Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 98(3): e0112923, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38305155

RESUMO

The global circulation of clade 2.3.4.4b H5Ny highly pathogenic avian influenza viruses (HPAIVs) in poultry and wild birds, increasing mammal infections, continues to pose a public health threat and may even form a pandemic. An efficacious vaccine against H5Ny HPAIVs is crucial for emergency use and pandemic preparedness. In this study, we developed a parainfluenza virus 5 (PIV5)-based vaccine candidate expressing hemagglutinin (HA) protein of clade 2.3.4.4b H5 HPAIV, termed rPIV5-H5, and evaluated its safety and efficacy in mice and ferrets. Our results demonstrated that intranasal immunization with a single dose of rPIV5-H5 could stimulate H5-specific antibody responses, moreover, a prime-boost regimen using rPIV5-H5 stimulated robust humoral, cellular, and mucosal immune responses in mice. Challenge study showed that rPIV5-H5 prime-boost regimen provided sterile immunity against lethal clade 2.3.4.4b H5N1 virus infection in mice and ferrets. Notably, rPIV5-H5 prime-boost regimen provided protection in mice against challenge with lethal doses of heterologous clades 2.2, 2.3.2, and 2.3.4 H5N1, and clade 2.3.4.4h H5N6 viruses. These results revealed that rPIV5-H5 can elicit protective immunity against a diverse clade of highly pathogenic H5Ny virus infection in mammals, highlighting the potential of rPIV5-H5 as a pan-H5 influenza vaccine candidate for emergency use.IMPORTANCEClade 2.3.4.4b H5Ny highly pathogenic avian influenza viruses (HPAIVs) have been widely circulating in wild birds and domestic poultry all over the world, leading to infections in mammals, including humans. Here, we developed a recombinant PIV5-vectored vaccine candidate expressing the HA protein of clade 2.3.4.4b H5 virus. Intranasal immunization with rPIV5-H5 in mice induced airway mucosal IgA responses, high levels of antibodies, and robust T-cell responses. Importantly, rPIV5-H5 conferred complete protection in mice and ferrets against clade 2.3.4.4b H5N1 virus challenge, the protective immunity was extended against heterologous H5Ny viruses. Taken together, our data demonstrate that rPIV5-H5 is a promising vaccine candidate against diverse H5Ny influenza viruses in mammals.


Assuntos
Virus da Influenza A Subtipo H5N1 , Virus da Influenza A Subtipo H5N6 , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Vírus da Parainfluenza 5 , Animais , Humanos , Camundongos , Furões/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunidade Celular , Imunidade Humoral , Imunidade nas Mucosas , Virus da Influenza A Subtipo H5N1/química , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N6/química , Virus da Influenza A Subtipo H5N6/classificação , Virus da Influenza A Subtipo H5N6/genética , Virus da Influenza A Subtipo H5N6/imunologia , Influenza Aviária/imunologia , Influenza Aviária/prevenção & controle , Influenza Aviária/transmissão , Influenza Aviária/virologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Vacinas contra Influenza/genética , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/virologia , Preparação para Pandemia/métodos , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/imunologia , Vírus da Parainfluenza 5/metabolismo , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Administração Intranasal , Aves Domésticas/virologia , Imunoglobulina A/imunologia , Linfócitos T/imunologia
2.
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
3.
Arch Virol ; 165(4): 799-807, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32100137

RESUMO

We previously found that infection with human parainfluenza virus type 2 (hPIV-2), a member of the genus Orthorubulavirus, family Paramyxoviridae, causes filamentous actin (F-actin) formation to promote viral growth. In the present study, we investigated whether similar regulation of F-actin formation is observed in infections with other rubulaviruses, such as parainfluenza virus type 5 (PIV-5) and simian virus 41 (SV41). Infection with these viruses caused F-actin formation and RhoA activation, which promoted viral growth. These results indicate that RhoA-induced F-actin formation is important for efficient growth of these rubulaviruses. Only SV41 and hPIV-2 V and P proteins bound to Graf1, while the V and P proteins of PIV-5, mumps virus, and hPIV-4 did not bind to Graf1. In contrast, the V proteins of these rubulaviruses bound to both inactive RhoA and profilin 2. These results suggest that there are common and unique mechanisms involved in regulation of F-actin formation by members of the genus Orthorubulavirus.


Assuntos
Actinas/metabolismo , Vírus da Parainfluenza 2 Humana/metabolismo , Vírus da Parainfluenza 5/metabolismo , Infecções por Rubulavirus/metabolismo , Rubulavirus/metabolismo , Actinas/química , Actinas/genética , Animais , Linhagem Celular , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 2 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/crescimento & desenvolvimento , Ligação Proteica , Rubulavirus/genética , Rubulavirus/crescimento & desenvolvimento , Infecções por Rubulavirus/genética , Infecções por Rubulavirus/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Int J Mol Sci ; 21(2)2020 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-31941046

RESUMO

The etiology of viral encephalitis in cattle often remains unresolved, posing a potential risk for animal and human health. In metagenomics studies of cattle with bovine non-suppurative encephalitis, parainfluenza virus 5 (PIV5) was identified in three brain samples. Interestingly, in two of these animals, bovine herpesvirus 6 and bovine astrovirus CH13 were additionally found. We investigated the role of PIV5 in bovine non-suppurative encephalitis and further characterized the three cases. With traditional sequencing methods, we completed the three PIV5 genomes, which were compared to one another. However, in comparison to already described PIV5 strains, unique features were revealed, like an 81 nucleotide longer open reading frame encoding the small hydrophobic (SH) protein. With in situ techniques, we demonstrated PIV5 antigen and RNA in one animal and found a broad cell tropism of PIV5 in the brain. Comparative quantitative analyses revealed a high viral load of PIV5 in the in situ positive animal and therefore, we propose that PIV5 was probably the cause of the disease. With this study, we clearly show that PIV5 is capable of naturally infecting different brain cell types in cattle in vivo and therefore it is a probable cause of encephalitis and neurological disease in cattle.


Assuntos
Antígenos Virais , Doenças dos Bovinos , Encefalite Viral , Genoma Viral , Vírus da Parainfluenza 5 , RNA Viral , Infecções por Rubulavirus , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Bovinos , Doenças dos Bovinos/genética , Doenças dos Bovinos/metabolismo , Doenças dos Bovinos/virologia , Encefalite Viral/genética , Encefalite Viral/metabolismo , Encefalite Viral/virologia , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Infecções por Rubulavirus/genética , Infecções por Rubulavirus/metabolismo
5.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31217248

RESUMO

Enveloped viruses utilize surface glycoproteins to bind and fuse with a target cell membrane. The zoonotic Hendra virus (HeV), a member of the family Paramyxoviridae, utilizes the attachment protein (G) and the fusion protein (F) to perform these critical functions. Upon triggering, the trimeric F protein undergoes a large, irreversible conformation change to drive membrane fusion. Previously, we have shown that the transmembrane (TM) domain of the F protein, separate from the rest of the protein, is present in a monomer-trimer equilibrium. This TM-TM association contributes to the stability of the prefusion form of the protein, supporting a role for TM-TM interactions in the control of F protein conformational changes. To determine the impact of disrupting TM-TM interactions, constructs expressing the HeV F TM with limited flanking sequences were synthesized. Coexpression of these constructs with HeV F resulted in dramatic reductions in the stability of F protein expression and fusion activity. In contrast, no effects were observed when the HeV F TM constructs were coexpressed with the nonhomologous parainfluenza virus 5 (PIV5) fusion protein, indicating a requirement for specific interactions. To further examine this, a TM peptide homologous to the PIV5 F TM domain was synthesized. Addition of the peptide prior to infection inhibited infection with PIV5 but did not significantly affect infection with human metapneumovirus, a related virus. These results indicate that targeted disruption of TM-TM interactions significantly impact viral fusion protein stability and function, presenting these interactions as a novel target for antiviral development.IMPORTANCE Enveloped viruses require virus-cell membrane fusion to release the viral genome and replicate. The viral fusion protein triggers from the pre- to the postfusion conformation, an essentially irreversible change, to drive membrane fusion. We found that small proteins containing the TM and a limited flanking region homologous to the fusion protein of the zoonotic Hendra virus reduced protein expression and fusion activity. The introduction of exogenous TM peptides may displace a TM domain, disrupting native TM-TM interactions and globally destabilizing the fusion protein. Supporting this hypothesis, we showed that a sequence-specific transmembrane peptide dramatically reduced viral infection in another enveloped virus model, suggesting a broader inhibitory mechanism. Viral fusion protein TM-TM interactions are important for protein function, and disruption of these interactions dramatically reduces protein stability.


Assuntos
Paramyxovirinae/metabolismo , Peptídeos/farmacologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Animais , Sítios de Ligação/efeitos dos fármacos , Chlorocebus aethiops , Vírus Hendra/química , Vírus Hendra/genética , Vírus Hendra/metabolismo , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , Paramyxovirinae/química , Paramyxovirinae/genética , Conformação Proteica/efeitos dos fármacos , Domínios Proteicos/efeitos dos fármacos , Estabilidade Proteica , Células Vero , Proteínas Virais de Fusão/efeitos dos fármacos
6.
Vaccine ; 37(22): 2925-2934, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31010715

RESUMO

Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in children under one year of age. In addition to causing severe respiratory diseases in children, it is also a major cause of morbidity and mortality among the elderly and immunocompromised individuals. RSV is the most common cause of lower respiratory tract infections, yet there are currently no licensed vaccines. A parainfluenza virus 5 (PIV5)-based amplifying virus-like particle (AVLP), which enables the use of PIV5 RNA transcription/replication machinery to express gene of interest, has recently been developed. We evaluated the PIV5-based AVLP system as a vaccine platform for RSV by incorporating the fusion protein (F) gene and the transcription factor protein (M2-1) gene of RSV into the PIV5-AVLP backbone (AVLP-F and AVLP-M2-1, respectively). Mice immunized with a single dose of the AVLP-F or AVLP-M2-1 developed RSV-F or RSV-M2-1-specific immune responses, respectively. Both vaccine candidates elicited antigen-specific cell-mediated responses at levels comparable to or higher than an RSV infection. Most importantly, each vaccine was able to induce protection against RSV A2 challenge in the mouse model. These results indicate the potential of the PIV5-based AVLP system as a platform for vaccines against RSV infection.


Assuntos
Antígenos Virais/imunologia , Vírus da Parainfluenza 5/metabolismo , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Antígenos Virais/genética , Western Blotting , Ensaio de Imunoadsorção Enzimática , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Vírus da Parainfluenza 5/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/genética
7.
Viruses ; 10(5)2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29693588

RESUMO

The complement system is a part of the innate immune system that viruses need to face during infections. Many viruses incorporate cellular regulators of complement activation (RCA) to block complement pathways and our prior work has shown that Parainfluenza virus 5 (PIV5) incorporates CD55 and CD46 to delay complement-mediated neutralization. In this paper, we tested the role of a third individual RCA inhibitor CD59 in PIV5 interactions with complement pathways. Using a cell line engineered to express CD59, we show that small levels of functional CD59 are associated with progeny PIV5, which is capable of blocking assembly of the C5b-C9 membrane attack complex (MAC). PIV5 containing CD59 (PIV5-CD59) showed increased resistance to complement-mediated neutralization in vitro comparing to PIV5 lacking regulators. Infection of A549 cells with PIV5 and RSV upregulated CD59 expression. TGF-beta treatment of PIV5-infected cells also increased cell surface CD59 expression and progeny virions were more resistant to complement-mediated neutralization. A comparison of individual viruses containing only CD55, CD46, or CD59 showed a potency of inhibiting complement-mediated neutralization, which followed a pattern of CD55 > CD46 > CD59.


Assuntos
Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Ativação do Complemento/imunologia , Complemento C9/metabolismo , Inativadores do Complemento/metabolismo , Proteína Cofatora de Membrana/metabolismo , Vírus da Parainfluenza 5/imunologia , Animais , Antígenos CD59/genética , Bovinos , Linhagem Celular , Membrana Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Testes de Neutralização , Vírus da Parainfluenza 5/metabolismo , Vírion/metabolismo
8.
J Mol Biol ; 430(5): 695-709, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29330069

RESUMO

Enveloped viruses enter cells by using their fusion proteins to merge the virus lipid envelope and the cell membrane. While crystal structures of the water-soluble ectodomains of many viral fusion proteins have been determined, the structure and assembly of the C-terminal transmembrane domain (TMD) remains poorly understood. Here we use solid-state NMR to determine the backbone conformation and oligomeric structure of the TMD of the parainfluenza virus 5 fusion protein. 13C chemical shifts indicate that the central leucine-rich segment of the TMD is α-helical in POPC/cholesterol membranes and POPE membranes, while the Ile- and Val-rich termini shift to the ß-strand conformation in the POPE membrane. Importantly, lipid mixing assays indicate that the TMD is more fusogenic in the POPE membrane than in the POPC/cholesterol membrane, indicating that the ß-strand conformation is important for fusion by inducing membrane curvature. Incorporation of para-fluorinated Phe at three positions of the α-helical core allowed us to measure interhelical distances using 19F spin diffusion NMR. The data indicate that, at peptide:lipid molar ratios of ~1:15, the TMD forms a trimeric helical bundle with inter-helical distances of 8.2-8.4Å for L493F and L504F and 10.5Å for L500F. These data provide high-resolution evidence of trimer formation of a viral fusion protein TMD in phospholipid bilayers, and indicate that the parainfluenza virus 5 fusion protein TMD harbors two functions: the central α-helical core is the trimerization unit of the protein, while the two termini are responsible for inducing membrane curvature by transitioning to a ß-sheet conformation.


Assuntos
Bicamadas Lipídicas/química , Vírus da Parainfluenza 5/química , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Membrana Celular/química , Colesterol/química , Simulação por Computador , Bicamadas Lipídicas/metabolismo , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular/métodos , Vírus da Parainfluenza 5/metabolismo , Peptídeos/química , Fosfatidilcolinas/química , Conformação Proteica , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Espalhamento a Baixo Ângulo , Proteínas Virais de Fusão/metabolismo
9.
Proc Natl Acad Sci U S A ; 113(27): E3844-51, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27335462

RESUMO

Parainfluenza virus 5 (PIV5) is an enveloped, single-stranded, negative-sense RNA virus of the Paramyxoviridae family. PIV5 fusion and entry are mediated by the coordinated action of the receptor-binding protein, hemagglutinin-neuraminidase (HN), and the fusion protein (F). Upon triggering by HN, F undergoes an irreversible ATP- and pH-independent conformational change, going down an energy gradient from a metastable prefusion state to a highly stable postfusion state. Previous studies have highlighted key conformational changes in the F-protein refolding pathway, but a detailed understanding of prefusion F-protein metastability remains elusive. Here, using two previously described F-protein mutations (S443D or P22L), we examine the capacity to modulate PIV5 F stability and the mechanisms by which these point mutants act. The S443D mutation destabilizes prefusion F proteins by disrupting a hydrogen bond network at the base of the F-protein globular head. The introduction of a P22L mutation robustly rescues destabilized F proteins through a local hydrophobic interaction between the N-terminal helix and a hydrophobic pocket. Prefusion stabilization conferred by a P22L-homologous mutation is demonstrated in the F protein of Newcastle disease virus, a paramyxovirus of a different genus, suggesting a conserved stabilizing structural element within the paramyxovirus family. Taken together, the available data suggest that movement of the N-terminal helix is a necessary early step for paramyxovirus F-protein refolding and presents a novel target for structure-based drug design.


Assuntos
Vírus da Parainfluenza 5/metabolismo , Proteínas Virais de Fusão/metabolismo , Substituição de Aminoácidos , Animais , Chlorocebus aethiops , Conformação Molecular , Mutação , Estabilidade Proteica , Células Vero
10.
J Virol ; 89(6): 3438-41, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25589638

RESUMO

The crystal structure of the F protein (prefusion form) of the paramyxovirus parainfluenza virus 5 (PIV5) WR isolate was determined. We investigated the basis by which point mutations affect fusion in PIV5 isolates W3A and WR, which differ by two residues in the F ectodomain. The P22 stabilizing site acts through a local conformational change and a hydrophobic pocket interaction, whereas the S443 destabilizing site appears sensitive to both conformational effects and amino acid charge/polarity changes.


Assuntos
Vírus da Parainfluenza 5/metabolismo , Proteínas Virais de Fusão/química , Motivos de Aminoácidos , Cristalografia por Raios X , Humanos , Mutação , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/genética , Estabilidade Proteica , Estrutura Terciária de Proteína , Infecções por Rubulavirus/virologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
11.
J Virol ; 89(6): 3427-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25552723

RESUMO

Rabies remains a major public health threat around the world. Once symptoms appear, there is no effective treatment to prevent death. In this work, we tested a recombinant parainfluenza virus 5 (PIV5) strain expressing the glycoprotein (G) of rabies (PIV5-G) as a therapy for rabies virus infection: we have found that PIV5-G protected mice as late as 6 days after rabies virus infection. PIV5-G is a promising vaccine for prevention and treatment of rabies virus infection.


Assuntos
Vetores Genéticos/genética , Vírus da Parainfluenza 5/genética , Vacina Antirrábica/administração & dosagem , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Proteínas do Envelope Viral/administração & dosagem , Animais , Anticorpos Antivirais/imunologia , Expressão Gênica , Vetores Genéticos/metabolismo , Humanos , Camundongos , Vírus da Parainfluenza 5/metabolismo , Raiva/imunologia , Raiva/virologia , Vacina Antirrábica/genética , Vacina Antirrábica/imunologia , Vírus da Raiva/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
12.
PLoS Pathog ; 5(7): e1000525, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19629176

RESUMO

The paramyxoviruses are a family of negative-sense RNA viruses that includes many important human and animal pathogens. Paramyxovirus RNA synthesis requires the viral phosphoprotein (P) and the large (L) protein. Phosphorylation of P is thought to regulate viral gene expression, though direct proof remains elusive. Recently, we reported that phosphorylation of a specific residue (Ser157) of the P protein of parainfluenza virus 5 (PIV5), a prototypical paramyxovirus, correlates with decreased viral gene expression and cytokine expression in infected cells. Here, we show that: Polo-like kinase 1 (PLK1), a serine/theronine kinase that plays a critical role in regulating the cell cycle, interacts with PIV5 P through the S157 residue; PLK1 inhibition increases viral gene expression; PLK1 over-expression inhibits viral gene expression; and PLK1 directly phosphorylates P in vitro, indicating that PLK1 down-regulates viral gene expression by phosphorylating P. Furthermore, we have determined the PLK1 phosphorylation site on P and found that mutant recombinant PIV5 whose P proteins cannot either bind to or be phosphorylated by PLK1 have similar phenotypes. Increased viral gene expression in PIV5 with mutations in the PLK1 binding/phosphorylation sites correlates with increased induction of cell death and cytokine expression, suggesting that PIV5 limits its viral gene expression to avoid these host effects. It is possible that targeting PLK1 will enhance host innate immune responses, leading to a novel strategy of clearing paramyxovirus infections quickly.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Infecções por Respirovirus/metabolismo , Respirovirus/genética , Motivos de Aminoácidos , Substituição de Aminoácidos , Apoptose/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Citocinas/metabolismo , Regulação para Baixo , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Vírus da Parainfluenza 5/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Pteridinas/farmacologia , Respirovirus/metabolismo , Transfecção , Proteínas Virais/metabolismo , Quinase 1 Polo-Like
13.
J Virol ; 82(19): 9369-80, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18667520

RESUMO

Viral fusogenic membrane proteins have been proposed as tools to increase the potency of oncolytic viruses, but there is a need for mechanisms to control the spread of fusogenic viruses in normal versus tumor cells. We have previously shown that a mutant of the paramyxovirus simian virus 5 (SV5) that harbors mutations in the P/V gene from the canine parainfluenza virus (P/V-CPI(-)) is a potent inducer of type I interferon (IFN) and apoptosis and is restricted for spread through normal but not tumor cells in vitro. Here, we have used the cytopathic P/V-CPI(-) as a backbone vector to test the hypothesis that a virus expressing a hyperfusogenic glycoprotein will be a more effective oncolytic vector but will retain sensitivity to IFN. A P/V mutant virus expressing an F protein with a glycine-to-alanine substitution in the fusion peptide (P/V-CPI(-)-G3A) was more fusogenic than the parental P/V-CPI(-) mutant. In two model prostate tumor cell lines which are defective in IFN production (LNCaP and DU145), the hyperfusogenic P/V-CPI(-)-G3A mutant had normal growth properties at low multiplicities of infection and was more effective than the parental P/V-CPI(-) mutant at cell killing in vitro. However, in PC3 cells which produce and respond to IFN, the hyperfusogenic P/V-CPI(-)-G3A mutant was attenuated for growth and spread. Killing of PC3 cells was equivalent between the parental P/V-CPI(-) mutant and the hyperfusogenic P/V-CPI(-)-G3A mutant. In a nude mouse model using LNCaP cells, the hyperfusogenic P/V-CPI(-)-G3A mutant was more effective than P/V-CPI(-) at reducing tumor burden. In the case of DU145 tumors, the two vectors based on P/V-CPI(-) were equally effective at limiting tumor growth. Together, our results provide proof of principle that a cytopathic SV5 P/V mutant can serve as an oncolytic virus and that the oncolytic effectiveness of P/V mutants can be enhanced by a fusogenic membrane protein without compromising sensitivity to IFN. The potential advantages of SV5-based oncolytic vectors are discussed.


Assuntos
Mutação , Vírus Oncolíticos/genética , Vírus da Parainfluenza 5/genética , Proteínas Estruturais Virais/fisiologia , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/virologia , Chlorocebus aethiops , Humanos , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Masculino , Camundongos , Camundongos Nus , Vírus Oncolíticos/metabolismo , Vírus da Parainfluenza 5/metabolismo , Células Vero , Proteínas Estruturais Virais/química
14.
Virology ; 368(2): 262-72, 2007 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-17692882

RESUMO

The V protein of parainfluenza virus 5 (PIV5) plays an important role in the evasion of host immune responses. The V protein blocks interferon (IFN) signaling in human cells by causing degradation of the STAT1 protein, a key component of IFN signaling, and blocks IFN-beta production by preventing nuclear translocation of IRF3, a key transcription factor for activating IFN-beta promoter. Interleukin-6 (IL-6), along with tumor necrosis factor (TNF)-alpha and IL-1beta, is a major proinflammatory cytokine that plays important roles in clearing virus infection through inflammatory responses. Many viruses have developed strategies to block IL-6 expression. Wild-type PIV5 infection induces little, if any, expression of cytokines such as IL-6 or TNF-alpha, whereas infection by a mutant PIV5 lacking the conserved C-terminal cysteine rich domain (rPIV5VDeltaC) induced high levels of IL-6 expression. Examination of mRNA levels of IL-6 indicated that the transcription activation of IL-6 played an important role in the increased IL-6 expression. Co-infection with wild-type PIV5 prevented the activation of IL-6 transcription by rPIV5VDeltaC, and a plasmid encoding the full-length PIV5 V protein prevented the activation of IL-6 promoter-driven reporter gene expression by rPIV5VDeltaC, indicating that the V protein played a role in inhibiting IL-6 transcription. The activation of IL-6 was independent of IFN-beta even though rPIV5VDeltaC-infected cells produced IFN-beta. Using reporter gene assays and chromatin immunoprecipitation (ChIP), it was found that NF-kappaB played an important role in activating expression of IL-6. We have proposed a model of activating and inhibiting IL-6 transcription by PIV5.


Assuntos
Regulação da Expressão Gênica , Interleucina-6/metabolismo , Vírus da Parainfluenza 5/patogenicidade , Proteínas Estruturais Virais/farmacologia , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Genes Reporter , Células HeLa , Humanos , Interleucina-6/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo
15.
J Virol ; 81(15): 8361-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17494063

RESUMO

Respiratory syncytial virus (RSV), a member of the Paramyxoviridae family, encodes a small hydrophobic (SH) protein of unknown function. Parainfluenza virus 5 (PIV5), a prototypical paramyxovirus, also encodes an SH protein, which inhibits tumor necrosis factor alpha (TNF-alpha) signaling. In this study, recombinant PIV5 viruses without their own SH but containing RSV SH (from RSV strain A2 or B1) in its place (PIV5DeltaSH-RSV SH) and RSV lacking its own SH (RSVDeltaSH) were generated and analyzed. The results indicate that the SH protein of RSV has a function similar to that of PIV5 SH and that it can inhibit TNF-alpha signaling.


Assuntos
Vírus Sinciciais Respiratórios/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Linhagem Celular , Humanos , Dados de Sequência Molecular , NF-kappa B/metabolismo , Vírus da Parainfluenza 5/metabolismo , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética , Alinhamento de Sequência
16.
J Virol ; 79(21): 13434-41, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16227264

RESUMO

The V protein of simian virus 5 (SV5) facilitates the ubiquitination and subsequent proteasome-mediated degradation of STAT1. Here we show, by visualizing direct protein-protein interactions and by using the yeast two-hybrid system, that while the SV5 V protein fails to bind to STAT1 directly, it binds directly and independently to both DDB1 and STAT2, two cellular proteins known to be essential for SV5-mediated degradation of STAT1. We also demonstrate that STAT1 and STAT2 interact independently of SV5 V and show that SV5 V protein acts as an adaptor molecule linking DDB1 to STAT2/STAT1 heterodimers, which in the presence of additional accessory cellular proteins, including Cullin 4a, can ubiquitinate STAT1. Additionally, we show that the avidity of STAT2 for V is relatively weak but is significantly enhanced by the presence of both STAT1 and DDB1, i.e., the complex of STAT1, STAT2, DDB1, and SV5 V is more stable than a complex of STAT2 and V. From these studies we propose a dynamic model in which SV5 V acts as a bridge, bringing together a DDB1/Cullin 4a-containing ubiquitin ligase complex and STAT1/STAT2 heterodimers, which leads to the degradation of STAT1. The loss of STAT1 results in a decrease in affinity of binding of STAT2 for V such that STAT2 either dissociates from V or is displaced from V by STAT1/STAT2 complexes, thereby ensuring the cycling of the DDB1 and SV5 V containing E3 complex for continued rounds of STAT1 ubiquitination and degradation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Vírus da Parainfluenza 5/fisiologia , Transativadores/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Proteínas Culina/metabolismo , Dimerização , Vírus da Parainfluenza 5/metabolismo , Ligação Proteica , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transdução de Sinais , Replicação Viral
17.
J Virol ; 79(3): 1543-51, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15650180

RESUMO

Formation of a six-helix bundle comprised of three C-terminal heptad repeat regions in antiparallel orientation in the grooves of an N-terminal coiled-coil is critical for promotion of membrane fusion by paramyxovirus fusion (F) proteins. We have examined the effect of mutations in four residues of the N-terminal heptad repeat in the simian virus 5 (SV5) F protein on protein folding, transport, and fusogenic activity. The residues chosen have previously been shown from study of isolated peptides to have differing effects on stability of the N-terminal coiled-coil and six-helix bundle (R. E. Dutch, G. P. Leser, and R. A. Lamb, Virology 254:147-159, 1999). The mutant V154M showed reduced proteolytic cleavage and surface expression, indicating a defect in intracellular transport, though this mutation had no effect when studied in isolated peptides. The mutation I137M, previously shown to lower thermostability of the six-helix bundle, resulted in an F protein which was properly processed and transported to the cell surface but which had reduced fusogenic activity. Finally, mutations at L140M and L161M, previously shown to disrupt alpha-helix formation of isolated N-1 peptides but not to affect six-helix bundle formation, resulted in F proteins that were properly processed. Interestingly, the L161M mutant showed increased syncytium formation and promoted fusion at lower temperatures than the wild-type F protein. These results indicate that interactions separate from formation of an N-terminal coiled-coil or six-helix bundle are important in the initial folding and transport of the SV5 F protein and that mutations that destabilize the N-terminal coiled-coil can result in stimulation of membrane fusion.


Assuntos
Fusão de Membrana , Vírus da Parainfluenza 5/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células Gigantes , Células HeLa , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 5/patogenicidade , Conformação Proteica , Dobramento de Proteína , Proteínas Recombinantes de Fusão , Células Vero , Proteínas Virais de Fusão/genética
18.
J Virol ; 78(16): 8513-23, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15280460

RESUMO

SER virus, a paramyxovirus that is closely related to simian virus 5 (SV5), is unusual in that it fails to induce syncytium formation. The SER virus F protein has an unusually long cytoplasmic tail (CT), and it was previously observed that truncations or specific mutations of this domain result in enhanced syncytium formation. In addition to the long CT, the SER F protein has nine amino acid differences from the F protein of SV5. We previously observed only a partial suppression of fusion in a chimeric SV5 F protein with a CT derived from SER virus, indicating that these other amino acid differences between the SER and SV5 F proteins also play a role in regulating the fusion phenotype. To examine the effects of individual amino acid differences, we mutated the nine SER residues individually to the respective residues of the SV5 F protein. We found that most of the mutants were expressed well and were transported to the cell surface at levels comparable to that of the wild-type SER F protein. Many of the mutants showed enhanced lipid mixing, calcein transfer, and syncytium formation even in the presence of the long SER F protein CT. Some mutants, such as the I310 M, T438S, M489I, T516V, and N529K mutants, also showed fusion at lower temperatures of 32, 25, and 18 degrees C. The residue Asn529 plays a critical role in the suppression of fusion activity, as the mutation of this residue to lysine caused a marked enhancement of fusion. The effect of the N529K mutation on the enhancement of fusion by a previously described mutant, L539,548A, as well as by chimeric SV5/SER F proteins was also dramatic. These results indicate that activation to a fusogenic conformation is dependent on the interplay of residues in the ectodomain, the transmembrane domain, and the CT domain of paramyxovirus F proteins.


Assuntos
Fusão de Membrana/fisiologia , Mutação , Paramyxovirinae/patogenicidade , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Fusão Celular , Regulação Viral da Expressão Gênica , Células Gigantes/fisiologia , Cobaias , Células HeLa , Humanos , Dados de Sequência Molecular , Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/metabolismo , Vírus da Parainfluenza 5/patogenicidade , Paramyxovirinae/genética , Paramyxovirinae/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais de Fusão/genética
19.
Cell Immunol ; 231(1-2): 14-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15919365

RESUMO

The increased potency of high avidity CD8+ T cells for the clearance of viral infections has been well documented. We have previously reported the novel finding that intranasal infection with the paramyxovirus SV5 induces a CD8+ T cell response to the SV5 P protein that is almost exclusively of high avidity. Based on our results that the level of peptide presentation is a critical factor in the selective expansion of high versus low avidity cells in vitro, we hypothesized that the avidity of the anti-viral response generated in vivo could be altered by increasing the turnover of the P protein during viral infection through linkage to ubiquitin (UbP). Infection with a virus expressing UbP (VV-UbP) elicited a significant increase in low avidity cells in both BALB/c and C3H mice compared to the almost exclusively high avidity response elicited by VV-P. Our results are the first demonstration of the control of avidity during the antiviral response through an engineered change to a viral antigen. The implications of our findings for vaccine development are discussed.


Assuntos
Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Vírus da Parainfluenza 5/imunologia , Infecções por Rubulavirus/imunologia , Infecções por Rubulavirus/virologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Meia-Vida , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Vírus da Parainfluenza 5/metabolismo , Fosfoproteínas/imunologia , Fosfoproteínas/metabolismo , Ligação Proteica , Proteínas de Ligação a RNA , Infecções por Rubulavirus/metabolismo , Ubiquitina/metabolismo , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA