Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(17): 6401-6, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24733924

RESUMO

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.


Assuntos
Influenza Humana/metabolismo , Influenza Humana/prevenção & controle , Engenharia de Proteínas , Receptores Virais/metabolismo , Animais , Peso Corporal , Quimiocinas/metabolismo , Cães , Humanos , Mediadores da Inflamação/metabolismo , Vírus da Influenza A Subtipo H1N1/fisiologia , Pulmão/patologia , Pulmão/virologia , Células Madin Darby de Rim Canino , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Receptores de Superfície Celular/metabolismo , Análise de Sobrevida
2.
J Gen Virol ; 96(10): 2951-2960, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26297234

RESUMO

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.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Influenza A/crescimento & desenvolvimento , Macrófagos/fisiologia , Macrófagos/virologia , Fenótipo , Animais , Sobrevivência Celular , Células Cultivadas , Feminino , Imunofenotipagem , Macrófagos/imunologia , Camundongos da Linhagem 129
3.
J Gen Virol ; 93(Pt 5): 980-986, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22258859

RESUMO

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.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Peptídeos/farmacologia , Ligação Viral/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Ligação Proteica , Mapeamento de Interação de Proteínas , Ensaio de Placa Viral
4.
J Virol ; 85(6): 2907-17, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191029

RESUMO

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.


Assuntos
Aedes/fisiologia , Aedes/virologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Aedes/imunologia , Animais , Linhagem Celular , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/genética , RNA Viral/genética , RNA Viral/metabolismo , Vírus da Floresta de Semliki/genética
5.
J Virol ; 83(5): 2321-6, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109392

RESUMO

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.


Assuntos
RNA de Transferência/metabolismo , RNA Viral/metabolismo , Rhadinovirus/metabolismo , Vírion/metabolismo , Animais , Linhagem Celular , Camundongos , RNA de Transferência/isolamento & purificação , RNA Viral/isolamento & purificação , Rhadinovirus/genética , Rhadinovirus/fisiologia , Vírion/genética , Vírion/fisiologia , Montagem de Vírus
6.
J Virol ; 83(11): 5735-48, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19297476

RESUMO

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.


Assuntos
Culicidae/virologia , Interferência de RNA , Vírus da Floresta de Semliki/genética , Animais , Linhagem Celular , Cricetinae , Regulação Viral da Expressão Gênica , RNA Viral/genética , Replicação Viral
7.
Hepatol Res ; 39(2): 187-94, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19208039

RESUMO

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.

8.
J Leukoc Biol ; 84(1): 50-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18436582

RESUMO

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.


Assuntos
Gammaherpesvirinae/fisiologia , Ativação de Macrófagos/imunologia , Macrófagos/virologia , Animais , Movimento Celular , Citocinas/genética , Fibrose , Células Germinativas/virologia , Cinética , Macrófagos/patologia , Camundongos , Receptores CCR4/metabolismo , Receptores de Interferon/deficiência , Baço/patologia , Baço/virologia , Células Th2/imunologia , Transcrição Gênica , Regulação para Cima , Receptor de Interferon gama
9.
Virology ; 526: 155-164, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30390564

RESUMO

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.


Assuntos
Vírus da Influenza A/patogenicidade , Infecções por Orthomyxoviridae/fisiopatologia , Receptores de Interferon/genética , Transdução de Sinais , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Neutrófilos/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Transdução de Sinais/genética , Carga Viral , Receptor de Interferon gama
10.
J Interferon Cytokine Res ; 22(11): 1081-8, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12513907

RESUMO

Infection of mice by murine gammaherpesvirus 68 (MHV-68) represents a suitable animal model in which to investigate the immune response against gammaherpesviruses and to test the efficacy of vaccination strategies. In this study, we evaluated the efficacy of heat-inactivated MHV-68 as a vaccine as well as the adjuvant activity of type I interferon (IFN-I) administered together with the vaccine. Mice vaccinated with inactivated MHV-68 and subsequently infected with the virus exhibited a significant augmentation of the virus-specific humoral immune response and a considerable inhibition of MHV-68 acute replication in the lungs compared with unvaccinated control mice. The coadministration of IFN-I with inactivated MHV-68 significantly enhanced the humoral immune response elicited by the vaccine by stimulating the production of virus-specific IgG2 antibodies but did not significantly enhance protection from viral challenge. We conclude that IFN-I, recently shown to exhibit a powerful adjuvant activity to a poorly immunogenic subunit vaccine in mice, can also enhance the humoral immune response when used as adjuvant of an inactivated viral vaccine, even though this effect is less marked as a result of the strong immune response elicited by the inactivated virus alone, which may also involve the contribution of endogenous IFN.


Assuntos
Anticorpos Antivirais/sangue , Formação de Anticorpos , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Interferon Tipo I/uso terapêutico , Vacinas de Produtos Inativados/farmacologia , Vacinas Virais/farmacologia , Animais , Linhagem Celular , Infecções por Herpesviridae/prevenção & controle , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos , Camundongos Endogâmicos C57BL
11.
PLoS One ; 7(1): e29443, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22238612

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Influenza Humana/patologia , Macrófagos/patologia , Monócitos/patologia , Animais , Apoptose/imunologia , Apoptose/fisiologia , Caspases/metabolismo , Caspases/fisiologia , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Citocinas/fisiologia , Citofagocitose/imunologia , Citofagocitose/fisiologia , Suscetibilidade a Doenças , Cães , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/virologia , Vírus da Influenza A Subtipo H3N2/patogenicidade , Influenza Humana/sangue , Influenza Humana/imunologia , Macrófagos/classificação , Macrófagos/fisiologia , Macrófagos/virologia , Monócitos/classificação , Monócitos/fisiologia , Monócitos/virologia
12.
PLoS One ; 6(10): e25333, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22031815

RESUMO

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.


Assuntos
Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Antivirais/uso terapêutico , Infecções por Orthomyxoviridae/tratamento farmacológico , Animais , Linhagem Celular , Cães , Feminino , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Catelicidinas
13.
Vaccine ; 29(23): 3935-44, 2011 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-21481326

RESUMO

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.


Assuntos
Infecções por Herpesviridae/prevenção & controle , Interferon-alfa/metabolismo , Organismos Geneticamente Modificados/fisiologia , Rhadinovirus/patogenicidade , Infecções Tumorais por Vírus/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Animais , Linhagem Celular , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Humanos , Interferon-alfa/genética , Camundongos , Camundongos Endogâmicos C57BL , Organismos Geneticamente Modificados/genética , Organismos Geneticamente Modificados/metabolismo , Recombinação Genética , Rhadinovirus/genética , Rhadinovirus/metabolismo , Baço , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Replicação Viral
14.
J Gen Virol ; 90(Pt 9): 2061-72, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19570957

RESUMO

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.


Assuntos
Infecções por Arbovirus/virologia , Arbovírus/imunologia , Culicidae/imunologia , Culicidae/virologia , Imunidade Inata , Insetos Vetores/imunologia , Animais , Infecções por Arbovirus/transmissão , Culicidae/genética , Humanos , Insetos Vetores/genética , Insetos Vetores/virologia
15.
PLoS One ; 4(8): e6492, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19652715

RESUMO

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.


Assuntos
Gammaherpesvirinae/isolamento & purificação , Infecções por Herpesviridae/virologia , Integrases/metabolismo , Animais , Linfócitos B/virologia , Sequência de Bases , Southern Blotting , Linhagem Celular , Cricetinae , Citometria de Fluxo , Gammaherpesvirinae/genética , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Recombinação Genética
16.
J Gen Virol ; 87(Pt 4): 803-807, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16528028

RESUMO

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.


Assuntos
Gammaherpesvirinae/patogenicidade , Regulação Viral da Expressão Gênica , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral , Animais , Linhagem Celular , Cricetinae , Gammaherpesvirinae/genética , Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/virologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Baço/virologia , Replicação Viral
17.
Pathol Int ; 55(9): 558-68, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16143031

RESUMO

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.


Assuntos
Gammaherpesvirinae/genética , Genoma Viral , Infecções por Herpesviridae/virologia , Neoplasias/virologia , Imunodeficiência Combinada Severa/genética , Infecções Tumorais por Vírus/virologia , Animais , Primers do DNA/química , DNA Viral/análise , Modelos Animais de Doenças , Feminino , Gammaherpesvirinae/classificação , Gammaherpesvirinae/patogenicidade , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/mortalidade , Hibridização In Situ , Camundongos , Camundongos SCID , Neoplasias/imunologia , Neoplasias/patologia , RNA Viral/análise , Homologia de Sequência do Ácido Nucleico , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Taxa de Sobrevida , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/mortalidade
18.
J Gen Virol ; 81(Pt 11): 2635-2643, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11038374

RESUMO

Murine gammaherpesvirus-68 (MHV-68) was originally isolated from a bank vole by passage through mouse brain. Given its ability to replicate in mouse brain and its subsequent reisolation from trigeminal ganglia, it was originally considered to be an alphaherpesvirus. Molecular studies have now firmly established MHV-68 to be a gammaherpesvirus. Other gammaherpesviruses have been suggested to cause and in some cases shown to cause neurological disease. Given the isolation history of MHV-68, we have studied the ability of this virus to gain access to, to replicate in and to persist in the mouse CNS. Following intranasal inoculation the virus was not generally neuroinvasive. However, in mice with a deletion of the type-I interferon receptor gene, peripheral virus titres are higher and perivascular CNS infection was observed. There was no evidence of virus spread via olfactory routes. Direct intracerebral inoculation of virus was fatal with widespread infection and destruction predominantly of meningeal and ependymal cells. Hippocampal pyramidal neurons, oligodendrocytes, Bergmann glia cells in the cerebellar cortex and neural progenitor cells in the rostral migratory stream were also infected. A similar infection was observed in younger mice. CNS infection following virus reactivation was investigated by implantation of infected glial cells. Implantation into a brain ventricle led to widespread fatal infection, principally involving ependymal and meningeal cells. Implantation into the striatum resulted in a predominantly neuronal infection. Implantation of cells into mice transiently treated with the antiviral thionucleoside analogue 2'-deoxy-5-ethyl-beta-4'-thiouridine resulted in survival with detection of virus-infected cells in the brain 1 year later.


Assuntos
Sistema Nervoso Central/virologia , Gammaherpesvirinae/isolamento & purificação , Infecções por Herpesviridae/virologia , Animais , Células Cultivadas , Gammaherpesvirinae/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Neuroglia/transplante , Neuroglia/virologia , Ativação Viral , Replicação Viral
19.
Vaccine ; 22(11-12): 1433-40, 2004 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15063566

RESUMO

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.


Assuntos
Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Herpesviridae/imunologia , Vacinas contra Herpesvirus/imunologia , Interferon Tipo I/fisiologia , Receptores de Interferon/fisiologia , Latência Viral/imunologia , Replicação Viral/fisiologia , Animais , Linfócitos B/imunologia , Linfócitos B/fisiologia , Linhagem Celular , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Testes de Neutralização , Receptores de Interferon/genética , Vacinação , Vacinas de Produtos Inativados/imunologia
20.
J Gen Virol ; 82(Pt 5): 1187-1197, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11297694

RESUMO

Murine gammaherpesvirus (MHV-68) is well established as a small animal model for the study of gammaherpesviruses. The MHV-68 genome contains an open reading frame (ORF74) that has significant sequence homology with mammalian G-protein coupled receptors (GPCRs) and the GPCR from the related Kaposi's sarcoma-associated herpesvirus (KSHV). Here we show that the MHV-68 ORF74 is predicted to encode a GPCR since it has seven potential transmembrane helices and that it has other sequence motifs in common with GPCRS: Of interest is the observation that the sequence around a conserved arginine at the start of the second intracellular loop suggests that the ORF74 product may signal constitutively (agonist independent). Given that the ORF74 product is predicted to encode a GPCR we named it MHV-GPCR. In studies on the transcription of the MHV-GPCR, we determined that it was encoded on multiple early transcripts of 3.4, 4.4, 6.6 and 8.7 kb in size. At least one of these transcripts was bicistronic, containing the ORF encoding the Bcl-2 homologue also. In vivo, we found that MHV GPCR was expressed during acute infection but also during persistence, particularly in the lungs of infected mice. Immunofluorescence studies indicated that the MHV GPCR protein was expressed on the surface of cells in patches. Finally, like the KSHV GPCR, expression of the MHV GPCR resulted in transformation of NIH 3T3 cells. We surmise, therefore, that the MHV GPCR may act in concert with genes with which it is expressed such as vBcl-2 to enhance the growth and survival of MHV-68-infected cells.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Gammaherpesvirinae , Proteínas Oncogênicas/fisiologia , Fases de Leitura Aberta , Receptores de Superfície Celular/fisiologia , Proteínas Virais/fisiologia , Células 3T3 , Sequência de Aminoácidos , Animais , Linhagem Celular , Transformação Celular Viral , Cricetinae , Feminino , Gammaherpesvirinae/genética , Expressão Gênica , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas Oncogênicas/genética , Receptores de Superfície Celular/genética , Transcrição Gênica , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa