Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Virol ; 95(9)2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33328306

RESUMO

Classical swine fever virus (CSFV) is the causative agent of classical swine fever, a notifiable disease of economic importance that causes severe leukopenia, fever and haemorrhagic disease in domesticated pigs and wild boar across the globe. CSFV has been shown to antagonise the induction of type I IFN, partly through a function of its N-terminal protease (Npro) which binds IRF3 and targets it for proteasomal degradation. Additionally, Npro has been shown to antagonise apoptosis triggered by the dsRNA-homolog poly(I:C), however the exact mechanism by which this is achieved has not been fully elucidated. In this study we confirm the ability of Npro to inhibit dsRNA-mediated apoptosis and show that Npro is also able to antagonise Sendai virus-mediated apoptosis in PK-15 cells. Gene edited PK-15 cell lines were used to show the dsRNA-sensing pathogen recognition receptors (PRRs) TLR3 and RIG-I specifically respond to poly(I:C) and SeV respectively, subsequently triggering apoptosis through pathways that converge on IRF3 and culminate in the cleavage of caspase-3. Importantly, this IRF3-mediated apoptosis was found to be dependent on transcription-independent functions of IRF3 and also on Bax, a pro-apoptotic Bcl-2 family protein, through a direct interaction between the two proteins. Deletion of IRF3, stable expression of Npro and infection with wild-type CSFV were found to antagonise the mitochondrial localisation of Bax, a key hallmark of the intrinsic, mitochondrial pathway of apoptosis. Together, these findings show that Npro's putative interaction with IRF3 is involved not only in its antagonism of type I IFN, but also dsRNA-mediated mitochondrial apoptosis.Importance Responsible for severe haemorrhagic disease in domestic pigs and wild boar, classical swine fever is recognised by the World Organisation for Animal Health (OIE) and European Union as a notifiable disease of economic importance. Persistent infection, immunotolerance and early dissemination of the virus at local sites of infection have been linked to the antagonism of type I IFN induction by Npro This protein may further contribute to these phenomena by antagonising the induction of dsRNA-mediated apoptosis. Ultimately, apoptosis is an important innate mechanism by which cells counter viruses at local sites of infection, thus preventing wider spread and dissemination within the host, potentially also contributing to the onset of persistence. Elucidation of the mechanism by which Npro antagonises the apoptotic response will help inform the development of rationally-designed live-attenuated vaccines and antivirals for control of outbreaks in typically CSFV-free countries.

2.
Nature ; 487(7408): 486-90, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22810585

RESUMO

Viruses must enter host cells to replicate, assemble and propagate. Because of the restricted size of their genomes, viruses have had to evolve efficient ways of exploiting host cell processes to promote their own life cycles and also to escape host immune defence mechanisms. Many viral open reading frames (viORFs) with immune-modulating functions essential for productive viral growth have been identified across a range of viral classes. However, there has been no comprehensive study to identify the host factors with which these viORFs interact for a global perspective of viral perturbation strategies. Here we show that different viral perturbation patterns of the host molecular defence network can be deduced from a mass-spectrometry-based host-factor survey in a defined human cellular system by using 70 innate immune-modulating viORFs from 30 viral species. The 579 host proteins targeted by the viORFs mapped to an unexpectedly large number of signalling pathways and cellular processes, suggesting yet unknown mechanisms of antiviral immunity. We further experimentally verified the targets heterogeneous nuclear ribonucleoprotein U, phosphatidylinositol-3-OH kinase, the WNK (with-no-lysine) kinase family and USP19 (ubiquitin-specific peptidase 19) as vulnerable nodes in the host cellular defence system. Evaluation of the impact of viral immune modulators on the host molecular network revealed perturbation strategies used by individual viruses and by viral classes. Our data are also valuable for the design of broad and specific antiviral therapies.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Vírus/imunologia , Endopeptidases/metabolismo , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Imunidade Inata/imunologia , Espectrometria de Massas , Fases de Leitura Aberta/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais , Especificidade por Substrato , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Vírus/metabolismo
3.
Vet Res ; 47(1): 75, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27494935

RESUMO

Viruses that infect birds pose major threats-to the global supply of chicken, the major, universally-acceptable meat, and as zoonotic agents (e.g. avian influenza viruses H5N1 and H7N9). Controlling these viruses in birds as well as understanding their emergence into, and transmission amongst, humans will require considerable ingenuity and understanding of how different species defend themselves. The type I interferon-coordinated response constitutes the major antiviral innate defence. Although interferon was discovered in chicken cells, details of the response, particularly the identity of hundreds of stimulated genes, are far better described in mammals. Viruses induce interferon-stimulated genes but they also regulate the expression of many hundreds of cellular metabolic and structural genes to facilitate their replication. This study focusses on the potentially anti-viral genes by identifying those induced just by interferon in primary chick embryo fibroblasts. Three transcriptomic technologies were exploited: RNA-seq, a classical 3'-biased chicken microarray and a high density, "sense target", whole transcriptome chicken microarray, with each recognising 120-150 regulated genes (curated for duplication and incorrect assignment of some microarray probesets). Overall, the results are considered robust because 128 of the compiled, curated list of 193 regulated genes were detected by two, or more, of the technologies.


Assuntos
Galinhas/genética , Genes/efeitos dos fármacos , Interferon-alfa/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos/veterinária , Animais , Embrião de Galinha , Galinhas/imunologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária
4.
Proc Natl Acad Sci U S A ; 110(8): 3053-8, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23391734

RESUMO

Severe infectious disease in children may be a manifestation of primary immunodeficiency. These genetic disorders represent important experiments of nature with the capacity to elucidate nonredundant mechanisms of human immunity. We hypothesized that a primary defect of innate antiviral immunity was responsible for unusually severe viral illness in two siblings; the proband developed disseminated vaccine strain measles following routine immunization, whereas an infant brother died after a 2-d febrile illness from an unknown viral infection. Patient fibroblasts were indeed abnormally permissive for viral replication in vitro, associated with profound failure of type I IFN signaling and absence of STAT2 protein. Sequencing of genomic DNA and RNA revealed a homozygous mutation in intron 4 of STAT2 that prevented correct splicing in patient cells. Subsequently, other family members were identified with the same genetic lesion. Despite documented infection by known viral pathogens, some of which have been more severe than normal, surviving STAT2-deficient individuals have remained generally healthy, with no obvious defects in their adaptive immunity or developmental abnormalities. These findings imply that type I IFN signaling [through interferon-stimulated gene factor 3 (ISGF3)] is surprisingly not essential for host defense against the majority of common childhood viral infections.


Assuntos
Predisposição Genética para Doença , Fator de Transcrição STAT2/genética , Viroses/genética , Sequência de Bases , Células Cultivadas , Pré-Escolar , Primers do DNA , Feminino , Humanos , Interferon Tipo I/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Viroses/metabolismo
5.
J Virol ; 87(9): 5028-40, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23427151

RESUMO

Viruses must be able to resist host innate responses, especially the type I interferon (IFN) response. They do so by preventing the induction or activity of IFN and/or by resisting the antiviral effectors that it induces. Poxviruses are no exception, with many mechanisms identified whereby mammalian poxviruses, notably, vaccinia virus (VACV), but also cowpox and myxoma viruses, are able to evade host IFN responses. Similar mechanisms have not been described for avian poxviruses (avipoxviruses). Restricted for permissive replication to avian hosts, they have received less attention; moreover, the avian host responses are less well characterized. We show that the prototypic avipoxvirus, fowlpox virus (FWPV), is highly resistant to the antiviral effects of avian IFN. A gain-of-function genetic screen identified fpv014 to contribute to increased resistance to exogenous recombinant chicken alpha IFN (ChIFN1). fpv014 is a member of the large family of poxvirus (especially avipoxvirus) genes that encode proteins containing N-terminal ankyrin repeats (ANKs) and C-terminal F-box-like motifs. By binding the Skp1/cullin-1 complex, the F box in such proteins appears to target ligands bound by the ANKs for ubiquitination. Mass spectrometry and immunoblotting demonstrated that tandem affinity-purified, tagged fpv014 was complexed with chicken cullin-1 and Skp1. Prior infection with an fpv014-knockout mutant of FWPV still blocked transfected poly(I·C)-mediated induction of the beta IFN (ChIFN2) promoter as effectively as parental FWPV, but the mutant was more sensitive to exogenous ChIFN1. Therefore, unlike the related protein fpv012, fpv014 does not contribute to the FWPV block to induction of ChIFN2 but does confer resistance to an established antiviral state.


Assuntos
Repetição de Anquirina , Vírus da Varíola das Aves Domésticas/imunologia , Varíola Aviária/imunologia , Interferon-alfa/imunologia , Doenças das Aves Domésticas/imunologia , Proteínas Virais/química , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Embrião de Galinha , Galinhas , Varíola Aviária/genética , Varíola Aviária/virologia , Vírus da Varíola das Aves Domésticas/química , Vírus da Varíola das Aves Domésticas/genética , Biblioteca Gênica , Interferon-alfa/genética , Dados de Sequência Molecular , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia , Estrutura Terciária de Proteína , Proteínas Virais/genética
6.
J Virol ; 87(9): 5041-52, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23427153

RESUMO

Mammalian poxviruses, including vaccinia virus (VACV), have evolved multiple mechanisms to evade the host type I interferon (IFN) responses at different levels, with viral proteins targeting IFN induction, signaling, and antiviral effector functions. Avian poxviruses (avipoxviruses), which have been developed as recombinant vaccine vectors for permissive (i.e., poultry) and nonpermissive (i.e., mammals, including humans) species, encode no obvious equivalents of any of these proteins. We show that fowlpox virus (FWPV) fails to induce chicken beta IFN (ChIFN2) and is able to block its induction by transfected poly(I·C), an analog of cytoplasmic double-stranded RNA (dsRNA). A broad-scale loss-of-function genetic screen was used to find FWPV-encoded modulators of poly(I·C)-mediated ChIFN2 induction. It identified fpv012, a member of a family of poxvirus genes highly expanded in the avipoxviruses (31 in FWPV; 51 in canarypox virus [CNPV], representing 15% of the total gene complement), encoding proteins containing N-terminal ankyrin repeats (ANKs) and C-terminal F-box-like motifs. Under ectopic expression, the first ANK of fpv012 is dispensable for inhibitory activity and the CNPV ortholog is also able to inhibit induction of ChIFN2. FWPV defective in fpv012 replicates well in culture and barely induces ChIFN2 during infection, suggesting that other factors are involved in blocking IFN induction and resisting the antiviral effectors. Nevertheless, unlike parental and revertant viruses, the mutants induce moderate levels of expression of interferon-stimulated genes (ISGs), suggesting either that there is sufficient ChIFN2 expression to partially induce the ISGs or the involvement of alternative, IFN-independent pathways that are also normally blocked by fpv012.


Assuntos
Repetição de Anquirina , Vírus da Varíola das Aves Domésticas/imunologia , Varíola Aviária/imunologia , Interferon beta/imunologia , Doenças das Aves Domésticas/imunologia , Proteínas Virais/química , Proteínas Virais/imunologia , Animais , Embrião de Galinha , Galinhas , Varíola Aviária/genética , Varíola Aviária/virologia , Vírus da Varíola das Aves Domésticas/química , Vírus da Varíola das Aves Domésticas/genética , Biblioteca Gênica , Interferon beta/genética , Mutação , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia , Estrutura Terciária de Proteína , Proteínas Virais/genética
7.
J Virol ; 86(7): 3411-21, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301134

RESUMO

RIG-I and mda-5 are activated by viral RNA and stimulate type I interferon production. Laboratory of genetics and physiology 2 (LGP2) shares homology with RIG-I and mda-5 but lacks the CARD domains required for signaling. The V proteins of paramyxoviruses limit interferon induction by binding mda-5 and preventing its activation; however, they do not bind RIG-I and have not been considered inhibitors of RIG-I signaling. Here we uncover a novel mechanism of RIG-I inhibition in which the V protein of parainfluenzavirus type 5 (PIV5; formerly known as simian virus type 5 [SV5]) interacts with LGP2 and cooperatively inhibits induction by RIG-I ligands. A complex between RIG-I and LGP2 is observed in the presence of PIV5-V, and we propose that this complex is refractory to activation by RIG-I ligands. The V proteins from other paramyxoviruses also bind LGP2 and demonstrate LGP2-dependent inhibition of RIG-I signaling. This is significant, because it demonstrates a general mechanism for the targeting of the RIG-I pathway by paramyxoviruses.


Assuntos
RNA Helicases DEAD-box/metabolismo , Interferon beta/metabolismo , Interferons/metabolismo , RNA Helicases/metabolismo , Infecções por Rubulavirus/enzimologia , Rubulavirus/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Humanos , Helicase IFIH1 Induzida por Interferon , Interferon beta/genética , Interferons/genética , Dados de Sequência Molecular , Ligação Proteica , RNA Helicases/química , RNA Helicases/genética , Receptores Imunológicos , Rubulavirus/genética , Infecções por Rubulavirus/genética , Infecções por Rubulavirus/virologia , Alinhamento de Sequência , Proteínas Virais/genética
8.
J Virol ; 84(20): 10681-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20702639

RESUMO

The African swine fever virus (ASFV) DP71L protein is present in all isolates as either a short form of 70 to 72 amino acids or a long form of about 184 amino acids, and both of these share sequence similarity to the C-terminal domain of the herpes simplex virus ICP34.5 protein and cellular protein GADD34. In the present study we expressed DP71L in different mammalian cells and demonstrated that DP71L causes dephosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) in resting cells and during chemical-induced endoplasmic reticulum stress and acts to enhance expression of cotransfected reporter genes. We showed that DP71L binds to all the three isoforms (α, ß, and γ) of the protein phosphatase 1 catalytic subunit (PP1c) and acts by recruiting PP1c to eIF2α. We also showed that DP71L inhibits the induction of ATF4 and its downstream target, CHOP. We investigated the eIF2α phosphorylation status and induction of CHOP in porcine macrophages infected by two ASFV field isolates, Malawi Lil20/1 and Benin 97/1, and two DP71L deletion mutants, MalawiΔNL and E70ΔNL. Our results showed that deletion of the DP71L gene did not cause an increase in the level of eIF2α phosphorylation or induction of CHOP, indicating that DP71L is not the only factor required by the virus to control the phosphorylation level of eIF2α during infection. We therefore hypothesize that ASFV has other mechanisms to prevent the eIF2α phosphorylation and the subsequent protein synthesis inhibition.


Assuntos
Vírus da Febre Suína Africana/fisiologia , Vírus da Febre Suína Africana/patogenicidade , Fator de Iniciação 2 em Eucariotos/metabolismo , Proteína Fosfatase 1/metabolismo , Fator de Transcrição CHOP/biossíntese , Proteínas Virais/fisiologia , Febre Suína Africana/genética , Febre Suína Africana/metabolismo , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Animais , Células Cultivadas , Técnicas de Inativação de Genes , Genes Virais , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Técnicas In Vitro , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Fosforilação , Ligação Proteica , Proteína Fosfatase 1/antagonistas & inibidores , Proteína Fosfatase 1/genética , RNA Interferente Pequeno/genética , Suínos , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética
9.
Viruses ; 13(4)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807175

RESUMO

Type I interferons (IFNs) are produced by most cells in response to virus infection and stimulate a program of anti-viral gene expression in neighboring cells to suppress virus replication. Type III IFNs have similar properties, however their effects are limited to epithelial cells at mucosal surfaces due to restricted expression of the type III IFN receptor. Rotavirus (RV) replicates in intestinal epithelial cells that respond predominantly to type III IFNs, and it has been shown that type III rather than type I IFNs are important for controlling RV infections in vivo. The RV NSP1 protein antagonizes the host type I IFN response by targeting IRF-3, IRF-5, IRF-7, or ß-TrCP for proteasome-mediated degradation in a strain-specific manner. Here we provide the first demonstration that NSP1 proteins from several human and animal RV strains antagonize type III as well as type I IFN induction. We also show that NSP1 is a potent inhibitor of IRF-1, a previously undescribed property of NSP1 which is conserved among human and animal RVs. Interestingly, all NSP1 proteins were substantially more effective inhibitors of IRF-1 than either IRF-3 or IRF-7 which has significance for evasion of basal anti-viral immunity and type III IFN induction in the intestinal epithelium.


Assuntos
Células Epiteliais/virologia , Interferon Tipo I/antagonistas & inibidores , Interferons/antagonistas & inibidores , Rotavirus/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Animais , Células Epiteliais/imunologia , Células HEK293 , Humanos , Fator Regulador 1 de Interferon/antagonistas & inibidores , Fator Regulador 1 de Interferon/imunologia , Interferon Tipo I/imunologia , Interferons/imunologia , Intestinos/citologia , Rotavirus/química , Rotavirus/isolamento & purificação
10.
J Gen Virol ; 91(Pt 3): 721-6, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19906943

RESUMO

Classical swine fever is a notifiable disease of pigs. The causative agent, classical swine fever virus (CSFV), is highly contagious and causes mild to severe haemorrhagic disease depending on the virulence of the strain. The RNA genome of CSFV is translated as a single polyprotein that is processed to yield 12 proteins. Like other pestiviruses, the first protein to be translated is the N-terminal autoprotease termed N(pro). A novel pestiviral protein with no known cellular homologues, N(pro) antagonizes type I interferon (IFN) induction by binding and targeting the transcription factor IFN regulatory factor 3 (IRF-3) for ubiquitin-dependent proteasomal degradation. In this study, CSFV-infected PK-15 cells and stable cell lines were used to show that N(pro) is itself an unstable protein that is targeted for proteasomal degradation in a ubiquitin-dependent manner. In addition, N(pro) is not degraded as a direct consequence of its ability to interact with IRF-3 or to target IRF-3 for proteasomal degradation.


Assuntos
Vírus da Febre Suína Clássica/imunologia , Endopeptidases/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Suínos , Ubiquitina/metabolismo
11.
PLoS One ; 12(5): e0177300, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28475628

RESUMO

Peste des petits ruminants virus (PPRV) is a morbillivirus that produces clinical disease in goats and sheep. We have studied the induction of interferon-ß (IFN-ß) following infection of cultured cells with wild-type and vaccine strains of PPRV, and the effects of such infection with PPRV on the induction of IFN-ß through both MDA-5 and RIG-I mediated pathways. Using both reporter assays and direct measurement of IFN-ß mRNA, we have found that PPRV infection induces IFN-ß only weakly and transiently, and the virus can actively block the induction of IFN-ß. We have also generated mutant PPRV that lack expression of either of the viral accessory proteins (V&C) to characterize the role of these proteins in IFN-ß induction during virus infection. Both PPRV_ΔV and PPRV_ΔC were defective in growth in cell culture, although in different ways. While the PPRV V protein bound to MDA-5 and, to a lesser extent, RIG-I, and over-expression of the V protein inhibited both IFN-ß induction pathways, PPRV lacking V protein expression can still block IFN-ß induction. In contrast, PPRV C bound to neither MDA-5 nor RIG-I, but PPRV lacking C protein expression lost the ability to block both MDA-5 and RIG-I mediated activation of IFN-ß. These results shed new light on the inhibition of the induction of IFN-ß by PPRV.


Assuntos
Fibroblastos/metabolismo , Doenças das Cabras/virologia , Interferon Tipo I/metabolismo , Peste dos Pequenos Ruminantes/virologia , Vírus da Peste dos Pequenos Ruminantes/metabolismo , Animais , Fibroblastos/virologia , Doenças das Cabras/metabolismo , Cabras , Peste dos Pequenos Ruminantes/metabolismo
12.
Viruses ; 9(7)2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28703784

RESUMO

Modern vaccinology has increasingly focused on non-living vaccines, which are more stable than live-attenuated vaccines but often show limited immunogenicity. Immunostimulatory substances, known as adjuvants, are traditionally used to increase the magnitude of protective adaptive immunity in response to a pathogen-associated antigen. Recently developed adjuvants often include substances that stimulate pattern recognition receptors (PRRs), essential components of innate immunity required for the activation of antigen-presenting cells (APCs), which serve as a bridge between innate and adaptive immunity. Nearly all PRRs are potential targets for adjuvants. Given the recent success of toll-like receptor (TLR) agonists in vaccine development, molecules with similar, but additional, immunostimulatory activity, such as defective interfering particles (DIPs) of viruses, represent attractive candidates for vaccine adjuvants. This review outlines some of the recent advances in vaccine development related to the use of TLR agonists, summarizes the current knowledge regarding DIP immunogenicity, and discusses the potential applications of DIPs in vaccine adjuvantation.


