Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 212
Filtrar
1.
Mol Immunol ; 136: 55-64, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34087624

RESUMO

Pseudorabies virus (PRV) is an enveloped double-stranded DNA virus that is the etiological agent of Aujeszky's disease in pigs. Vaccination is currently available to prevent PRV infection, but there is still an urgent need for new strategies to control this infectious disease. Histone deacetylases (HDACs) are epigenetic regulators that regulate the histone tail, chromatin conformation, protein-DNA interaction and even transcription. Viral transcription and protein activities are intimately linked to regulation by histone acetyltransferases and HDACs that remodel chromatin and regulate gene expression. We reported here that genetic and pharmacological inhibition of HDAC1 significantly influenced PRV replication. Moreover, we demonstrated that inhibition of HDAC1 induced a DNA damage response and antiviral innate immunity. Mechanistically, the HDAC1 inhibition-induced DNA damage response resulted in the release of double-strand DNA into the cytosol to activate cyclic GMP-AMP synthase and the downstream STING/TBK1/IRF3 innate immune signaling pathway. Our results demonstrate that an HDAC1 inhibitor may be used as a new strategy to prevent Aujeszky's disease in pigs.


Assuntos
Herpesvirus Suídeo 1/efeitos dos fármacos , Histona Desacetilase 1/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Pseudorraiva/tratamento farmacológico , Células 3T3 , Animais , Linhagem Celular , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/genética , Células HEK293 , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Histona Desacetilase 1/genética , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Nucleotidiltransferases/metabolismo , Pseudorraiva/imunologia , Células RAW 264.7 , Interferência de RNA , RNA Interferente Pequeno/genética , Suínos , Doenças dos Suínos/virologia , Replicação Viral/efeitos dos fármacos
2.
Virology ; 553: 70-80, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33242760

RESUMO

Many viruses utilize molecular chaperone heat shock protein 90 (Hsp90) for protein folding and stabilization, however, the role of Hsp90 in herpesvirus lifecycle is obscure. Here, we provide evidence that Hsp90 participates in pseudorabies virus (PRV) replication. Viral growth kinetics assays show that Hsp90 inhibitor geldanamycin (GA) abrogates PRV replication at the post-penetration step. Transmission electron microscopy demonstrates that dysfunction of Hsp90 diminishes the quantity of PRV nucleocapsids. Overexpression and knockdown of Hsp90 suggest that de novo Hsp90 is involved in PRV replication. Mechanismly, dysfunction of Hsp90 inhibits PRV major capsid protein VP5 expression. Co-immunoprecipitation and indirect immunofluorescence assays indicate that Hsp90 interacts with VP5. Interestingly, Hsp70, a collaborator of Hsp90, also interacts with VP5, but doesn't affect PRV growth. Finally, inhibition of Hsp90 results in PRV VP5 degradation in a proteasome-dependent manner. Collectively, our data suggest that Hsp90 contributes to PRV virion assembly and replication via stabilization of VP5.


Assuntos
Proteínas do Capsídeo/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Herpesvirus Suídeo 1/fisiologia , Montagem de Vírus , Animais , Benzoquinonas/farmacologia , Proteínas do Capsídeo/química , Linhagem Celular , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/ultraestrutura , Humanos , Lactamas Macrocíclicas/farmacologia , Nucleocapsídeo/ultraestrutura , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Dobramento de Proteína , Estabilidade Proteica , Suínos , Vírion/crescimento & desenvolvimento , Vírion/fisiologia , Replicação Viral/efeitos dos fármacos
3.
Biotechnol Lett ; 42(12): 2551-2560, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32816175

RESUMO

Fixed-bed bioreactors packed with macrocarriers show great potential to be used for vaccine process development and large-scale production due to distinguishing features of low shear force, high cell adhering surface area, and easy replacement of culture media in situ. As an initial step of utilizing this type of bioreactors for Pseudorabies virus production (PRV) by African green monkey kidney (Vero) cells, we developed a tube-fixed-bed bioreactor in the previous study, which represents a scale-down model for further process optimization. By using this scale-down model, here we evaluated impacts of two strategies (use of serum-free medium and low cell inoculum density) on PRV production, which have benefits of simplifying downstream process and reducing risk of contamination. We first compared Vero cell cultures with different media, bioreactors and inoculum densities, and conclude that cell growth with serum-free medium is comparable to that with serum-containing medium in tube-fixed-bed bioreactor, and low inoculum density supports cell growth only in this bioreactor. Next, we applied serum-free medium and low inoculum cell density for PRV production. By optimization of time of infection (TOI), multiplicity of infection (MOI) and the harvesting strategy, we obtained total amount of virus particles ~ 9 log10 TCID50 at 5 days post-infection (dpi) in the tube-fixed-bed bioreactor. This process was then scaled up by 25-fold to a Xcell 1-L fixed-bed bioreactor, which yields totally virus particles of 10.5 log10 TCID50, corresponding to ~ 3 × 105 doses of vaccine. The process studied in this work holds promise to be developed as a generic platform for the production of vaccines for animal and human health.


Assuntos
Reatores Biológicos , Contagem de Células , Herpesvirus Suídeo 1/genética , Células Vero/virologia , Animais , Chlorocebus aethiops/genética , Chlorocebus aethiops/crescimento & desenvolvimento , Meios de Cultura/química , Meios de Cultura/farmacologia , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Cultura de Vírus/métodos
4.
J Neurovirol ; 26(4): 556-564, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32572833

RESUMO

Pseudorabies virus (PRV) is known to cause severe encephalitis in juvenile pigs and various non-native hosts; recent evidences suggest that PRV might cause encephalitis in humans. In a multicenter cohort study in China, next-generation sequencing of cerebrospinal fluid (CSF) was performed to detect pathogens in all patients with clinically suspected central nervous system infections. This study involved all the patients whose CSF samples were positive for PRV-DNA; their clinical features were evaluated, and species-specific PCR and serological tests were sequentially applied for validation. Among the 472 patients tested from June 1, 2016, to December 1, 2018, six were positive for PRV-DNA, which were partially validated by PCR and serological tests. Additionally, we retrospectively examined another case with similar clinical and neuroimaging appearance and detected the presence of PRV-DNA. These patients had similar clinical manifestations, including a rapid progression of panencephalitis, and similar neuroimaging features of symmetric lesions in the basal ganglia and bilateral hemispheres. Six of the patients were engaged in occupations connected with swine production. PRV infection should be suspected in patients with rapidly progressive panencephalitis and characteristic neuroimaging features, especially with exposure to swine.


Assuntos
Gânglios da Base/patologia , Cérebro/patologia , DNA Viral/genética , Encefalite Viral/patologia , Herpesvirus Suídeo 1/genética , Carne/virologia , Pseudorraiva/patologia , Adulto , Animais , Anticorpos Antivirais/líquido cefalorraquidiano , Gânglios da Base/diagnóstico por imagem , Gânglios da Base/virologia , Cérebro/diagnóstico por imagem , Cérebro/virologia , China , DNA Viral/líquido cefalorraquidiano , Encefalite Viral/líquido cefalorraquidiano , Encefalite Viral/diagnóstico , Encefalite Viral/virologia , Feminino , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/patogenicidade , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Pseudorraiva/líquido cefalorraquidiano , Pseudorraiva/diagnóstico , Pseudorraiva/virologia , Suínos
5.
Vet Microbiol ; 240: 108543, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31902487

RESUMO

Since 2011, to control the spread of pseudorabies (PR), US7/US8/UL23-deleted recombinant PRV (rPRV) vaccines based on current variants have been developed. The vaccines can provide effective immune protection to pigs, but fur-bearing animals, such as dogs, foxes, and minks, are increasingly infected by PRV due to consuming contaminated raw meat or offal from immunized pigs. It is suspected that the attenuated PRV vaccine strain is not safe for these fur-bearing animals. To confirm this, we construct a US7/US8/UL23-deleted and a US7/US8/UL23/US3-deleted rPRV based on PRV GL isolated from fox using the CRISPR/Cas9 method. Growth kinetics in vitro and pathogenicity in dogs were compared between the wild type and both rPRVs. The results showed that the growth kinetics of wild-type PRV and US7/US8/UL23-deleted rPRV were faster than those of US7/US8/UL23/US3-deleted recombinant PRV from 24 h to 48 h post infection. Moreover, PRV GL- and rPRVdelUS7/US8/UL23-infected cells formed cell-cell fusion, but the rPRVdelUS7/US8/UL23/US3-infected cells did not. Dogs challenged with wild-type PRV or US7/US8/UL23-deleted rPRV showed obvious nervous symptoms, and all the dogs died, but the group challenged with the US7/US8/UL23/US3-deleted rPRV did not show any nervous symptoms, and all the dogs survived for the duration of the experiment. Tissue viral load analyses also showed that the virulence of the US7/US8/UL23/US3-deleted rPRV was significantly reduced in dogs. This study provides evidence that the US7/US8/UL23-deleted rPRV variant still exhibits high virulence for dogs and also highlights the role of the US3 gene in the pathogenicity of PRV in dogs and provides a strategy for developing a safer vaccine.


Assuntos
Deleção de Genes , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Pseudorraiva/virologia , Vacina Antirrábica/imunologia , Proteínas Virais/genética , Animais , Anticorpos Antivirais/sangue , Sistemas CRISPR-Cas , Cães , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Pseudorraiva/imunologia , Vacina Antirrábica/administração & dosagem , Vacina Antirrábica/genética , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/genética , Virulência
6.
Intervirology ; 62(3-4): 116-123, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31430757

RESUMO

BACKGROUND: Pseudorabies virus (PRV) infection induces apoptosis in swine cells both in vitro and in vivo; however, the mechanism associated with host-cell signaling has not been studied. This study investigated the role of free radicals caused by cellular oxidative stress after viral infection and examined whether the DNA damage response plays an important role in PRV-induced apoptosis. METHODS: Several apoptosis assays and western blotting confirmed PRV-induced apoptosis. PRV-mediated oxidative stress was evaluated by reactive oxygen species (ROS) assay. RESULTS: Our results showed that PRV caused apoptosis in a porcine kidney cell line, PK15, and induced expressions of proapoptotic Bcl family proteins in a dose- and time-dependent manner. Expressions of specific DNA damage sensors and phosphorylation of histone H2AX were also significantly increased, which subsequently activated the expressions of checkpoint kinase 1/2 and proapoptotic p53. Caffeine, a known DNA damage inhibitor, was found to inhibit caspase-3 activation and protect cells from PRV-induced apoptosis. Additionally, the antioxidant N-acetyl-L-cysteine was shown to prevent the production of cellular ROS, protecting DNA from cleavage. CONCLUSIONS: Our results confirmed that oxidative stress and free radicals arising from PRV infection cause DNA damage, which consequently triggers apoptosis.


Assuntos
Apoptose , Dano ao DNA , Células Epiteliais/virologia , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Estresse Oxidativo , Transdução de Sinais , Animais , Western Blotting , Linhagem Celular , Células Epiteliais/patologia , Espécies Reativas de Oxigênio/análise , Suínos
7.
Biomed Res Int ; 2019: 7398208, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30941371

RESUMO

Ribonuclease L (RNase L) is an important antiviral endoribonuclease regulated by type I IFN. RNase L is activated by viral infection and dsRNA. Because the role of swine RNase L (sRNase L) is not fully understood, in this study, we generated a sRNase L knockout PK-15 (KO-PK) cell line through the CRISPR/Cas9 gene editing system to evaluate the function of sRNase L. After transfection with CRISPR-Cas9 followed by selection using puromycin, sRNase L knockout in PK-15 cells was further validated by agarose gel electrophoresis, DNA sequencing, and Western blotting. The sRNase L KO-PK cells failed to trigger RNA degradation and induced less apoptosis than the parental PK-15 cells after transfected with poly (I: C). Furthermore, the levels of ISGs mRNA in sRNase L KO-PK cells were higher than those in the parental PK-15 cells after treated with poly (I: C). Finally, both wild type and attenuated pseudorabies viruses (PRV) replicated more efficiently in sRNase L KO-PK cells than the parental PK-15 cells. Taken together, these findings suggest that sRNase L has multiple biological functions including cellular single-stranded RNA degradation, induction of apoptosis, downregulation of transcript levels of ISGs, and antiviral activity against PRV. The sRNase L KO-PK cell line will be a valuable tool for studying functions of sRNase L as well as for producing PRV attenuated vaccine.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Endorribonucleases/metabolismo , Técnicas de Inativação de Genes , Herpesvirus Suídeo 1/fisiologia , Replicação Viral/fisiologia , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Linhagem Celular , Edição de Genes , Herpesvirus Suídeo 1/efeitos dos fármacos , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Poli I-C/farmacologia , Pseudorraiva/virologia , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Ribossômico/genética , Suínos , Vacinas Virais/imunologia , Replicação Viral/efeitos dos fármacos
8.
Protein Expr Purif ; 153: 53-58, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30165247

RESUMO

Recombinant interferon-α (rIFN-α) has been widely used for treating viral infections. However, the clinical efficacy of unmodified rIFN-α is limited due to small molecular size and rapid clearance from circulation. In this study we developed a novel strategy for half-life extension of porcine IFN-α (PoIFN-α) by fusion to the immunoglobulin (Ig)-binding C2 domain of streptococcal protein G (SPG). The coding sequences for PoIFN-α6 and SPG C2 domain, with a tobacco etch virus (TEV) protease recognition sequence introduced at the 5-end, were cloned into an elastin-like polypeptide (ELP) fusion expression vector and expressed as an ELP-PoIFNα-C2 fusion protein. After optimization of the conditions for soluble protein expression and purification, the fusion protein was purified to more than 90% purity by two rounds of inverse transition cycling (ITC) in the presence of 0.5% Triton X-100. After cleavage with self-aggregating peptide ELK-16-tagged tobacco etch virus protease, the protease was removed by quick centrifugation and PoIFNα-C2 protein was recovered by an additional round of ITC with 98% purity. Western blotting analysis showed that PoIFNα-C2 protein had the specific affinity for pig IgG binding. The antiviral assay showed that PoIFNα-C2 protein had potent antiviral activities against vesicular stomatitis virus and porcine pseudorabies virus. After single intravenous or subcutaneous injection into rats, PoIFNα-C2 protein showed 16- or 4-fold increase in serum half-life with significantly improved bioavailability.


Assuntos
Proteínas de Bactérias/farmacocinética , Herpesvirus Suídeo 1/efeitos dos fármacos , Interferon-alfa/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Vesiculovirus/efeitos dos fármacos , Motivos de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Bioensaio , Disponibilidade Biológica , Linhagem Celular , Clonagem Molecular , Elastina/genética , Elastina/metabolismo , Endopeptidases/química , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Meia-Vida , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/imunologia , Humanos , Interferon-alfa/genética , Interferon-alfa/imunologia , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Suínos , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/imunologia
9.
Virol J ; 15(1): 195, 2018 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-30594230

RESUMO

BACKGROUND: Pseudorabies virus (PRV) of the family Herpesviridae is the causative agent of Aujeszky's disease. Attenuation of PRV by serial passaging in vitro is a well-established method; however, the dynamic variations occurring on viral genome during this process have not been characterized. METHODS: Genome sequencing and comparative genomic analyses of a virulent pseudorabies virus and a series of its plaque-purified strains via serial passaging in vitro were performed, and the properties in vitro and in vivo of which were further characterized. RESULTS: Compared to the parental virus, replication in vitro was enhanced in the highly passaged F50, F91, and F120. In contrast, lethality in mice decreased gradually with passage number. Genome sequencing of F50, F91, and F120 showed deletion of a large fragment containing gE, which is likely related to their attenuation. In addition, single nucleotide variations were identified in many genes of F50, F91, and F120. In-frame and frameshift indels were also detected in specific genes of passaged strains. Particularly frameshift mutations were observed in highly passaged strains, resulting in a truncated but overexpressed pUL46. CONCLUSION: During attenuation of PRV by serial passaging in Vero cells, dynamic variation patterns including a large deletion, single nucleotide variations, small in-frame indels, and also frameshifts mutations successively emerged, contributing to evolution of the viral population and enabling the gradual attenuation of the virus. These data provide clues to better understand PRV attenuation during passaging.


Assuntos
Genoma Viral , Genômica , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/genética , Inoculações Seriadas , Animais , Chlorocebus aethiops , Modelos Animais de Doenças , Mutação da Fase de Leitura , Herpesvirus Suídeo 1/patogenicidade , Mutação INDEL , Camundongos , Polimorfismo de Nucleotídeo Único , Pseudorraiva/patologia , Pseudorraiva/virologia , Análise de Sobrevida , Células Vero , Virulência , Replicação Viral , Sequenciamento Completo do Genoma
10.
J Virol Methods ; 259: 106-115, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29894711

RESUMO

Since late 2011, a pseudorabies virus (PRV) variant with increased virulence in old pigs had caused major disease outbreaks and great economic losses to the pig industry in China. The gene mutations that contributed to the increased virulence were unclear. To study the basis of the enhanced pathogenicity, an infectious bacterial artificial chromosome (BAC) clone consisting of the complete genome of the PRV variant was developed. Using homologous recombination and Cre/LoxP recombination, the recombinant virus rJS-2012-BAC carrying a BAC insertion downstream of the open reading frame (ORF) of gG was constructed. The circular genome of rJS-2012-BAC was extracted from infected Vero cells and transformed into Escherichia coli DH10B, generating the BAC clone pBAC-JS2012. The loxP sites flanking the BAC vector were used to excise the BAC sequences using Cre recombinase. The reconstituted BAC-excision virus, vJS2012 L, from pBAC-JS2012 exhibited similar biological properties to the wild-type virulent strain JS-2012. To manipulate the BAC clone pBAC-JS2012, the galK selection system was adopted to delete the gI/gE gene from pBAC-JS2012 in E. coli and to generate the gI/gE-deleted virus vJS2012-ΔgE/gI. The BAC clone, pBAC-JS2012, retained the same level of virulence as its parent strain and was easily manipulated using a galK system which would facilitate the study of the enhanced pathogenicity of the PRV variant as well as other studies on PRV.


Assuntos
Cromossomos Artificiais Bacterianos , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/genética , Animais , China , Chlorocebus aethiops , Clonagem Molecular , Escherichia coli/genética , Infecções por Escherichia coli , Deleção de Genes , Herpesvirus Suídeo 1/patogenicidade , Síndrome Respiratória e Reprodutiva Suína , Recombinação Genética , Suínos , Células Vero , Proteínas Virais/genética , Virulência , Fatores de Virulência/genética
11.
Virology ; 520: 59-66, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29777914

RESUMO

Pseudorabies virus (PRV), the etiological pathogen of Aujeszky's disease, belongs to the Alphaherpesvirus subfamily. Large latency transcript (LLT), the most abundant PRV transcript, harbors a ~ 4.6 kb microRNA (miRNA) cluster-encoding intron. To investigate the function of the LLT miRNA cluster during the life cycle of PRV, we generated a miRNA cluster mutation virus (PRV-∆miR cluster) and revertant virus. Analysis of the growth kinetics of PRV-ΔmiR cluster-infected cells revealed significantly smaller plaques and lower titers than the wild-type and revertant viruses. The mutation virus exhibited increased IE180 and decreased EP0 expression. The clinical symptoms observed in mice infected with PRV-ΔmiR cluster revealed that the miRNA cluster is involved in the pathogenesis of PRV. Physical parameters, virus shedding assays, and the SN50 titers revealed that the miRNA cluster enhances PRV virulence in pigs. Collectively, our findings suggest that the full-length miRNA cluster is involved in PRV replication and virulence.


Assuntos
Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Íntrons , MicroRNAs/genética , Animais , Linhagem Celular , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Camundongos , Mutação , Pseudorraiva/virologia , Suínos , Virulência , Latência Viral/genética , Replicação Viral/genética , Eliminação de Partículas Virais
12.
Virus Res ; 251: 56-67, 2018 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-29634995

RESUMO

Pseudorabies virus (PRV) is the causative agent of pseudorabies (PR) which causes large economic losses for Chinese swine industry since breaking out in late 2011. As a member of herpesviruses, PRV is able to escape the host immune elimination and establish latency, resulting in persistent infection. Here, we report that a currently prevalent Chinese PRV variant down-regulated swine leukocyte antigen class I (SLA-I) molecules on the surface of PK-15 cells and targeted them for degradation through lysosome pathway. Viral pUL56 protein, independent of other viral proteins, was associated with this function by inducing degradation of cellular SLA-I heavy chain (HC) in a manner that was dependent on the lysosome machinery. In addition, pUL56 interacted with SLA-I HC and increased its ubiquitination. Further studies demonstrated that the late domains (PPXY motifs) of pUL56 were required for the ubiquitination and degradation of SLA-I HC by pUL56. Together, our findings reveal the mechanisms by which PRV interferes with cytotoxic T lymphocyte (CTL) responses and provide novel insights into the roles of PRV pUL56.


Assuntos
Regulação para Baixo , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Lisossomos/metabolismo , Pseudorraiva/patologia , Doenças dos Suínos/virologia , Animais , Linhagem Celular , China , Modelos Biológicos , Proteólise , Pseudorraiva/virologia , Suínos , Doenças dos Suínos/patologia
13.
Virus Res ; 244: 199-207, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29155034

RESUMO

2', 5'-Oligoadenylate synthetase-lilke (OASL) protein is an atypical oligoadenylate synthetase (OAS) family member, which possesses antiviral activity but lacks 2', 5'-oligoadenylate synthetase activity. Here, a novel variant of porcine OASL (pOASL2) was identified through RT-PCR amplification. This gene is distinguishable from the previously described wild-type porcine OASL (pOASL1). The gene appears to be derived from a truncation of exon 4 plus 8 nucleotides of exon 5 with a premature termination, measuring only 633 bp in length, although its position corresponds to that of pOASL1. Given this novel gene appears to be a variant of pOASL, we assayed for antiviral activity of the protein. We demonstrated that pOASL2 could inhibit Japanese encephalitis virus (JEV) proliferation as well as pOASL1 in a transient overexpression assay of pOASL1 and pOASL2 in PK-15 and Vero cells. In addition to JEV, pOASL1 and pOASL2 also decreased the proliferations of Porcine reproductive and respiratory syndrome virus (PRRSV) and vesicular stomatitis virus (VSV), but did not exhibit antiviral activity against pseudorabies virus (PRV). Structural analysis showed that the pOASL2 gene retained only the first three exons at the 5'-. To investigate the role of the αN4 helix in pOASL in antiviral responses like that in hOASL, we mutated key residues in the anchor domain of the αN4 helix in pOASL2, based on the domain's location in hOASL. However, the antiviral activity of pOASL2 was not affected. Thus, the αN4 helix of pOASL likely does not play a significant role in its antiviral activity. In conclusion, pOASL2 acts as a new splice isoform of pOASL that plays a role in resistance to infection of several kinds of RNA viruses.


Assuntos
2',5'-Oligoadenilato Sintetase/farmacologia , Processamento Alternativo , Antivirais/farmacologia , Vírus da Encefalite Japonesa (Espécie)/efeitos dos fármacos , Vírus da Síndrome Respiratória e Reprodutiva Suína/efeitos dos fármacos , Vesiculovirus/efeitos dos fármacos , 2',5'-Oligoadenilato Sintetase/biossíntese , 2',5'-Oligoadenilato Sintetase/química , 2',5'-Oligoadenilato Sintetase/genética , Sequência de Aminoácidos , Animais , Antivirais/química , Linhagem Celular , Chlorocebus aethiops , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/crescimento & desenvolvimento , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/virologia , Éxons , Herpesvirus Suídeo 1/efeitos dos fármacos , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/metabolismo , Isoenzimas/biossíntese , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/farmacologia , Rim/efeitos dos fármacos , Rim/patologia , Rim/virologia , Fases de Leitura Aberta , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/crescimento & desenvolvimento , Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Conformação Proteica em alfa-Hélice , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Suínos , Células Vero , Vesiculovirus/genética , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/metabolismo
14.
J Gen Virol ; 98(6): 1467-1476, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28631596

RESUMO

Cholesterol 25-hydroxylase (CH25H) catalyses the production of 25-hydroxycholesterol (25HC) from cholesterol by adding a second hydroxyl group at position 25. The aim of this study was to examine the antiviral effect of CH25H on pseudorabies virus (PRV), a swine pathogen that can cause devastating disease and economic losses worldwide. The results showed that porcine ch25h was induced by either interferon or PRV infection. PRV infection of porcine alveolar macrophages (3D4/21 cells) was attenuated by CH25H overexpression and enhanced by silencing of CH25H. Furthermore, treatment of 3D4/21 cells with 25HC inhibited the growth of PRV in vitro, suggesting that CH25H may restrict PRV replication by 25HC production. We further identified that the anti-PRV role of CH25H and 25HC was subject to their inhibitory effect on PRV attachment and entry. Collectively, these findings demonstrate that CH25H is an intrinsic host restriction factor in PRV infection of porcine alveolar macrophages.


Assuntos
Antivirais/metabolismo , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/imunologia , Interações Hospedeiro-Patógeno , Hidroxicolesteróis/metabolismo , Esteroide Hidroxilases/metabolismo , Replicação Viral , Animais , Células Cultivadas , Imunidade Inata , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/virologia , Suínos , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
15.
Intervirology ; 59(1): 20-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27362774

RESUMO

OBJECTIVE: The aim of this study was to use partial Ul44 sequences (glycoprotein C) of Suid herpesvirus 1 to examine the evolution and dynamics of the virus in different periods and hosts. METHODS: Phylogenetic trees were constructed using the software MrBayes after analysis in the software jModelTest to evaluate the best phylogenetic models. The software SplitsTree 4.0 was used to create phylogenetic networks, and the BEAST program was used to generate data on phylogeography. Replication kinetics and serum neutralization tests were applied to tree strains from different phylogenetic groups. RESULTS: Ul44 sequences derived from domestic swine and wild swine clustered in different clades and had different selective pressures depending on the host. We found no differences in replication kinetics and serum neutralization tests in the strains tested. Data show that the evolution of herpesviruses is complex, and different genetic groups may be evolving at different rates. Ul44 is an important marker for molecular evolution and epidemiology studies, but it is not useful for biological information.


Assuntos
Evolução Molecular , Herpesvirus Suídeo 1/genética , Pseudorraiva/virologia , Doenças dos Suínos/virologia , Animais , Animais Selvagens/virologia , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Herpesvirus Suídeo 1/fisiologia , Interações Hospedeiro-Patógeno , Testes de Neutralização , Filogenia , Filogeografia , Pseudorraiva/sangue , Pseudorraiva/epidemiologia , Alinhamento de Sequência , Software , Sus scrofa/virologia , Suínos/virologia , Replicação Viral
16.
Virus Res ; 213: 274-282, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26756577

RESUMO

We describe a simple and efficient method to obtain recombinant pseudorabies virus (PRV) in mammalian cells by using the PRV BACs, PBAC80 deficient in pac sequences and PBAC90 deficient in the IE180 gene. These essential viral sequences were used as targets to obtain viable recombinant viruses. PBAC80 was constructed, confirmed to encode a copy of the IE180 gene regulated by the inducible Ptet promoter, and used to obtain recombinant attenuated PRV viruses that express the EGFP protein (PRV-BT80GF virus). PBAC90 was used to obtain the vBAC90D virus, deficient in IE180 and free of replication-competent revertants, and which can be used as a helper in the production of PRV amplicons.


Assuntos
Cromossomos Artificiais Bacterianos , DNA Viral/genética , Herpesvirus Suídeo 1/genética , Recombinação Genética , Animais , Linhagem Celular , Genes Essenciais , Genes Virais , Vírus Auxiliares/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Humanos , Virulência
17.
J Virol Methods ; 228: 10-5, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26562056

RESUMO

Only very few comparative studies have been performed that evaluate general trends of virus growth under 3D in comparison with 2D cell culture conditions. The aim of this study was to investigate differences when four animal viruses are cultured in 2D and 3D. Suid herpesvirus 1 (SuHV-1), Vesicular stomatitis virus (VSIV), Bovine adenovirus (BAdV) and Bovine parainfluenza 3 virus (BPIV-3) were cultivated in 3D rotating wall vessels (RWVs) and conventional 2D cultures. The production of virus particles, the portion of infectious particles, and the infectious growth curves were compared. For all viruses, the production of virus particles (related to cell density), including the non-infectious ones, was lower in 3D than in 2D culture. The production of only infectious particles was significantly lower in BAdV and BPIV-3 in 3D cultures in relation to cell density. The two cultivation approaches resulted in significantly different virus particle-to-TCID50 ratios in three of the four viruses: lower in SuHV-1 and BPIV-3 and higher in BAdV in 3D culture. The infectious virus growth rates were not significantly different in all viruses. Although 3D RWV culture resulted in lower production of virus particles compared to 2D systems, the portion of infectious particles was higher for some viruses.


Assuntos
Atadenovirus/crescimento & desenvolvimento , Técnicas de Cultura de Células , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Bovina/crescimento & desenvolvimento , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Cultura de Vírus/métodos , Animais , Atadenovirus/fisiologia , Atadenovirus/ultraestrutura , Bovinos , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Chlorocebus aethiops , Cães , Herpesvirus Suídeo 1/fisiologia , Herpesvirus Suídeo 1/ultraestrutura , Células Madin Darby de Rim Canino , Vírus da Parainfluenza 3 Bovina/fisiologia , Vírus da Parainfluenza 3 Bovina/ultraestrutura , Suínos , Células Vero , Vírus da Estomatite Vesicular Indiana/fisiologia , Vírus da Estomatite Vesicular Indiana/ultraestrutura , Replicação Viral
18.
J Gen Virol ; 97(2): 487-495, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26589961

RESUMO

Although pseudorabies virus (PRV) has been eradicated in domestic swine in many countries, its presence in wild boars remains a threat for a reintroduction into the currently unprotected swine population. To assess the possible impact of such a reintroduction in a naive herd, an in vivo infection study using two genetically characterized wild boar PRV isolates (BEL24043 and BEL20075) representative for wild boar strains circulating in south-western and central Europe and the virulent NIA3 reference strain was performed in 2- and 15-week-old domestic pigs. Our study revealed an attenuated nature of both wild boar strains in 15-week-old pigs. In contrast, it showed the capacity of strain BEL24043 to induce severe clinical symptoms and mortality in young piglets, thereby confirming that the known age dependency of disease outcome after PRV infection also holds for wild boar isolates. Despite the absence of clinical disease in 15-week-old sows, both wild boar PRV strains were able to induce seroconversion, but to a different extent. Importantly, differences in infection and transmission capacity of both strains were observed in 15-week-old sows. Strain BEL24043 induced a more prolonged and disseminated infection than strain BEL20075 and was able to spread efficiently to contact animals, indicative of its capacity to induce a sustained infection. In conclusion, it was shown that a reintroduction of a wild boar isolate into the domestic swine population could have serious economic consequences due to the induction of clinical symptoms in piglets and by jeopardizing the PRV-negative status.


Assuntos
Herpesvirus Suídeo 1/crescimento & desenvolvimento , Pseudorraiva/patologia , Pseudorraiva/virologia , Sus scrofa/virologia , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Animais , Modelos Animais de Doenças , Transmissão de Doença Infecciosa , Europa (Continente) , Herpesvirus Suídeo 1/isolamento & purificação , Herpesvirus Suídeo 1/patogenicidade , Pseudorraiva/transmissão , Suínos , Doenças dos Suínos/transmissão , Resultado do Tratamento
19.
Vaccine ; 33(43): 5733-5740, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26428456

RESUMO

One of the distinct features of the emerging Chinese pseudorabies virus (PRV) variant is its ability to cause severe neurological signs and high mortality in growing pigs in Bartha-K61-vaccinated pig farms. Either single- or multiple-gene-deleted live vaccine candidates have been developed; however, none was evaluated thoroughly in growing pigs. Here, we generated rSMXΔgI/gEΔTK, an attenuated PRV variant with defects in TK, gI and gE genes. The growth kinetics of the attenuated virus was similar to the wild type (wt) strain. It was safe for 1-day-old piglets. Twenty one-day-old weaned pigs were immunized intramuscularly either with 10(6.0) TCID50 of rSMXΔgI/gEΔTK or one dose of commercial Bartha-K61 vaccine, or with DMEM, and were challenged intranasally with 10(7.0) TCID50 wt virus at 28 days post vaccination. rSMXΔgI/gEΔTK elicited higher level neutralization antibody against both PRV variant SMX and Bartha-K61 strain, while Bartha-K61 vaccine elicited lower neutralization activity of antibody against SMX. After challenge, all pigs in rSMXΔgI/gEΔTK group survived without any clinical signs, while unvaccinated group showed 100% mortality, and Bartha-K61 group showed severe respiratory symptoms and 3 out of 5 pigs exhibited severe neurological signs. Pigs in rSMXΔgI/gEΔTK group gained significantly higher body weight and diminished viral excretion titer and period, compared with Bartha-K61 group. Furthermore, the safety and efficacy of rSMXΔgI/gEΔTK was also evaluated in sheep and compared with local vaccine in growing pigs. These data suggest that the attenuated strain rSMXΔgI/gEΔTK is a promising live marker vaccine candidate for PR control in the context of emerging PRV variants.


Assuntos
Deleção de Genes , Herpesvirus Suídeo 1/imunologia , Pseudorraiva/prevenção & controle , Doenças dos Suínos/prevenção & controle , Proteínas Virais/genética , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Peso Corporal , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Injeções Intramusculares , Pseudorraiva/imunologia , Pseudorraiva/patologia , Ovinos , Análise de Sobrevida , Suínos , Doenças dos Suínos/imunologia , Resultado do Tratamento , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Vacinas Marcadoras/administração & dosagem , Vacinas Marcadoras/efeitos adversos , Vacinas Marcadoras/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos
20.
mBio ; 6(2)2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25805728

RESUMO

UNLABELLED: Infection by alphaherpesviruses invariably results in invasion of the peripheral nervous system (PNS) and establishment of either a latent or productive infection. Infection begins with long-distance retrograde transport of viral capsids and tegument proteins in axons toward the neuronal nuclei. Initial steps of axonal entry, retrograde transport, and replication in neuronal nuclei are poorly understood. To better understand how the mode of infection in the PNS is determined, we utilized a compartmented neuron culturing system where distal axons of PNS neurons are physically separated from cell bodies. We infected isolated axons with fluorescent-protein-tagged pseudorabies virus (PRV) particles and monitored viral entry and transport in axons and replication in cell bodies during low and high multiplicities of infection (MOIs of 0.01 to 100). We found a threshold for efficient retrograde transport in axons between MOIs of 1 and 10 and a threshold for productive infection in the neuronal cell bodies between MOIs of 1 and 0.1. Below an MOI of 0.1, the viral genomes that moved to neuronal nuclei were silenced. These genomes can be reactivated after superinfection by a nonreplicating virus, but not by a replicating virus. We further showed that viral particles at high-MOI infections compete for axonal proteins and that this competition determines the number of viral particles reaching the nuclei. Using mass spectrometry, we identified axonal proteins that are differentially regulated by PRV infection. Our results demonstrate the impact of the multiplicity of infection and the axonal milieu on the establishment of neuronal infection initiated from axons. IMPORTANCE: Alphaherpesvirus genomes may remain silent in peripheral nervous system (PNS) neurons for the lives of their hosts. These genomes occasionally reactivate to produce infectious virus that can reinfect peripheral tissues and spread to other hosts. Here, we use a neuronal culture system to investigate the outcome of axonal infection using different numbers of viral particles and coinfection assays. We found that the dynamics of viral entry, transport, and replication change dramatically depending on the number of virus particles that infect axons. We demonstrate that viral genomes are silenced when the infecting particle number is low and that these genomes can be reactivated by superinfection with UV-inactivated virus, but not with replicating virus. We further show that viral invasion rapidly changes the profiles of axonal proteins and that some of these axonal proteins are rate limiting for efficient infection. Our study provides new insights into the establishment of silent versus productive alphaherpesvirus infections in the PNS.


Assuntos
Axônios/química , Axônios/virologia , Núcleo Celular/virologia , Herpesvirus Suídeo 1/fisiologia , Internalização do Vírus , Latência Viral , Replicação Viral , Animais , Células Cultivadas , Herpesvirus Suídeo 1/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Proteínas/análise , Ratos Sprague-Dawley , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA