Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 12(5): e1005654, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27223694

RESUMEN

Gamma-herpesviruses colonise lymphocytes. Murid Herpesvirus-4 (MuHV-4) infects B cells via epithelial to myeloid to lymphoid transfer. This indirect route entails exposure to host defences, and type I interferons (IFN-I) limit infection while viral evasion promotes it. To understand how IFN-I and its evasion both control infection outcomes, we used Mx1-cre mice to tag floxed viral genomes in IFN-I responding cells. Epithelial-derived MuHV-4 showed low IFN-I exposure, and neither disrupting viral evasion nor blocking IFN-I signalling markedly affected acute viral replication in the lungs. Maximising IFN-I induction with poly(I:C) increased virus tagging in lung macrophages, but the tagged virus spread poorly. Lymphoid-derived MuHV-4 showed contrastingly high IFN-I exposure. This occurred mainly in B cells. IFN-I induction increased tagging without reducing viral loads; disrupting viral evasion caused marked attenuation; and blocking IFN-I signalling opened up new lytic spread between macrophages. Thus, the impact of IFN-I on viral replication was strongly cell type-dependent: epithelial infection induced little response; IFN-I largely suppressed macrophage infection; and viral evasion allowed passage through B cells despite IFN-I responses. As a result, IFN-I and its evasion promoted a switch in infection from acutely lytic in myeloid cells to chronically latent in B cells. Murine cytomegalovirus also showed a capacity to pass through IFN-I-responding cells, arguing that this is a core feature of herpesvirus host colonization.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Evasión Inmune/inmunología , Interferón Tipo I/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Gammaherpesvirinae/inmunología , Gammaherpesvirinae/patogenicidad , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Células RAW 264.7
2.
J Gen Virol ; 96(8): 2314-2327, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25872742

RESUMEN

Lymphocyte proliferation, mobility and longevity make them prime targets for virus infection. Myeloid cells that process and present environmental antigens to lymphocytes are consequently an important line of defence. Subcapsular sinus macrophages (SSMs) filter the afferent lymph and communicate with B-cells. How they interact with B-cell-tropic viruses is unknown. We analysed their encounter with murid herpesvirus-4 (MuHV-4), an experimentally accessible gammaherpesvirus related to Kaposi's sarcoma-associated herpesvirus. MuHV-4 disseminated via lymph nodes, and intranasally or subcutaneously inoculated virions readily infected SSMs. However, this infection was poorly productive. SSM depletion with clodronate-loaded liposomes or with diphtheria toxin in CD169-diphtheria toxin receptor transgenic mice increased B-cell infection and hastened virus spread to the spleen. Dendritic cells provided the main route to B-cells, and SSMs slowed host colonization, apparently by absorbing virions non-productively from the afferent lymph.


Asunto(s)
Infecciones por Herpesviridae/virología , Macrófagos/virología , Rhadinovirus/fisiología , Animales , Infecciones por Herpesviridae/inmunología , Humanos , Ganglios Linfáticos/virología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Rhadinovirus/genética , Rhadinovirus/inmunología
3.
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
4.
Cell Host Microbe ; 15(4): 457-70, 2014 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-24721574

RESUMEN

Gamma-herpesviruses (γHVs) are widespread oncogenic pathogens that chronically infect circulating lymphocytes. How they subvert the immune check-point function of the spleen to promote persistent infection is not clear. We show that Murid Herpesvirus-4 (MuHV-4) enters the spleen by infecting marginal zone (MZ) macrophages, which provided a conduit to MZ B cells. Relocation of MZ B cells to the white pulp allowed virus transfer to follicular dendritic cells. From here the virus reached germinal center B cells to establish persistent infection. Mice lacking MZ B cells, or treated with a sphingosine-1-phosphate receptor agonist to dislocate them, were protected against MuHV-4 colonization. MuHV-4 lacking ORF27, which encodes a glycoprotein necessary for efficient intercellular spread, could infect MZ macrophages but was impaired in long-term infection. Thus, MuHV-4, a γHV, exploits normal immune communication routes to spread by serial lymphoid/myeloid exchange.


Asunto(s)
Células Dendríticas Foliculares/virología , Centro Germinal/virología , Infecciones por Herpesviridae/inmunología , Macrófagos/virología , Rhadinovirus/inmunología , Animales , Linfocitos B/virología , Línea Celular , Cricetinae , Centro Germinal/citología , Infecciones por Herpesviridae/transmisión , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Sistemas de Lectura Abierta/genética , Receptores de Lisoesfingolípidos/metabolismo , Rhadinovirus/genética , Bazo/patología , Bazo/virología
5.
J Virol ; 88(8): 3965-75, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24501409

RESUMEN

UNLABELLED: Lymphocyte colonization by gammaherpesviruses (γHVs) is an important target for cancer prevention. However, how it works is not clear. Epstein-Barr virus drives autonomous B cell proliferation in vitro but in vivo may more subtly exploit the proliferative pathways provided by lymphoid germinal centers (GCs). Murid herpesvirus 4 (MuHV-4), which realistically infects inbred mice, provides a useful tool with which to understand further how a γHV colonizes B cells in vivo. Not all γHVs necessarily behave the same, but common events can with MuHV-4 be assigned an importance for host colonization and so a potential as therapeutic targets. MuHV-4-driven B cell proliferation depends quantitatively on CD4(+) T cell help. Here we show that it also depends on T cell-independent survival signals provided by the B cell-activating factor (BAFF) receptor (BAFF-R). B cells could be infected in BAFF-R(-/-) mice, but virus loads remained low. This corresponded to a BAFF-R-dependent defect in GC colonization. The close parallels between normal, antigen-driven B cell responses and virus-infected B cell proliferation argue that in vivo, γHVs mostly induce infected B cells into normal GC reactions rather than generating large numbers of autonomously proliferating blasts. IMPORTANCE: γHVs cause cancers by driving the proliferation of infected cells. B cells are a particular target. Thus, we need to know how virus-driven B cell proliferation works. Controversy exists as to whether viral genes drive it directly or less directly orchestrate the engagement of normal, host-driven pathways. Here we show that the B cell proliferation driven by a murid γHV requires BAFF-R. This supports the idea that γHVs exploit host proliferation pathways and suggests that interfering with BAFF-R could more generally reduce γHV-associated B cell proliferation.


Asunto(s)
Factor Activador de Células B/metabolismo , Receptor del Factor Activador de Células B/deficiencia , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Rhadinovirus/fisiología , Animales , Receptor del Factor Activador de Células B/genética , Linfocitos B/metabolismo , Linfocitos B/virología , Femenino , Gammaherpesvirinae/genética , Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Rhadinovirus/genética
6.
J Virol ; 87(19): 10828-42, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23903840

RESUMEN

Glycoprotein B (gB) is a conserved herpesvirus virion component implicated in membrane fusion. As with many-but not all-herpesviruses, the gB of murid herpesvirus 4 (MuHV-4) is cleaved into disulfide-linked subunits, apparently by furin. Preventing gB cleavage for some herpesviruses causes minor infection deficits in vitro, but what the cleavage contributes to host colonization has been unclear. To address this, we mutated the furin cleavage site (R-R-K-R) of the MuHV-4 gB. Abolishing gB cleavage did not affect its expression levels, glycosylation, or antigenic conformation. In vitro, mutant viruses entered fibroblasts and epithelial cells normally but had a significant entry deficit in myeloid cells such as macrophages and bone marrow-derived dendritic cells. The deficit in myeloid cells was not due to reduced virion binding or endocytosis, suggesting that gB cleavage promotes infection at a postendocytic entry step, presumably viral membrane fusion. In vivo, viruses lacking gB cleavage showed reduced lytic spread in the lungs. Alveolar epithelial cell infection was normal, but alveolar macrophage infection was significantly reduced. Normal long-term latency in lymphoid tissue was established nonetheless.


Asunto(s)
Glicoproteínas/metabolismo , Pulmón/virología , Células Mieloides/virología , Rhadinovirus/fisiología , Proteínas del Envoltorio Viral/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Secuencia de Bases , Western Blotting , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Furina/metabolismo , Glicoproteínas/genética , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Mutación/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Proteínas del Envoltorio Viral/genética , Virión , Replicación Viral
7.
J Virol ; 87(19): 10477-88, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23903843

RESUMEN

Herpes simplex virus 1 (HSV-1) is a ubiquitous and important human pathogen. It is known to persist in trigeminal ganglia (TG), but how it reaches this site has been difficult to determine, as viral transmission is sporadic, pathogenesis is complicated, and early infection is largely asymptomatic. We used mice to compare the most likely natural HSV-1 host entry routes: oral and nasal. Intranasal infection was 100-fold more efficient than oral and targeted predominantly the olfactory neuroepithelium. Live imaging of HSV-1-expressed luciferase showed infection progressing from the nose to the TG and then reemerging in the facial skin. The brain remained largely luciferase negative throughout. Infected cell tagging by viral Cre recombinase expression in floxed reporter gene mice showed nasal virus routinely reaching the TG and only rarely reaching the olfactory bulbs. Thus, HSV-1 spread from the olfactory neuroepithelium to the TG and reemerged peripherally without causing significant neurological disease. This recapitulation of typical clinical infection suggests that HSV-1 might sometimes also enter humans via the respiratory tract.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/patogenicidad , Células Neuroepiteliales/virología , Bulbo Olfatorio/virología , Ganglio del Trigémino/virología , Internalización del Virus , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Herpes Simple/genética , Herpes Simple/patología , Humanos , Técnicas para Inmunoenzimas , Riñón/metabolismo , Riñón/patología , Riñón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/patología , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/patología , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/patología , Replicación Viral
8.
PLoS Pathog ; 8(9): e1002935, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028329

RESUMEN

Gamma-herpesviruses persist in lymphocytes and cause disease by driving their proliferation. Lymphocyte infection is therefore a key pathogenetic event. Murid Herpesvirus-4 (MuHV-4) is a rhadinovirus that like the related Kaposi's Sarcoma-associated Herpesvirus persists in B cells in vivo yet infects them poorly in vitro. Here we used MuHV-4 to understand how virion tropism sets the path to lymphocyte colonization. Virions that were highly infectious in vivo showed a severe post-binding block to B cell infection. Host entry was accordingly an epithelial infection and B cell infection a secondary event. Macrophage infection by cell-free virions was also poor, but improved markedly when virion binding improved or when macrophages were co-cultured with infected fibroblasts. Under the same conditions B cell infection remained poor; it improved only when virions came from macrophages. This reflected better cell penetration and correlated with antigenic changes in the virion fusion complex. Macrophages were seen to contact acutely infected epithelial cells, and cre/lox-based virus tagging showed that almost all the virus recovered from lymphoid tissue had passed through lysM(+) and CD11c(+) myeloid cells. Thus MuHV-4 reached B cells in 3 distinct stages: incoming virions infected epithelial cells; infection then passed to myeloid cells; glycoprotein changes then allowed B cell infection. These data identify new complexity in rhadinovirus infection and potentially also new vulnerability to intervention.


Asunto(s)
Linfocitos B/virología , Células Epiteliales/virología , Infecciones por Herpesviridae/virología , Macrófagos/virología , Rhadinovirus/fisiología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/virología , Células 3T3 , Animales , Antígeno CD11c , Línea Celular , Cricetinae , Fibroblastos/virología , Células HEK293 , Infecciones por Herpesviridae/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Células Mieloides/virología , Rhadinovirus/inmunología , Infecciones Tumorales por Virus/inmunología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
9.
PLoS Pathog ; 7(11): e1002346, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22102809

RESUMEN

Dendritic cells (DCs) play a central role in initiating immune responses. Some persistent viruses infect DCs and can disrupt their functions in vitro. However, these viruses remain strongly immunogenic in vivo. Thus what role DC infection plays in the pathogenesis of persistent infections is unclear. Here we show that a persistent, B cell-tropic gamma-herpesvirus, Murid Herpesvirus-4 (MuHV-4), infects DCs early after host entry, before it establishes a substantial infection of B cells. DC-specific virus marking by cre-lox recombination revealed that a significant fraction of the virus latent in B cells had passed through a DC, and a virus attenuated for replication in DCs was impaired in B cell colonization. In vitro MuHV-4 dramatically altered the DC cytoskeleton, suggesting that it manipulates DC migration and shape in order to spread. MuHV-4 therefore uses DCs to colonize B cells.


Asunto(s)
Linfocitos B/virología , Células Dendríticas/virología , Infecciones por Herpesviridae/inmunología , Rhadinovirus/patogenicidad , Animales , Presentación de Antígeno , Antígenos Virales/inmunología , Linfocitos B/inmunología , Línea Celular , Cricetinae , Células Dendríticas/inmunología , Infecciones por Herpesviridae/virología , Integrasas , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Rhadinovirus/inmunología , Rhadinovirus/fisiología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología
10.
J Gen Virol ; 92(Pt 7): 1550-1560, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21471322

RESUMEN

The difficulty of eliminating herpesvirus carriage makes host entry a key target for infection control. However, its viral requirements are poorly defined. Murid herpesvirus-4 (MuHV-4) can potentially provide insights into gammaherpesvirus host entry. Upper respiratory tract infection requires the MuHV-4 thymidine kinase (TK) and ribonucleotide reductase large subunit (RNR-L), suggesting a need for increased nucleotide production. However, both TK and RNR-L are likely to be multifunctional. We therefore tested further the importance of nucleotide production by disrupting the MuHV-4 ribonucleotide reductase small subunit (RNR-S). This caused a similar attenuation to RNR-L disruption: despite reduced intra-host spread, invasive inoculations still established infection, whereas a non-invasive upper respiratory tract inoculation did so only at high dose. Histological analysis showed that RNR-S(-), RNR-L(-) and TK(-) viruses all infected cells in the olfactory neuroepithelium but unlike wild-type virus then failed to spread. Thus captured host nucleotide metabolism enzymes, up to now defined mainly as important for alphaherpesvirus reactivation in neurons, also have a key role in gammaherpesvirus host entry. This seemed to reflect a requirement for lytic replication to occur in a terminally differentiated cell before a viable pool of latent genomes could be established.


Asunto(s)
Rhadinovirus/enzimología , Ribonucleótido Reductasas/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Femenino , Infecciones por Herpesviridae/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Rhadinovirus/genética , Rhadinovirus/fisiología , Ribonucleótido Reductasas/genética , Proteínas Virales/genética , Replicación Viral
11.
J Immunol ; 185(6): 3669-76, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20720208

RESUMEN

A critical factor influencing the ability of the host to mount a robust immune response against a virus depends on the rapid recruitment of dendritic cells (DCs) presenting Ags. From the outset, this step sets the tempo for subsequent activation of virus-specific T cells. Despite this, how induction of the immune response might be modified by pathogens with the capacity to establish persistence is unclear. In this study, we have characterized the in vivo influence of murine gamma-herpesvirus K3-mediated interference with MHC class I in DCs that drive the initial adaptive immune response. We observed that gamma-herpesvirus could interfere with the very earliest phase of Ag presentation through K3 by directly targeting migratory and lymph node-resident DCs. These results show that a pathogen with the capacity to interfere with early Ag presentation can establish suboptimal conditions for rapid induction of the adaptive immune response and thus favor establishment of viral persistence.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Infecciones por Herpesviridae/inmunología , Rhadinovirus/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Enfermedad Crónica , Reactividad Cruzada/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , Infecciones por Herpesviridae/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Rhadinovirus/patogenicidad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/virología , Infecciones Tumorales por Virus/metabolismo , Interferencia Viral/inmunología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/biosíntesis
12.
J Virol ; 84(20): 10937-42, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668075

RESUMEN

Viral enzymes that process small molecules provide potential chemotherapeutic targets. A key constraint-the replicative potential of spontaneous enzyme mutants-has been hard to define with human gammaherpesviruses because of their narrow species tropisms. Here, we disrupted the murid herpesvirus 4 (MuHV-4) ORF61, which encodes its ribonucleotide reductase (RNR) large subunit. Mutant viruses showed delayed in vitro lytic replication, failed to establish infection via the upper respiratory tract, and replicated to only a very limited extent in the lower respiratory tract without reaching lymphoid tissue. RNR could therefore provide a good target for gammaherpesvirus chemotherapy.


Asunto(s)
Rhadinovirus/enzimología , Rhadinovirus/patogenicidad , Ribonucleótido Reductasas/fisiología , Animales , Secuencia de Bases , Línea Celular , Cricetinae , ADN Viral/genética , Genes Virales , Infecciones por Herpesviridae/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutagénesis Insercional , Sistema Respiratorio/virología , Rhadinovirus/genética , Ribonucleótido Reductasas/química , Ribonucleótido Reductasas/genética , Infecciones Tumorales por Virus/virología , Virulencia/genética , Virulencia/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
13.
J Gen Virol ; 91(Pt 9): 2176-85, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20573854

RESUMEN

Cancers with viral aetiologies can potentially be prevented by antiviral vaccines. Therefore, it is important to understand how viral infections and cancers might be linked. Some cancers frequently carry gammaherpesvirus genomes. However, they generally express the same viral genes as non-transformed cells, and differ mainly in also carrying oncogenic host mutations. Infection, therefore, seems to play a triggering or accessory role in disease. The hit-and-run hypothesis proposes that cumulative host mutations can allow viral genomes to be lost entirely, such that cancers remaining virus-positive represent only a fraction of those to which infection contributes. This would have considerable implications for disease control. However, the hit-and-run hypothesis has so far lacked experimental support. Here, we tested it by using Cre-lox recombination to trigger transforming mutations in virus-infected cells. Thus, 'floxed' oncogene mice were infected with Cre recombinase-positive murid herpesvirus-4 (MuHV-4). The emerging cancers showed the expected genetic changes but, by the time of presentation, almost all lacked viral genomes. Vaccination with a non-persistent MuHV-4 mutant nonetheless conferred complete protection. Equivalent human gammaherpesvirus vaccines could therefore potentially prevent not only viral genome-positive cancers, but possibly also some cancers less suspected of a viral origin because of viral genome loss.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Rhadinovirus/inmunología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/prevención & control , Vacunas Virales/farmacología , Animales , Secuencia de Bases , Cartilla de ADN/genética , Genes p53 , Genes ras , Genoma Viral , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Humanos , Ratones , Ratones Mutantes , Ratones Transgénicos , Modelos Biológicos , Mutagénesis Insercional , Mutación , Rhadinovirus/genética , Sarcoma Experimental/genética , Sarcoma Experimental/inmunología , Sarcoma Experimental/prevención & control , Sarcoma Experimental/virología , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/virología , Vacunación/métodos
14.
PLoS One ; 5(6): e11080, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20552028

RESUMEN

The narrow species tropisms of Epstein-Barr Virus (EBV) and the Kaposi's Sarcoma -associated Herpesvirus (KSHV) have made Murid Herpesvirus-4 (MuHV-4) an important tool for understanding how gammaherpesviruses colonize their hosts. However, while MuHV-4 pathogenesis studies can assign a quantitative importance to individual genes, the complexity of in vivo infection can make the underlying mechanisms hard to discern. Furthermore, the lack of good in vitro MuHV-4 latency/reactivation systems with which to dissect mechanisms at the cellular level has made some parallels with EBV and KSHV hard to draw. Here we achieved control of the MuHV-4 lytic/latent switch in vitro by modifying the 5' untranslated region of its major lytic transactivator gene, ORF50. We terminated normal ORF50 transcripts by inserting a polyadenylation signal and transcribed ORF50 instead from a down-stream, doxycycline-inducible promoter. In this way we could establish fibroblast clones that maintained latent MuHV-4 episomes without detectable lytic replication. Productive virus reactivation was then induced with doxycycline. We used this system to show that the MuHV-4 K3 gene plays a significant role in protecting reactivating cells against CD8(+) T cell recognition.


Asunto(s)
Rhadinovirus/fisiología , Activación Viral , Latencia del Virus , Animales , Línea Celular , ADN Viral , Citometría de Flujo , Humanos , Técnicas In Vitro , Ratones , Microscopía Fluorescente , Mutagénesis , Sistemas de Lectura Abierta , Rhadinovirus/genética
15.
J Gen Virol ; 91(Pt 10): 2542-52, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20519454

RESUMEN

Herpesviruses characteristically disseminate from immune hosts. Therefore in the context of natural infection, antibody neutralizes them poorly. Murid herpesvirus-4 (MuHV-4) provides a tractable model with which to understand gammaherpesvirus neutralization. MuHV-4 virions blocked for cell binding by immune sera remain infectious for IgG-Fc receptor(+) myeloid cells, so broadly neutralizing antibodies must target the virion fusion complex - glycoprotein B (gB) or gH/gL. While gB-specific neutralizing antibodies are rare, its domains I+II (gB-N) contain at least one potent neutralization epitope. Here, we tested whether immunization with recombinant gB presenting this epitope could induce neutralizing antibodies in naive mice and protect them against MuHV-4 challenge. Immunizing with the full-length gB extracellular domain induced a strong gB-specific antibody response and reduced MuHV-4 lytic replication but did not induce detectable neutralization. gB-N alone, which more selectively displayed pre-fusion epitopes including neutralization epitopes, also failed to induce neutralizing responses, and while viral lytic replication was again reduced this depended completely on IgG Fc receptors. gB and gB-N also boosted neutralizing responses in only a minority of carrier mice. Therefore, it appears that neutralizing epitopes on gB are intrinsically difficult for the immune response to target.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Glicoproteínas/inmunología , Infecciones por Herpesviridae/prevención & control , Vacunas contra Herpesvirus/inmunología , Rhadinovirus/inmunología , Proteínas Virales/inmunología , Replicación Viral/inmunología , Animales , Anticuerpos Antivirales/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Pruebas de Neutralización , Receptores de IgG/inmunología , Vacunas Sintéticas/inmunología
16.
J Gen Virol ; 90(Pt 5): 1202-1214, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19264603

RESUMEN

Antibodies readily neutralize acute, epidemic viruses, but are less effective against more indolent pathogens such as herpesviruses. Murid herpesvirus 4 (MuHV-4) provides an accessible model for tracking the fate of antibody-exposed gammaherpesvirus virions. Glycoprotein L (gL) plays a central role in MuHV-4 entry: it allows gH to bind heparan sulfate and regulates fusion-associated conformation changes in gH and gB. However, gL is non-essential: heparan sulfate binding can also occur via gp70, and the gB-gH complex alone seems to be sufficient for membrane fusion. Here, we investigated how gL affects the susceptibility of MuHV-4 to neutralization. Immune sera neutralized gL(-) virions more readily than gL(+) virions, chiefly because heparan sulfate binding now depended on gp70 and was therefore easier to block. However, there were also post-binding effects. First, the downstream, gL-independent conformation of gH became a neutralization target; gL normally prevents this by holding gH in an antigenically distinct heterodimer until after endocytosis. Second, gL(-) virions were more vulnerable to gB-directed neutralization. This covered multiple epitopes and thus seemed to reflect a general opening up of the gH-gB entry complex, which gL again normally restricts to late endosomes. gL therefore limits MuHV-4 neutralization by providing redundancy in cell binding and by keeping key elements of the virion fusion machinery hidden until after endocytosis.


Asunto(s)
Anticuerpos Antivirales/inmunología , Glicoproteínas/inmunología , Rhadinovirus/inmunología , Rhadinovirus/metabolismo , Proteínas del Envoltorio Viral/inmunología , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Células Epiteliales , Femenino , Fibroblastos , Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Unión Proteica
17.
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
18.
J Gen Virol ; 90(Pt 3): 602-613, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19218205

RESUMEN

Many herpesviruses bind to heparan sulfate (HS). Murid herpesvirus-4 (MuHV-4) does so via its envelope glycoproteins gp70 and gH/gL. MuHV-4 gp150 further regulates an HS-independent interaction to make that HS-dependent too. Cell binding by MuHV-4 virions is consequently strongly HS-dependent. Gp70 and gH/gL show some in vitro redundancy: an antibody-mediated blockade of HS binding by one is well tolerated, whereas a blockade of both severely impairs infection. In order to understand the importance of HS binding for MuHV-4 in vivo, we generated mutants lacking both gL and gp70. As expected, gL(-)gp70(-) MuHV-4 showed very poor cell binding. It infected mice at high dose but not at low dose, indicating defective host entry. But once entry occurred, host colonization, which for MuHV-4 is relatively independent of the infection dose, was remarkably normal. The gL(-)gp70(-) entry deficit was much greater than that of gL(-) or gp70(-) single knockouts. And gp150 disruption, which allows HS-independent cell binding, largely rescued the gL(-)gp70(-) cell binding and host entry deficits. Thus, it appeared that MuHV-4 HS binding is important in vivo, principally for efficient host entry.


Asunto(s)
Glicoproteínas/metabolismo , Heparitina Sulfato/metabolismo , Receptores Virales/metabolismo , Rhadinovirus/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Cricetinae , Femenino , Glicoproteínas/genética , Riñón/citología , Riñón/virología , Ratones , Células 3T3 NIH , Unión Proteica , Rhadinovirus/genética , Rhadinovirus/metabolismo , Proteínas del Envoltorio Viral/genética
19.
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
20.
PLoS One ; 3(7): e2781, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18648660

RESUMEN

All gamma-herpesviruses encode at least one homolog of the cellular enzyme formyl-glycineamide-phosphoribosyl-amidotransferase. Murid herpesvirus-4 (MuHV-4) encodes 3 (ORFs 75a, 75b and 75c), suggesting that at least some copies have acquired new functions. Here we show that the corresponding proteins are all present in virions and localize to infected cell nuclei. Despite these common features, ORFs 75a and 75b did not substitute functionally for a lack of ORF75c, as ORF75c virus knockouts were severely impaired for lytic replication in vitro and for host colonization in vivo. They showed 2 defects: incoming capsids failed to migrate to the nuclear margin following membrane fusion, and genomes that did reach the nucleus failed to initiate normal gene expression. The latter defect was associated with a failure of in-coming virions to disassemble PML bodies. The capsid transport deficit seemed to be functionally more important, since ORF75c(-) MuHV-4 infected both PML(+) and PML(-) cells poorly. The original host enzyme has therefore evolved into a set of distinct and multi-functional viral tegument proteins. One important function is moving incoming capsids to the nuclear margin for viral genome delivery.


Asunto(s)
Regulación Viral de la Expresión Génica , Sistemas de Lectura Abierta , Rhadinovirus/genética , Rhadinovirus/fisiología , Células 3T3 , Animales , Cápside , Núcleo Celular/metabolismo , Cricetinae , Fibroblastos/metabolismo , Genoma Viral , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Rhadinovirus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...