Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 7(1): 17485, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29235573

RESUMO

The spontaneously immortalised DF-1 cell line is rapidly replacing its progenitor primary chicken embryo fibroblasts (CEFs) for studies on avian viruses such as avian influenza but no comprehensive study has as yet been reported comparing their innate immunity phenotypes. We conducted microarray analyses of DF-1 and CEFs, under both normal and stimulated conditions using chicken interferon-α (chIFN-α) and the attenuated infectious bursal disease virus vaccine strain PBG98. We found that DF-1 have an attenuated innate response compared to CEFs. Basal expression levels of Suppressor of Cytokine Signalling 1 (chSOCS1), a negative regulator of cytokine signalling in mammals, are 16-fold higher in DF-1 than in CEFs. The chSOCS1 "SOCS box" domain (which in mammals, interacts with an E3 ubiquitin ligase complex) is not essential for the inhibition of cytokine-induced JAK/STAT signalling activation in DF-1. Overexpression of SOCS1 in chIFN-α-stimulated DF-1 led to a relative decrease in expression of interferon-stimulated genes (ISGs; MX1 and IFIT5) and increased viral yield in response to PBG98 infection. Conversely, knockdown of SOCS1 enhanced induction of ISGs and reduced viral yield in chIFN-α-stimulated DF-1. Consequently, SOCS1 reduces induction of the IFN signalling pathway in chicken cells and can potentiate virus replication.


Assuntos
Proteínas Aviárias/metabolismo , Fibroblastos/imunologia , Imunidade Inata/fisiologia , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Animais , Proteínas Aviárias/genética , Doenças das Aves/imunologia , Infecções por Birnaviridae/imunologia , Infecções por Birnaviridae/veterinária , Linhagem Celular , Proliferação de Células/fisiologia , Galinhas , Expressão Gênica , Técnicas de Silenciamento de Genes , Vírus da Doença Infecciosa da Bursa , Interferon-alfa/metabolismo , Janus Quinases/metabolismo , Análise em Microsséries , Cultura Primária de Células , Fatores de Transcrição STAT/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/genética
2.
J Virol ; 90(20): 9446-56, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27512068

RESUMO

UNLABELLED: We have previously shown that IFIT1 is primarily responsible for the antiviral action of interferon (IFN) alpha/beta against parainfluenza virus type 5 (PIV5), selectively inhibiting the translation of PIV5 mRNAs. Here we report that while PIV2, PIV5, and mumps virus (MuV) are sensitive to IFIT1, nonrubulavirus members of the paramyxoviridae such as PIV3, Sendai virus (SeV), and canine distemper virus (CDV) are resistant. The IFIT1 sensitivity of PIV5 was not rescued by coinfection with an IFIT1-resistant virus (PIV3), demonstrating that PIV3 does not specifically inhibit the antiviral activity of IFIT1 and that the inhibition of PIV5 mRNAs is regulated by cis-acting elements. We developed an in vitro translation system using purified human IFIT1 to further investigate the mechanism of action of IFIT1. While the translations of PIV2, PIV5, and MuV mRNAs were directly inhibited by IFIT1, the translations of PIV3, SeV, and CDV mRNAs were not. Using purified human mRNA-capping enzymes, we show biochemically that efficient inhibition by IFIT1 is dependent upon a 5' guanosine nucleoside cap (which need not be N7 methylated) and that this sensitivity is partly abrogated by 2'O methylation of the cap 1 ribose. Intriguingly, PIV5 M mRNA, in contrast to NP mRNA, remained sensitive to inhibition by IFIT1 following in vitro 2'O methylation, suggesting that other structural features of mRNAs may influence their sensitivity to IFIT1. Thus, surprisingly, the viral polymerases (which have 2'-O-methyltransferase activity) of rubulaviruses do not protect these viruses from inhibition by IFIT1. Possible biological consequences of this are discussed. IMPORTANCE: Paramyxoviruses cause a wide variety of diseases, and yet most of their genes encode structural proteins and proteins involved in their replication cycle. Thus, the amount of genetic information that determines the type of disease that paramyxoviruses cause is relatively small. One factor that will influence disease outcomes is how they interact with innate host cell defenses, including the interferon (IFN) system. Here we show that different paramyxoviruses interact in distinct ways with cells in a preexisting IFN-induced antiviral state. Strikingly, all the rubulaviruses tested were sensitive to the antiviral action of ISG56/IFIT1, while all the other paramyxoviruses tested were resistant. We developed novel in vitro biochemical assays to investigate the mechanism of action of IFIT1, demonstrating that the mRNAs of rubulaviruses can be directly inhibited by IFIT1 and that this is at least partially because their mRNAs are not correctly methylated.


Assuntos
Proteínas de Transporte/farmacologia , Paramyxoviridae/genética , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , Rubulavirus/genética , Células A549 , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular Tumoral , Humanos , Interferon-alfa/metabolismo , Metilação , Vírus da Caxumba/genética , Vírus da Parainfluenza 5/genética , Capuzes de RNA/genética , RNA Viral/genética , Proteínas de Ligação a RNA , Vírus Sendai/genética , Replicação Viral/genética
3.
J Virol ; 87(9): 4798-807, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23449801

RESUMO

Preparations of parainfluenza virus 5 (PIV5) that are potent activators of the interferon (IFN) induction cascade were generated by high-multiplicity passage in order to accumulate defective interfering virus genomes (DIs). Nucleocapsid RNA from these virus preparations was extracted and subjected to deep sequencing. Sequencing data were analyzed using methods designed to detect internal deletion and "copyback" DIs in order to identify and characterize the different DIs present and to approximately quantify the ratio of defective to nondefective genomes. Trailer copybacks dominated the DI populations in IFN-inducing preparations of both the PIV5 wild type (wt) and PIV5-VΔC (a recombinant virus that does not encode a functional V protein). Although the PIV5 V protein is an efficient inhibitor of the IFN induction cascade, we show that nondefective PIV5 wt is unable to prevent activation of the IFN response by coinfecting copyback DIs due to the interfering effects of copyback DIs on nondefective virus protein expression. As a result, copyback DIs are able to very rapidly activate the IFN induction cascade prior to the expression of detectable levels of V protein by coinfecting nondefective virus.


Assuntos
Vírus Defeituosos/genética , Genoma Viral , Infecções por Rubulavirus/imunologia , Infecções por Rubulavirus/virologia , Rubulavirus/genética , Animais , Linhagem Celular , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Interferons/genética , Interferons/imunologia , Infecções por Rubulavirus/genética , Proteínas Virais/genética
4.
J Gen Virol ; 94(Pt 1): 59-68, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23052390

RESUMO

Interferon (IFN) induces an antiviral state in cells that results in alterations of the patterns and levels of parainfluenza virus type 5 (PIV5) transcripts and proteins. This study reports that IFN-stimulated gene 56/IFN-induced protein with tetratricopeptide repeats 1 (ISG56/IFIT1) is primarily responsible for these effects of IFN. It was shown that treating cells with IFN after infection resulted in an increase in virus transcription but an overall decrease in virus protein synthesis. As there was no obvious decrease in the overall levels of cellular protein synthesis in infected cells treated with IFN, these results suggested that ISG56/IFIT1 selectively inhibits the translation of viral mRNAs. This conclusion was supported by in vitro translation studies. Previous work has shown that ISG56/IFIT1 can restrict the replication of viruses lacking a 2'-O-methyltransferase activity, an enzyme that methylates the 2'-hydroxyl group of ribose sugars in the 5'-cap structures of mRNA. However, the data in the current study strongly suggested that PIV5 mRNAs are methylated at the 2'-hydroxyl group and thus that ISG56/IFIT1 selectively inhibits the translation of PIV5 mRNA by some as yet unrecognized mechanism. It was also shown that ISG56/IFIT1 is primarily responsible for the IFN-induced inhibition of PIV5.


Assuntos
Proteínas de Transporte/biossíntese , Interferon-alfa/farmacologia , Infecções por Respirovirus/virologia , Respirovirus/efeitos dos fármacos , Respirovirus/genética , Proteínas Virais/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Replicação do DNA , Técnicas de Silenciamento de Genes , Humanos , Interferon alfa-2 , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , Proteínas de Ligação a RNA , Proteínas Recombinantes/farmacologia , Respirovirus/metabolismo , Infecções por Respirovirus/tratamento farmacológico , Infecções por Respirovirus/metabolismo , Transcrição Gênica , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
5.
J Gen Virol ; 93(Pt 2): 299-307, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22049094

RESUMO

Conflicting reports exist regarding the requirement for virus replication in interferon (IFN) induction by paramyxoviruses. Our previous work has demonstrated that pathogen-associated molecular patterns capable of activating the IFN-induction cascade are not normally generated during virus replication, but are associated instead with the presence of defective interfering (DI) viruses. We demonstrate here that DIs of paramyxoviruses, including parainfluenza virus 5, mumps virus and Sendai virus, can activate the IFN-induction cascade and the IFN-ß promoter in the absence of virus protein synthesis. As virus protein synthesis is an absolute requirement for paramyxovirus genome replication, our results indicate that these DI viruses do not require replication to activate the IFN-induction cascade.


Assuntos
Interferon beta/biossíntese , Interferon beta/genética , Paramyxoviridae/imunologia , Paramyxoviridae/fisiologia , Regiões Promotoras Genéticas , Ativação Transcricional , Replicação Viral , Animais , Linhagem Celular , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Humanos , Paramyxoviridae/genética , Rubulavirus , Proteínas Virais/biossíntese
6.
Virology ; 415(1): 39-46, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21511322

RESUMO

It is generally thought that pathogen-associated molecular patterns (PAMPs) responsible for triggering interferon (IFN) induction are produced during virus replication and, to limit the activation of the IFN response by these PAMPs, viruses encode antagonists of IFN induction. Here we have studied the induction of IFN by parainfluenza virus type 5 (PIV5) at the single-cell level, using a cell line expressing GFP under the control of the IFN-ß promoter. We demonstrate that a recombinant PIV5 (termed PIV5-VΔC) that lacks a functional V protein (the viral IFN antagonist) does not activate the IFN-ß promoter in the majority of infected cells. We conclude that viral PAMPs capable of activating the IFN induction cascade are not produced or exposed during the normal replication cycle of PIV5, and suggest instead that defective viruses are primarily responsible for inducing IFN during PIV5 infection in this system.


Assuntos
Interferon beta/antagonistas & inibidores , Interferon beta/genética , Regiões Promotoras Genéticas , Rubulavirus/fisiologia , Proteínas Virais/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Imunofluorescência , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Humanos , Immunoblotting , Interferon beta/metabolismo , Mutação , Rubulavirus/genética , Células Vero , Proteínas Virais/genética , Replicação Viral
7.
Virology ; 407(2): 247-55, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20833406

RESUMO

The infection of cells by RNA viruses is associated with the recognition of virus PAMPs (pathogen-associated molecular patterns) and the production of type I interferon (IFN). To counter this, most, if not all, RNA viruses encode antagonists of the IFN system. Here we present data on the dynamics of IFN production and response during developing infections by paramyxoviruses, influenza A virus and bunyamwera virus. We show that only a limited number of infected cells are responsible for the production of IFN, and that this heterocellular production is a feature of the infecting virus as opposed to an intrinsic property of the cells.


Assuntos
Vírus Bunyamwera/patogenicidade , Vírus da Influenza A/patogenicidade , Interferon Tipo I/metabolismo , Rim/virologia , Pulmão/virologia , Paramyxoviridae/patogenicidade , Animais , Vírus Bunyamwera/imunologia , Linhagem Celular Tumoral/virologia , Chlorocebus aethiops , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/imunologia , Interferon Tipo I/genética , Interferon-alfa/genética , Interferon-alfa/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Rim/citologia , Rim/imunologia , Pulmão/citologia , Pulmão/imunologia , Paramyxoviridae/imunologia , Especificidade da Espécie , Células Vero/virologia , Replicação Viral
8.
J Virol ; 83(3): 1465-73, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19019954

RESUMO

The RNA helicases encoded by melanoma differentiation-associated gene 5 (mda-5) and retinoic acid-inducible gene I (RIG-I) detect foreign cytoplasmic RNA molecules generated during the course of a virus infection, and their activation leads to induction of type I interferon synthesis. Paramyxoviruses limit the amount of interferon produced by infected cells through the action of their V protein, which binds to and inhibits mda-5. Here we show that activation of both mda-5 and RIG-I by double-stranded RNA (dsRNA) leads to the formation of homo-oligomers through self-association of the helicase domains. We identify a region within the helicase domain of mda-5 that is targeted by all paramyxovirus V proteins and demonstrate that they inhibit activation of mda-5 by blocking dsRNA binding and consequent self-association. In addition to this commonly targeted domain, some paramyxovirus V proteins target additional regions of mda-5. In contrast, V proteins cannot bind to RIG-I and consequently have no effect on the ability of RIG-I to bind dsRNA or to form oligomers.


Assuntos
RNA Helicases DEAD-box/antagonistas & inibidores , Paramyxoviridae/fisiologia , Proteínas Virais/fisiologia , Animais , Biopolímeros , Linhagem Celular , Chlorocebus aethiops , RNA Helicases DEAD-box/metabolismo , Humanos , Hidrólise , Helicase IFIH1 Induzida por Interferon , Técnicas do Sistema de Duplo-Híbrido , Células Vero
9.
Virology ; 368(1): 114-21, 2007 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-17640695

RESUMO

A dynamic model of STAT1 degradation by the V protein of parainfluenza virus 5 (PIV5; formerly SV5) has been proposed. In it, the V protein functions as a bipartite adaptor linking DDB1, a component of a cellular SCF-like ubiquitin E3 ligase complex, to STAT2, which in turn binds STAT1 and presents STAT1 to the E3 ligase complex for ubiquitination and subsequent degradation. Furthermore, it appears that loss of STAT1 from the complex results in decreased affinity of V for STAT2 such that STAT2 either dissociates from V or is displaced by STAT1/STAT2 complexes, facilitating the cycling of the DDB1/PIV5 V containing E3 complex for further rounds of STAT1 ubiquitination and degradation. By determining the approximate number of molecules of V, DDB1, STAT1 and STAT2 present in IFN-treated 2fTGH cells, we provide additional evidence for this dynamic model of STAT1 degradation. These results show that (i) in IFN-treated cells there is approximately 4-fold less STAT2 and 15-fold less DDB1 than STAT1 per cell and thus DDB1 and STAT2 must repeatedly acquire more STAT1 for degradation to go to completion, and (ii) approximately 600 molecules of V protein per cell can target as many as 120,000 molecules of STAT1 for degradation in the absence of either viral or cellular protein synthesis. The importance of this mechanism in terms of the ability of the virus to dismantle the IFN-induced anti-viral state of cells is discussed.


Assuntos
Interferons/imunologia , Vírus da Parainfluenza 5/imunologia , Fator de Transcrição STAT1/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Proteínas de Ligação a DNA/metabolismo , Humanos , Ligação Proteica , Fator de Transcrição STAT2/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
10.
J Gen Virol ; 88(Pt 3): 956-966, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17325370

RESUMO

Mapuera virus (MPRV) is a paramyxovirus that was originally isolated from bats, but its host range remains unknown. It was classified as a member of the genus Rubulavirus on the basis of structural and genetic features. Like other rubulaviruses it encodes a V protein (MPRV/V) that functions as an interferon (IFN) antagonist. Here we show that MPRV/V differs from the IFN antagonists of other rubulaviruses in that it does not induce the proteasomal degradation of STAT proteins, key factors in the IFN signalling cascade. Rather, MPRV/V prevents the nuclear translocation of STATs in response to IFN stimulation and inhibits the formation of the transcription factor complex ISGF3. We also show that MPRV/V blocks IFN signalling in cells from diverse mammalian species and discuss the IFN response as a barrier to cross-species infections.


Assuntos
Interferons/antagonistas & inibidores , Rubulavirus/imunologia , Fatores de Transcrição STAT/metabolismo , Proteínas Virais/fisiologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Fator Gênico 3 Estimulado por Interferon/metabolismo , Interferons/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Fosforilação , Proteínas Virais/genética
11.
J Virol ; 81(5): 2318-27, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17182679

RESUMO

Many viruses, including human influenza A virus, have developed strategies for counteracting the host type I interferon (IFN) response. We have explored whether avian influenza viruses were less capable of combating the type I IFN response in mammalian cells, as this might be a determinant of host range restriction. A panel of avian influenza viruses isolated between 1927 and 1997 was assembled. The selected viruses showed variation in their ability to activate the expression of a reporter gene under the control of the IFN-beta promoter and in the levels of IFN induced in mammalian cells. Surprisingly, the avian NS1 proteins expressed alone or in the genetic background of a human influenza virus controlled IFN-beta induction in a manner similar to the NS1 protein of human strains. There was no direct correlation between the IFN-beta induction and replication of avian influenza viruses in human A549 cells. Nevertheless, human cells deficient in the type I IFN system showed enhanced replication of the avian viruses studied, implying that the human type I IFN response limits avian influenza viruses and can contribute to host range restriction.


Assuntos
Vírus da Influenza A/patogenicidade , Interferon Tipo I/biossíntese , Proteínas não Estruturais Virais/fisiologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Cães , Expressão Gênica , Humanos , Imunidade Inata , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/genética , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Replicação Viral
12.
Virology ; 347(1): 52-64, 2006 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16378631

RESUMO

We investigated the ability of a selection of human influenza A viruses, including recent clinical isolates, to induce IFN-beta production in cultured cell lines. In contrast to the well-characterized laboratory strain A/PR/8/34, several, but not all, recent isolates of H3N2 viruses resulted in moderate IFN-beta stimulation. Through the generation of recombinant viruses, we were able to show that this is not due to a loss of the ability of the NS1 genes to suppress IFN-beta induction; indeed, the NS1 genes behaved similarly with respect to their abilities to block dsRNA signaling. Interestingly, replication of A/Sydney/5/97 virus was less susceptible to pre-treatment with IFN-alpha than the other viruses. In contrast to the universal effect on dsRNA signaling, we noted differences in the effect of NS1 proteins on expression of interferon stimulated genes and also genes induced by a distinct pathway. The majority of NS1 proteins blocked expression from both IFN-dependent and TNF-dependent promoters by an apparent post-transcriptional mechanism. The NS1 gene of A/PR/8/34 NS1 did not confer these blocks. We noted striking differences in the cellular localization of different influenza A virus NS1 proteins during infection, which might explain differences in biological activity.


Assuntos
Vírus da Influenza A/imunologia , Interferon beta/biossíntese , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , DNA Viral/genética , Genes Reporter , Genes Virais , Humanos , Imunidade Inata , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/imunologia , Vírus da Influenza A Subtipo H3N2/patogenicidade , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Interferon beta/genética , Regiões Promotoras Genéticas , RNA de Cadeia Dupla/genética , Recombinação Genética , Transdução de Sinais , Células Vero , Proteínas não Estruturais Virais/genética
13.
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
14.
J Gen Virol ; 86(Pt 1): 151-158, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15604442

RESUMO

Previous work has documented that the V protein of simian virus 5 (SV5) targets STAT1 for proteasome-mediated degradation, whilst the V protein of human parainfluenza virus type 2 (hPIV2) targets STAT2. Here, it was shown that the processes of ubiquitination and degradation could be reconstructed in vitro by using programmed rabbit reticulocyte lysates. Using this system, the addition of bacterially expressed and purified SV5 V protein to programmed lysates was demonstrated to result in the polyubiquitination and degradation of in vitro-translated STAT1, but only if human STAT2 was also present. Surprisingly, in the same assay, purified hPIV2 V protein induced the polyubiquitination of both STAT1 and STAT2. In the light of these in vitro results, the specificity of degradation of STAT1 and STAT2 by SV5 and hPIV2 in tissue-culture cells was re-examined. As previously reported, STAT1 could not be detected in human cells that expressed SV5 V protein constitutively, whilst STAT2 could not be detected in human cells that expressed hPIV2 V protein, although the levels of STAT1 may also have been reduced in some human cells infected with hPIV2. In contrast, STAT1 could not be detected, whereas STAT2 remained present, in a variety of animal cells, including canine (MDCK) cells, that expressed the V protein of either SV5 or hPIV2. Thus, the V protein of SV5 appears to be highly specific for STAT1 degradation, but the V protein of hPIV2 is more promiscuous.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Respirovirus/metabolismo , Transativadores/metabolismo , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Humanos , Coelhos , Reticulócitos/metabolismo , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transdução de Sinais , Especificidade da Espécie , Ubiquitina/metabolismo
15.
Proc Natl Acad Sci U S A ; 101(49): 17264-9, 2004 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-15563593

RESUMO

Most paramyxoviruses circumvent the IFN response by blocking IFN signaling and limiting the production of IFN by virus-infected cells. Here we report that the highly conserved cysteine-rich C-terminal domain of the V proteins of a wide variety of paramyxoviruses binds melanoma differentiation-associated gene 5 (mda-5) product. mda-5 is an IFN-inducible host cell DExD/H box helicase that contains a caspase recruitment domain at its N terminus. Overexpression of mda-5 stimulated the basal activity of the IFN-beta promoter in reporter gene assays and significantly enhanced the activation of the IFN-beta promoter by intracellular dsRNA. Both these activities were repressed by coexpression of the V proteins of simian virus 5, human parainfluenza virus 2, mumps virus, Sendai virus, and Hendra virus. Similar results to the reporter assays were obtained by measuring IFN production. Inhibition of mda-5 by RNA interference or by dominant interfering forms of mda-5 significantly inhibited the activation of the IFN-beta promoter by dsRNA. It thus appears that mda-5 plays a central role in an intracellular signal transduction pathway that can lead to the activation of the IFN-beta promoter, and that the V proteins of paramyxoviruses interact with mda-5 to block its activity.


Assuntos
Interferon beta/genética , Paramyxoviridae/química , RNA Helicases/antagonistas & inibidores , Ativação Transcricional/efeitos dos fármacos , Proteínas Virais/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , RNA Helicases DEAD-box , Humanos , Helicase IFIH1 Induzida por Interferon , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais , Transfecção , Proteínas Virais/genética , Proteínas Virais/farmacologia
16.
J Virol ; 77(3): 2174-81, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12525652

RESUMO

The V protein of the paramyxovirus simian virus 5 blocks interferon (IFN) signaling by targeting STAT1 for proteasome-mediated degradation. Here we report on the isolation of human cell lines that express the V protein and can no longer respond to IFN. A variety of viruses, particularly slow-growing wild-type viruses and vaccine candidate viruses (which are attenuated due to mutations that affect virus replication, virus spread, or ability to circumvent the IFN response), form bigger plaques and grow to titers that are increased as much as 10- to 4,000-fold in these IFN-nonresponsive cells. We discuss the practical applications of using such cells in vaccine development and manufacture, virus diagnostics and isolation of newly emerging viruses, and studies on host cell tropism and pathogenesis.


Assuntos
Interferons/farmacologia , Transfecção , Replicação Viral , Vírus/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Humanos , Vacinas Sintéticas/imunologia , Células Vero , Vacinas Virais/imunologia
17.
J Virol ; 76(22): 11379-86, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388698

RESUMO

The V protein of simian virus 5 (SV5) blocks interferon signaling by targeting STAT1 for proteasome-mediated degradation. Here we present three main pieces of evidence which demonstrate that the p127 subunit (DDB1) of the UV damage-specific DNA binding protein (DDB) plays a central role in this degradation process. First, the V protein of an SV5 mutant which fails to target STAT1 for degradation does not bind DDB1. Second, mutations in the N and C termini of V which abolish the binding of V to DDB1 also prevent V from blocking interferon (IFN) signaling. Third, treatment of HeLa/SV5-V cells, which constitutively express the V protein of SV5 and thus lack STAT1, with short interfering RNAs specific for DDB1 resulted in a reduction in DDB1 levels with a concomitant increase in STAT1 levels and a restoration of IFN signaling. Furthermore, STAT1 is degraded in GM02415 (2RO) cells, which have a mutation in DDB2 (the p48 subunit of DDB) which abolishes its ability to interact with DDB1, thereby demonstrating that the role of DDB1 in STAT1 degradation is independent of its association with DDB2. Evidence is also presented which demonstrates that STAT2 is required for the degradation of STAT1 by SV5. These results suggest that DDB1, STAT1, STAT2, and V may form part of a large multiprotein complex which leads to the targeted degradation of STAT1 by the proteasome.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Rubulavirus/patogenicidade , Transativadores/metabolismo , Proteínas Virais , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Células HeLa , Humanos , Interferons/metabolismo , Vírus da Parainfluenza 2 Humana/patogenicidade , Vírus da Parainfluenza 2 Humana/fisiologia , Rubulavirus/fisiologia , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transdução de Sinais , Replicação Viral , Xeroderma Pigmentoso/metabolismo
18.
Virology ; 293(2): 234-42, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11886243

RESUMO

CPI(+) and CPI(-) are two canine isolates of simian virus 5 (SV5). CPI(+) was originally isolated from the cerebrospinal fluid of a dog with temporary posterior paralysis and CPI(-) was recovered at 12 days p.i. from the brain tissue of a dog experimentally infected with CPI(+). We have previously shown that the V protein of SV5 blocks interferon (IFN) signalling by targeting STAT1 for degradation. Here we report that whilst CPI(+) targets STAT1 for degradation, CPI(-) fails to and as a consequence, CPI(+) blocks IFN signalling but CPI(-) does not. Three amino acid differences in the P/V N-terminal common domain of the V protein are responsible for the observed difference in the abilities of CPI(+) and CPI(-) to block IFN signalling. In cells persistently infected with CPI(-) the virus may become repressed in response to IFN, under which circumstances virus glycoproteins are lost from the surface of infected cells and virus nucleocapsid proteins accumulate in cytoplasmic inclusion bodies. We suggest that in vivo cells infected with IFN-resistant viruses (in which there would be continuous virus protein synthesis) may be more susceptible to killing by cytotoxic T cells than cells infected with IFN-sensitive viruses (in which virus protein synthesis was repressed), and a model of virus persistence is put forward in which there is alternating selection of IFN-resistant and IFN-sensitive viruses depending upon the state of the adaptive immune response.


Assuntos
Interferon Tipo I/farmacologia , Paramyxoviridae/fisiologia , Transdução de Sinais , Substituição de Aminoácidos , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Cães , Humanos , Camundongos , Paramyxoviridae/metabolismo , Proteínas Recombinantes , Fator de Transcrição STAT1 , Especificidade da Espécie , Transativadores/metabolismo , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/metabolismo
19.
J Virol ; 76(5): 2159-67, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11836393

RESUMO

Human cell lines were isolated that express the V protein of either simian virus 5 (SV5) or human parainfluenza virus type 2 (hPIV2); the cell lines were termed 2f/SV5-V and 2f/PIV2-V, respectively. STAT1 was not detectable in 2f/SV5-V cells, and the cells failed to signal in response to either alpha/beta interferons (IFN-alpha and IFN-beta, or IFN-alpha/beta) or gamma interferon (IFN-gamma). In contrast, STAT2 was absent from 2f/PIV2-V cells, and IFN-alpha/beta but not IFN-gamma signaling was blocked in these cells. Treatment of both 2f/SV5-V and 2f/PIV2-V cells with a proteasome inhibitor allowed the respective STAT levels to accumulate at rates similar to those seen in 2fTGH cells, indicating that the V proteins target the STATs for proteasomal degradation. Infection with SV5 can lead to a complete loss of both phosphorylated and nonphosphorylated forms of STAT1 by 6 h postinfection. Since the turnover of STAT1 in uninfected cells is longer than 24 h, we conclude that degradation of STAT1 is the main mechanism by which SV5 blocks interferon (IFN) signaling. Pretreatment of 2fTGH cells with IFN-alpha severely inhibited both SV5 and hPIV2 protein synthesis. However, and in marked contrast, pretreatment of 2fTGH cells with IFN-gamma had little obvious effect on SV5 protein synthesis but did significantly reduce the replication of hPIV2. Pretreament with IFN-alpha or IFN-gamma did not induce an antiviral state in 2f/SV5-V cells, indicating either that the induction of an antiviral state is completely dependent on STAT signaling or that the V protein interferes with other, STAT-independent cell signaling pathways that may be induced by IFNs. Even though SV5 blocked IFN signaling, the addition of exogenous IFN-alpha to the culture medium of 2fTGH cells 12 h after a low-multiplicity infection with SV5 significantly reduced the subsequent cell-to-cell spread of virus. The significance of the results in terms of the strategy that these viruses have evolved to circumvent the IFN response is discussed.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Vírus da Parainfluenza 2 Humana/fisiologia , Rubulavirus/fisiologia , Transativadores/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Antivirais/farmacologia , Divisão Celular , Linhagem Celular , Clonagem Molecular , Humanos , Interferons/farmacologia , Vírus da Parainfluenza 2 Humana/efeitos dos fármacos , Rubulavirus/efeitos dos fármacos , Fator de Transcrição STAT1 , Fator de Transcrição STAT2 , Transfecção , Raios Ultravioleta , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/genética , Replicação Viral
20.
J Virol ; 75(7): 3363-70, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11238862

RESUMO

Previous work has demonstrated that the V protein of simian virus 5 (SV5) targets STAT1 for proteasome-mediated degradation (thereby blocking interferon [IFN] signaling) in human but not in murine cells. In murine BF cells, SV5 establishes a low-grade persistent infection in which the virus fluxes between active and repressed states in response to local production of IFN. Upon passage of persistently infected BF cells, virus mutants were selected that were better able to replicate in murine cells than the parental W3 strain of SV5 (wild type [wt]). Viruses with mutations in the Pk region of the N-terminal domain of the V protein came to predominate the population of viruses carried in the persistently infected cell cultures. One of these mutant viruses, termed SV5 mci-2, was isolated. Sequence analysis of the V/P gene of SV5 mci-2 revealed two nucleotide differences compared to wt SV5, only one of which resulted in an amino acid substitution (asparagine [N], residue 100, to aspartic acid [D]) in V. Unlike the protein of wt SV5, the V protein of SV5 mci-2 blocked IFN signaling in murine cells. Since the SV5 mci-2 virus had additional mutations in genes other than the V/P gene, a recombinant virus (termed rSV5-V/P N(100)D) was constructed that contained this substitution alone within the wt SV5 backbone to evaluate what effect the asparagine-to-aspartic-acid substitution in V had on the virus phenotype. In contrast to wt SV5, rSV5-V/P N(100)D blocked IFN signaling in murine cells. Furthermore, rSV5-V/P N(100)D virus protein synthesis in BF cells continued for significantly longer periods than that for wt SV5. However, even in cells infected with rSV5-V/P N(100)D, there was a late, but significant, inhibition in virus protein synthesis. Nevertheless, there was an increase in virus yield from BF cells infected with rSV5-V/P N(100)D compared to wt SV5, demonstrating a clear selective advantage to SV5 in being able to block IFN signaling in these cells.


Assuntos
Interferons/farmacologia , Respirovirus/fisiologia , Proteínas Estruturais Virais/fisiologia , Animais , Sequência de Bases , Proteínas de Ligação a DNA/análise , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Fases de Leitura Aberta , Mutação Puntual , Respirovirus/genética , Fator de Transcrição STAT1 , Relação Estrutura-Atividade , Transativadores/análise , Proteínas Virais/biossíntese , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...