Assuntos
Adjuvantes Imunológicos , Vírus Defeituosos/imunologia , Receptores de Reconhecimento de Padrão/agonistas , Receptores de Reconhecimento de Padrão/imunologia , Vacinas/química , Imunidade Adaptativa , Animais , Células Apresentadoras de Antígenos/imunologia , Genoma Viral , Humanos , Imunidade Inata , Imunogenicidade da Vacina , Camundongos , Receptores Toll-Like/agonistas , Vacinas/administração & dosagem
13.
Nucleic Acids Res ; 31(12): 3016-26, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12799427

RESUMO

We have identified two novel proteins that interact specifically with the C-terminal repression domain of Interferon Regulatory Factor-2 (IRF-2). These proteins, which we term IRF-2 binding proteins 1 and 2 (IRF-2BP1 and IRF-2BP2, the latter having two splicing isoforms, A and B), are nuclear proteins, and have the properties of IRF-2-dependent transcriptional co-repressors that can inhibit both enhancer-activated and basal transcription in a manner that is not dependent upon histone deacetylation. IRF-2BP1 and IRF-2BP2A/B contain an N-terminal zinc finger and a C-terminal RING finger domain of the C3HC4 subclass, but show no homology to other known transcriptional regulators; they therefore define a new family of co- repressor proteins. An alternatively spliced form of IRF-2 that lacks two amino acids (valines 177 and 178) in the central portion of the protein (IRF-2[S]) cannot bind to these co-repressors and cannot mediate repression despite having the same C- terminal repression domain as IRF-2, suggesting that the relative conformation of the DNA binding domain and the C-terminal region of IRF-2 is crucial for transcriptional repression.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição , Processamento Alternativo , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte/química , Clonagem Molecular , DNA Complementar/isolamento & purificação , Proteínas de Ligação a DNA/química , Inativação Gênica , Células HeLa , Inibidores de Histona Desacetilases , Humanos , Fator Regulador 2 de Interferon , Dados de Sequência Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Alinhamento de Sequência , Transcrição Gênica , Ubiquitina-Proteína Ligases
14.
Vet Immunol Immunopathol ; 100(3-4): 135-44, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15207451

RESUMO

Non-cytopathogenic bovine viral diarrhoea virus (ncpBVDV) has previously been shown to inhibit the function of interferon regulatory factor-3 in cultured cells [J. Virol. 76 (2002) 8979]. In this study, we show that, like ncpBVDV, when cells were previously exposed to cytopathogenic BVDV (cpBVDV) the appearance of an IRF-3-DNA complex from nuclear extracts that can be induced by heterologous virus infection was not observed. Infection of cells with ncpBVDV or cpBVDV resulted in neither the translocation of IRF-7 from the cytoplasm to the nucleus of infected cells, nor an inhibition of its nuclear translocation in cells super-infected by Semliki Forest Virus. We conclude that cpBVDV and ncpBVDV both share the ability to inhibit the full function of IRF-3 but neither stimulate or block the nuclear uptake of IRF-7.


Assuntos
Doença das Mucosas por Vírus da Diarreia Viral Bovina/imunologia , Proteínas de Ligação a DNA/imunologia , Vírus da Diarreia Viral Bovina/imunologia , Fatores de Transcrição/imunologia , Animais , Apoptose/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/metabolismo , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Bovinos , Células Cultivadas , Efeito Citopatogênico Viral/imunologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Vírus da Diarreia Viral Bovina/patogenicidade , Ensaio de Desvio de Mobilidade Eletroforética/veterinária , Imunofluorescência/veterinária , Fator Regulador 3 de Interferon , Fator Regulador 7 de Interferon , Proteínas Proto-Oncogênicas c-jun/imunologia , Vírus da Floresta de Semliki/imunologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Virulência
15.
PLoS One ; 8(5): e64202, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23671710

RESUMO

The DExD/H box RNA helicases retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation associated gene-5 (mda-5) sense viral RNA in the cytoplasm of infected cells and activate signal transduction pathways that trigger the production of type I interferons (IFNs). Laboratory of genetics and physiology 2 (LGP2) is thought to influence IFN production by regulating the activity of RIG-I and mda-5, although its mechanism of action is not known and its function is controversial. Here we show that expression of LGP2 potentiates IFN induction by polyinosinic-polycytidylic acid [poly(I:C)], commonly used as a synthetic mimic of viral dsRNA, and that this is particularly significant at limited levels of the inducer. The observed enhancement is mediated through co-operation with mda-5, which depends upon LGP2 for maximal activation in response to poly(I:C). This co-operation is dependent upon dsRNA binding by LGP2, and the presence of helicase domain IV, both of which are required for LGP2 to interact with mda-5. In contrast, although RIG-I can also be activated by poly(I:C), LGP2 does not have the ability to enhance IFN induction by RIG-I, and instead acts as an inhibitor of RIG-I-dependent poly(I:C) signaling. Thus the level of LGP2 expression is a critical factor in determining the cellular sensitivity to induction by dsRNA, and this may be important for rapid activation of the IFN response at early times post-infection when the levels of inducer are low.


Assuntos
RNA Helicases DEAD-box/metabolismo , RNA Helicases/metabolismo , RNA de Cadeia Dupla/metabolismo , Sítios de Ligação/genética , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células HEK293 , Humanos , Immunoblotting , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon , Poli I-C/farmacologia , Ligação Proteica , RNA Helicases/genética , Interferência de RNA , RNA de Cadeia Dupla/genética , Receptores Imunológicos , Ativação Transcricional/efeitos dos fármacos , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/genética , Proteínas Virais/metabolismo
16.
Insect Biochem Mol Biol ; 41(8): 628-36, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21501684

RESUMO

Regulation of the Aedes albopictus cecropin A1 promoter was studied to provide insight into the transcriptional control of this antimicrobial peptide (AMP) gene in mosquitoes. Gene expression levels of cecropin A1 increased in A. albopictus C6/36 cells in response to heat-killed Escherichiacoli. Reporter gene assays incorporating -757 to +32 of the A. albopictus cecropin A1 promoter revealed that E. coli could induce expression in these cells with more pronounced expression than that seen with lipopolysaccharide (LPS). Analysis of deletion constructs demonstrated that the 5' boundary of the regulatory region for the activation of this AMP was located between -173 and -64. Western blotting with anti-phospho-specific antibodies demonstrated that p38 mitogen-activated protein kinase (p38 MAPK) and c-Jun N-terminal kinase (JNK) were activated by LPS, whereas only p38 MAPK was activated by E. coli. Moreover, pharmacological experiments revealed that pre-incubation of cells with the p38 MAPK inhibitor SB203580 resulted in a striking activation of the cecropin A1 promoter following immune challenge, demonstrating that p38 MAPK negatively regulates cecropin A1 promoter activity. Finally the region required for the negative regulation by p38 MAPK was identified as being between -173 and -64. This report is the first to show involvement of the p38 MAPK pathway in the negative regulation of AMP production in a mosquito.


Assuntos
Aedes/genética , Cecropinas/genética , Expressão Gênica/efeitos dos fármacos , Proteínas de Insetos/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Aedes/imunologia , Aedes/metabolismo , Animais , Anticorpos Fosfo-Específicos/análise , Cecropinas/imunologia , Cecropinas/metabolismo , Células Cultivadas , Escherichia coli/química , Expressão Gênica/imunologia , Regulação da Expressão Gênica , Genes Reporter , Temperatura Alta , Imidazóis/farmacologia , Imunidade Inata , Proteínas de Insetos/imunologia , Proteínas de Insetos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/farmacologia , Luciferases/análise , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Viabilidade Microbiana , Regiões Promotoras Genéticas/imunologia , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
17.
J Interferon Cytokine Res ; 29(9): 539-47, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19702509

RESUMO

Experimentally, paramyxoviruses are conventionally considered good inducers of type I interferons (IFN-alpha/beta), and have been used as agents in the commercial production of human IFN-alpha. However, in the last few years it has become clear that viruses in general mount a major challenge to the IFN system, and paramyxoviruses are no exception. Indeed, most paramyxoviruses encode mechanisms to inhibit both the production of, and response to, type I IFN. Here we review our knowledge of the type I IFN-inducing signals (by so-called pathogen-associated molecular patterns, or PAMPs) produced during paramyxovirus infections, and discuss how paramyxoviruses limit the production of PAMPs and inhibit the cellular responses to PAMPs by interfering with the activities of the pattern recognition receptors (PRRs), mda-5, and RIG-I, as well as downstream components in the type I IFN induction cascades.


Assuntos
Antígenos de Superfície/metabolismo , Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Paramyxoviridae/imunologia , Animais , RNA Helicases DEAD-box/metabolismo , Proteínas Ligadas por GPI , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon , Paramyxoviridae/patogenicidade , Infecções por Paramyxoviridae/genética , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Proteínas Recombinantes , Transdução de Sinais
18.
J Gen Virol ; 89(Pt 1): 1-47, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18089727

RESUMO

The interferon (IFN) system is an extremely powerful antiviral response that is capable of controlling most, if not all, virus infections in the absence of adaptive immunity. However, viruses can still replicate and cause disease in vivo, because they have some strategy for at least partially circumventing the IFN response. We reviewed this topic in 2000 [Goodbourn, S., Didcock, L. & Randall, R. E. (2000). J Gen Virol 81, 2341-2364] but, since then, a great deal has been discovered about the molecular mechanisms of the IFN response and how different viruses circumvent it. This information is of fundamental interest, but may also have practical application in the design and manufacture of attenuated virus vaccines and the development of novel antiviral drugs. In the first part of this review, we describe how viruses activate the IFN system, how IFNs induce transcription of their target genes and the mechanism of action of IFN-induced proteins with antiviral action. In the second part, we describe how viruses circumvent the IFN response. Here, we reflect upon possible consequences for both the virus and host of the different strategies that viruses have evolved and discuss whether certain viruses have exploited the IFN response to modulate their life cycle (e.g. to establish and maintain persistent/latent infections), whether perturbation of the IFN response by persistent infections can lead to chronic disease, and the importance of the IFN system as a species barrier to virus infections. Lastly, we briefly describe applied aspects that arise from an increase in our knowledge in this area, including vaccine design and manufacture, the development of novel antiviral drugs and the use of IFN-sensitive oncolytic viruses in the treatment of cancer.


Assuntos
Interferons/fisiologia , Viroses/tratamento farmacológico , Fenômenos Fisiológicos Virais , Vírus/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Humanos , Interferons/genética , Interferons/uso terapêutico , RNA Viral/genética , Transdução de Sinais , Vírus/genética , Vírus/crescimento & desenvolvimento
19.
Virology ; 359(1): 190-200, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17049367

RESUMO

The induction of IFN-beta by the paramyxovirus PIV5 (formerly known as SV5) is limited by the action of the viral V protein that targets the cellular RNA helicase mda-5. Here we show that 12 other paramyxoviruses also target mda-5 by a direct interaction between the conserved cysteine-rich C-terminus of their V proteins and the helicase domain of mda-5. The inhibition of IFN-beta induction is not species-restricted, being observed in a range of mammalian cells as well as in avian cells, and we show that the inhibition of mda-5 function is also not restricted to mammalian cells. In contrast, the V proteins do not bind to the related RNA helicase RIG-I and do not inhibit its activity. The relative contributions of mda-5 and RIG-I to IFN-beta induction are discussed.


Assuntos
RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/metabolismo , Vírus da Parainfluenza 5/fisiologia , Proteínas Estruturais Virais/metabolismo , Sequência de Aminoácidos , Animais , Aves , Bovinos , Linhagem Celular , Chlorocebus aethiops , RNA Helicases DEAD-box/química , Humanos , Imunoprecipitação , Interferon beta/biossíntese , Dados de Sequência Molecular , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Técnicas do Sistema de Duplo-Híbrido , Proteínas Estruturais Virais/química
20.
J Gen Virol ; 88(Pt 11): 3002-3006, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17947522

RESUMO

Classical swine fever virus (CSFV) is a member of the genus Pestivirus in the family Flaviviridae. The N(pro) product of CSFV targets the host's innate immune response and can prevent the production of type I interferon (IFN). The mechanism by which CSFV orchestrates this inhibition was investigated and it is shown that, like the related pestivirus bovine viral diarrhea virus (BVDV), this involves the N(pro) protein targeting interferon regulatory factor-3 (IRF-3) for degradation by proteasomes and thus preventing IRF-3 from activating transcription from the IFN-beta promoter. Like BVDV, the steady-state levels of IRF-3 mRNA are not reduced markedly by CSFV infection or N(pro) overexpression. Moreover, IFN-alpha stimulation of CSFV-infected cells induces the antiviral protein MxA, indicating that, as in BVDV-infected cells, the JAK/STAT pathway is not targeted for inhibition.


Assuntos
Vírus da Febre Suína Clássica/imunologia , Endopeptidases/imunologia , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/imunologia , Proteínas Virais/imunologia , Animais , Western Blotting , Linhagem Celular , Complexo de Endopeptidases do Proteassoma/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA