Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 16(8): e1008775, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32866218

RESUMO

Small RNA viruses only have a very limited coding capacity, thus most viral proteins have evolved to fulfill multiple functions. The highly conserved matrix protein 1 (M1) of influenza A viruses is a prime example for such a multifunctional protein, as it acts as a master regulator of virus replication whose different functions have to be tightly regulated. The underlying mechanisms, however, are still incompletely understood. Increasing evidence points towards an involvement of posttranslational modifications in the spatio-temporal regulation of M1 functions. Here, we analyzed the role of M1 tyrosine phosphorylation in genuine infection by using recombinant viruses expressing M1 phosphomutants. Presence of M1 Y132A led to significantly decreased viral replication compared to wildtype and M1 Y10F. Characterization of phosphorylation dynamics by mass spectrometry revealed the presence of Y132 phosphorylation in M1 incorporated into virions that is most likely mediated by membrane-associated Janus kinases late upon infection. Molecular dynamics simulations unraveled a potential phosphorylation-induced exposure of the positively charged linker domain between helices 4 and 5, supposably acting as interaction platform during viral assembly. Consistently, M1 Y132A showed a defect in lipid raft localization due to reduced interaction with viral HA protein resulting in a diminished structural stability of viral progeny and the formation of filamentous particles. Importantly, reduced M1-RNA binding affinity resulted in an inefficient viral genome incorporation and the production of non-infectious virions that interferes with virus pathogenicity in mice. This study advances our understanding of the importance of dynamic phosphorylation as a so far underestimated level of regulation of multifunctional viral proteins and emphasizes the potential feasibility of targeting posttranslational modifications of M1 as a novel antiviral intervention.


Assuntos
Vírus da Influenza A/metabolismo , Mutação de Sentido Incorreto , Proteínas da Matriz Viral/metabolismo , Células A549 , Substituição de Aminoácidos , Animais , Cães , Feminino , Células HEK293 , Humanos , Vírus da Influenza A/genética , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação , Proteínas da Matriz Viral/genética
2.
FASEB J ; 26(3): 1290-300, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22106366

RESUMO

Guanylate-binding proteins (GBPs) belong to the family of large GTPases that are induced in response to interferons. GBPs contain an N-terminal globular GTPase domain and a C-terminal α-helical regulatory domain that are connected by a short middle domain. Antiviral activity against vesicular stomatitis virus and encephalomyocarditis virus has been shown for hGBP-1; however, no anti-influenza virus properties for GBPs have been described to date. Here we show that hGBP-1 and hGBP-3 possess anti-influenza viral activity. Furthermore, we have identified a novel splice variant of hGBP-3, named hGBP-3ΔC, with a largely modified C-terminal α-helical domain. While all three GBP isoforms were up-regulated on influenza virus infection, hGBP-3ΔC showed the most prominent antiviral activity in epithelial cells. Mutational analysis of hGBPs revealed that the globular domain is the principal antiviral effector domain, and GTP-binding, but not hydrolysis, is necessary for antiviral action. Furthermore, we showed that hGBP-3ΔC strongly represses the activity of the viral polymerase complex, which results in decreased synthesis of viral vRNA, cRNA, mRNA, and viral proteins, as well.


Assuntos
Proteínas de Ligação ao GTP/genética , Transcrição Gênica , Replicação Viral , Vírus/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Citocinas/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Proteínas de Ligação ao GTP/imunologia , Proteínas de Ligação ao GTP/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Dados de Sequência Molecular , Mutação , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , Orthomyxoviridae/fisiologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Vírus/imunologia
3.
Biol Chem ; 392(12): 1135-44, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22050228

RESUMO

PB1-F2 is a nonstructural protein of influenza viruses encoded by the PB1 gene segment from a +1 open reading frame. It has been shown that PB1-F2 contributes to viral pathogenicity, although the underlying mechanisms are still unclear. Induction of type I interferon (IFN) and the innate immune response are the first line of defense against viral infection. Here we show that influenza A viruses (IAVs) lacking the PB1-F2 protein induce an enhanced expression of IFN-ß and IFN-stimulated genes in infected epithelial cells. Studying molecular mechanisms underlying the PB1-F2-mediated IFN antagonistic activity showed that PB1-F2 interferes with the RIG-I/MAVS protein complex thereby inhibiting the activation of the downstream transcription factor IFN regulatory factor 3. These findings were also reflected in in vivo studies demonstrating that infection with PR8 wild-type (wt) virus resulted in higher lung titers and a more severe onset of disease compared with infection with its PB1-F2-deficient counterpart. Accordingly, a much more pronounced infiltration of lungs with immune cells was detected in mice infected with the PB1-F2 wt virus. In summary, we demonstrate that the PB1-F2 protein of IAVs exhibits a type I IFN-antagonistic function by interfering with the RIG-I/MAVS complex, which contributes to an enhanced pathogenicity in vivo.


Assuntos
Interferon Tipo I/antagonistas & inibidores , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Cães , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Biol Chem ; 392(3): 239-48, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21194374

RESUMO

The Mss4 (mammalian suppressor of yeast Sec4) is an evolutionarily highly conserved protein and is expressed in all mammalian tissues. Although its precise biological function is still elusive, it has been shown to associate with a subset of secretory Rab proteins (Rab1b, Rab3a, Rab8a, Rab10) and to possess a rather low guanine nucleotide exchange factor (GEF) activity towards them in vitro (Rab1, Rab3a and Rab8a). By screening a human placenta cDNA library with Mss4 as bait, we identified several Rab GTPases (Rab12, Rab13 and Rab18) as novel Mss4-binding Rab proteins. Only exocytic but no endocytic Rab GTPases were found in our search. The binding of Mss4 to Rab proteins was confirmed by direct yeast two-hybrid interaction, by co-immunoprecipitation from lysates of mammalian cells, by immunofluorescence colocalisation as well as by direct in vitro binding studies. Analysis of Mss4 catalytic activity towards different Rab substrates confirmed that it is a somewhat inefficient GEF. These data, together with our mutational analysis of Mss4-Rab binding capacity, support the already proposed idea that Mss4 functions rather as a chaperone for exocytic Rab GTPases than as a GEF.


Assuntos
Chaperonas Moleculares/metabolismo , Proteínas rab de Ligação ao GTP/isolamento & purificação , Proteínas rab de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Imunofluorescência , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Humanos , Camundongos , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Mutação , Células NIH 3T3 , Ligação Proteica , Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/genética
5.
Hybridoma (Larchmt) ; 29(4): 321-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20715990

RESUMO

In this report we describe the generation of a mouse monoclonal antibody (MAb) against the influenza A virus PB1-F2 protein that is derived from a +1 reading frame of the polymerase basic protein (PB1) gene segment. We further present data that the hybridoma subclone F2-6G10 produces antibodies that specifically recognize the PB1-F2 protein of H1N1 influenza virus types only. The antibody can be used for immunodetection of the PB1-F2 protein in ELISA, Western blot, immunoprecipitation, and immunofluorescence assays.


Assuntos
Anticorpos Monoclonais/imunologia , Imunoglobulina G/imunologia , Proteínas Recombinantes/imunologia , Proteínas Virais/imunologia , Animais , Células Cultivadas , Cães , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HeLa , Humanos , Hibridomas , Immunoblotting , Técnicas Imunoenzimáticas , Imunoprecipitação , Rim/citologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
Cell Microbiol ; 11(10): 1502-16, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19523156

RESUMO

The 11(th) influenza A virus (IAV) protein PB1-F2 is encoded by an alternative reading frame of the PB1 polymerase gene and found in the nucleus, cytosol and at the mitochondria of infected cells, the latter is consistent with experimental evidence for its pro-apoptotic function. Here, the function of PB1-F2 as a phosphoprotein was characterized. PB1-F2 derived from isolate IAV(PR8) and synthetic fragments thereof were phosphorylated in vitro by purified protein kinase C (PKC) and cellular extract. Constitutively active PKCalpha interacts with PB1-F2 in yeast two-hybrid assays. (32)P radiolabelling of transfected 293T cells revealed that phosphorylation of PB1-F2 is sensitive to inhibitors of PKC and could be increased by the PKC activator PMA. ESI-MS analysis and cellular expression of PB1-F2 mutants identified the positions Ser-35 as the major and the Thr-27 as an alternative PKC phosphorylation site. Infection of MDCK cells with recombinant IAV(PR8) lacking these PKC sites abrogated phosphorylation of PB1-F2 in vivo. Furthermore, infection of primary human monocytes with mutant viruses lacking these PB1-F2 phosphorylation sites resulted in impaired caspase 3 activation and reduced progeny virus titres, indicating that the integrity of the identified phosphorylation sites is crucial for a cell-specific function of PB1-F2 during virus replication.


Assuntos
Apoptose , Vírus da Influenza A/patogenicidade , Monócitos/imunologia , Proteína Quinase C/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Humanos , Fosforilação , Mapeamento de Interação de Proteínas , Saccharomyces cerevisiae , Técnicas do Sistema de Duplo-Híbrido
7.
Cell Microbiol ; 10(5): 1140-52, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18182088

RESUMO

The 11th influenza A virus protein PB1-F2 was previously shown to enhance apoptosis in response to cytotoxic stimuli. The 87 amino acid protein that is encoded by an alternative reading frame of the PB1 polymerase gene was described to localize to mitochondria consistent with its proapoptotic function. However, PB1-F2 is also found diffusely distributed in the cytoplasm and in the nucleus suggesting additional functions of the protein. Here we show that PB1-F2 colocalizes and directly interacts with the viral PB1 polymerase protein. Lack of PB1-F2 during infection resulted in an altered localization of PB1 and decreased viral polymerase activity. Consequently, mutant viruses devoid of a functional PB1-F2 reading frame exhibited a small plaque phenotype. Thus, we have identified a novel function of PB1-F2 as an indirect regulator of the influenza virus polymerase activity via its interaction with PB1.


Assuntos
RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Citoplasma/química , Citoplasma/metabolismo , Cães , Humanos , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/análise , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...