Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vet Microbiol ; 259: 109154, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34237497

RESUMO

Parainfluenza virus 5 (PIV5) belongs to the genus Orthorubulavirus in the family Paramyxoviridae. PIV5 can infect a range of mammals, but induce mild or even unobservable clinical signs in some animals, except kennel cough in dogs. It is also able to infect a variety of cell lines, but causes minimal or even invisible cytopathic effects on many cells. Sometimes, owing to neither observable cytopathic effects in vitro nor typical clinical signs in vivo, the PIV5 is not easily usable for screening antiviral drugs. To solve this issue, we used reverse genetics to recover a dual reporter-tagged recombinant PIV5 that could simultaneously express enhanced green fluorescence protein (eGFP) and NanoLuc® luciferase (NLuc) in virus-infected cells. Both reporters were genetically stable during twenty serial passages of virus in MDBK cells. The eGFP allowed us to observe virus-infected MDBK cells in real time, and moreover the NLuc made it possible to quantify the degree of viral replication for determining antiviral activity of a given drug. Subsequently, the recombinant PIV5 was used for antiviral assays on five common drugs, i.e., ribavirin, apigenin, 1-adamantylamine hydrochloride, moroxydine hydrochloride and tea polyphenol. The results showed that only the ribavirin had an anti-PIV5 effect in MDBK cells. This study proposed a novel method for rapid screening (or prescreening) of anti-PIV5 drugs.


Assuntos
Antivirais/farmacologia , Genes Reporter , Ensaios de Triagem em Larga Escala/métodos , Vírus da Parainfluenza 5/efeitos dos fármacos , Vírus da Parainfluenza 5/metabolismo , Linhagem Celular , Efeito Citopatogênico Viral , Proteínas de Fluorescência Verde/metabolismo , Replicação Viral/efeitos dos fármacos
2.
Mol Ther ; 29(3): 1174-1185, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33352107

RESUMO

Self-amplifying RNA (saRNA) is a cutting-edge platform for both nucleic acid vaccines and therapeutics. saRNA is self-adjuvanting, as it activates types I and III interferon (IFN), which enhances the immunogenicity of RNA vaccines but can also lead to inhibition of translation. In this study, we screened a library of saRNA constructs with cis-encoded innate inhibiting proteins (IIPs) and determined the effect on protein expression and immunogenicity. We observed that the PIV-5 V and Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a proteins enhance protein expression 100- to 500-fold in vitro in IFN-competent HeLa and MRC5 cells. We found that the MERS-CoV ORF4a protein partially abates dose nonlinearity in vivo, and that ruxolitinib, a potent Janus kinase (JAK)/signal transducer and activator of transcription (STAT) inhibitor, but not the IIPs, enhances protein expression of saRNA in vivo. Both the PIV-5 V and MERS-CoV ORF4a proteins were found to enhance the percentage of resident cells in human skin explants expressing saRNA and completely rescued dose nonlinearity of saRNA. Finally, we observed that the MERS-CoV ORF4a increased the rabies virus (RABV)-specific immunoglobulin G (IgG) titer and neutralization half-maximal inhibitory concentration (IC50) by ∼10-fold in rabbits, but not in mice or rats. These experiments provide a proof of concept that IIPs can be directly encoded into saRNA vectors and effectively abate the nonlinear dose dependency and enhance immunogenicity.


Assuntos
Imunidade Inata/efeitos dos fármacos , Imunogenicidade da Vacina , Biossíntese de Proteínas/efeitos dos fármacos , Vacinas Sintéticas/farmacologia , Proteínas do Envelope Viral/administração & dosagem , Animais , Linhagem Celular , Vírus da Encefalite Equina Venezuelana/efeitos dos fármacos , Vírus da Encefalite Equina Venezuelana/imunologia , Vírus da Encefalite Equina Venezuelana/patogenicidade , Fibroblastos , Regulação da Expressão Gênica , Células HeLa , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoglobulina G/biossíntese , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/genética , Janus Quinases/imunologia , Camundongos , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/patogenicidade , NF-kappa B/genética , NF-kappa B/imunologia , Nitrilas , Vírus da Parainfluenza 5/efeitos dos fármacos , Vírus da Parainfluenza 5/imunologia , Vírus da Parainfluenza 5/patogenicidade , Pirazóis/farmacologia , Pirimidinas , Coelhos , Vírus da Raiva/efeitos dos fármacos , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Ratos , Fatores de Transcrição STAT/antagonistas & inibidores , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Vacinas Sintéticas/biossíntese , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas de mRNA
3.
Med Mol Morphol ; 50(2): 103-111, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28283804

RESUMO

It remains unclear why gastric disease does not develop in all cases of Helicobacter pylori infection. In this study, we analyzed whether simian virus 5 (SV5) enhanced adherence of H. pylori to adenocarcinoma epithelial cells (AGS). H. pylori in AGS (harboring SV5) and SV5-infected Vero cells, and an agglutination of H. pylori mixed with SV5 were observed by light microscopy, scanning and transmission electron microscopies. The adherent rate of H. pylori to SV5-infected Vero cells and treated with an anti-SV5 antibody was determined. H. pylori adhered to the surface of AGS cells near SV5 particles, as shown by scanning and transmission electron microscopies. The adherence of H. pylori to SV5-infected Vero cells was significantly enhanced compared with that to Vero cells. In contrast, the adherence of H. pylori to Vero cells was decreased by treatment with the anti-SV5 antibody. Agglutination of H. pylori mixed with SV5 was observed by scanning and transmission electron microscopies. Agglutination did not occur when SV5 was treated with the anti-SV5 antibody before mixing. These findings demonstrated that SV5 enhanced the adherence of H. pylori to host cells, suggesting that a persistently infected virus may be a factor enhancing the pathogenicity of H. pylori in humans.


Assuntos
Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/farmacologia , Células Epiteliais/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Interações Microbianas , Vírus da Parainfluenza 5/efeitos dos fármacos , Aglutinação/efeitos dos fármacos , Animais , Aderência Bacteriana/efeitos dos fármacos , Linhagem Celular Tumoral , Chlorocebus aethiops , Células Epiteliais/microbiologia , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/patogenicidade , Helicobacter pylori/ultraestrutura , Humanos , Vírus da Parainfluenza 5/crescimento & desenvolvimento , Células Vero
4.
PLoS One ; 7(11): e50144, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23185558

RESUMO

Parainfluenza virus 5 (PIV5), formerly known as simian virus 5 (SV5), is a paramyxovirus often referred to as canine parainfluenza virus (CPI) in the veterinary field. PIV5 is thought to be a contributing factor to kennel cough. Kennel cough vaccines containing live PIV5 have been used in dogs for many decades. PIV5 is not known to cause any diseases in humans or other animals. PIV5 has been used as a vector for vaccine development for humans and animals. One critical question concerning the use of PIV5 as a vector is whether prior exposure to PIV5 would prevent the use of PIV5-based vaccines. In this work, we have examined immunogenicity of a recombinant PIV5 expressing hemagglutinin (HA) of influenza A virus subtype 3 (rPIV5-H3) in dogs that were immunized against PIV5. We found that vaccination of the dogs containing neutralizing antibodies against PIV5 with rPIV5-H3 generated immunity against influenza A virus, indicting that PIV5-based vaccine is immunogenic in dogs with prior exposure. Furthermore, we have examined exposure of PIV5 in human populations. We have detected neutralizing antibody (nAb) against PIV5 in 13 out of 45 human serum samples (about 29 percent). The nAb titers in humans were lower than that in vaccinated dogs, suggesting that nAb in humans is unlikely to prevent PIV5 from being an efficacious vector in humans.


Assuntos
Anticorpos Antivirais/sangue , Doenças do Cão/prevenção & controle , Imunização Secundária , Vírus da Parainfluenza 5/imunologia , Infecções por Rubulavirus/prevenção & controle , Infecções por Rubulavirus/veterinária , Vacinação , Animais , Anticorpos Neutralizantes , Linhagem Celular , Doenças do Cão/imunologia , Doenças do Cão/virologia , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Vírus da Influenza A/química , Vírus da Parainfluenza 5/efeitos dos fármacos , Infecções por Rubulavirus/imunologia , Infecções por Rubulavirus/virologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
5.
Virology ; 335(1): 131-44, 2005 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-15823612

RESUMO

A paramyxovirus SV5 mutant (rSV5-P/V-CPI-) that encodes 6 naturally-occurring P/V gene substitutions is a potent inducer of type I interferon (IFN) and is restricted for low moi growth, two phenotypes not seen with WT SV5. In this study, we have compared the IFN sensitivity of WT SV5 and the rSV5-P/V-CPI- mutant in tumor cell lines and in cultures of normal primary cells. We have tested the hypothesis that differences in IFN induction elicited by WT rSV5 and rSV5-P/V-CPI- are responsible for differences in low moi growth and spread. In contrast to WT SV5, low moi infection of A549 lung carcinoma cells with rSV5-P/V-CPI- resulted in a plateau of virus production by 24-48 h pi when secreted IFN levels were between approximately 100 and 1000 U/ml. Gene microarray and RT-PCR analyses identified IFN genes and IFN-stimulated genes whose expression were increased by infection of A549 cells with WT and P/V mutant viruses. Restricted low moi growth and spread of rSV5-P/V-CPI- in A549 cells was relieved in the presence of neutralizing antibodies to IFN-beta but not TNF-alpha. When A549 or MDA-MB-435 breast tumor cells were pretreated with IFN, both WT and P/V mutant viruses showed delayed spread and approximately 10-fold reduction in virus yield, but infections were not eliminated. Using normal primary human epithelial cells that have undergone limited passage in culture, WT rSV5 and rSV5-P/V-CPI- displayed high moi growth properties that were similar to that seen in A549 cells. However, IFN pretreatment of these primary cells as well as normal human lung cells eliminated low moi spread of both mutant and WT rSV5 infections. Together, these data demonstrate that SV5 growth in normal primary human cells is highly sensitive to IFN compared to growth in some tumor cell lines, regardless of whether the P/V gene is WT or mutant. These results suggest a model in which spread of WT SV5 in normal human cells is dependent on the ability of the virus to prevent IFN synthesis. The implications of these results for the use of recombinant paramyxoviruses as vectors are discussed.


Assuntos
Interferon Tipo I/farmacologia , Mutação , Vírus da Parainfluenza 5/crescimento & desenvolvimento , Fosfoproteínas/genética , Recombinação Genética , Proteínas Virais/genética , Proteínas Estruturais Virais/genética , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Interferon Tipo I/metabolismo , Vírus da Parainfluenza 5/efeitos dos fármacos , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/patogenicidade , Proteínas de Ligação a RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...