that shapes immune responses to respiratory pathogens.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Trasplante de Médula Ósea/efectos adversos , Fibrosis Pulmonar/etiología , Receptores de Hidrocarburo de Aril/metabolismo , Rhadinovirus/patogenicidad , Triptófano Oxigenasa/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Dendríticas/patología , Células Dendríticas/fisiología , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Indolamina-Pirrol 2,3,-Dioxigenasa/deficiencia , Interleucina-17/biosíntesis , Quinurenina/metabolismo , Ligandos , Pulmón/inmunología , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Rhadinovirus/inmunología , Transducción de Señal , Células Th17/inmunología
3.
Acta Virol ; 64(2): 167-176, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32551785

RESUMEN

The aim of this work was to give an overview of murid herpesvirus 4 (MuHV-4) (synonyms: murine gammaherpesvirus 68, mouse herpesvirus strain 68), the first model for the study of human and animal oncogenic gammaherpesviruses. Based on our results confirming similarity of murine gammaherpesvirus 68 (MHV-68) to another gammaherpesvirus, human oncogenic Epstein-Barr virus (EBV), we were able to interpret some processes observed in the course of MHV-68 infection in analogy to EBV infection. In particular, that were the processes occurring during MHV-68-induced persistent infection in mice accompanied by tumor formation and leukemia following immunosuppression. Since EBV is a highly species specific virus, infecting humans only, these processes cannot be experimentally examined at the molecular, cellular, and tissue levels in natural host. However, they can be investigated in BALB/c mice infected with MHV-68, which is nowadays generally accepted model mainly thanks to experimental results achieved by our research team. The important mouse model MHV-68 is a prototype strain of MuHV-4 species and is classified as a member of the order Herpesvirales, family Herpesviridae, subfamily Gammaherpesvirinae and genus Rhadinovirus. During 40 years since its isolation from wild rodents, the virus was distributed into many virological laboratories in Europe (such as England, Slovakia, Germany, Italy, Portugal, Belgium, Denmark, Spain, Switzerland, Hungary, Russia, Sweden), USA, Canada, China, Korea, Japan and Australia. Global research of this virus, which has become an irreplaceable animal model, has expanded our understanding of the pathogenesis and immunology of human and animal gammaherpesvirus infections as well as the gammaherpesvirus-associated oncogenesis. No less important fact is that MHV-68 provides an excellent model to explore methods for controlling gammaherpesvirus infections through vaccination and chemotherapy. Keywords: MHV-68; EBV; KSHV; immunology; pathogenesis; oncogenesis; genome.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Herpesviridae/virología , Rhadinovirus/patogenicidad , Animales , Carcinogénesis , Herpesvirus Humano 4 , Humanos , Ratones , Ratones Endogámicos BALB C
4.
PLoS One ; 15(2): e0228484, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32017809

RESUMEN

Rhesus macaque (RM) rhadinovirus (RRV) is a simian gamma-2 herpesvirus closely related to human Kaposi's sarcoma-associated herpesvirus (KSHV). RRV is associated with the development of diseases in simian immunodeficiency virus (SIV) co-infected RM that resemble KSHV-associated pathologies observed in HIV-infected humans, including B cell lymphoproliferative disorders (LPD) and lymphoma. Importantly, how de novo KSHV infection affects the expression of host genes in humans, and how these alterations in gene expression affect viral replication, latency, and disease is unknown. The utility of the RRV/RM infection model provides a novel approach to address these questions in vivo, and utilizing the RRV bacterial artificial chromosome (BAC) system, the effects of specific viral genes on host gene expression patterns can also be explored. To gain insight into the effects of RRV infection on global host gene expression patterns in vivo, and to simultaneously assess the contributions of the immune inhibitory viral CD200 (vCD200) molecule to host gene regulation, RNA-seq was performed on pre- and post-infection lymph node (LN) biopsy samples from RM infected with either BAC-derived WT (n = 4) or vCD200 mutant RRV (n = 4). A variety of genes were identified as being altered in LN tissue samples due to RRV infection, including cancer-associated genes activation-induced cytidine deaminase (AICDA), glypican-1 (GPC1), CX3C chemokine receptor 1 (CX3CR1), and Ras dexamethasone-induced 1 (RasD1). Further analyses also indicate that GPC1 may be associated with lymphomagenesis. Finally, comparison of infection groups identified the differential expression of host gene thioredoxin interacting protein (TXNIP), suggesting a possible mechanism by which vCD200 negatively affects RRV viral loads in vivo.


Asunto(s)
Perfilación de la Expresión Génica/veterinaria , Infecciones por Herpesviridae/veterinaria , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/veterinaria , Animales , Receptor 1 de Quimiocinas CX3C/genética , Transformación Celular Neoplásica/genética , Citidina Desaminasa/genética , Regulación Neoplásica de la Expresión Génica , Glipicanos/genética , Infecciones por Herpesviridae/genética , Tejido Linfoide/metabolismo , Macaca mulatta , Análisis de Secuencia de ARN/veterinaria , Infecciones Tumorales por Virus/genética , Latencia del Virus , Replicación Viral , Proteínas ras/genética
5.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597758

RESUMEN

Gammaherpesviruses are ubiquitous pathogens that establish lifelong infections in the majority of adults worldwide. Chronic gammaherpesvirus infection has been implicated in both lymphomagenesis and, somewhat controversially, autoimmune disease development. Pathogenesis is largely associated with the unique ability of gammaherpesviruses to usurp B cell differentiation, specifically, the germinal center response, to establish long-term latency in memory B cells. The host tyrosine phosphatase SHP1 is known as a brake on immune cell activation and is downregulated in several gammaherpesvirus-driven malignancies. However, here we demonstrate that B cell- but not T cell-intrinsic SHP1 expression supports the gammaherpesvirus-driven germinal center response and the establishment of viral latency. Furthermore, B cell-intrinsic SHP1 deficiency cooperated with gammaherpesvirus infection to increase the levels of double-stranded DNA-reactive antibodies at the peak of viral latency. Thus, in spite of decreased SHP1 levels in gammaherpesvirus-driven B cell lymphomas, B cell-intrinsic SHP1 expression plays a proviral role during the establishment of chronic infection, suggesting that the gammaherpesvirus-SHP1 interaction is more nuanced and is modified by the stage of infection and pathogenesis.IMPORTANCE Gammaherpesviruses establish lifelong infection in a majority of adults worldwide and are associated with a number of malignancies, including B cell lymphomas. These viruses infect naive B cells and manipulate B cell differentiation to achieve a lifelong infection of memory B cells. The germinal center stage of B cell differentiation is important as both an amplifier of the viral latent reservoir and the target of malignant transformation. In this study, we demonstrate that expression of tyrosine phosphatase SHP1, a negative regulator that normally limits the activation and proliferation of hematopoietic cells, enhances the gammaherpesvirus-driven germinal center response and the establishment of chronic infection. The results of this study uncover an intriguing beneficial interaction between gammaherpesviruses that are presumed to profit from B cell activation and a cellular phosphatase that is traditionally perceived to be a negative regulator of the same processes.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Infecciones por Herpesviridae/genética , Interacciones Huésped-Patógeno/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Rhadinovirus/genética , Infecciones Tumorales por Virus/genética , Animales , Anticuerpos Antinucleares/biosíntesis , Linfocitos B/virología , Enfermedad Crónica , ADN/genética , ADN/inmunología , Femenino , Centro Germinal/virología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Memoria Inmunológica , Activación de Linfocitos , Masculino , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Rhadinovirus/inmunología , Rhadinovirus/patogenicidad , Linfocitos T/inmunología , Linfocitos T/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Latencia del Virus/genética , Latencia del Virus/inmunología
6.
mBio ; 9(5)2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30377280

RESUMEN

Misincorporation of uracil or spontaneous cytidine deamination is a common mutagenic insult to DNA. Herpesviruses encode a viral uracil-DNA glycosylase (vUNG) and a viral dUTPase (vDUT), each with enzymatic and nonenzymatic functions. However, the coordinated roles of these enzymatic activities in gammaherpesvirus pathogenesis and viral genomic stability have not been defined. In addition, potential compensation by the host UNG has not been examined in vivo The genetic tractability of the murine gammaherpesvirus 68 (MHV68) system enabled us to delineate the contribution of host and viral factors that prevent uracilated DNA. Recombinant MHV68 lacking vUNG (ORF46.stop) was not further impaired for acute replication in the lungs of UNG-/- mice compared to wild-type (WT) mice, indicating host UNG does not compensate for the absence of vUNG. Next, we investigated the separate and combinatorial consequences of mutating the catalytic residues of the vUNG (ORF46.CM) and vDUT (ORF54.CM). ORF46.CM was not impaired for replication, while ORF54.CM had a slight transient defect in replication in the lungs. However, disabling both vUNG and vDUT led to a significant defect in acute expansion in the lungs, followed by impaired establishment of latency in the splenic reservoir. Upon serial passage of the ORF46.CM/ORF54.CM mutant in either fibroblasts or the lungs of mice, we noted rapid loss of the nonessential yellow fluorescent protein (YFP) reporter gene from the viral genome, due to recombination at repetitive elements. Taken together, our data indicate that the vUNG and vDUT coordinate to promote viral genomic stability and enable viral expansion prior to colonization of latent reservoirs.IMPORTANCE Unrepaired uracils in DNA can lead to mutations and compromise genomic stability. Herpesviruses have hijacked host processes of DNA repair and nucleotide metabolism by encoding a viral UNG that excises uracils and a viral dUTPase that initiates conversion of dUTP to dTTP. To better understand the impact of these processes on gammaherpesvirus pathogenesis, we examined the separate and collaborative roles of vUNG and vDUT upon MHV68 infection of mice. Simultaneous disruption of the enzymatic activities of both vUNG and vDUT led to a severe defect in acute replication and establishment of latency, while also revealing a novel, combinatorial function in promoting viral genomic stability. We propose that herpesviruses require these enzymatic processes to protect the viral genome from damage, possibly triggered by misincorporated uracil. This reveals a novel point of therapeutic intervention to potentially block viral replication and reduce the fitness of multiple herpesviruses.


Asunto(s)
Eliminación de Gen , Inestabilidad Genómica , Pirofosfatasas/metabolismo , Recombinación Genética , Rhadinovirus/enzimología , Rhadinovirus/patogenicidad , Uracil-ADN Glicosidasa/metabolismo , Animales , Genoma Viral , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Pulmón/virología , Ratones , Pirofosfatasas/genética , Rhadinovirus/genética , Enfermedades de los Roedores/virología , Uracil-ADN Glicosidasa/genética , Virulencia
7.
Eur J Immunol ; 48(2): 250-257, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28980305

RESUMEN

Viruses, particularly the Epstein-Barr virus (EBV) has long been suspected to exacerbate acute arthritic symptoms. However, the cell populations that contribute to import viruses into the inflamed tissues remain to be identified. In the present study, we have investigated the role of monocytes in the transport of Murid herpesvirus 68 (MHV-68), a mouse virus closely related to EBV, using the serum transfer-induced arthritis (STIA) model. We found compelling evidence that MHV-68 infection markedly increased disease severity in NR4A1-/- mice, which are deficient for Ly6Clow monocytes. In contrast, the MHV-68-induced enhancement of joint inflammation was lessened in CCR2-/- mice, suggesting the involvement of inflammatory Ly6Chigh monocytes in viral transport. We also observed that following selective depletion of monocyte subsets by administration of clodronate, MHV-68 transport into the synovium occurs only in the presence of Ly6Chigh monocytes. Tracking of adoptively transferred Ly6Chigh GFP infected monocytes into arthritic CCR2-/- mice by two-photon intravital microscopy showed that this monocyte subset has the capacity to deliver viruses to inflamed AR joints, as confirmed by the detection of viral DNA in inflamed tissues of recipient mice. We thus conclude that Ly6Chigh monocytes import MHV-68 when they are mobilized to the inflamed arthritic joint.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 4/fisiología , Monocitos/inmunología , Rhadinovirus/fisiología , Infecciones Tumorales por Virus/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Artritis Experimental/virología , Artritis Reumatoide/virología , Células Cultivadas , ADN Viral/análisis , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/virología , Femenino , Infecciones por Herpesviridae/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/trasplante , Muridae , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Receptores CCR2/genética , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/virología
8.
Proc Natl Acad Sci U S A ; 113(36): 10085-90, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27528682

RESUMEN

Viruses are intracellular pathogens that hijack host cell machinery and resources to replicate. Rather than being constant, host physiology is rhythmic, undergoing circadian (∼24 h) oscillations in many virus-relevant pathways, but whether daily rhythms impact on viral replication is unknown. We find that the time of day of host infection regulates virus progression in live mice and individual cells. Furthermore, we demonstrate that herpes and influenza A virus infections are enhanced when host circadian rhythms are abolished by disrupting the key clock gene transcription factor Bmal1. Intracellular trafficking, biosynthetic processes, protein synthesis, and chromatin assembly all contribute to circadian regulation of virus infection. Moreover, herpesviruses differentially target components of the molecular circadian clockwork. Our work demonstrates that viruses exploit the clockwork for their own gain and that the clock represents a novel target for modulating viral replication that extends beyond any single family of these ubiquitous pathogens.


Asunto(s)
Factores de Transcripción ARNTL/genética , Relojes Circadianos/genética , Herpes Simple/virología , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno , Infecciones por Orthomyxoviridae/virología , Infecciones Tumorales por Virus/virología , Factores de Transcripción ARNTL/deficiencia , Animales , Transporte Biológico , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Línea Celular , Ensamble y Desensamble de Cromatina , Ritmo Circadiano/genética , Cricetinae , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Herpes Simple/genética , Herpes Simple/metabolismo , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 1/patogenicidad , Herpesvirus Humano 1/fisiología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H1N1 del Virus de la Influenza A/fisiología , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Rhadinovirus/patogenicidad , Rhadinovirus/fisiología , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/metabolismo , Replicación Viral
9.
J Virol ; 90(17): 8013-28, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27356898

RESUMEN

UNLABELLED: Nuclear domain 10 (ND10) components restrict herpesviral infection, and herpesviruses antagonize this restriction by a variety of strategies, including degradation or relocalization of ND10 proteins. The rhesus monkey rhadinovirus (RRV) shares many key biological features with the closely related Kaposi's sarcoma-associated herpesvirus (KSHV; human herpesvirus 8) and readily infects cells of both human and rhesus monkey origin. We used the clustered regularly interspaced short palindromic repeat-Cas9 (CRISPR-Cas9) technique to generate knockout (ko) cells for each of the four ND10 components, PML, SP100, DAXX, and ATRX. These ko cells were analyzed with regard to permissiveness for RRV infection. In addition, we analyzed the fate of the individual ND10 components in infected cells by immunofluorescence and Western blotting. Knockout of the ND10 component DAXX markedly increased RRV infection, while knockout of PML or SP100 had a less pronounced effect. In line with these observations, RRV infection resulted in rapid degradation of SP100, followed by degradation of PML and the loss of ND10 structures, whereas the protein levels of ATRX and DAXX remained constant. Notably, inhibition of the proteasome but not inhibition of de novo gene expression prevented the loss of SP100 and PML in cells that did not support lytic replication, compatible with proteasomal degradation of these ND10 components through the action of a viral tegument protein. Expression of the RRV FGARAT homolog ORF75 was sufficient to effect the loss of SP100 and PML in transfected or transduced cells, implicating ORF75 as the viral effector protein. IMPORTANCE: Our findings highlight the antiviral role of ND10 and its individual components and further establish the viral FGARAT homologs of the gammaherpesviruses to be important viral effectors that counteract ND10-instituted intrinsic immunity. Surprisingly, even closely related viruses like KSHV and RRV evolved to use different strategies to evade ND10-mediated restriction. RRV first targets SP100 for degradation and then targets PML with a delayed kinetic, a strategy which clearly differs from that of other gammaherpesviruses. Despite efficient degradation of these two major ND10 components, RRV is still restricted by DAXX, another abundant ND10 component, as evidenced by a marked increase in RRV infection and replication upon knockout of DAXX. Taken together, our findings substantiate PML, SP100, and DAXX as key antiviral proteins, in that the first two are targeted for degradation by RRV and the last one still potently restricts replication of RRV.


Asunto(s)
Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Interacciones Huésped-Patógeno , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Rhadinovirus/patogenicidad , Proteínas Estructurales Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Western Blotting , Línea Celular , Proteínas Co-Represoras , ADN Helicasas/metabolismo , Humanos , Microscopía Fluorescente , Chaperonas Moleculares , Proteolisis , Proteína Nuclear Ligada al Cromosoma X
10.
PLoS One ; 10(11): e0142540, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26544979

RESUMEN

Establishment of persistent infection in memory B cells by murid herpesvirus-4 (MuHV-4) depends on the proliferation of latently infected germinal center B cells, for which T cell help is essential. Whether the virus is capable of modulating B-T helper cell interaction for its own benefit is still unknown. Here, we investigate if the MuHV-4 latency associated M2 protein, which assembles multiprotein complexes with B cell signaling proteins, plays a role. We observed that M2 led to the upregulation of adhesion and co-stimulatory molecules in transduced B cell lines. In an MHC-II restricted OVA peptide-specific system, M2 polarized to the B-T helper contact zone. Furthermore, it promoted B cell polarization, as demonstrated by the increased proximity of the B cell microtubule organizing center to the interface. Consistent with these data, M2 promoted the formation of B-T helper cell conjugates. In an in vitro competition assay, this translated into a competitive advantage, as T cells preferentially conjugated with M2-expressing B cells. However, expression of M2 alone in B cells was not sufficient to lead to T cell activation, as it only occurred in the presence of specific peptide. Taken together, these findings support that M2 promotes the formation of B-T helper cell conjugates. In an in vivo context this may confer a competitive advantage to the infected B cell in acquisition of T cell help and initiation of a germinal center reaction, hence host colonization.


Asunto(s)
Linfoma de Células B/inmunología , Linfoma de Células B/virología , Rhadinovirus/patogenicidad , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/virología , Proteínas Virales/inmunología , Animales , Moléculas de Adhesión Celular/fisiología , Línea Celular , Línea Celular Tumoral , Polaridad Celular , Proliferación Celular , Interacciones Huésped-Patógeno , Memoria Inmunológica , Activación de Linfocitos , Linfoma de Células B/patología , Ratones , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Rhadinovirus/inmunología , Rhadinovirus/fisiología , Linfocitos T Colaboradores-Inductores/patología , Latencia del Virus
11.
PLoS Pathog ; 11(3): e1004761, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25790477

RESUMEN

Rhadinoviruses establish chronic infections of clinical and economic importance. Several show respiratory transmission and cause lung pathologies. We used Murid Herpesvirus-4 (MuHV-4) to understand how rhadinovirus lung infection might work. A primary epithelial or B cell infection often is assumed. MuHV-4 targeted instead alveolar macrophages, and their depletion reduced markedly host entry. While host entry was efficient, alveolar macrophages lacked heparan - an important rhadinovirus binding target - and were infected poorly ex vivo. In situ analysis revealed that virions bound initially not to macrophages but to heparan⁺ type 1 alveolar epithelial cells (AECs). Although epithelial cell lines endocytose MuHV-4 readily in vitro, AECs did not. Rather bound virions were acquired by macrophages; epithelial infection occurred only later. Thus, host entry was co-operative - virion binding to epithelial cells licensed macrophage infection, and this in turn licensed AEC infection. An antibody block of epithelial cell binding failed to block host entry: opsonization provided merely another route to macrophages. By contrast an antibody block of membrane fusion was effective. Therefore co-operative infection extended viral tropism beyond the normal paradigm of a target cell infected readily in vitro; and macrophage involvement in host entry required neutralization to act down-stream of cell binding.


Asunto(s)
Células Epiteliales/virología , Infecciones por Herpesviridae/virología , Macrófagos/virología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Alveolos Pulmonares/virología , Rhadinovirus/patogenicidad , Virión/patogenicidad
12.
mBio ; 6(1): e01670-14, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25691585

RESUMEN

UNLABELLED: Many viruses express noncoding RNAs (ncRNAs). The gammaherpesviruses (γHVs), including Epstein-Barr virus, Kaposi's sarcoma-associated herpesvirus, and murine γHV68, each contain multiple ncRNA genes, including microRNAs (miRNAs). While these ncRNAs can regulate multiple host and viral processes in vitro, the genetic contribution of these RNAs to infection and pathogenesis remains largely unknown. To study the functional contribution of these RNAs to γHV infection, we have used γHV68, a small-animal model of γHV pathogenesis. γHV68 encodes eight small hybrid ncRNAs that contain both tRNA-like elements and functional miRNAs. These genes are transcribed by RNA polymerase III and are referred to as the γHV68 TMERs (tRNA-miRNA-encoded RNAs). To determine the total concerted genetic contribution of these ncRNAs to γHV acute infection and pathogenesis, we generated and characterized a recombinant γHV68 strain devoid of all eight TMERs. TMER-deficient γHV68 has wild-type levels of lytic replication in vitro and normal establishment of latency in B cells early following acute infection in vivo. In contrast, during acute infection of immunodeficient mice, TMER-deficient γHV68 has reduced virulence in a model of viral pneumonia, despite having an enhanced frequency of virus-infected cells. Strikingly, expression of a single viral tRNA-like molecule, in the absence of all other virus-encoded TMERs and miRNAs, reverses both attenuation in virulence and enhanced frequency of infected cells. These data show that γHV ncRNAs play critical roles in acute infection and virulence in immunocompromised hosts and identify these RNAs as a new potential target to modulate γHV-induced infection and pathogenesis. IMPORTANCE: The gammaherpesviruses (γHVs) are a subfamily of viruses associated with chronic inflammatory diseases and cancer, particularly in immunocompromised individuals. These viruses uniformly encode multiple types of noncoding RNAs (ncRNAs) that are not translated into proteins. It remains unclear how virus-expressed ncRNAs influence the course and outcome of infection in vivo. Here, we generated a mouse γHV that lacks the expression of multiple ncRNAs. Notably, this mutant virus is critically impaired in the ability to cause disease in immunocompromised hosts yet shows a paradoxical increase in infected cells early during infection in these hosts. While the original mouse virus encodes multiple ncRNAs, the expression of a single domain of one ncRNA can partially reverse the defects of the mutant virus. These studies demonstrate that γHV ncRNAs can directly contribute to virus-induced disease in vivo and that these RNAs may be multifunctional, allowing the opportunity to specifically interfere with different functional domains of these RNAs.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , ARN no Traducido/metabolismo , Rhadinovirus/genética , Rhadinovirus/patogenicidad , Factores de Virulencia/metabolismo , Animales , Línea Celular , Fibroblastos/fisiología , Fibroblastos/virología , Eliminación de Gen , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN no Traducido/genética , Virulencia , Factores de Virulencia/genética
13.
J Immunotoxicol ; 12(4): 330-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25412621

RESUMEN

Murine gammaherpesvirus-68 (MHV-68), a natural pathogen of mice, is being evaluated as a model of Epstein Barr Virus (EBV) infection for use in investigation of the effects of immunomodulatory therapy on herpesvirus pathogenesis in humans. Immunosuppressive agents are used for treatment of a variety of autoimmune diseases as well as for prevention of tissue rejection after organ transplantation and can result in recrudescence of latent herpesvirus infections. Prior to examination of MHV-68 as a suitable model for EBV, better characterization of the MHV-68 model was desirable. Characterization of the MHV-68 model involved development of assays for detecting virus and for demonstration of safety when present in murine colonies. Limited information is available in the literature regarding MHV-68 transmission, although recent reports indicate the virus is not horizontally spread in research facilities. To further determine transmission potential, immunocompetent and immunodeficient mice were infected with MHV-68 and co-habitated with naïve animals. Molecular pathology assays were developed to characterize the MHV-68 model and to determine viral transmission. Horizontal transmission of virus was not observed from infected animals to naïve cagemates after fluorescence microscopy assays and quantitative PCR (qPCR). Serologic analysis complemented these studies and was used as a method of monitoring infection amongst murine colonies. Overall, these findings demonstrate that MHV-68 infection can be controlled and monitored in murine research facilities, and the potential for unintentional infection is low.


Asunto(s)
Infecciones por Herpesviridae/transmisión , Modelos Biológicos , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/transmisión , Animales , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Rhadinovirus/inmunología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/patología
14.
PLoS Pathog ; 10(7): e1004269, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25079788

RESUMEN

Murid γ-herpesvirus-4 (MuHV-4) promotes polyclonal B cell activation and establishes latency in memory B cells via unclear mechanisms. We aimed at exploring whether B cell receptor specificity plays a role in B cell susceptibility to viral latency and how this is related to B cell activation. We first observed that MuHV-4-specific B cells represent a minority of the latent population, and to better understand the influence of the virus on non-MuHV-4 specific B cells we used the SWHEL mouse model, which produce hen egg lysozyme (HEL)-specific B cells. By tracking HEL+ and HEL- B cells, we showed that in vivo latency was restricted to HEL- B cells while the two populations were equally sensitive to the virus in vitro. Moreover, MuHV-4 induced two waves of B cell activation. While the first wave was characterized by a general B cell activation, as shown by HEL+ and HEL- B cells expansion and upregulation of CD69 expression, the second wave was restricted to the HEL- population, which acquired germinal center (GC) and plasma cell phenotypes. Antigenic stimulation of HEL+ B cells led to the development of HEL+ GC B cells where latent infection remained undetectable, indicating that MuHV-4 does not benefit from acute B cell responses to establish latency in non-virus specific B cells but relies on other mechanisms of the humoral response. These data support a model in which the establishment of latency in B cells by γ-herpesviruses is not stochastic in terms of BCR specificity and is tightly linked to the formation of GCs.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Herpesviridae/inmunología , Muramidasa/inmunología , Infecciones Tumorales por Virus/inmunología , Latencia del Virus/inmunología , Animales , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Infecciones por Herpesviridae/virología , Inmunidad Celular , Inmunización , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/virología
15.
Vet Microbiol ; 172(1-2): 318-22, 2014 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-24803260

RESUMEN

Malignant catarrhal fever (MCF), due to ovine herpesvirus 2 (OvHV-2), causes appreciable death loss in ranched bison (Bison bison) throughout North America. No vaccine exists to protect animals from disease. Since OvHV-2 has not been propagated in vitro, one strategy to develop a modified live vaccine is to use a closely related, non-pathogenic member of the malignant catarrhal fever virus family as a vector expressing potentially protective OvHV-2 epitopes. To date, no controlled experimental challenge studies with alcelaphine herpesvirus 2 (AlHV-2) derived from topi (Damaliscus lunatus jimela) have been reported The unique or light DNA segment of the AlHV-2 genome was sequenced and annotated and the virus was tested for its ability to infect and induce disease in American bison. Yearling bison were inoculated intranasally (n=4) or intramuscularly (n=3) with 2 × 10(-4.7) TCID50 of AlHV-2, and monitored for infection and the development of disease. Six inoculated bison became infected with AlHV-2. Two of the six animals developed clinical signs and had gross and histological lesions consistent with terminal MCF, which differed in distribution from those in bison with MCF due to OvHV-2. One other animal developed minor clinical signs and had gross and histological pulmonary lesions consistent with early (pre-clinical) stages of MCF. Unmodified low cell culture passage AlHV-2 derived from topi is an unsuitable vaccine vector for the prevention of MCF. However, the annotated genome might be useful in identifying genes which could be deleted to potentially attenuate the virus for bison.


Asunto(s)
Bison/virología , Gammaherpesvirinae/patogenicidad , Genoma Viral , Infecciones por Herpesviridae/veterinaria , Fiebre Catarral Maligna/virología , Rhadinovirus/patogenicidad , Animales , Bison/inmunología , Femenino , Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Masculino , Fiebre Catarral Maligna/inmunología , Fiebre Catarral Maligna/patología , Anotación de Secuencia Molecular , Rhadinovirus/fisiología , Análisis de Secuencia de ADN , Estados Unidos , Carga Viral
16.
Curr Opin Virol ; 3(3): 245-50, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23747119

RESUMEN

Rhesus macaque rhadinovirus (RRV) is a gamma-2 herpesvirus that naturally infects rhesus macaque (RM) monkeys and is closely related to human herpesvirus-8 (HHV-8)/Kaposi's sarcoma-associated herpesvirus (KSHV). Infection of immunodeficient RM induces disease in infected RM that resembles KSHV-associated pathologies. Importantly, RRV possesses homologues of KSHV ORFs that are postulated to play a role in disease development. As such, RRV has emerged as a prominent in vivo model system for examining mechanisms of infection and disease of these pathogenic herpesviruses, and has provided unique insight into how these viruses cause disease.


Asunto(s)
Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/patología , Infecciones Tumorales por Virus/virología , Animales , Macaca mulatta
17.
J Gen Virol ; 94(Pt 7): 1613-1623, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23535573

RESUMEN

To better understand the role of the M2 protein of the murine herpes virus strain 68 (MHV-68) in vivo, B-lymphocyte-restricted, M2-transgenic mice were constructed. The transgenic mice contained normal B-cell subpopulations in bone marrow, lymph nodes and spleen. After immunization with sheep red blood cells, spleens from M2-transgenic mice had increased germinal centres. Transgenic mice responded to the T-cell-dependent antigen keyhole limpet haemocyanin (KLH) with higher levels of secondary IgM and IgG2a antibodies than WT mice. Normal and M2-transgenic mice were infected with WT and M2 frame-shift mutant (M2FS) MHV-68 viruses. The pathogenesis of M2-transgenic mice infected with the M2-deficient mutant virus did not revert to that observed upon infection of normal mice with WT virus. However, the higher reactivation levels late after M2-transgenic mice were infected with WT virus reflected the importance of M2 as a target for the immune response, and thus with an impact on the establishment of latency. Finally, there was markedly less apoptosis in B-cells from M2-transgenic mice infected with either WT or M2FS mutant than from similarly infected WT mice, consistent with the published inhibitory influence of M2 on apoptosis in vitro. Thus, M2 provides a strategy to increase the pool of germinal centre B-cells through inhibition of apoptosis in the infected cell.


Asunto(s)
Formación de Anticuerpos/inmunología , Apoptosis/inmunología , Linfocitos B/metabolismo , Rhadinovirus/patogenicidad , Proteínas Virales/metabolismo , Latencia del Virus , Animales , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Linfocitos B/virología , Regulación Viral de la Expresión Génica , Centro Germinal , Hemocianinas/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Ratones , Ratones Transgénicos , Rhadinovirus/genética , Rhadinovirus/metabolismo , Linfocitos T/inmunología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Proteínas Virales/genética , Proteínas Virales/inmunología , Replicación Viral
18.
J Virol ; 87(10): 6051-4, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23514885

RESUMEN

There has been extensive research regarding T cell recognition of Epstein-Barr virus-transformed cells; however, less is known regarding the recognition of B cells immortalized by gamma-2 herpesviruses. Here we show that B cells immortalized by murine gammaherpesvirus 68 (MHV-68, γHV-68) can be controlled by either CD4 or CD8 T cells in vivo. We present evidence for the direct recognition of infected B cells by CD4 and CD8 T cells. These data will help in the development of immunotherapeutic approaches combating gamma-2 herpesvirus-related disease.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/virología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Transformación Celular Viral , Rhadinovirus/patogenicidad , Animales , Línea Celular Tumoral , Ratones , Ratones Endogámicos C57BL
19.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA