Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Virology ; 526: 155-164, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30390564

RESUMEN

IFNγ is a key regulator of inflammatory responses but its role in influenza A virus (IAV) pathogenesis is unclear. Our studies show that infection of mice lacking the IFNγ receptor (IFNγR-/-) at a dose which caused severe disease in wild type 129 Sv/Ev (WT) mice resulted in milder clinical symptoms and significantly lower lung virus titers by 6 days post-infection (dpi). Viral spread was reduced in IFNγR-/- lungs at 2 and 4 dpi. Levels of inflammatory cytokines and chemokines were lower in IFNγR-/- mice at 2 dpi and there was less infiltration of monocyte/macrophage lineage cells than in WT mice. There was no difference in CD4+ and CD8+ T cells and alveolar macrophages in the bronchoalveolar lavage fluid (BALF) at 2 and 4 dpi but by 4 dpi IFNγR-/- mice had significantly higher percentages of neutrophils. Our data strongly suggest that IAV can use the inflammatory response to promote viral spread.


Asunto(s)
Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/fisiopatología , Receptores de Interferón/genética , Transducción de Señal , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/metabolismo , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Transducción de Señal/genética , Carga Viral , Receptor de Interferón gamma
2.
J Gen Virol ; 96(10): 2951-2960, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26297234

RESUMEN

The role of the macrophage in influenza virus infection is complex. Macrophages are critical for resolution of influenza virus infections but implicated in morbidity and mortality in severe infections. They can be infected with influenza virus and consequently macrophage infection is likely to have an impact on the host immune response. Macrophages display a range of functional phenotypes, from the prototypical pro-inflammatory classically activated cell to alternatively activated anti-inflammatory macrophages involved in immune regulation and wound healing. We were interested in how macrophages of different phenotype respond to influenza virus infection and therefore studied the infection of bone marrow-derived macrophages (BMDMs) of classical and alternative phenotype in vitro. Our results show that alternatively activated macrophages are more readily infected and killed by the virus than classically activated. Classically activated BMDMs express the pro-inflammatory markers inducible nitric oxide synthase (iNOS) and TNF-α, and TNF-α expression was further upregulated following infection. Alternatively activated macrophages express Arginase-1 and CD206; however, following infection, expression of these markers was downregulated whilst expression of iNOS and TNF-α was upregulated. Thus, infection can override the anti-inflammatory state of alternatively activated macrophages. Importantly, however, this results in lower levels of pro-inflammatory markers than those produced by classically activated cells. Our results showed that macrophage phenotype affects the inflammatory macrophage response following infection, and indicated that modulating the macrophage phenotype may provide a route to develop novel strategies to prevent and treat influenza virus infection.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/crecimiento & desarrollo , Macrófagos/fisiología , Macrófagos/virología , Fenotipo , Animales , Supervivencia Celular , Células Cultivadas , Femenino , Inmunofenotipificación , Macrófagos/inmunología , Ratones de la Cepa 129
3.
Proc Natl Acad Sci U S A ; 111(17): 6401-6, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24733924

RESUMEN

There is a need for new approaches for the control of influenza given the burden caused by annual seasonal outbreaks, the emergence of viruses with pandemic potential, and the development of resistance to current antiviral drugs. We show that multivalent biologics, engineered using carbohydrate-binding modules specific for sialic acid, mask the cell-surface receptor recognized by the influenza virus and protect mice from a lethal challenge with 2009 pandemic H1N1 influenza virus. The most promising biologic protects mice when given as a single 1-µg intranasal dose 7 d in advance of viral challenge. There also is sufficient virus replication to establish an immune response, potentially protecting the animal from future exposure to the virus. Furthermore, the biologics appear to stimulate inflammatory mediators, and this stimulation may contribute to their protective ability. Our results suggest that this host-targeted approach could provide a front-line prophylactic that has the potential to protect against any current and future influenza virus and possibly against other respiratory pathogens that use sialic acid as a receptor.


Asunto(s)
Gripe Humana/metabolismo , Gripe Humana/prevención & control , Ingeniería de Proteínas , Receptores Virales/metabolismo , Animales , Peso Corporal , Quimiocinas/metabolismo , Perros , Humanos , Mediadores de Inflamación/metabolismo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Pulmón/patología , Pulmón/virología , Células de Riñón Canino Madin Darby , Ratones , Ácido N-Acetilneuramínico/metabolismo , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Receptores de Superficie Celular/metabolismo , Análisis de Supervivencia
4.
J Gen Virol ; 93(Pt 5): 980-986, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22258859

RESUMEN

The emergence of drug-resistant strains of influenza virus has catalysed a search for new antiviral agents to supplement or replace existing drugs. Following the success of the human immunodeficiency virus entry blocker Enfuvirtide, there has been a resurgence of interest in peptide-based antivirals. In this paper, we report on the discovery of a novel family of peptides (FluPep, FP) that function as inhibitors of influenza A virus infection. The prototype peptide (FP1, also known as Tkip) interacts with haemagglutinin and inhibits the binding of the virus to cell membranes. Using a plaque-reduction assay, we have demonstrated that a variety of influenza A virus subtypes (including H1N1, H3N2 and H5N1) are inhibited by FluPep and its derivatives at nanomolar concentrations. By truncating FluPep we have identified a minimal sequence of 6 aa that binds to haemagglutinin and inhibits infection. Using a mouse model of intranasal influenza virus infection, we observed potent inhibition of virus infection when peptide is given at the time of virus administration. These data indicate that FluPep is a highly effective anti-influenza agent with the potential to translate to the clinic.


Asunto(s)
Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Péptidos/farmacología , Acoplamiento Viral/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Unión Proteica , Mapeo de Interacción de Proteínas , Ensayo de Placa Viral
5.
PLoS One ; 7(1): e29443, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238612

RESUMEN

Influenza virus infection accounts for significant morbidity and mortality world-wide. Interactions of the virus with host cells, particularly those of the macrophage lineage, are thought to contribute to various pathological changes associated with poor patient outcome. Development of new strategies to treat disease therefore requires a detailed understanding of the impact of virus infection upon cellular responses. Here we report that human blood-derived monocytes could be readily infected with the H3N2 influenza virus A/Udorn/72 (Udorn), irrespective of their phenotype (CD14(++)/CD16(-), CD14(++)/CD16(+) or CD14(dim)CD16(++)), as determined by multi-colour flow cytometry for viral haemagglutinin (HA) expression and cell surface markers 8-16 hours post infection. Monocytes are relatively resistant to influenza-induced cell death early in infection, as approximately 20% of cells showed influenza-induced caspase-dependent apoptosis. Infection of monocytes with Udorn also induced the release of IL-6, IL-8, TNFα and IP-10, suggesting that NS1 protein of Udorn does not (effectively) inhibit this host defence response in human monocytes. Comparative analysis of human monocyte-derived macrophages (Mph) demonstrated greater susceptibility to human influenza virus than monocytes, with the majority of both pro-inflammatory Mph1 and anti-inflammatory/regulatory Mph2 cells expressing viral HA after infection with Udorn. Influenza infection of macrophages also induced cytokine and chemokine production. However, both Mph1 and Mph2 phenotypes released comparable amounts of TNFα, IL-12p40 and IP-10 after infection with H3N2, in marked contrast to differential responses to LPS-stimulation. In addition, we found that influenza virus infection augmented the capacity of poorly phagocytic Mph1 cells to phagocytose apoptotic cells by a mechanism that was independent of either IL-10 or the Mer receptor tyrosine kinase/Protein S pathway. In summary, our data reveal that influenza virus infection of human macrophages causes functional alterations that may impact on the process of resolution of inflammation, with implications for viral clearance and lung pathology.


Asunto(s)
Diferenciación Celular/fisiología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Gripe Humana/patología , Macrófagos/patología , Monocitos/patología , Animales , Apoptosis/inmunología , Apoptosis/fisiología , Caspasas/metabolismo , Caspasas/fisiología , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Citocinas/fisiología , Citofagocitosis/inmunología , Citofagocitosis/fisiología , Susceptibilidad a Enfermedades , Perros , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Gripe Humana/sangre , Gripe Humana/inmunología , Macrófagos/clasificación , Macrófagos/fisiología , Macrófagos/virología , Monocitos/clasificación , Monocitos/fisiología , Monocitos/virología
6.
PLoS One ; 6(10): e25333, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22031815

RESUMEN

The extensive world-wide morbidity and mortality caused by influenza A viruses highlights the need for new insights into the host immune response and novel treatment approaches. Cationic Host Defense Peptides (CHDP, also known as antimicrobial peptides), which include cathelicidins and defensins, are key components of the innate immune system that are upregulated during infection and inflammation. Cathelicidins have immunomodulatory and anti-viral effects, but their impact on influenza virus infection has not been previously assessed. We therefore evaluated the effect of cathelicidin peptides on disease caused by influenza A virus in mice. The human cathelicidin, LL-37, and the murine cathelicidin, mCRAMP, demonstrated significant anti-viral activity in vivo, reducing disease severity and viral replication in infected mice to a similar extent as the well-characterized influenza virus-specific antiviral drug zanamivir. In vitro and in vivo experiments suggested that the peptides may act directly on the influenza virion rather than via receptor-based mechanisms. Influenza virus-infected mice treated with LL-37 had lower concentrations of pro-inflammatory cytokines in the lung than did infected animals that had not been treated with cathelicidin peptides. These data suggest that treatment of influenza-infected individuals with cathelicidin-derived therapeutics, or modulation of endogenous cathelicidin production may provide significant protection against disease.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/uso terapéutico , Antivirales/uso terapéutico , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Línea Celular , Perros , Femenino , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Catelicidinas
7.
Vaccine ; 29(23): 3935-44, 2011 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-21481326

RESUMEN

Human gammaherpesviruses such as Epstein-Barr virus (EBV) cause lifelong infections and associated diseases, by virtue of their ability to establish latent infection. Many studies performed in the past years in murine herpesvirus 68 (MHV-68) model of infection suggested that the limited immunity generated against isolated viral components by subunit vaccines cannot counteract the multiple immune evasion strategies operated by gammaherpesviruses. Indeed, a significant inhibition of long-term latency establishment could be observed in mice vaccinated with strains of genetically modified MHV-68 defective in reactivation or establishment of latency. In this study, we focused on the effects of interferon-α (IFN-α) on both the lytic and latent phase of MHV-68 infection, as exerted by the constitutive release of IFN-α1 by a clone of MHV-68 genetically modified to produce this cytokine (MHV-68mIFNα1). Although the MHV-68mIFNα1 recombinant virus exhibited in vitro replication features indistinguishable from those of the wild type MHV-68, its pathological properties were severely attenuated in vivo in immunocompetent mice and not in mice rendered genetically unresponsive to type I IFN, suggesting that a stronger immune response was primed in the presence of the cytokine. Notably, MHV-68mIFNα1 attenuation did not result in a reduced level of long-term spleen latency establishment. These results prompted us to evaluate the efficacy of MHV-68mIFNα1 in a prophylactic vaccination regimen aimed at inhibiting the symptoms of acute virus infection and the establishment of long-term latency after MHV-68 challenge. Our results show that mice vaccinated with MHV-68mIFNα1, administered as a live-attenuated or partially inactivated (by Psoralen and UV treatment) vaccine, were protected against the challenge with wt MHV-68 from all phases of infection. The ability of MHV-68mIFNα1 to produce IFN-α at the site of the infection, thus efficiently stimulating the immune system in case of virus reactivation from latency, makes this recombinant virus a safer live-attenuated vaccine as compared to the previously reported latency-deficient clones.


Asunto(s)
Infecciones por Herpesviridae/prevención & control , Interferón-alfa/metabolismo , Organismos Modificados Genéticamente/fisiología , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/prevención & control , Vacunas Atenuadas/administración & dosificación , Animales , Línea Celular , Femenino , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Humanos , Interferón-alfa/genética , Ratones , Ratones Endogámicos C57BL , Organismos Modificados Genéticamente/genética , Organismos Modificados Genéticamente/metabolismo , Recombinación Genética , Rhadinovirus/genética , Rhadinovirus/metabolismo , Bazo , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Replicación Viral
8.
J Virol ; 85(6): 2907-17, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191029

RESUMEN

RNA interference (RNAi) is an important mosquito defense mechanism against arbovirus infection. In this paper we study the processes underlying antiviral RNAi in Aedes albopictus-derived U4.4 mosquito cells infected with Semliki Forest virus (SFV) (Togaviridae; Alphavirus). The production of virus-derived small interfering RNAs (viRNAs) from viral double-stranded RNA (dsRNA) is a key event in this host response. dsRNA could be formed by RNA replication intermediates, by secondary structures in RNA genomes or antigenomes, or by both. Which of these dsRNAs is the substrate for the generation of viRNAs is a fundamental question. Here we used deep sequencing of viRNAs and bioinformatic analysis of RNA secondary structures to gain insights into the characteristics and origins of viRNAs. An asymmetric distribution of SFV-derived viRNAs with notable areas of high-level viRNA production (hot spots) and no or a low frequency of viRNA production (cold spots) along the length of the viral genome with a slight bias toward the production of genome-derived viRNAs over antigenome-derived viRNAs was observed. Bioinformatic analysis suggests that hot spots of viRNA production are rarely but not generally associated with putative secondary structures in the SFV genome, suggesting that most viRNAs are derived from replicative dsRNA. A pattern of viRNAs almost identical to those of A. albopictus cells was observed for Aedes aegypti-derived Aag2 cells, suggesting common mechanisms that lead to viRNA production. Hot-spot viRNAs were found to be significantly less efficient at mediating antiviral RNAi than cold-spot viRNAs, pointing toward a nucleic acid-based viral decoy mechanism to evade the RNAi response.


Asunto(s)
Aedes/fisiología , Aedes/virología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Virus de los Bosques Semliki/crecimiento & desarrollo , Aedes/inmunología , Animales , Línea Celular , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/genética , ARN Viral/genética , ARN Viral/metabolismo , Virus de los Bosques Semliki/genética
9.
PLoS One ; 4(8): e6492, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19652715

RESUMEN

BACKGROUND: Detection, isolation, and identification of individual virus infected cells during long term infection are critical to advance our understanding of mechanisms of pathogenesis for latent/persistent viruses. However, current approaches to study these viruses in vivo have been hampered by low sensitivity and effects of cell-type on expression of viral encoded reporter genes. We have designed a novel Cre recombinase (Cre)-based murine system to overcome these problems, and thereby enable tracking and isolation of individual in vivo infected cells. METHODOLOGY/PRINCIPAL FINDINGS: Murine gammaherpesvirus 68 (MHV-68) was used as a prototypic persistent model virus. A Cre expressing recombinant virus was constructed and characterised. The virus is attenuated both in lytic virus replication, producing ten-fold lower lung virus titres than wild type virus, and in the establishment of latency. However, despite this limitation, when the sEGFP7 mouse line containing a Cre-activated enhanced green fluorescent protein (EGFP) was infected with the Cre expressing virus, sites of latent and persistent virus infection could be identified within B cells and macrophages of the lymphoid system on the basis of EGFP expression. Importantly, the use of the sEGFP7 mouse line which expresses high levels of EGFP allowed individual virus positive cells to be purified by FACSorting. Virus gene expression could be detected in these cells. Low numbers of EGFP positive cells could also be detected in the bone marrow. CONCLUSIONS/SIGNIFICANCE: The use of this novel Cre-based virus/mouse system allowed identification of individual latently infected cells in vivo and may be useful for the study and long-term monitoring of other latent/persistent virus infections.


Asunto(s)
Gammaherpesvirinae/aislamiento & purificación , Infecciones por Herpesviridae/virología , Integrasas/metabolismo , Animales , Linfocitos B/virología , Secuencia de Bases , Southern Blotting , Línea Celular , Cricetinae , Citometría de Flujo , Gammaherpesvirinae/genética , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Recombinación Genética
10.
J Gen Virol ; 90(Pt 9): 2061-72, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570957

RESUMEN

Arthropod-borne viruses - arboviruses - are a significant threat to public health. Whilst there is considerable knowledge about arbovirus interactions with vertebrate immunity, relatively little is known about how vectors such as mosquitoes control arbovirus infections. In this review, we discuss novel findings in the field of mosquito antiviral responses to arboviruses, in particular RNA interference, the up-and-coming field of general immune-signalling pathways, and cell death/apoptosis.


Asunto(s)
Infecciones por Arbovirus/virología , Arbovirus/inmunología , Culicidae/inmunología , Culicidae/virología , Inmunidad Innata , Insectos Vectores/inmunología , Animales , Infecciones por Arbovirus/transmisión , Culicidae/genética , Humanos , Insectos Vectores/genética , Insectos Vectores/virología
11.
J Virol ; 83(11): 5735-48, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19297476

RESUMEN

In their vertebrate hosts, arboviruses such as Semliki Forest virus (SFV) (Togaviridae) generally counteract innate defenses and trigger cell death. In contrast, in mosquito cells, following an early phase of efficient virus production, a persistent infection with low levels of virus production is established. Whether arboviruses counteract RNA interference (RNAi), which provides an important antiviral defense system in mosquitoes, is an important question. Here we show that in Aedes albopictus-derived mosquito cells, SFV cannot prevent the establishment of an antiviral RNAi response or prevent the spread of protective antiviral double-stranded RNA/small interfering RNA (siRNA) from cell to cell, which can inhibit the replication of incoming virus. The expression of tombusvirus siRNA-binding protein p19 by SFV strongly enhanced virus spread between cultured cells rather than virus replication in initially infected cells. Our results indicate that the spread of the RNAi signal contributes to limiting virus dissemination.


Asunto(s)
Culicidae/virología , Interferencia de ARN , Virus de los Bosques Semliki/genética , Animales , Línea Celular , Cricetinae , Regulación Viral de la Expresión Génica , ARN Viral/genética , Replicación Viral
12.
Hepatol Res ; 39(2): 187-94, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19208039

RESUMEN

AIM: Infection of gamma interferon receptor defective mice with murid herpesvirus-4 also known as murine gammaherpesvirus-68 results in multi-organ fibrosis. In this paper we characterise the pathological changes occurring in the liver in this model. METHODS: Standard immunohistochemistry and in situ hybridisation techniques were used to identify the cellular changes and the presence of virus at different times post infection. RESULTS: In liver sections from infected gamma interferon receptor defective mice sampled on day 16 to at least day 120, 79% showed proliferating intrahepatic bile ducts associated with a chronic mononuclear cell inflammation. Only 8% of wild type mice showed similar lesions. Coincident with the inflammatory response bile duct epithelial cells were positive for arginase 1. Around day 50 post infection onwards focal fibrotic lesions appeared in approximately 30% of gamma interferon receptor defective mice resulting in destruction of intrahepatic bile ducts. In contrast to the chronic persisting inflammatory response the presence of virus infected cells were only observed between day 12-20 post-infection. CONCLUSION: Infection of gamma interferon receptor defective mice with a murine gammaherpesvirus initiates a chronic persisting inflammatory response with a pathological profile similar to the human fibrotic liver disorder Primary Sclerosing Cholangitis.

13.
J Virol ; 83(5): 2321-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19109392

RESUMEN

Noncoding RNAs are a feature of many herpesvirus genomes. They include microRNAs, whose function is the subject of intense investigation, in addition to longer RNA molecules such as the Epstein-Barr virus-encoded RNAs and herpesvirus saimiri U RNAs, which have been known for some time but whose function is still not well defined. Murine gammaherpesvirus 68 (MHV-68) encodes eight viral tRNA-like molecules (vtRNAs) of unknown function. Investigating the kinetics of expression of the vtRNAs, we observed that they were present directly after infection with the virus. This strongly suggested that vtRNAs were part of the virion structure, which was confirmed by their detection within various purified, RNase-treated preparations. Although both viral and cellular mRNAs were also detected within the MHV-68 virion, the major RNA species present were small RNAs of around 70 nucleotides in length. Interestingly, incorporation of viral mRNA was not related to the relative abundance in infected cells, as M11 mRNA, which is present at low abundance, was found in virions. MHV-76, which lacks the genes encoding the vtRNAs, also incorporated small RNA molecules within the virion, suggesting a requirement for these molecules for virion maturation. In productively infected cells the vtRNAs localized predominantly within the cytoplasm, although they also exhibited a globular pattern of nuclear staining. Their presence in the cytoplasm is consistent with interaction with virion components prior to maturation of virus particles. The significance of these findings for virion architecture and function is discussed.


Asunto(s)
ARN de Transferencia/metabolismo , ARN Viral/metabolismo , Rhadinovirus/metabolismo , Virión/metabolismo , Animales , Línea Celular , Ratones , ARN de Transferencia/aislamiento & purificación , ARN Viral/aislamiento & purificación , Rhadinovirus/genética , Rhadinovirus/fisiología , Virión/genética , Virión/fisiología , Ensamble de Virus
14.
J Leukoc Biol ; 84(1): 50-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18436582

RESUMEN

Murine gammaherpesvirus 68 (MHV-68) is a natural pathogen of rodents closely related to the human gammaherpesviruses Kaposi's sarcoma-associated herpesvirus and EBV. Following intranasal infection, the virus replicates in the lung epithelium prior to establishing latent infection in lymphoid tissue. Infection of mice deficient in IFN-gammaR signaling (IFN-gammaR-/-) results in a multiple organ fibrosis, in which the spleen is severely affected. We show here that by Day 12 postinfection, prior to development of fibrosis in the spleens of IFN-gammaR-/- mice, different subsets of splenic macrophages (Mvarphis) are morphologically activated and enter latently infected germinal centers (GCs). Mvarphis coexpressing arginase I (ARG1), a marker of alternative activation of Mvarphis, and murine Mvarphi markers F4/80, ER-TR9, and MOMA-1 are found in GCs of IFN-gammaR-/- mice but not of wild-type mice. Quantitative RT-PCR of spleen RNA confirms induction of ARG1 and in addition, shows up-regulation of found in inflammatory zone 1/resistin-like molecule-alpha, tissue inhibitor of metalloproteinase-1, matrix metalloproteinase-12, fibronectin, and factor XIIIA in IFN-gammaR-/- mice. In contrast, inducible NO synthase, associated with classical Mvarphi activation, is up-regulated following infection of wild-type mice but not IFN-gammaR(-/-) mice. Concomitant with the aaMvarphis, transcription of the Th2 cytokines IL-13, IL-21, and IL-5 is up-regulated. Thus, in the absence of IFN-gammaR signaling, MHV-68 initiates a Th2 immune response, leading to alternative activation of macrophages and induction of fibrosis. This system provides an important model for studying the pathogenesis of fibrosis initiated by a latent herpesvirus infection.


Asunto(s)
Gammaherpesvirinae/fisiología , Activación de Macrófagos/inmunología , Macrófagos/virología , Animales , Movimiento Celular , Citocinas/genética , Fibrosis , Células Germinativas/virología , Cinética , Macrófagos/patología , Ratones , Receptores CCR4/metabolismo , Receptores de Interferón/deficiencia , Bazo/patología , Bazo/virología , Células Th2/inmunología , Transcripción Genética , Regulación hacia Arriba , Receptor de Interferón gamma
15.
J Gen Virol ; 87(Pt 4): 803-807, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16528028

RESUMEN

Murine gammaherpesvirus 68 (MHV-68) encodes a set of unique genes, M1, M2, M3 and M4, and eight non-translated tRNA-like molecules that are thought to be important in virus-host interactions and latent infection. The M4 gene is predicted to encode a novel secreted protein. To investigate the role of M4 in viral pathogenesis, a mutant MHV-68 that did not express M4 was constructed and its replication was characterized in vitro and in vivo. Virus replication was identical to the wild type in vitro and no difference could be detected in virus replication in the lung following intranasal infection. However, in the spleen, virus deficient in M4 expression was severely attenuated in the establishment of latency. These results indicate a critical role for M4 in MHV-68 pathogenesis.


Asunto(s)
Gammaherpesvirinae/patogenicidad , Regulación Viral de la Expresión Génica , Proteínas Virales/genética , Proteínas Virales/metabolismo , Latencia del Virus , Animales , Línea Celular , Cricetinae , Gammaherpesvirinae/genética , Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/virología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Mutación , Bazo/virología , Replicación Viral
16.
Pathol Int ; 55(9): 558-68, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16143031

RESUMEN

Murine gammaherpesvirus (MHV)-68-infected mice are well-known as models for Epstein-Barr virus (EBV)-related lymphoproliferative diseases. MHV-72 may be a relative of MHV-68, but any genetic comparison between the two (except for the M7 gene) has never been reported. The genetic compositions of MHV-72 and MHV-68 were compared and the pathology of MHV-72 infection studied in CB-17 severe combined immunodeficiency (scid/scid; SCID) and CB17 wild-type (CB17+/+) mice. The MHV-72 DNA sequence was almost identical to MHV-68 except for approximately 7000 bp corresponding to the MHV-68 M1-M3 genes. Twenty-seven of 30 MHV-72-infected SCID mice (90%) died from generalized infection with intranuclear viral inclusions for approximately 1 month, while MHV-72-infected CB17+/+ mice recovered from acute infection. Long observation and pathological study of 68 MHV-72-infected mice for up to 24 months revealed that the survival rate (29.4%) and survival time (21.3 months) of MHV-72-infected CB17+/+ mice were significantly lower (P = 0.0127) and shorter (P = 0.0065) than those of the controls (61.1% and 22.9 months), respectively. The malignancy development rate (60.3%) of the infected CB17+/+ mice was also significantly higher (P = 0.004) than those of the controls (22.2%). However, no MHV-72 DNA was detected in the tumors of infected mice. MHV-72 may have some tumor-promoting effects but the tumorigenesis in infected CB17+/+ mice is different from EBV-associated tumors.


Asunto(s)
Gammaherpesvirinae/genética , Genoma Viral , Infecciones por Herpesviridae/virología , Neoplasias/virología , Inmunodeficiencia Combinada Grave/genética , Infecciones Tumorales por Virus/virología , Animales , Cartilla de ADN/química , ADN Viral/análisis , Modelos Animales de Enfermedad , Femenino , Gammaherpesvirinae/clasificación , Gammaherpesvirinae/patogenicidad , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/mortalidad , Hibridación in Situ , Ratones , Ratones SCID , Neoplasias/inmunología , Neoplasias/patología , ARN Viral/análisis , Homología de Secuencia de Ácido Nucleico , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Tasa de Supervivencia , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/mortalidad
17.
J Gen Virol ; 85(Pt 6): 1393-1400, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15166421

RESUMEN

Infection with the murine gammaherpesvirus MHV-68 has profound effects on splenic and mediastinal lymph node pathology in mice which lack the interferon-gamma receptor (IFN-gamma R(-/-)). In these mice MHV-68 infection causes fibrosis and loss of lymphocytes in the spleen and the mediastinal lymph node as well as interstitial pulmonary fibrosis and fibrotic changes in the liver. The changes are associated with transient elevated latent virus loads in the spleen. Four independent virus mutants with insertions and/or deletions in the left end of the genome fail to induce the pathological changes and establish latency at normal levels in the spleen. The data indicate that the pathology does not correlate with any of the known genes encoded within this region of the genome, genes M1-M4 and the eight vtRNAs. Northern analysis of mRNAs transcribed by wild-type and mutant viruses shows that at least two uncharacterized transcripts are encoded within this region. These transcripts are absent in the mutant viruses and are candidates for the virus genes responsible for the aberrant pathology in IFN-gamma R(-/-) mice.


Asunto(s)
Genoma Viral , Rhadinovirus/genética , Bazo/patología , Animales , Ratones , Receptores de Interferón/fisiología , Rhadinovirus/patogenicidad , Receptor de Interferón gamma
18.
Vaccine ; 22(11-12): 1433-40, 2004 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-15063566

RESUMEN

Human gammaherpesviruses such as Epstein-Barr virus (EBV) cause lifelong infections and associated diseases, including malignancies, and the development of an effective vaccine against this class of viral infections is of considerable interest. The murine herpesvirus 68 (MHV-68) model provides a useful experimental setting to investigate the immune response to gammaherpesvirus infections and to evaluate the efficacy of vaccination strategies. In this study, we tested a heat-inactivated MHV-68 vaccine in immunocompetent mice as well as in B cell-deficient or type I IFN receptor knockout mice. Vaccination with heat-inactivated MHV-68 protected immunocompetent mice from the acute MHV-68 infection in the lung and strongly reduced the expansion of latently infected cells in the spleen and the development of splenomegaly. A similar inhibition of the acute viral replication in the lung was also observed in vaccinated B cell-deficient mice. Of note, the inactivated MHV-68 vaccine completely protected type I IFN receptor knockout mice from the infection with a lethal dose of MHV-68.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Herpesviridae/inmunología , Vacunas contra Herpesvirus/inmunología , Interferón Tipo I/fisiología , Receptores de Interferón/fisiología , Latencia del Virus/inmunología , Replicación Viral/fisiología , Animales , Linfocitos B/inmunología , Linfocitos B/fisiología , Línea Celular , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Receptores de Interferón/genética , Vacunación , Vacunas de Productos Inactivados/inmunología
19.
J Virol ; 77(17): 9700-9, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12915582

RESUMEN

This work describes analyses of the function of the murid herpesvirus 4 strain 68 (MHV-68) M2 gene. A frameshift mutation was made in the M2 open reading frame that caused premature termination of translation of M2 after amino acid residue 90. The M2 mutant showed no defect in productive replication in vitro or in lungs after infection of mice. Likewise, the characteristic transient increase in spleen cell number, Vbeta4 T-cell-receptor-positive CD8(+) T-cell mononucleosis, and establishment of latency were unaffected. However, the M2 mutant virus was defective in its ability to cause the transient sharp rise in latently infected cells normally seen in the spleen after infection of mice. We also demonstrate that expression of M2 is restricted to B cells in the spleen and that M2 encodes a 30-kDa protein localizing predominantly in the cytoplasm and plasma membrane of B cells.


Asunto(s)
Antígenos Virales/genética , Linfocitos B/inmunología , Linfocitos B/virología , Rhadinovirus/inmunología , Rhadinovirus/patogenicidad , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Cricetinae , ADN Viral/genética , Genes Virales , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Técnicas In Vitro , Pulmón/inmunología , Pulmón/virología , Linfocitosis/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Rhadinovirus/genética , Rhadinovirus/fisiología , Bazo/inmunología , Bazo/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/virología , Transfección , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Replicación Viral
20.
J Gen Virol ; 84(Pt 1): 99-109, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12533705

RESUMEN

The murine gammaherpesvirus-68 genome encodes 73 protein-coding open reading frames with extensive similarities to human gamma(2) herpesviruses, as well as unique genes and cellular homologues. We performed transcriptome analysis of stage-specific viral RNA during permissive infection using an oligonucleotide-based microarray. Using this approach, M4, K3, ORF38, ORF50, ORF57 and ORF73 were designated as immediate-early genes based on cycloheximide treatment. The microarray analysis also identified 10 transcripts with early expression kinetics, 32 transcripts with early-late expression kinetics and 29 transcripts with late expression kinetics. The latter group consisted mainly of structural proteins, and showed high expression levels relative to other viral transcripts. Moreover, we detected all eight tRNA-like transcripts in the presence of cycloheximide and phosphonoacetic acid. Lytic infection with MHV-68 also resulted in a significant reduction in the expression of cellular transcripts included in the DNA chip. This global approach to viral transcript analysis offers a powerful system for examining molecular transitions between lytic and latent virus infections associated with disease pathogenesis.


Asunto(s)
Gammaherpesvirinae/fisiología , Gammaherpesvirinae/patogenicidad , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Proteoma , Proteínas Virales/metabolismo , Animales , Células Cultivadas , Cicloheximida/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/virología , Gammaherpesvirinae/genética , Infecciones por Herpesviridae/virología , Ratones , Sistemas de Lectura Abierta/genética , Ácido Fosfonoacético/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Proteínas Virales/genética , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA