Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 18598, 2024 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-39127765

RESUMO

Feline mesenchymal stem cells (fMSCs) are well known for their robust differentiation capabilities and are commonly used in studying immune-related diseases in cats. Despite their importance, the susceptibility of fMSCs to viral infections remains uncertain. This study aimed to assess the susceptibility of feline adipose-derived mesenchymal stem cells (fAD-MSCs) and feline umbilical cord-derived mesenchymal stem cells (fUC-MSCs) to common feline viruses, including feline coronavirus (FCoV), feline herpesvirus type 1 (FHV-1), and feline panleukopenia virus (FPV). The results demonstrated that both FCoV and FHV-1 were able to infect both types of cells, while FPV did not exhibit cytopathic effects on fUC-MSCs. Furthermore, all three viruses were successfully isolated from fAD-MSCs. These findings suggest that certain feline viruses can replicate in fMSCs, indicating potential limitations in using fMSCs for treating viral diseases caused by these specific viruses. This study has important clinical implications for veterinarians, particularly in the management of viral diseases.


Assuntos
Coronavirus Felino , Células-Tronco Mesenquimais , Animais , Gatos , Células-Tronco Mesenquimais/virologia , Células-Tronco Mesenquimais/citologia , Coronavirus Felino/fisiologia , Vírus da Panleucopenia Felina , Células Cultivadas , Varicellovirus/fisiologia , Replicação Viral , Diferenciação Celular , Tecido Adiposo/citologia , Doenças do Gato/virologia
2.
Front Immunol ; 15: 1408212, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38887303

RESUMO

Introduction: Varicella zoster virus (VZV) causes varicella and can reactivate as herpes zoster, and both diseases present a significant burden worldwide. However, the mechanisms by which VZV establishes latency in the sensory ganglia and disseminates to these sites remain unclear. Methods: We combined a single-cell sequencing approach and a well-established rhesus macaque experimental model using Simian varicella virus (SVV), which recapitulates the VZV infection in humans, to define the acute immune response to SVV in the lung as well as compare the transcriptome of infected and bystander lung-resident T cells and macrophages. Results and discussion: Our analysis showed a decrease in the frequency of alveolar macrophages concomitant with an increase in that of infiltrating macrophages expressing antiviral genes as well as proliferating T cells, effector CD8 T cells, and T cells expressing granzyme A (GZMA) shortly after infection. Moreover, infected T cells harbored higher numbers of viral transcripts compared to infected macrophages. Furthermore, genes associated with cellular metabolism (glycolysis and oxidative phosphorylation) showed differential expression in infected cells, suggesting adaptations to support viral replication. Overall, these data suggest that SVV infection remodels the transcriptome of bystander and infected lung-resident T cells and macrophages.


Assuntos
Pulmão , Macaca mulatta , Animais , Pulmão/imunologia , Pulmão/virologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/virologia , Transcriptoma , Linfócitos T/imunologia , Varicellovirus/fisiologia , Varicellovirus/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/fisiologia , Modelos Animais de Doenças , Análise de Célula Única
3.
Viruses ; 13(12)2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34960727

RESUMO

Malignant mesothelioma (MM) is an aggressive asbestos-related cancer, against which no curative modalities exist. Oncolytic virotherapy is a promising therapeutic approach, for which MM is an ideal candidate; indeed, the pleural location provides direct access for the intra-tumoral injection of oncolytic viruses (OVs). Some non-human OVs offer advantages over human OVs, including the non-pathogenicity in humans and the absence of pre-existing immunity. We previously showed that caprine herpesvirus 1 (CpHV-1), a non-pathogenic virus for humans, can kill different human cancer cell lines. Here, we assessed CpHV-1 effects on MM (NCI-H28, MSTO, NCI-H2052) and non-tumor mesothelial (MET-5A) cells. We found that CpHV-1 reduced cell viability and clonogenic potential in all MM cell lines without affecting non-tumor cells, in which, indeed, we did not detect intracellular viral DNA after treatment. In particular, CpHV-1 induced MM cell apoptosis and accumulation in G0/G1 or S cell cycle phases. Moreover, CpHV-1 strongly synergized with cisplatin, the drug currently used in MM chemotherapy, and this agent combination did not affect normal mesothelial cells. Although further studies are required to elucidate the mechanisms underlying the selective CpHV-1 action on MM cells, our data suggest that the CpHV-1-cisplatin combination could be a feasible strategy against MM.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cisplatino/farmacologia , Mesotelioma Maligno/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Varicellovirus/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Humanos , Mesotelioma Maligno/tratamento farmacológico , Mesotelioma Maligno/fisiopatologia , Mesotelioma Maligno/virologia , Vírus Oncolíticos/genética , Varicellovirus/genética
4.
Viruses ; 13(7)2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34372499

RESUMO

Caprine herpesvirus 1 (CpHV-1) is a member of the alpha subfamily of herpesviruses, which is responsible for genital lesions and latent infections in goat populations worldwide. In this study, for the first time, the transcriptome and proteomics of CpHV-1 infected Madin Darby bovine kidney (MDBK) cells were explored using RNA-Sequencing (RNA-Seq) and isobaric tags for relative and absolute quantitation-liquid chromatography tandem mass spectrometry (iTRAQ-LC-MS/MS) technology, respectively. RNA-Seq analysis revealed 81 up-regulated and 19 down-regulated differentially expressed genes (DEGs) between infected and mock-infected MDBK cells. Bioinformatics analysis revealed that most of these DEGs were mainly involved in the innate immune response, especially the interferon stimulated genes (ISGs). Gene Ontology (GO) enrichment analysis results indicated that the identified DEGs were significantly mainly enriched for response to virus, defense response to virus, response to biotic stimulus and regulation of innate immune response. Viral carcinogenesis, the RIG-I-like receptor signaling pathway, the cytosolic DNA-sensing pathway and pathways associated with several viral infections were found to be significantly enriched in the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. Eleven selected DEGs (Mx1, RSAD2, IFIT1, IFIT2, IFIT5, IFIH1, IFITM3, IRF7, IRF9, OAS1X and OAS1Y) associated with immune responses were selected, and they exhibited a concordant direction both in RNA-Seq and quantitative real-time RT-PCR analysis. Proteomic analysis also showed significant up-regulation of innate immunity-related proteins. GO analysis showed that the differentially expressed proteins were mostly enriched in defense response and response to virus, and the pathways associated with viral infection were enriched under KEGG analysis. Protein-protein interaction network analysis indicated most of the DEGs related to innate immune responses, as DDX58(RIG-I), IFIH1(MDA5), IRF7, Mx1, RSAD2, OAS1 and IFIT1, were located in the core of the network and highly connected with other DGEs. Our findings support the notion that CpHV-1 infection induced the transcription and protein expression alterations of a series of genes related to host innate immune response, which helps to elucidate the resistance of host cells to viral infection and to clarify the pathogenesis of CpHV-1.


Assuntos
Perfilação da Expressão Gênica , Hepatócitos/virologia , Imunidade Inata/genética , Proteômica , Regulação para Cima , Varicellovirus/genética , Animais , Bovinos , Linhagem Celular , Cromatografia Líquida , Biologia Computacional , Imunidade Inata/imunologia , Rim/citologia , Rim/virologia , Análise de Sequência de RNA , Espectrometria de Massas em Tandem , Transcriptoma , Varicellovirus/fisiologia , Replicação Viral/genética
5.
Viruses ; 13(2)2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33499363

RESUMO

Felid herpesvirus-1 (FeHV-1) is an important respiratory and ocular pathogen of cats and current vaccines are limited in duration and efficacy because they do not prevent infection, viral nasal shedding and latency. To address these shortcomings, we have constructed FeHV-1 gE-TK- and FeHV-1 PK- deletion mutants (gE-TK- and PK-) using bacterial artificial chromosome (BAC) mutagenesis and shown safety and immunogenicity in vitro. Here, we compare the safety and efficacy of a prime boost FeHV-1 gE-TK- and FeHV-1 PK- vaccination regimen with commercial vaccination in cats. Cats in the vaccination groups were vaccinated at 3-week intervals and all cats were challenge infected 3 weeks after the last vaccination. Evaluations included clinical signs, nasal shedding, virus neutralizing antibodies (VN), cytokine mRNA gene expression, post-mortem histology and detection of latency establishment. Vaccination with gE-TK- and PK- mutants was safe and resulted in significantly reduced clinical disease scores, pathological changes, viral nasal shedding, and viral DNA in the trigeminal ganglia (the site of latency) following infection. Both mutants induced VN antibodies and interferons after immunization. In addition, after challenge infection, we observed a reduction of IL-1ß expression, and modulation of TNFα, TGFß and IL10 expression. In conclusion, this study shows the merits of using FeHV-1 deletion mutants for prevention of FeHV-1 infection in cats.


Assuntos
Doenças do Gato/prevenção & controle , Infecções por Herpesviridae/veterinária , Imunidade Inata , Varicellovirus/genética , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Doenças do Gato/virologia , Gatos , Linhagem Celular , Citocinas/genética , Citocinas/imunologia , Deleção de Genes , Infecções por Herpesviridae/prevenção & controle , Imunização Secundária/veterinária , Masculino , Varicellovirus/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/genética , Virulência/genética , Replicação Viral , Eliminação de Partículas Virais
6.
J Feline Med Surg ; 22(6): 492-499, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31246133

RESUMO

OBJECTIVES: The aim of this study was to assess the effects of famciclovir administration in cats with spontaneously acquired acute upper respiratory tract disease. METHODS: Twenty-four kittens with clinical signs of acute upper respiratory tract disease were randomly allocated to receive doxycycline (5 mg/kg PO q12h) alone (group D; n = 12) or with famciclovir (90 mg/kg PO q12h; group DF; n = 12) for up to 3 weeks. Clinical disease severity was scored at study entry and daily thereafter. Oculo-oropharyngeal swabs collected at study entry and exit were assessed using quantitative PCR for nucleic acids of feline herpesvirus type 1 (FHV-1), feline calicivirus (FCV), Chlamydia felis, Bordetella bronchiseptica and Mycoplasma felis. RESULTS: The median (range) age of cats was 1.5 (1-6) months in group D vs 1.6 (1-5) months in group DF (P = 0.54). Pathogens detected in oculo-oropharyngeal swabs at study entry included FCV (n = 13/24; 54%), M felis (n = 8/24; 33%), FHV-1 (n = 7/24; 29%), C felis (n = 7/24; 29%) and B bronchiseptica (n = 3/24; 12%). Median (range) duration of clinical signs was 11.5 (3-21) days in group DF and 11 (3-21) days in group D (P = 0.75). Median (range) total disease score at the end of the study did not differ between groups (group D 1 [1-1] vs group DF 1 [1-3]; P = 0.08). CONCLUSIONS AND RELEVANCE: This study revealed no significant difference in response to therapy between cats treated with doxycycline alone or with famciclovir; cats improved rapidly in both groups. However, identification of FHV-1 DNA was relatively uncommon in this study and clinical trials focused on FHV-1-infected cats are warranted to better evaluate famciclovir efficacy.


Assuntos
Antivirais/administração & dosagem , Doenças do Gato/tratamento farmacológico , Famciclovir/administração & dosagem , Infecções Respiratórias/veterinária , Animais , Infecções por Bordetella/tratamento farmacológico , Infecções por Bordetella/microbiologia , Infecções por Bordetella/veterinária , Bordetella bronchiseptica/isolamento & purificação , Bordetella bronchiseptica/fisiologia , Infecções por Caliciviridae/tratamento farmacológico , Infecções por Caliciviridae/veterinária , Infecções por Caliciviridae/virologia , Calicivirus Felino/isolamento & purificação , Calicivirus Felino/fisiologia , Doenças do Gato/microbiologia , Doenças do Gato/virologia , Gatos , Chlamydia/isolamento & purificação , Chlamydia/fisiologia , Infecções por Chlamydia/tratamento farmacológico , Infecções por Chlamydia/microbiologia , Infecções por Chlamydia/veterinária , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Mycoplasma/isolamento & purificação , Mycoplasma/fisiologia , Infecções por Mycoplasma/tratamento farmacológico , Infecções por Mycoplasma/microbiologia , Infecções por Mycoplasma/veterinária , Ácidos Nucleicos/análise , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/microbiologia , Infecções Respiratórias/virologia , Varicellovirus/isolamento & purificação , Varicellovirus/fisiologia
7.
Virus Res ; 264: 56-67, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30796929

RESUMO

Feline herpesvirus-1 (FHV-1) infection occurs worldwide and is a leading cause of respiratory and ocular diseases in cats. Current vaccines reduce the severity of symptoms but do not prevent infection and, therefore, do not provide defense against an establishment of latency and reactivation. We hypothesize that immunomodulation of FHV-1 is the cause of lack in protection and that deletion of virulence/immune modulatory genes of FHV-1 will enhance safety and immunogenicity. Our objective was to use feline respiratory epithelial cell (FREC) cultures to define in vitro growth characteristics and immunomodulation resulting from infection of FRECs with the virulent FHV-1 strain C27 (WT) and glycoprotein C-deletion (gC-), glycoprotein E-deletion (gE-), serine/threonine protein kinase-deletion (PK-), as well as gE and thymidine kinase-double-deletion (gE-TK-) mutants generated by bacterial artificial chromosome mutagenesis. Differentiated FRECs were mock inoculated or inoculated with WT, gC-, gE-, PK-, or gE-TK- mutants. Virus titration and real-time quantitative PCR assays were performed on samples collected at 1 hpi followed by 24 h intervals between 24 and 96 hpi to determine growth kinetics. Real-time PCR was used to quantitate IFNα, TNFα, IL-1ß, IL-10, and TGFß-specific mRNA levels. Immunoassays were performed to measure the protein levels of subsets of cytokines/chemokines secreted by FRECs. Inoculation of FRECs with gE-TK- resulted in significantly lower end-point titers than inoculation with WT or gE-. Both PK- and gC- inoculated FRECs also produced significantly lower end-point titers at 96 hpi than WT. Overall, intracellular virus titers were higher than those of extracellular virus. PCR results for viral DNA paralleled the virus titration results. Further, in contrast to WT inoculation, an increase in IFNα and IL-10 mRNA expression was not observed following inoculation with gE-TK- and PK-, but inoculation with gE-TK- and PK- did result in increased TGFß expression in FRECs compared to responses following infection with WT. Moreover, gE-TK- and PK- blocked the inhibition of IL-8 and neutrophil chemoattractant (KC), which was observed following inoculation with WT. In summary, the results obtained in FRECs may be used to predict the safety and immunogenicity characteristics of these mutants in vivo. Our study highlights the value of the FREC system for studying replication kinetics/immune modulation factors of FHV-1 and screening prospective vaccine candidates before their use in experimental cats.


Assuntos
Células Epiteliais/imunologia , Imunidade Inata , Varicellovirus/fisiologia , Replicação Viral , Animais , Gatos , Linhagem Celular , Citocinas/genética , Citocinas/imunologia , Células Epiteliais/virologia , Deleção de Genes , Glicoproteínas de Membrana/genética , Mutação , Reação em Cadeia da Polimerase , Estudos Prospectivos , Timidina Quinase/genética , Timidina Quinase/imunologia , Varicellovirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Virulência/genética
8.
J Gen Virol ; 99(8): 1115-1128, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29916804

RESUMO

Anti-microbial compounds typically exert their action by directly interfering with one or more stages of the pathogen's life cycle. However, some compounds also have secondary effects on the host that aid in pathogen clearance. Raltegravir is a human immunodeficiency virus (HIV)-integrase inhibitor that has been shown to alter the host immune response to HIV in addition to its direct antiviral effect. Interestingly, raltegravir can also directly inhibit the replication of various herpesviruses. However, the host-targeted effects of this drug in the context of a herpesvirus infection have not been explored. Here, we used felid alphaherpesvirus 1 (FHV-1), a close relative of human alphaherpesvirus 1 (HHV-1) that similarly causes ocular herpes, to characterize the host-targeted effects of raltegravir on corneal epithelial cells during an alphaherpesvirus infection. Using RNA deep sequencing, we found that raltegravir specifically boosts the expression of anti-angiogenic factors and promotes metabolic homeostasis in FHV-1-infected cells. In contrast, few changes in host gene transcription were found in uninfected cells. Importantly, we were able to demonstrate that these effects were specific to raltegravir and independent of the direct-acting antiviral effect of the drug, since treatment with the DNA polymerase inhibitor phosphonoacetic acid did not induce these host-targeted effects. Taken together, these results indicate that raltegravir has profound and specific effects on the host transcription profile of herpesvirus-infected cells that may contribute to the overall antiviral activity of the drug and could provide therapeutic benefits in vivo. Furthermore, this study provides a framework for future efforts evaluating the host-targeted effects of anti-microbial compounds.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Inibidores de Integrase de HIV/farmacologia , Raltegravir Potássico/farmacologia , Transcriptoma/efeitos dos fármacos , Varicellovirus/efeitos dos fármacos , Animais , Gatos , Células Cultivadas , Epitélio Corneano/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Amplificação de Ácido Nucleico , Reprodutibilidade dos Testes , Replicação de Sequência Autossustentável , Organismos Livres de Patógenos Específicos , Varicellovirus/fisiologia
9.
Viruses ; 10(4)2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29597335

RESUMO

The pathogenesis of enteric zoster, a rare debilitating complication of reactivation of latent varicella-zoster virus (VZV) in the enteric nervous system (ENS), is largely unknown. Infection of monkeys with the closely related Varicellovirus simian varicella virus (SVV) mimics VZV disease in humans. In this study, we determined the applicability of the SVV nonhuman primate model to study Varicellovirus infection of the ENS. We confirmed VZV infection of the gut in latently infected adults and demonstrated that SVV DNA was similarly present in gut of monkeys latently infected with SVV using quantitative real-time PCR. In situ analyses showed that enteric neurons expressed SVV open reading frame (ORF) 63 RNA, but not viral nucleocapsid proteins, suggestive of latent ENS infection. During primary infection, SVV-infected T-cells were detected in gut-draining mesenteric lymph nodes and located in close vicinity to enteric nerves in the gut. Furthermore, flow cytometric analysis of blood from acutely SVV-infected monkeys demonstrated that virus-infected T-cells expressed the gut-homing receptor α4ß7 integrin. Collectively, the data demonstrate that SVV infects ENS neurons during primary infection and supports the role of T-cells in virus dissemination to the gut. Because SVV reactivation can be experimentally induced, the SVV nonhuman primate model holds great potential to study the pathogenesis of enteric zoster.


Assuntos
Expressão Gênica , Integrinas/genética , Neurônios/metabolismo , Neurônios/virologia , Linfócitos T/fisiologia , Linfócitos T/virologia , Varicellovirus/fisiologia , Adulto , Idoso , Animais , Biomarcadores , Biópsia , Sistema Nervoso Entérico/virologia , Feminino , Imunofluorescência , Infecções por Herpesviridae/veterinária , Herpesvirus Humano 3/fisiologia , Humanos , Integrinas/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Linfonodos/virologia , Macaca mulatta , Masculino , Pessoa de Meia-Idade , Doenças dos Macacos/genética , Doenças dos Macacos/imunologia , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Nódulos Linfáticos Agregados/virologia , Infecção pelo Vírus da Varicela-Zoster/genética , Infecção pelo Vírus da Varicela-Zoster/imunologia , Infecção pelo Vírus da Varicela-Zoster/patologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Carga Viral
10.
Virus Res ; 214: 39-48, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26795546

RESUMO

Infection with feline herpesvirus-1 (FHV-1) accounts for 50% of viral upper respiratory diseases in domestic cats and is a significant cause of ocular diseases. Despite the clinical significance and high prevalence of FHV-1 infection, currently available vaccines cannot completely protect cats from infection and lifelong latency. FHV-1 infects via the mucous membranes and replicates in respiratory epithelial cells, but very little is known about the early innate immunity at this site. To address questions about immunity to FHV-1, feline respiratory epithelial cells cultured at air-liquid interface (ALI-FRECs) were established by collecting respiratory tracts from 6 healthy cats after euthanasia. Cells were isolated, cultured and characterized histologically and immunologically before infection with FHV-1. The expression of Toll-like receptors (TLRs), cytokine and chemokine responses were measured by real time PCR. ALI-FRECs morphologically resembled the natural airways of cats with multilayered columnar epithelial cells and cilia. Immunological properties of the natural airways were maintained in ALI-FRECs, as evidenced by the expression of TLRs, cytokines, chemokines, interferons, beta-defensins, and other regulatory genes. Furthermore, ALI-FRECs were able to support infection and replication of FHV-1, as well as modulate transcriptional regulation of various immune genes in response to infection. IL-1ß and TNFα were increased in ALI-FRECs by 24hpi, whereas expression levels of IFN-α and TLR9 were not increased until 36hpi. In contrast, TLR3, GM-CSF and TGF-1ß expression was down-regulated at 36hpi. The data presented show the development of a system ideal for investigating the molecular pathogenesis and immunity of FHV-1 or other respiratory pathogens.


Assuntos
Doenças do Gato/imunologia , Doenças do Gato/virologia , Imunidade Inata , Varicellovirus/fisiologia , Animais , Doenças do Gato/metabolismo , Gatos , Linhagem Celular , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Epiteliais , Expressão Gênica , Imunidade Inata/genética , Mediadores da Inflamação/metabolismo , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
11.
J Virol ; 89(19): 9817-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26178993

RESUMO

UNLABELLED: Like varicella-zoster virus (VZV), simian varicella virus (SVV) reactivates to produce zoster. In the present study, 5 rhesus macaques were inoculated intrabronchially with SVV, and 5 months later, 4 monkeys were immunosuppressed; 1 monkey was not immunosuppressed but was subjected to the stress of transportation. In 4 monkeys, a zoster rash developed 7 to 12 weeks after immunosuppression, and a rash also developed in the monkey that was not immunosuppressed. Analysis at 24 to 48 h after zoster revealed SVV antigen in the lung alveolar wall, in ganglionic neurons and nonneuronal cells, and in skin and in lymph nodes. In skin, SVV was found primarily in sweat glands. In lymph nodes, the SVV antigen colocalized mostly with macrophages, dendritic cells, and, to a lesser extent, T cells. The presence of SVV in lymph nodes, as verified by quantitative PCR detection of SVV DNA, might reflect the sequestration of virus by macrophages and dendritic cells in lymph nodes or the presentation of viral antigens to T cells to initiate an immune response against SVV, or both. IMPORTANCE: VZV causes varicella (chickenpox), becomes latent in ganglia, and reactivates to produce zoster and multiple other serious neurological disorders. SVV in nonhuman primates has proved to be a useful model in which the pathogenesis of the virus parallels the pathogenesis of VZV in humans. Here, we show that SVV antigens are present in sweat glands in skin and in macrophages and dendritic cells in lymph nodes after SVV reactivation in monkeys, raising the possibility that macrophages and dendritic cells in lymph nodes serve as antigen-presenting cells to activate T cell responses against SVV after reactivation.


Assuntos
Herpes Zoster/patologia , Herpes Zoster/virologia , Linfonodos/virologia , Varicellovirus/fisiologia , Ativação Viral/fisiologia , Animais , Chlorocebus aethiops , DNA Viral/análise , Células Dendríticas/virologia , Imunofluorescência , Imuno-Histoquímica , Terapia de Imunossupressão , Linfonodos/citologia , Macaca mulatta , Macrófagos/virologia , Reação em Cadeia da Polimerase em Tempo Real , Pele/patologia , Pele/virologia , Linfócitos T/virologia , Células Vero
12.
Prev Vet Med ; 116(1-2): 56-62, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24985155

RESUMO

Water buffalo (Bubalus bubalis) are susceptible to bovine herpesvirus type 1 (BoHV-1) and a species-specific herpesvirus, bubaline herpesvirus type 1 (BuHV-1). In this study, an attenuated marker BoHV-1 based vaccine against BuHV-1 challenge was evaluated to determine whether it induces protection from viral replication. One group of water buffalo calves was immunized with an attenuated BoHV-1 marker vaccine. A second group was not vaccinated and used as the control. During the post-vaccination period, we monitored the humoral immune response. The efficacy of the vaccine was tested after intranasal challenge of the calves with a BuHV-1 strain. The experiment showed that after vaccination, BuHV-1 replication was significantly reduced by approximately three titer points compared to the controls. The control animals showed high levels of viral shedding and mild signs associated with BuHV-1 infection. Therefore, our study provides evidence for the existence of cross-protection between BoHV-1 and BuHV-1 in buffalo calves.


Assuntos
Búfalos , Proteção Cruzada , Infecções por Herpesviridae/veterinária , Vacinas contra Herpesvirus/imunologia , Varicellovirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Feminino , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/imunologia , Herpesvirus Bovino 1/fisiologia , Vacinas contra Herpesvirus/uso terapêutico , Masculino , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Vacinas Marcadoras/imunologia , Vacinas Marcadoras/uso terapêutico , Varicellovirus/fisiologia , Replicação Viral
13.
J Virol ; 82(24): 12441-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18829759

RESUMO

Porcine endogenous retrovirus (PERV), porcine cytomegalovirus (PCMV), and porcine lymphotropic herpesvirus (PLHV) are common porcine viruses that may be activated with immunosuppression for xenotransplantation. Studies of viral replication or transmission are possible due to prolonged survival of xenografts in baboon recipients from human decay-accelerating factor transgenic or alpha-1,3-galactosyltransferase gene knockout miniature swine. Ten baboons underwent xenotransplantation with transgenic pig organs. Graft survival was 32 to 179 days. Recipient serial samples of peripheral blood mononuclear cells (PBMC) and plasma were analyzed for PCMV, PERV, and PLHV-1 nucleic acids and viral replication using quantitative PCR assays. The PBMC contained PERV proviral DNA in 10 animals, PLHV-1 DNA in 6, and PCMV in 2. PERV RNA was not detected in any PBMC or serum samples. Plasma PLHV-1 DNA was detected in one animal. Pig cell microchimerism (pig major histocompatibility complex class I and pig mitochondrial cytochrome c oxidase subunit II sequences) was present in all recipients with detectable PERV or PLHV-1 (85.5%). Productive infection of PERV or PLHV-1 could not be demonstrated. The PLHV-1 viral load did not increase in serum over time, despite prolonged graft survival and pig cell microchimerism. There was no association of viral loads with the nature of exogenous immune suppression. In conclusion, PERV provirus and PLHV-1 DNA were detected in baboons following porcine xenotransplantation. Viral detection appeared to be due to persistent pig cell microchimerism. There was no evidence of productive infection in recipient baboons for up to 6 months of xenograft function.


Assuntos
Quimerismo , Linfócitos , Retroviridae/fisiologia , Transplante Heterólogo , Varicellovirus/fisiologia , Replicação Viral , Animais , Animais Geneticamente Modificados , Papio , Sus scrofa
14.
PLoS Pathog ; 4(5): e1000080, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18516302

RESUMO

Cytotoxic T-lymphocytes play an important role in the protection against viral infections, which they detect through the recognition of virus-derived peptides, presented in the context of MHC class I molecules at the surface of the infected cell. The transporter associated with antigen processing (TAP) plays an essential role in MHC class I-restricted antigen presentation, as TAP imports peptides into the ER, where peptide loading of MHC class I molecules takes place. In this study, the UL 49.5 proteins of the varicelloviruses bovine herpesvirus 1 (BHV-1), pseudorabies virus (PRV), and equine herpesvirus 1 and 4 (EHV-1 and EHV-4) are characterized as members of a novel class of viral immune evasion proteins. These UL 49.5 proteins interfere with MHC class I antigen presentation by blocking the supply of antigenic peptides through inhibition of TAP. BHV-1, PRV, and EHV-1 recombinant viruses lacking UL 49.5 no longer interfere with peptide transport. Combined with the observation that the individually expressed UL 49.5 proteins block TAP as well, these data indicate that UL 49.5 is the viral factor that is both necessary and sufficient to abolish TAP function during productive infection by these viruses. The mechanisms through which the UL 49.5 proteins of BHV-1, PRV, EHV-1, and EHV-4 block TAP exhibit surprising diversity. BHV-1 UL 49.5 targets TAP for proteasomal degradation, whereas EHV-1 and EHV-4 UL 49.5 interfere with the binding of ATP to TAP. In contrast, TAP stability and ATP recruitment are not affected by PRV UL 49.5, although it has the capacity to arrest the peptide transporter in a translocation-incompetent state, a property shared with the BHV-1 and EHV-1 UL 49.5. Taken together, these results classify the UL 49.5 gene products of BHV-1, PRV, EHV-1, and EHV-4 as members of a novel family of viral immune evasion proteins, inhibiting TAP through a variety of mechanisms.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Herpesvirus Bovino 1/imunologia , Herpesvirus Equídeo 1/imunologia , Herpesvirus Suídeo 1/imunologia , Varicellovirus/fisiologia , Proteínas do Envelope Viral/imunologia , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apresentação de Antígeno , Bovinos , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Cães , Herpesvirus Bovino 1/genética , Herpesvirus Equídeo 1/genética , Herpesvirus Suídeo 1/genética , Cavalos , Humanos , Transporte Proteico , Recombinação Genética , Suínos , Transdução Genética , Varicellovirus/patogenicidade , Proteínas do Envelope Viral/genética
15.
Virology ; 368(1): 50-9, 2007 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17651776

RESUMO

SVV infection of primates closely resembles VZV infection of humans. Like VZV, SVV becomes latent in ganglionic neurons. We used this model to study the effect of immunosuppression on varicella reactivation. Cynomolgus monkeys latently infected with SVV were irradiated and treated with tacrolimus and prednisone. Of four latently infected monkeys that were immunosuppressed and subjected to the stress of transportation and isolation, one developed zoster, and three others developed features of subclinical reactivation. Another non-immunosuppressed latently infected monkey that was subjected to the same stress of travel and isolation showed features of subclinical reactivation. Virus reactivation was confirmed not only by the occurrence of zoster in one monkey, but also by the presence of late SVV RNA in ganglia, and the detection of SVV DNA in non-ganglionic tissue, and SVV antigens in skin, ganglia and lung.


Assuntos
Varicela/imunologia , Varicela/virologia , Varicellovirus/imunologia , Varicellovirus/fisiologia , Ativação Viral , Animais , Varicela/patologia , DNA Viral/análise , Gânglios/patologia , Gânglios/virologia , Terapia de Imunossupressão , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Pulmão/patologia , Pulmão/virologia , Macaca fascicularis , Prednisona/administração & dosagem , Prednisona/farmacologia , RNA Viral/análise , Pele/patologia , Pele/virologia , Estresse Psicológico , Tacrolimo/administração & dosagem , Tacrolimo/farmacologia , Raios X
16.
Arch Virol ; 152(3): 553-63, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17115302

RESUMO

Simian varicella virus (SVV) is closely related to varicella-zoster virus (VZV), the causative agent of chickenpox and shingles. The SVV and VZV gene 61 polypeptides are homologs of the HSV-1 ICP0, a viral transactivator which appears to play a role in viral latency and reactivation. In this study, the molecular properties of the SVV 61 were characterized. The SVV open reading frame (ORF) 61 encodes a 54.1-kDa polypeptide with 37% amino acid identity to the VZV 61. Homology to the HSV-1 ICP-0 is limited to a conserved RING finger motif at the amino terminus of the protein. A nuclear localization sequence (nls) at the carboxy-terminus directs the SVV 61 to the cell nucleus, while a SVV 61nls(-) mutant is confined to the cell cytoplasm. The SVV 61 transactivates its own promoter as well as SVV immediate early (IE, ORF 62), early (ORFs 28 and 29), and late (ORF 68) gene promoters in transfected Vero cells. The RING finger and nls motifs are required for efficient SVV 61 transactivation. The SVV 61 has no effect on the ability of the major SVV transactivator (IE62) to induce SVV promoters. Generation and propagation of a SVV gene 61 deletion mutant demonstrated that the SVV 61 is non-essential for in vitro replication. SVV gene 61 is expressed in liver, lung, and neural ganglia of infected monkeys during acute simian varicella.


Assuntos
Genes Virais , Transativadores/genética , Varicellovirus/genética , Replicação Viral/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Chlorocebus aethiops , DNA Viral/genética , DNA Viral/isolamento & purificação , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/virologia , Herpesvirus Humano 3/genética , Humanos , Rim , Dados de Sequência Molecular , Fases de Leitura Aberta , Doenças dos Primatas/virologia , Primatas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Varicellovirus/fisiologia , Células Vero
17.
Virology ; 345(1): 244-50, 2006 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-16242745

RESUMO

Simian varicella virus (SVV) infection in primates closely resembles varicella-zoster virus (VZV) infection in humans. SVV ORF 63 has 51.6% homology at the amino acid level to VZV ORF 63. We cloned SVV ORFs 63 and 62, transcribed and translated in vitro, and immunoprecipitated the expected proteins with rabbit polyclonal antibodies. Immunoprecipitation analysis revealed that SVV ORF 63 is expressed as a 43-kDa phosphorylated protein in virus-infected cells. In both neuronal and non-neuronal cells, SVV ORF 62 protein alone upregulated the SVV 21 promoter, while SVV ORF 63 protein alone did not have any effect. SVV ORF 62-mediated transactivation of the SVV ORF 21 promoter was upregulated in neuronal cells, but downregulated in non-neuronal cells, by SVV ORF 63 protein. This is the first study in which a varicella protein (ORF 63) expressed during latency has been shown to have a differential effect on a promoter that is also active during latency, in neuronal as compared to non-neuronal cells.


Assuntos
Regulação Viral da Expressão Gênica , Genes Virais , Neurônios/virologia , Regiões Promotoras Genéticas , Ativação Transcricional , Varicellovirus/genética , Sequência de Aminoácidos , Animais , Fusão Gênica Artificial , Linhagem Celular , Chlorocebus aethiops , Regulação para Baixo , Humanos , Proteínas Imediatamente Precoces/genética , Luciferases/análise , Luciferases/genética , Dados de Sequência Molecular , Fases de Leitura Aberta , Biossíntese de Proteínas , Homologia de Sequência de Aminoácidos , Regulação para Cima , Varicellovirus/fisiologia , Células Vero , Proteínas do Envelope Viral/genética
18.
J Gen Virol ; 86(Pt 12): 3209-3214, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16298965

RESUMO

Recently, glycoprotein G (gG) of several alphaherpesviruses infecting large herbivores was shown to belong to a new family of chemokine-binding proteins (vCKBPs). In the present study, the function of Felid herpesvirus 1 (FeHV-1) gG as a vCKBP was investigated and the following conclusions were reached: (i) FeHV-1 secreted gG is a high-affinity broad-spectrum vCKBP that binds CC, CXC and C chemokines; (ii) gG is the only vCKBP expressed by FeHV-1 that binds CCL3 and CXCL1; (iii) secreted gG blocks chemokine activity by preventing their interaction with high-affinity cellular receptors; (iv) the membrane-anchored form of gG expressed on the surface of infected cells is also able to bind chemokines; and (v) the vCKBP activity is conserved among different field isolates of FeHV-1. Altogether, these data demonstrate that FeHV-1 gG is a new member of the vCKBP-4 family. Moreover, this study is the first to demonstrate that gG expressed at the surface of FeHV-1-infected cells can also bind chemokines.


Assuntos
Quimiocinas/metabolismo , Varicellovirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Quimiocinas/antagonistas & inibidores , Quimiocinas C/metabolismo , Quimiocinas CC/metabolismo , Quimiocinas CXC/metabolismo , DNA Viral/química , Dados de Sequência Molecular , Ligação Proteica , Análise de Sequência de DNA
19.
Oral Microbiol Immunol ; 18(2): 131-4, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12654105

RESUMO

Oral mucosal salivary samples were collected from 25 cats with chronic gingivostomatitis and 24 cats with periodontal disease. Viral culture and isolation of feline calicivirus and feline herpesvirus 1 were performed. Eighty-eight per cent of cats with chronic gingivostomatitis were shedding both viruses, compared to 21% of cats without chronic oral inflammatory disease. Cats with chronic gingivostomatitis are significantly more likely to concurrently shed both feline calicivirus and feline herpesvirus 1 than are cats with classical periodontal disease.


Assuntos
Infecções por Caliciviridae/veterinária , Calicivirus Felino/fisiologia , Doenças do Gato/virologia , Gengivite/veterinária , Infecções por Herpesviridae/veterinária , Estomatite/veterinária , Varicellovirus/fisiologia , Eliminação de Partículas Virais/fisiologia , Animais , Gatos , Doença Crônica , Intervalos de Confiança , Gengivite/virologia , Doenças Periodontais/virologia , Estomatite/virologia
20.
Arch Virol ; 148(3): 537-45, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12607104

RESUMO

The simian varicella virus (SVV) glycoprotein C (gC), which may play an important role in viral pathogenesis, shares extensive homology to the varicella-zoster virus (VZV) gC. The SVV gC gene includes two identical 83 base pair repeat elements which are conserved within the gC genes of epidemiologically distinct SVV isolates. Expression of the gC gene was confirmed by detection of viral gene products. Deletion of the gC gene and replacement with the green fluorescent protein (GFP) gene yields a SVVgC(-)/GFP mutant which replicates as efficiently as wild type virus, demonstrating the SVV gC gene is nonessential for in vitro replication.


Assuntos
Varicellovirus/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Deleção de Genes , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de DNA , Varicellovirus/genética , Varicellovirus/patogenicidade , Proteínas do Envelope Viral/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA