Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
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
2.
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
3.
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
4.
Viruses ; 12(9)2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32917018

RESUMO

Infectious keratoconjunctivitis (IKC) is a common transmissible ocular disease in semi-domesticated Eurasian tundra reindeer (Rangifer tarandus tarandus). In large outbreaks, IKC may affect tens of animals in a herd, with the most severe cases often requiring euthanasia due to the destruction of the affected eyes and permanent blindness. An experimental inoculation with cervid herpesvirus 2 (CvHV2), alone or in combination with Moraxella bovoculi, demonstrated that CvHV2 has the ability to cause clinical signs of IKC in previously unexposed reindeer. Tissues collected from upper and lower eyelids, lacrimal gland and cornea, were processed for light and transmission electron microscopy. Histopathological analysis of the eyes inoculated with CvHV2 showed widespread and severe pathological findings. Mucosal tissues from these eyes showed fibrinous and purulent exudates, hyperemia, hemorrhages, necrosis, vascular thrombosis, vascular necrosis, infiltration of mononuclear cells and neutrophils, and lymphoid follicle reaction, which matches the described histopathology of IKC in reindeer. Characteristic alpha-herpesvirus particles matching the size and morphology of CvHV2 were identified by transmission electron microscopy in the conjunctival tissue. The quantification of viral particles by qPCR revealed high copy numbers of viral DNA in all CvHV2 inoculated eyes, but also in the non-inoculated eyes of the same animals. The histopathology of eye tissues obtained from the CvHV2 inoculated reindeer and the lack of inflammation from bacterial infection, together with the detection of CvHV2 DNA in swabs from the inoculated and non-inoculated eyes of the same animals, verified that CvHV2 was the primary cause of the observed histopathological changes.


Assuntos
Olho/virologia , Infecções por Herpesviridae/veterinária , Ceratoconjuntivite Infecciosa/virologia , Rena/virologia , Varicellovirus/fisiologia , Animais , DNA Viral/genética , Olho/patologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Ceratoconjuntivite Infecciosa/patologia , Varicellovirus/genética
5.
Vet Pathol ; 57(5): 687-699, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32744164

RESUMO

Epizootic epitheliotropic disease virus (salmonid herpesvirus-3; EEDV) is responsible for the death of millions of hatchery-raised lake trout (Salvelinus namaycush) in the Laurentian Great Lakes Basin. However, little is known about its biology, pathology, tropism, and host interactions. In this study, the presence and disease progression of EEDV were evaluated following exposure of naïve juvenile lake trout to EEDV via bath immersion under controlled laboratory conditions (n = 84 infected; n = 44 control). Individual tissues (n = 10 per fish), collected over 6 weeks, were analyzed for viral load by quantitative polymerase chain reaction, gross and histopathologic changes, and virus cellular targets using in situ hybridization. Skin, fin, and ocular tissues were the earliest viral targets and yielded the highest viral loads throughout the course of infection. Early gross lesions included exophthalmia, ocular hemorrhage, fin congestion, and hyperemia of visceral blood vessels. Advanced disease was characterized by multifocal to coalescing erosions and ulcerations of the skin, and congestion of visceral organs. Microscopically, there was cellular degeneration and necrosis in the epidermis and spleen, and lymphohistiocytic perivasculitis of the dermis, omentum, and the epicardium. EEDV DNA was first detected by in situ hybridization in epithelial cells of the epidermis, with subsequent labeling in the epithelial lining of primary and secondary gill lamellae. During advanced disease, EEDV was detected in endothelial and dendritic cells as well as blood monocytes. This study characterized EEDV tissue tropism and associated pathologic features, to guide research aimed at understanding EEDV disease ecology and improving strategies for disease control.


Assuntos
Doenças dos Peixes/patologia , Infecções por Herpesviridae/veterinária , Truta/virologia , Varicellovirus/fisiologia , Animais , Progressão da Doença , Doenças dos Peixes/virologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia
6.
Vet Microbiol ; 245: 108707, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32456815

RESUMO

Feline viral rhinotracheitis is a prevalent disease among cats caused by feline herpesvirus 1 (FHV-1). microRNAs (miRNAs), which serve as important regulatory factors in the host, participate in the regulation of the host innate immune response to virus infection. However, the roles of miRNAs in the FHV-1 life cycle remain unclear. In this study, we found that a new miRNA, miR-101, could suppress FHV-1 replication. FHV-1 infection upregulated the expression level of miR-101 in a cGAS-dependent manner. Furthermore, miR-101 could significantly enhance type I interferon antiviral signaling by targeting suppressor of cytokine signaling 5 (SOCS5), a negative regulator of the JAK-STAT pathway. Likewise, knockdown of cellular SOCS5 also suppressed FHV-1 replication due to the enhancement of IFN-I-induced signaling cascades. Taken together, our data demonstrated a new strategy for miR-101-mediated defense against FHV-1 infection by enhancing IFN-I antiviral signaling and increased the knowledge of miRNAs regulating innate immune signaling pathways.


Assuntos
Infecções por Herpesviridae/veterinária , Interações Hospedeiro-Patógeno , MicroRNAs/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Varicellovirus/fisiologia , Replicação Viral , Animais , Gatos , Linhagem Celular , Infecções por Herpesviridae/virologia , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/imunologia , Varicellovirus/patogenicidade
7.
J Zoo Wildl Med ; 51(1): 210-216, 2020 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-32212565

RESUMO

Cheetahs (Acinonyx jubatus) are particularly susceptible to feline herpesvirus-1 (FHV-1). Recommendations for preventive health care in cheetahs include vaccination against FHV-1 using killed and modified live virus (MLV) vaccines. Although MLV vaccines tend to induce a more robust immune response than killed vaccines, they can induce disease. This case series details an FHV-1 outbreak in four adult cheetahs following the use of MLV vaccine in one of them. All four cheetahs developed severe FHV-1 clinical signs and were euthanized. Clinical signs included depression, anorexia, nasal discharge, ocular discharge, sneezing, and ulcerative dermatitis. Herpesvirus infection was diagnosed using history, clinical signs, polymerase chain reaction, and histologic evaluation. The timeline of events suggests the MLV vaccine was the inciting cause, although this was not conclusively proven. Outcome of this case suggests that when considering MLV vaccines for cheetahs, careful risk and benefit discussions are merited.


Assuntos
Acinonyx , Infecções por Herpesviridae/veterinária , Vacinação/veterinária , Vacinas Atenuadas/efeitos adversos , Varicellovirus/fisiologia , Animais , Animais de Zoológico , Feminino , Infecções por Herpesviridae/diagnóstico , Infecções por Herpesviridae/etiologia , Infecções por Herpesviridae/prevenção & controle , Masculino , Resultado do Tratamento , Vacinação/efeitos adversos , Varicellovirus/efeitos dos fármacos
8.
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
9.
Viruses ; 12(1)2019 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-31861450

RESUMO

In response to viral infection, host cells activate various antiviral responses to inhibit virus replication. While feline herpesvirus 1 (FHV-1) manipulates the host early innate immune response in many different ways, the host could activate the antiviral response to counteract it through some unknown mechanisms. MicroRNAs (miRNAs) which serve as a class of regulatory factors in the host, participate in the regulation of the host innate immune response against virus infection. In this study, we found that the expression levels of miR-26a were significantly upregulated upon FHV-1 infection. Furthermore, FHV-1 infection induced the expression of miR-26a via a cGAS-dependent pathway, and knockdown of cellular cGAS significantly blocked the expression of miR-26a induced by poly (dA:dT) or FHV-1 infection. Next, we investigated the biological function of miR-26a during viral infection. miR-26a was able to increase the phosphorylation of STAT1 and promote type I IFN signaling, thus inhibiting viral replication. The mechanism study showed that miR-26a directly targeted host SOCS5. Knockdown of SOCS5 increased the phosphorylation of STAT1 and enhanced the type I IFN-mediated antiviral response, and overexpression of suppressor of the cytokine signalling 5 (SOCS5) decreased the phosphorylation of STAT1 and inhibited the type I IFN-mediated antiviral response. Meanwhile, with the knockdown of SOCS5, the upregulated expression of phosphorylated STAT1 and the anti-virus effect induced by miR-26a were significantly inhibited. Taken together, our data demonstrated a new strategy of host miRNAs against FHV-1 infection by enhancing IFN antiviral signaling.


Assuntos
Regulação da Expressão Gênica , Interferon Tipo I/metabolismo , MicroRNAs/genética , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/genética , Varicellovirus/fisiologia , Replicação Viral/genética , Regiões 3' não Traduzidas , Animais , Doenças do Gato/genética , Doenças do Gato/metabolismo , Doenças do Gato/virologia , Gatos , Linhagem Celular , Células Cultivadas , Infecções por Herpesviridae/veterinária , Interações Hospedeiro-Patógeno/genética , Humanos , Interferon Tipo I/biossíntese , Janus Quinases/metabolismo , Nucleotidiltransferases/metabolismo , Interferência de RNA , Fatores de Transcrição STAT/metabolismo
10.
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
11.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30045987

RESUMO

Alphaherpesvirus-associated ocular infections in humans caused by human alphaherpesvirus 1 (HHV-1) remain challenging to treat due to the frequency of drug application required and the potential for the selection of drug-resistant viruses. Repurposing on-the-market drugs is a viable strategy to accelerate the pace of drug development. It has been reported that the human immunodeficiency virus (HIV) integrase inhibitor raltegravir inhibits HHV-1 replication by targeting the DNA polymerase accessory factor and limits terminase-mediated genome cleavage of human betaherpesvirus 5 (HHV-5). We have previously shown, both in vitro and in vivo, that raltegravir can also inhibit the replication of felid alphaherpesvirus 1 (FeHV-1), a common ocular pathogen of cats with a pathogenesis similar to that of HHV-1 ocular disease. In contrast to what was reported for HHV-1, we were unable to select for a raltegravir-resistant FeHV-1 strain in order to define any basis for drug action. A candidate-based approach to explore the mode of action of raltegravir against FeHV-1 showed that raltegravir did not impact FeHV-1 terminase function, as described for HHV-5. Instead, raltegravir inhibited DNA replication, similarly to HHV-1, but by targeting the initiation of viral DNA replication rather than elongation. In addition, we found that raltegravir specifically repressed late gene expression independently of DNA replication, and both activities are consistent with inhibition of ICP8. Taken together, these results suggest that raltegravir could be a valuable therapeutic agent against herpesviruses.IMPORTANCE The rise of drug-resistant herpesviruses is a longstanding concern, particularly among immunocompromised patients. Therefore, therapies targeting viral proteins other than the DNA polymerase that may be less likely to lead to drug-resistant viruses are urgently needed. Using FeHV-1, an alphaherpesvirus closely related to HHV-1 that similarly causes ocular herpes in its natural host, we found that the HIV integrase inhibitor raltegravir targets different stages of the virus life cycle beyond DNA replication and that it does so without developing drug resistance under the conditions tested. This shows that the drug could provide a viable strategy for the treatment of herpesvirus infections.


Assuntos
Inibidores de Integrase de HIV/farmacologia , Raltegravir Potássico/farmacologia , Varicellovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Gatos , Linhagem Celular , DNA Viral/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Varicellovirus/efeitos dos fármacos , Proteínas Virais/metabolismo
12.
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
13.
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
14.
J Virol ; 90(23): 10823-10843, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27681124

RESUMO

Primary infection with varicella-zoster virus (VZV), a neurotropic alphaherpesvirus, results in varicella. VZV establishes latency in the sensory ganglia and can reactivate later in life to cause herpes zoster. The relationship between VZV and its host during acute infection in the sensory ganglia is not well understood due to limited access to clinical specimens. Intrabronchial inoculation of rhesus macaques with simian varicella virus (SVV) recapitulates the hallmarks of VZV infection in humans. We leveraged this animal model to characterize the host-pathogen interactions in the ganglia during both acute and latent infection by measuring both viral and host transcriptomes on days postinfection (dpi) 3, 7, 10, 14, and 100. SVV DNA and transcripts were detected in sensory ganglia 3 dpi, before the appearance of rash. CD4 and CD8 T cells were also detected in the sensory ganglia 3 dpi. Moreover, lung-resident T cells isolated from the same animals 3 dpi also harbored SVV DNA and transcripts, suggesting that T cells may be responsible for trafficking SVV to the ganglia. Transcriptome sequencing (RNA-Seq) analysis showed that cessation of viral transcription 7 dpi coincides with a robust antiviral innate immune response in the ganglia. Interestingly, a significant number of genes that play a critical role in nervous system development and function remained downregulated into latency. These studies provide novel insights into host-pathogen interactions in the sensory ganglia during acute varicella and demonstrate that SVV infection results in profound and sustained changes in neuronal gene expression. IMPORTANCE: Many aspects of VZV infection of sensory ganglia remain poorly understood, due to limited access to human specimens and the fact that VZV is strictly a human virus. Infection of rhesus macaques with simian varicella virus (SVV), a homolog of VZV, provides a robust model of the human disease. Using this model, we show that SVV reaches the ganglia early after infection, most likely by T cells, and that the induction of a robust innate immune response correlates with cessation of virus transcription. We also report significant changes in the expression of genes that play an important role in neuronal function. Importantly, these changes persist long after viral replication ceases. Given the homology between SVV and VZV, and the genetic and physiological similarities between rhesus macaques and humans, our results provide novel insight into the interactions between VZV and its human host and explain some of the neurological consequences of VZV infection.


Assuntos
Gânglios Sensitivos/metabolismo , Gânglios Sensitivos/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Varicellovirus/patogenicidade , Doença Aguda , Animais , Transporte Axonal , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Varicela/virologia , DNA Viral/genética , DNA Viral/metabolismo , Modelos Animais de Doenças , Gânglios Sensitivos/imunologia , Expressão Gênica , Infecções por Herpesviridae/imunologia , Herpesvirus Humano 3/patogenicidade , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macaca mulatta , Neurogênese , Varicellovirus/genética , Varicellovirus/fisiologia , Replicação Viral
15.
Virus Res ; 221: 15-22, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27157860

RESUMO

Felid herpesvirus 1 (FHV-1) mutants were constructed using two-step Red-mediated recombination techniques based on a virulent full-length FHV-1 BAC clone. The individual mutant viruses generated were deficient in glycoprotein C (gC), glycoprotein E (gE), US3 serine/threonine protein kinase (PK), or both gE and thymidine kinase (TK). The gC- mutant virus produced plaques that were similar in size to those resulting from infection with the C-27 parent strain. In contrast, the gE(-), PK(-), and gE(-)PK(-) deletion mutants produced plaques that were significantly smaller. Multistep in vitro growth kinetics of the gE(-), PK(-), and gE(-)PK(-) viruses were slightly delayed compared to those of the C-27 parent strain. Peak progeny titers of these three mutants were approximately 10-fold lower than those generated with the C-27 strain. There was no delay in the growth kinetics of the gC- mutant, but the progeny virus titer obtained with this mutant was at least 3 logs lower compared to the parental strain titer. Based upon their in vitro characteristics, these mutants will be useful for the development of novel immunization strategies against this important feline pathogen.


Assuntos
Recombinação Genética , Varicellovirus/genética , Varicellovirus/fisiologia , Animais , Gatos , Linhagem Celular , Cromossomos Artificiais Bacterianos , Mutagênese , Carga Viral , Ensaio de Placa Viral , Proteínas Virais/genética , Replicação Viral
16.
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
17.
Elife ; 42015 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-26259874

RESUMO

Varicella-zoster virus (VZV) causes chickenpox and reactivation of latent VZV causes herpes zoster (HZ). VZV reactivation is subject to the opposing mechanisms of declining and boosted VZV-specific cellular mediated immunity (CMI). A reduction in exogenous re-exposure 'opportunities' through universal chickenpox vaccination could therefore lead to an increase in HZ incidence. We present the first individual-based model that integrates within-host data on VZV-CMI and between-host transmission data to simulate HZ incidence. This model allows estimating currently unknown pivotal biomedical parameters, including the duration of exogenous boosting at 2 years, with a peak threefold to fourfold increase of VZV-CMI; the VZV weekly reactivation probability at 5% and VZV subclinical reactivation having no effect on VZV-CMI. A 100% effective chickenpox vaccine given to 1 year olds would cause a 1.75 times peak increase in HZ 31 years after implementation. This increase is predicted to occur mainly in younger age groups than is currently assumed.


Assuntos
Vacina contra Varicela/administração & dosagem , Vacina contra Varicela/imunologia , Herpes Zoster/epidemiologia , Herpes Zoster/imunologia , Imunidade Celular , Varicellovirus/imunologia , Ativação Viral , Humanos , Modelos Biológicos , Modelos Estatísticos , Medição de Risco , Varicellovirus/fisiologia
18.
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
19.
PLoS Pathog ; 11(5): e1004901, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25973608

RESUMO

Varicella zoster virus (VZV) causes chickenpox in humans and, subsequently, establishes latency in the sensory ganglia from where it reactivates to cause herpes zoster. Infection of rhesus macaques with simian varicella virus (SVV) recapitulates VZV pathogenesis in humans thus representing a suitable animal model for VZV infection. While the type I interferon (IFN) response has been shown to affect VZV replication, the virus employs counter mechanisms to prevent the induction of anti-viral IFN stimulated genes (ISG). Here, we demonstrate that SVV inhibits type I IFN-activated signal transduction via the JAK-STAT pathway. SVV-infected rhesus fibroblasts were refractory to IFN stimulation displaying reduced protein levels of IRF9 and lacking STAT2 phosphorylation. Since previous work implicated involvement of the VZV immediate early gene product ORF63 in preventing ISG-induction we studied the role of SVV ORF63 in generating resistance to IFN treatment. Interestingly, SVV ORF63 did not affect STAT2 phosphorylation but caused IRF9 degradation in a proteasome-dependent manner, suggesting that SVV employs multiple mechanisms to counteract the effect of IFN. Control of SVV ORF63 protein levels via fusion to a dihydrofolate reductase (DHFR)-degradation domain additionally confirmed its requirement for viral replication. Our results also show a prominent reduction of IRF9 and inhibition of STAT2 phosphorylation in VZV-infected cells. In addition, cells expressing VZV ORF63 blocked IFN-stimulation and displayed reduced levels of the IRF9 protein. Taken together, our data suggest that varicella ORF63 prevents ISG-induction both directly via IRF9 degradation and indirectly via transcriptional control of viral proteins that interfere with STAT2 phosphorylation. SVV and VZV thus encode multiple viral gene products that tightly control IFN-induced anti-viral responses.


Assuntos
Infecções por Herpesviridae/metabolismo , Interações Hospedeiro-Patógeno , Interferon Tipo I/metabolismo , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Varicellovirus/fisiologia , Animais , Linhagem Celular , Cercopithecinae , Varicela/imunologia , Varicela/metabolismo , Varicela/patologia , Varicela/virologia , DNA Recombinante/metabolismo , Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/fisiologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Imunidade Inata , Interferon Tipo I/antagonistas & inibidores , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/antagonistas & inibidores , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma , Processamento de Proteína Pós-Traducional , Proteólise , Proteínas Recombinantes/metabolismo , Fatores de Transcrição STAT/genética , Varicellovirus/imunologia
20.
J Neurovirol ; 20(5): 526-30, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25139181

RESUMO

Simian varicella virus (SVV) infection of non-human primates models human varicella zoster virus (VZV) infection. Assessment of cell signaling immune responses in monkeys after primary SVV infection, after immunosuppression and during reactivation revealed strong pro-inflammatory responses and lesser anti-inflammatory components during varicella and reactivation. Pro-inflammatory mediators elevated during varicella included interferon-gamma (IFN-γ), interleukin (IL)-6, monocyte chemoattractant protein (MCP-1), interferon inducible T-cell α chemoattractant protein (I-TAC), interferon processing protein (IP-10), and anti-inflammatory interleukin-1 Receptor antagonist (IL-1Ra). After immunosuppression and at reactivation, levels of pro-inflammatory mediators MCP-1, eotaxin, IL-6, IL-8, MIF, RANTES (regulated-on-activation normal T-cell expressed and secreted), and HGF (hepatocyte growth factor) were elevated, as was the anti-inflammatory mediator IL-1Ra. Characterization of cytokine, chemokine and growth factor responses during different stages of varicella virus infection will facilitate immunotherapeutic and vaccine strategies.


Assuntos
Infecções por Herpesviridae/imunologia , Ativação Viral/imunologia , Latência Viral/imunologia , Animais , Quimiocinas/imunologia , Modelos Animais de Doenças , Inflamação/imunologia , Inflamação/virologia , Macaca mulatta , Masculino , Varicellovirus/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...