Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
PLoS Pathog ; 19(2): e1011170, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36802406

RESUMO

Viruses have evolved countless mechanisms to subvert and impair the host innate immune response. Measles virus (MeV), an enveloped, non-segmented, negative-strand RNA virus, alters the interferon response through different mechanisms, yet no viral protein has been described as directly targeting mitochondria. Among the crucial mitochondrial enzymes, 5'-aminolevulinate synthase (ALAS) is an enzyme that catalyzes the first step in heme biosynthesis, generating 5'-aminolevulinate from glycine and succinyl-CoA. In this work, we demonstrate that MeV impairs the mitochondrial network through the V protein, which antagonizes the mitochondrial enzyme ALAS1 and sequesters it to the cytosol. This re-localization of ALAS1 leads to a decrease in mitochondrial volume and impairment of its metabolic potential, a phenomenon not observed in MeV deficient for the V gene. This perturbation of the mitochondrial dynamics demonstrated both in culture and in infected IFNAR-/- hCD46 transgenic mice, causes the release of mitochondrial double-stranded DNA (mtDNA) in the cytosol. By performing subcellular fractionation post infection, we demonstrate that the most significant source of DNA in the cytosol is of mitochondrial origin. Released mtDNA is then recognized and transcribed by the DNA-dependent RNA polymerase III. The resulting double-stranded RNA intermediates will be captured by RIG-I, ultimately initiating type I interferon production. Deep sequencing analysis of cytosolic mtDNA editing divulged an APOBEC3A signature, primarily analyzed in the 5'TpCpG context. Finally, in a negative feedback loop, APOBEC3A an interferon inducible enzyme will orchestrate the catabolism of mitochondrial DNA, decrease cellular inflammation, and dampen the innate immune response.


Assuntos
Interferons , Mitocôndrias , Camundongos , Animais , Mitocôndrias/metabolismo , Vírus do Sarampo , 5-Aminolevulinato Sintetase/genética , 5-Aminolevulinato Sintetase/metabolismo , DNA Mitocondrial
2.
J Virol ; 97(2): e0194322, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36722971

RESUMO

Virus replication depends on a complex interplay between viral and host proteins. In the case of African swine fever virus (ASFV), a large DNA virus, only a few virus-host protein-protein interactions have been identified to date. In this study, we demonstrate that the ASFV protein CP204L interacts with the cellular homotypic fusion and protein sorting (HOPS) protein VPS39, blocking its association with the lysosomal HOPS complex, which modulates endolysosomal trafficking and promotes lysosome clustering. Instead, CP204L and VPS39 are targeted to virus factories and localized at the periphery of the virus DNA replication sites. Furthermore, we show that loss of VPS39 reduces the levels of virus proteins synthesized in the early phase of infection and delays ASFV replication but does not completely inhibit it. Collectively, these results identify a novel virus-host protein interaction that modulates host membrane rearrangement during infection and provide evidence that CP204L is a multifunctional protein engaged in distinct steps of the ASFV life cycle. IMPORTANCE African swine fever virus (ASFV) was first identified over a hundred years ago. Since then, much effort has been made to understand the pathogenesis of ASFV. However, the specific roles of many individual ASFV proteins during the infection remain enigmatic. This study provides evidence that CP204L, one of the most abundant ASFV proteins, modulates endosomal trafficking during virus infection. Through protein-protein interaction, CP204L prevents the recruitment of VPS39 to the endosomal and lysosomal membranes, resulting in their accumulation. Consequently, CP204L and VPS39 become sequestered in the ASFV replication and assembly site, known as the virus factory. These results uncover a novel function of viral protein CP204L and extend our understanding of complex interaction between virus and host.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Proteínas Virais , Replicação Viral , Animais , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/fisiologia , Lisossomos/metabolismo , Transporte Proteico , Suínos , Vacúolos/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
3.
J Biol Chem ; 297(3): 101081, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34403699

RESUMO

The human APOBEC3A (A3A) cytidine deaminase is a powerful DNA mutator enzyme recognized as a major source of somatic mutations in tumor cell genomes. However, there is a discrepancy between APOBEC3A mRNA levels after interferon stimulation in myeloid cells and A3A detection at the protein level. To understand this difference, we investigated the expression of two novel alternative "A3Alt" proteins encoded in the +1-shifted reading frame of the APOBEC3A gene. A3Alt-L and its shorter isoform A3Alt-S appear to be transmembrane proteins targeted to the mitochondrial compartment that induce membrane depolarization and apoptosis. Thus, the APOBEC3A gene represents a new example wherein a single gene encodes two proapoptotic proteins, A3A cytidine deaminases that target the genome and A3Alt proteins that target mitochondria.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/fisiologia , Mitocôndrias/genética , Proteínas/genética , Proteínas/fisiologia , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Citidina Desaminase/metabolismo , DNA/genética , Mutação da Fase de Leitura/genética , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Genoma/genética , Humanos , Mitocôndrias/metabolismo , Mutação/genética , Proteínas/metabolismo , RNA Mensageiro/genética , Fases de Leitura/genética
4.
Virologie (Montrouge) ; 26(5): 387-400, 2022 09 01.
Artigo em Francês | MEDLINE | ID: mdl-36413123

RESUMO

African swine fever (ASF) is a highly pathogenic disease causing haemorrhagic fever in domestic and wild swine. It is responsible for numerous epizootics, particularly in Europe and Asia, causing major economic losses for the pig industry. African Swine Fever virus (ASFV) is the etiological agent responsible for this disease. It is a very large double-stranded DNA virus, encoding for over 150 proteins. Various studies have shown that there is a close relationship between the ability of some viral proteins to inhibit the type I interferon (IFNI) response and the attenuation and virulence processes of ASFV. This review describes the mechanisms of inhibition of the IFN-I response by ASFV proteins, which provide a molecular explanation of how ASFV escapes the innate immune response.


La peste porcine africaine (PPA) est une maladie hautement pathogène causant une fièvre hémorragique chez les suidés domestiques et sauvages. Elle est responsable de nombreuses épizooties notamment en Europe et en Asie, causant de grandes pertes économiques pour la filière porcine. Le virus de la peste porcine africaine (ASFV) est l'agent étiologique responsable de cette maladie. C'est un virus avec un génome à ADN double brin de grande taille, codant pour plus de 150 protéines. Différents travaux ont montré qu'il existe une étroite relation entre la capacité de certaines protéines virales à inhiber la réponse interféron de type I (IFN-I) et les processus d'atténuation et de virulence pour l'ASFV. Cette revue décrit les mécanismes d'inhibition de la réponse IFN-I par les protéines d'ASFV permettant d'expliquer sur le plan moléculaire l'échappement à la réponse immunitaire innée.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Suínos , Animais , Vírus da Febre Suína Africana/genética , Imunidade Inata/genética , Virulência
5.
J Immunol ; 202(5): 1479-1493, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30683700

RESUMO

Herpes simplex encephalitis (HSE), caused by HSV type 1 (HSV-1) infection, is an acute neuroinflammatory condition of the CNS and remains the most common type of sporadic viral encephalitis worldwide. Studies in humans have shown that susceptibility to HSE depends in part on the genetic make-up of the host, with deleterious mutations in the TLR3/type I IFN axis underlying some cases of childhood HSE. Using an in vivo chemical mutagenesis screen for HSV-1 susceptibility in mice, we identified a susceptible pedigree carrying a causal truncating mutation in the Rel gene (RelC307X ), encoding for the NF-κB transcription factor subunit c-Rel. Like Myd88-/- and Irf3-/- mice, RelC307X mice were susceptible to intranasal HSV-1 infection. Reciprocal bone marrow transfers into lethally irradiated hosts suggested that defects in both hematopoietic and CNS-resident cellular compartments contributed together to HSE susceptibility in RelC307X mice. Although the RelC307X mutation maintained cell-intrinsic antiviral control, it drove increased apoptotic cell death in infected fibroblasts. Moreover, reduced numbers of CD4+CD25+Foxp3+ T regulatory cells, and dysregulated NK cell and CD4+ effector T cell responses in infected RelC307X animals, indicated that protective immunity was also compromised in these mice. In the CNS, moribund RelC307X mice failed to control HSV-1 viral replication in the brainstem and cerebellum, triggering cell death and elevated expression of Ccl2, Il6, and Mmp8 characteristic of HSE neuroinflammation and pathology. In summary, our work implicates c-Rel in both CNS-resident cell survival and lymphocyte responses to HSV-1 infection and as a novel cause of HSE disease susceptibility in mice.


Assuntos
Sistema Nervoso Central/imunologia , Encefalite por Herpes Simples/imunologia , Inflamação/imunologia , Replicação Viral/imunologia , Animais , Chlorocebus aethiops , Encefalite por Herpes Simples/virologia , Inflamação/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Vero
6.
J Virol ; 93(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31167915

RESUMO

Bluetongue virus (BTV) is an arbovirus transmitted by blood-feeding midges to a wide range of wild and domestic ruminants. In this report, we showed that BTV, through its nonstructural protein NS3 (BTV-NS3), is able to activate the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway, as assessed by phosphorylation levels of ERK1/2 and the translation initiation factor eukaryotic translation initiation factor 4E (eIF4E). By combining immunoprecipitation of BTV-NS3 and mass spectrometry analysis from both BTV-infected and NS3-transfected cells, we identified the serine/threonine-protein kinase B-Raf (BRAF), a crucial player in the MAPK/ERK pathway, as a new cellular interactor of BTV-NS3. BRAF silencing led to a significant decrease in the MAPK/ERK activation by BTV, supporting a model wherein BTV-NS3 interacts with BRAF to activate this signaling cascade. This positive regulation acts independently of the role of BTV-NS3 in counteracting the induction of the alpha/beta interferon response. Furthermore, the intrinsic ability of BTV-NS3 to bind BRAF and activate the MAPK/ERK pathway is conserved throughout multiple serotypes/strains but appears to be specific to BTV compared to other members of Orbivirus genus. Inhibition of MAPK/ERK pathway with U0126 reduced viral titers, suggesting that BTV manipulates this pathway for its own replication. Altogether, our data provide molecular mechanisms that unravel a new essential function of NS3 during BTV infection.IMPORTANCE Bluetongue virus (BTV) is responsible of the arthropod-borne disease bluetongue (BT) transmitted to ruminants by blood-feeding midges. In this report, we found that BTV, through its nonstructural protein NS3 (BTV-NS3), interacts with BRAF, a key component of the MAPK/ERK pathway. In response to growth factors, this pathway promotes cell survival and increases protein translation. We showed that BTV-NS3 enhances the MAPK/ERK pathway, and this activation is BRAF dependent. Treatment of MAPK/ERK pathway with the pharmacologic inhibitor U0126 impairs viral replication, suggesting that BTV manipulates this pathway for its own benefit. Our results illustrate, at the molecular level, how a single virulence factor has evolved to target a cellular function to increase its viral replication.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/metabolismo , Bluetongue/virologia , Interações Hospedeiro-Patógeno , Sistema de Sinalização das MAP Quinases , Proteínas não Estruturais Virais/metabolismo , Animais , Vírus Bluetongue/patogenicidade , Linhagem Celular , Proteínas de Ligação a DNA , Humanos , Interferons/metabolismo , Fosforilação , Ligação Proteica , Transporte Proteico , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Fatores de Transcrição , Fatores de Virulência , Replicação Viral
7.
Int J Mol Sci ; 21(15)2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32722052

RESUMO

Microvascular endothelial cells constitute potential targets for exogenous microorganisms, in particular for vector-borne pathogens. Their phenotypic and functional variations according to the organs they are coming from provide an explanation of the organ selectivity expressed in vivo by pathogens. In order to make available relevant tools for in vitro studies of infection mechanisms, our aim was to immortalize bovine organospecific endothelial cells but also to assess their permissivity to viral infection. Using transfection with SV40 large T antigen, six bovine microvascular endothelial cell lines from various organs and one macrovascular cell line from an umbilical cord were established. They display their own panel of endothelial progenitor/mature markers, as assessed by flow cytometry and RT-qPCR, as well as the typical angiogenesis capacity. Using both Bluetongue and foot-and-mouth disease viruses, we demonstrate that some cell lines are preferentially infected. In addition, they can be transfected and are able to express viral proteins such as BTV8-NS3. Such microvascular endothelial cell lines bring innovative tools for in vitro studies of infection by viruses or bacteria, allowing for the study of host-pathogen interaction mechanisms with the actual in vivo target cells. They are also suitable for applications linked to microvascularization, such as anti-angiogenic and anti-tumor research, growing fields in veterinary medicine.


Assuntos
Células Endoteliais/metabolismo , Microvasos/metabolismo , Modelos Biológicos , Viroses , Animais , Bovinos , Linhagem Celular , Células Endoteliais/patologia , Células Endoteliais/virologia , Microvasos/patologia , Microvasos/virologia
8.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795408

RESUMO

Schmallenberg virus (SBV) was discovered in Germany in late 2011 and then spread rapidly to many European countries. SBV is an orthobunyavirus that causes abortion and congenital abnormalities in ruminants. A virus-encoded nonstructural protein, termed NSs, is a major virulence factor of SBV, and it is known to promote the degradation of Rpb1, a subunit of the RNA polymerase II (Pol II) complex, and therefore hampers global cellular transcription. In this study, we found that NSs is mainly localized in the nucleus of infected cells and specifically appears to target the nucleolus through a nucleolar localization signal (NoLS) localized between residues 33 and 51 of the protein. NSs colocalizes with nucleolar markers such as B23 (nucleophosmin) and fibrillarin. We observed that in SBV-infected cells, B23 undergoes a nucleolus-to-nucleoplasm redistribution, evocative of virus-induced nucleolar disruption. In contrast, the nucleolar pattern of B23 was unchanged upon infection with an SBV recombinant mutant with NSs lacking the NoLS motif (SBVΔNoLS). Interestingly, unlike wild-type SBV, the inhibitory activity of SBVΔNoLS toward RNA Pol II transcription is impaired. Overall, our results suggest that a putative link exists between NSs-induced nucleolar disruption and its inhibitory function on cellular transcription, which consequently precludes the cellular antiviral response and/or induces cell death. IMPORTANCE: Schmallenberg virus (SBV) is an emerging arbovirus of ruminants that spread in Europe between 2011 and 2013. SBV induces fetal abnormalities during gestation, with the central nervous system being one of the most affected organs. The virus-encoded NSs protein acts as a virulence factor by impairing host cell transcription. Here, we show that NSs contains a nucleolar localization signal (NoLS) and induces disorganization of the nucleolus. The NoLS motif in the SBV NSs is absolutely necessary for virus-induced inhibition of cellular transcription. To our knowledge, this is the first report of nucleolar functions for NSs within the Bunyaviridae family.


Assuntos
Nucléolo Celular/virologia , Células Ependimogliais/virologia , Interações Hospedeiro-Patógeno , Orthobunyavirus/patogenicidade , RNA Polimerase II/química , Proteínas não Estruturais Virais/química , Animais , Linhagem Celular Transformada , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , Plexo Corióideo/citologia , Plexo Corióideo/metabolismo , Plexo Corióideo/virologia , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Células Ependimogliais/metabolismo , Células Ependimogliais/ultraestrutura , Regulação da Expressão Gênica , Células HeLa , Humanos , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Orthobunyavirus/genética , Orthobunyavirus/metabolismo , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteólise , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Ovinos , Transdução de Sinais , Transcrição Gênica , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
9.
PLoS Pathog ; 9(10): e1003678, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098125

RESUMO

Searching for stimulators of the innate antiviral response is an appealing approach to develop novel therapeutics against viral infections. Here, we established a cell-based reporter assay to identify compounds stimulating expression of interferon-inducible antiviral genes. DD264 was selected out of 41,353 compounds for both its immuno-stimulatory and antiviral properties. While searching for its mode of action, we identified DD264 as an inhibitor of pyrimidine biosynthesis pathway. This metabolic pathway was recently identified as a prime target of broad-spectrum antiviral molecules, but our data unraveled a yet unsuspected link with innate immunity. Indeed, we showed that DD264 or brequinar, a well-known inhibitor of pyrimidine biosynthesis pathway, both enhanced the expression of antiviral genes in human cells. Furthermore, antiviral activity of DD264 or brequinar was found strictly dependent on cellular gene transcription, nuclear export machinery, and required IRF1 transcription factor. In conclusion, the antiviral property of pyrimidine biosynthesis inhibitors is not a direct consequence of pyrimidine deprivation on the virus machinery, but rather involves the induction of cellular immune response.


Assuntos
Infecções por Alphavirus/metabolismo , Antivirais/farmacologia , Vírus Chikungunya/metabolismo , Imunidade Inata/efeitos dos fármacos , Pirimidinas/biossíntese , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/genética , Infecções por Alphavirus/imunologia , Animais , Antivirais/química , Febre de Chikungunya , Vírus Chikungunya/genética , Vírus Chikungunya/imunologia , Chlorocebus aethiops , Células HeLa , Humanos , Imunidade Inata/imunologia , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 1 de Interferon/metabolismo , Pirimidinas/imunologia , Células Vero
10.
PLoS Pathog ; 9(9): e1003637, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068938

RESUMO

Herpes simplex encephalitis (HSE) is a lethal neurological disease resulting from infection with Herpes Simplex Virus 1 (HSV-1). Loss-of-function mutations in the UNC93B1, TLR3, TRIF, TRAF3, and TBK1 genes have been associated with a human genetic predisposition to HSE, demonstrating the UNC93B-TLR3-type I IFN pathway as critical in protective immunity to HSV-1. However, the TLR3, UNC93B1, and TRIF mutations exhibit incomplete penetrance and represent only a minority of HSE cases, perhaps reflecting the effects of additional host genetic factors. In order to identify new host genes, proteins and signaling pathways involved in HSV-1 and HSE susceptibility, we have implemented the first genome-wide mutagenesis screen in an in vivo HSV-1 infectious model. One pedigree (named P43) segregated a susceptible trait with a fully penetrant phenotype. Genetic mapping and whole exome sequencing led to the identification of the causative nonsense mutation L3X in the Receptor-type tyrosine-protein phosphatase C gene (Ptprc(L3X)), which encodes for the tyrosine phosphatase CD45. Expression of MCP1, IL-6, MMP3, MMP8, and the ICP4 viral gene were significantly increased in the brain stems of infected Ptprc(L3X) mice accounting for hyper-inflammation and pathological damages caused by viral replication. Ptprc(L3X) mutation drastically affects the early stages of thymocytes development but also the final stage of B cell maturation. Transfer of total splenocytes from heterozygous littermates into Ptprc(L3X) mice resulted in a complete HSV-1 protective effect. Furthermore, T cells were the only cell population to fully restore resistance to HSV-1 in the mutants, an effect that required both the CD4⁺ and CD8⁺ T cells and could be attributed to function of CD4⁺ T helper 1 (Th1) cells in CD8⁺ T cell recruitment to the site of infection. Altogether, these results revealed the CD45-mediated T cell function as potentially critical for infection and viral spread to the brain, and also for subsequent HSE development.


Assuntos
Códon sem Sentido , Encefalite por Herpes Simples/genética , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Imunidade Celular , Antígenos Comuns de Leucócito/metabolismo , Células Th1/imunologia , Animais , Tronco Encefálico/imunologia , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Tronco Encefálico/virologia , Células Cultivadas , Cruzamentos Genéticos , Suscetibilidade a Doenças , Encefalite por Herpes Simples/etiologia , Feminino , Estudo de Associação Genômica Ampla , Herpes Simples/patologia , Herpes Simples/fisiopatologia , Herpes Simples/virologia , Antígenos Comuns de Leucócito/genética , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutagênese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/virologia , Análise de Sobrevida , Células Th1/metabolismo , Células Th1/patologia , Células Th1/virologia
12.
Front Immunol ; 15: 1358219, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38529285

RESUMO

African swine fever virus represents a significant reemerging threat to livestock populations, as its incidence and geographic distribution have surged over the past decade in Europe, Asia, and Caribbean, resulting in substantial socio-economic burdens and adverse effects on animal health and welfare. In a previous report, we described the protective properties of our newly thermo-attenuated strain (ASFV-989) in pigs against an experimental infection of its parental Georgia 2007/1 virulent strain. In this new study, our objective was to characterize the molecular mechanisms underlying the attenuation of ASFV-989. We first compared the activation of type I interferon pathway in response to ASFV-989 and Georgia 2007/1 infections, employing both in vivo and in vitro models. Expression of IFN-α was significantly increased in porcine alveolar macrophages infected with ASFV-989 while pigs infected with Georgia 2007/1 showed higher IFN-α than those infected by ASFV-989. We also used a medium-throughput transcriptomic approach to study the expression of viral genes by both strains, and identified several patterns of gene expression. Subsequently, we investigated whether proteins encoded by the eight genes deleted in ASFV-989 contribute to the modulation of the type I interferon signaling pathway. Using different strategies, we showed that MGF505-4R interfered with the induction of IFN-α/ß pathway, likely through interaction with TRAF3. Altogether, our data reveal key differences between ASFV-989 and Georgia 2007/1 in their ability to control IFN-α/ß signaling and provide molecular mechanisms underlying the role of MGF505-4R as a virulence factor.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Suínos , Animais , Virulência , Macrófagos
13.
Mol Cell Proteomics ; 10(12): M110.007443, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21911578

RESUMO

RNA viruses exhibit small-sized genomes encoding few proteins, but still establish complex networks of interactions with host cell components to achieve replication and spreading. Ideally, these virus-host protein interactions should be mapped directly in infected cell culture, but such a high standard is often difficult to reach when using conventional approaches. We thus developed a new strategy based on recombinant viruses expressing tagged viral proteins to capture both direct and indirect physical binding partners during infection. As a proof of concept, we engineered a recombinant measles virus (MV) expressing one of its virulence factors, the MV-V protein, with a One-STrEP amino-terminal tag. This allowed virus-host protein complex analysis directly from infected cells by combining modified tandem affinity chromatography and mass spectrometry analysis. Using this approach, we established a prosperous list of 245 cellular proteins interacting either directly or indirectly with MV-V, and including four of the nine already known partners of this viral factor. These interactions were highly specific of MV-V because they were not recovered when the nucleoprotein MV-N, instead of MV-V, was tagged. Besides key components of the antiviral response, cellular proteins from mitochondria, ribosomes, endoplasmic reticulum, protein phosphatase 2A, and histone deacetylase complex were identified for the first time as prominent targets of MV-V and the critical role of the later protein family in MV replication was addressed. Most interestingly, MV-V showed some preferential attachment to essential proteins in the human interactome network, as assessed by centrality and interconnectivity measures. Furthermore, the list of MV-V interactors also showed a massive enrichment for well-known targets of other viruses. Altogether, this clearly supports our approach based on reverse genetics of viruses combined with high-throughput proteomics to probe the interaction network that viruses establish in infected cells.


Assuntos
Interações Hospedeiro-Patógeno , Vírus do Sarampo/fisiologia , Sarampo/virologia , Animais , Chlorocebus aethiops , RNA Helicases DEAD-box/isolamento & purificação , RNA Helicases DEAD-box/metabolismo , Células HEK293 , Histona Desacetilases/metabolismo , Humanos , Helicase IFIH1 Induzida por Interferon , Sarampo/metabolismo , Vírus do Sarampo/genética , Complexos Multiproteicos/isolamento & purificação , Complexos Multiproteicos/metabolismo , Organismos Geneticamente Modificados , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Genética Reversa , Fator de Transcrição STAT1/isolamento & purificação , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/isolamento & purificação , Fator de Transcrição STAT2/metabolismo , Sensibilidade e Especificidade , Espectrometria de Massas em Tandem , Células Vero , Fatores de Virulência/genética , Fatores de Virulência/isolamento & purificação , Fatores de Virulência/metabolismo , Replicação Viral
14.
Pathogens ; 12(6)2023 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-37375491

RESUMO

Bluetongue virus (BTV), a double-stranded RNA virus belonging to the Sedoreoviridae family, provokes an economically important disease in ruminants. In this study, we show that the production of activated caspase-1 and interleukin 1 beta (IL-1ß) is induced in BTV-infected cells. This response seems to require virus replication since a UV-inactivated virus is unable to activate this pathway. In NLRP3-/- cells, BTV could not trigger further IL-1ß synthesis, indicating that it occurs through NLRP3 inflammasome activation. Interestingly, we observed differential activation levels in bovine endothelial cells depending on the tissue origin. In particular, inflammasome activation was stronger in umbilical cord cells, suggesting that these cells are more prone to induce the inflammasome upon BTV infection. Finally, the strength of the inflammasome activation also depends on the BTV strain, which points to the importance of viral origin in inflammasome modulation. This work reports the crucial role of BTV in the activation of the NLRP3 inflammasome and further shows that this activation relies on BTV replication, strains, and cell types, thus providing new insights into BTV pathogenesis.

15.
Viruses ; 15(3)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36992375

RESUMO

Foot-and-mouth disease (FMD) is a highly contagious viral disease affecting cloven-hoofed animals. One of the issues related to this disease is the persistence of its causative agent, foot-and-mouth disease virus (FMDV). While the mechanisms of FMDV persistence remain unclear, there are clues that it may be related to protein-protein interactions (PPI) between viral proteins and cellular proteins involved in the interferon (IFN) response. Since FMDV persistence has been described in cattle, sheep and goats but not in swine, we screened PPI involving FMDV proteins and sixteen major type-I IFN pathway proteins from these four species by nanoluciferase-2-hybrid complementation assay, in order to identify new PPI and determine their host specificity. As the results concerning the 3Dpol were the most interesting in view of the limited data concerning its role in immune escape, we decided to focus particularly on this protein. The identified PPI were confirmed by GST pull-down. We identified PPI between 3Dpol and seven IFN pathway proteins, namely, IKKα, IKKε, IRF3, IRF7, NEMO, MDA5 and MAVS. These PPI are conserved among the four studied species, with the exception of the one between 3Dpol and MAVS, which was only found with the swine protein. We also showed, using luciferase reporter assays, that 3Dpol could inhibit the induction phase of the IFN pathway. These results demonstrate, for the first time, a putative role for 3Dpol in FMDV innate immune escape.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Interferon Tipo I , Suínos , Animais , Bovinos , Ovinos , Proteínas Virais/genética , Proteínas Virais/metabolismo
17.
Virologie (Montrouge) ; 16(5): 286-298, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-33065913

RESUMO

Host immunity against viruses integrates both innate and adaptive responses. Innate immunity represents the first line of defense triggered by viral infections whereas adaptive immunity allows for the establishment of long-term memory. Initially described for their antiviral activity, type I interferons are also involved in the induction of all immune responses. Since these cytokines are crucial for the establishment of an efficient antiviral response, viruses have developped mechanisms to block their expression and signals. The family of Paramyxoviridae presents a wonderful example of how viruses escape the antiviral response. Evasion strategies mainly involve the non structural proteins encoded by the gene of the phosphoprotein (P). These virulence factors can simultaneously or sequentially interact with cellular proteins through multiple interfaces and sophisticated mechanisms. This review describes the different mechanisms developped by paramyxoviruses to hijack the type I IFN response and molecular details, in particular protein-protein interactions, will be highlighted.

18.
Viruses ; 14(2)2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-35215776

RESUMO

Bluetongue virus (BTV) is the etiologic agent of a non-contagious arthropod-borne disease transmitted to wild and domestic ruminants. BTV induces a large panel of clinical manifestations ranging from asymptomatic infection to lethal hemorrhagic fever. Despite the fact that BTV has been studied extensively, we still have little understanding of the molecular determinants of BTV virulence. In our report, we have performed a comparative yeast two-hybrid (Y2H) screening approach to search direct cellular targets of the NS4 virulence factor encoded by two different serotypes of BTV: BTV8 and BTV27. This led to identifying Wilms' tumor 1-associated protein (WTAP) as a new interactor of the BTV-NS4. In contrast to BTV8, 1, 4 and 25, NS4 proteins from BTV27 and BTV30 are unable to interact with WTAP. This interaction with WTAP is carried by a peptide of 34 amino acids (NS422-55) within its putative coil-coiled structure. Most importantly, we showed that binding to WTAP is restored with a chimeric protein where BTV27-NS4 is substituted by BTV8-NS4 in the region encompassing residue 22 to 55. We also demonstrated that WTAP silencing reduces viral titers and the expression of viral proteins, suggesting that BTV-NS4 targets a cellular function of WTAP to increase its viral replication.


Assuntos
Vírus Bluetongue/metabolismo , Bluetongue/metabolismo , Bluetongue/virologia , Doenças dos Bovinos/metabolismo , Fatores de Processamento de RNA/metabolismo , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Bluetongue/genética , Vírus Bluetongue/química , Vírus Bluetongue/genética , Vírus Bluetongue/patogenicidade , Bovinos , Doenças dos Bovinos/genética , Doenças dos Bovinos/virologia , Interações Hospedeiro-Patógeno , Ligação Proteica , Fatores de Processamento de RNA/genética , Alinhamento de Sequência , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Fatores de Virulência/genética , Replicação Viral
19.
PLoS Pathog ; 5(9): e1000587, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19806178

RESUMO

A number of paramyxoviruses are responsible for acute respiratory infections in children, elderly and immuno-compromised individuals, resulting in airway inflammation and exacerbation of chronic diseases like asthma. To understand the molecular pathogenesis of these infections, we searched for cellular targets of the virulence protein C of human parainfluenza virus type 3 (hPIV3-C). We found that hPIV3-C interacts directly through its C-terminal domain with STAT1 and GRB2, whereas C proteins from measles or Nipah viruses failed to do so. Binding to STAT1 explains the previously reported capacity of hPIV3-C to block type I interferon signaling, but the interaction with GRB2 was unexpected. This adaptor protein bridges Epidermal Growth Factor (EGF) receptor to MAPK/ERK pathway, a signaling cascade recently found to be involved in airway inflammatory response. We report that either hPIV3 infection or transient expression of hPIV3-C both increase cellular response to EGF, as assessed by Elk1 transactivation and phosphorylation levels of ERK1/2, 40S ribosomal subunit protein S6 and translation initiation factor 4E (eIF4E). Furthermore, inhibition of MAPK/ERK pathway with U0126 prevented viral protein expression in infected cells. Altogether, our data provide molecular basis to explain the role of hPIV3-C as a virulence factor and determinant of pathogenesis and demonstrate that Paramyxoviridae have evolved a single virulence factor to block type I interferon signaling and to boost simultaneous cellular response to growth factors.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Interferon Tipo I/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Fatores de Virulência/metabolismo , Animais , Sítios de Ligação , Contagem de Células , Linhagem Celular , Chlorocebus aethiops , Fator de Iniciação 4E em Eucariotos/metabolismo , Citometria de Fluxo , Proteína Adaptadora GRB2/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Fosforilação , Mapeamento de Interação de Proteínas , Reprodutibilidade dos Testes , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Células Vero , Proteínas Virais/metabolismo
20.
Viruses ; 13(2)2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33540654

RESUMO

Bluetongue virus (BTV), an arbovirus transmitted by Culicoides biting midges, is a major concern of wild and domestic ruminants. While BTV induces type I interferon (alpha/beta interferon [IFN-α/ß]) production in infected cells, several reports have described evasion strategies elaborated by this virus to dampen this intrinsic, innate response. In the present study, we suggest that BTV VP3 is a new viral antagonist of the IFN-ß synthesis. Indeed, using split luciferase and coprecipitation assays, we report an interaction between VP3 and both the mitochondrial adapter protein MAVS and the IRF3-kinase IKKε. Overall, this study describes a putative role for the BTV structural protein VP3 in the control of the antiviral response.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Vírus Bluetongue/metabolismo , Bluetongue/metabolismo , Proteína DEAD-box 58/metabolismo , Receptores Imunológicos/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Bluetongue/genética , Bluetongue/virologia , Vírus Bluetongue/genética , Proteína DEAD-box 58/genética , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Ligação Proteica , Receptores Imunológicos/genética , Transdução de Sinais , Proteínas do Core Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA