Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Virologie (Montrouge) ; 27(1): 16-17, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36896771

RESUMEN

Epizootic hemorrhagic disease (EHD) is a non-contagious arthropod-borne disease transmitted by blood-sucking midges of the genus Culicoides. It affects domestic and wild ruminants, mainly white-tailed deer and cattle. At the end of October and in November 2022, outbreaks of EHD were confirmed in several cattle farms in Sardinia and Sicily. This is the first detection of EHD in Europe. The loss of free status and the lack of effective prophylactic measures could have significant economic consequences for infected countries.


Asunto(s)
Ciervos , Trastornos Hemorrágicos , Infecciones por Reoviridae , Animales , Bovinos , Infecciones por Reoviridae/epidemiología , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/diagnóstico , Rumiantes , Europa (Continente)/epidemiología , Sicilia
2.
Virologie (Montrouge) ; 27(1): 27-29, 2023 02 01.
Artículo en Francés | MEDLINE | ID: mdl-36891778

RESUMEN

Epizootic hemorrhagic disease (EHD) is a non-contagious arthropod-borne disease transmitted by blood-sucking midges of the genus Culicoides. It affects domestic and wild ruminants, mainly white-tailed deer and cattle. At the end of October and in November 2022, outbreaks of EHD were confirmed in several cattle farms in Sardinia and Sicily. This is the first detection of EHD in Europe. The loss of free status and the lack of effective prophylactic measures could have significant economic consequences for infected countries.


Asunto(s)
Ciervos , Trastornos Hemorrágicos , Infecciones por Reoviridae , Animales , Bovinos , Infecciones por Reoviridae/epidemiología , Infecciones por Reoviridae/veterinaria , Infecciones por Reoviridae/diagnóstico , Rumiantes , Europa (Continente)/epidemiología , Sicilia
3.
J Biol Chem ; 297(3): 101081, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34403699

RESUMEN

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.


Asunto(s)
Citidina Desaminasa/genética , Citidina Desaminasa/fisiología , Mitocondrias/genética , Proteínas/genética , Proteínas/fisiología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Citidina Desaminasa/metabolismo , ADN/genética , Mutación del Sistema de Lectura/genética , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Genoma/genética , Humanos , Mitocondrias/metabolismo , Mutación/genética , Proteínas/metabolismo , ARN Mensajero/genética , Sistemas de Lectura/genética
4.
Virologie (Montrouge) ; 26(5): 375-386, 2022 09 01.
Artículo en Francés | MEDLINE | ID: mdl-36413122

RESUMEN

African horse sickness (AHS) is a major arthropod-borne disease that causes significant losses in horses in sub-Saharan Africa. It is caused by the African horse sickness virus (AHSV), which is transmitted during a blood meal by Culicoides biting midges. The distribution of historical African culicoid vectors increases due to global warming. In addition, recent (Thailand, 2020) and earlier (Iberian Peninsula, 1965-66/1987-90) AHS outbreaks outside Africa demonstrate the adaptation of the virus to endogenous species in AHS-free regions, similar to what has been observed for bluetongue disease in recent decades. Therefore, many regions are considered at risk of introduction of AHS which could have important economic consequences for the equine industry. Overall, this prone the European Union to launch research programs to get better diagnostic and prophylactic tools.


La peste équine est une arbovirose majeure qui entraîne des pertes importantes chez les chevaux en Afrique subsaharienne. Elle est provoquée par le virus de la peste équine (African horse sickness virus, AHSV) dont la transmission s'effectue au cours d'un repas sanguin par des petits moucherons hématophages appartenant au genre Culicoides. En outre, les espèces vectrices historiques de culicoïdes présentes en Afrique voient leur aire de répartition s'étendre en lien avec le réchauffement climatique à l'échelle mondiale. Par ailleurs, des épisodes épizootiques récents (Thaïlande, 2020) ou un peu plus anciens (péninsule ibérique, 1965-66/1987-90) en dehors du continent africain soulignent la capacité d'adaptation du virus à des espèces vectrices autochtones, à l'instar de ce qui a été observé pour la fièvre catarrhale ovine ces dernières décennies. Ces facteurs laissent craindre à tout moment une introduction de la peste équine dans des régions indemnes. L'urgence est donc donnée actuellement par l'Union européenne pour se doter de meilleurs outils diagnostiques et prophylactiques afin de prévenir des conséquences économiques brutales pour l'industrie équine.


Asunto(s)
Virus de la Enfermedad Equina Africana , Enfermedad Equina Africana , Lengua Azul , Ceratopogonidae , Ovinos , Animales , Caballos , Enfermedad Equina Africana/epidemiología , Enfermedad Equina Africana/prevención & control , África del Sur del Sahara
5.
Virologie (Montrouge) ; 26(5): 387-400, 2022 09 01.
Artículo en Francés | MEDLINE | ID: mdl-36413123

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Interferón Tipo I , Porcinos , Animales , Virus de la Fiebre Porcina Africana/genética , Inmunidad Innata/genética , Virulencia
6.
J Virol ; 93(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31167915

RESUMEN

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.


Asunto(s)
Virus de la Lengua Azul/fisiología , Lengua Azul/metabolismo , Lengua Azul/virología , Interacciones Huésped-Patógeno , Sistema de Señalización de MAP Quinasas , Proteínas no Estructurales Virales/metabolismo , Animales , Virus de la Lengua Azul/patogenicidad , Línea Celular , Proteínas de Unión al ADN , Humanos , Interferones/metabolismo , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Factores de Transcripción , Factores de Virulencia , Replicación Viral
7.
Int J Mol Sci ; 21(15)2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32722052

RESUMEN

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.


Asunto(s)
Células Endoteliales/metabolismo , Microvasos/metabolismo , Modelos Biológicos , Virosis , Animales , Bovinos , Línea Celular , Células Endoteliales/patología , Células Endoteliales/virología , Microvasos/patología , Microvasos/virología
10.
J Virol ; 91(1)2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27795408

RESUMEN

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.


Asunto(s)
Nucléolo Celular/virología , Células Ependimogliales/virología , Interacciones Huésped-Patógeno , Orthobunyavirus/patogenicidad , ARN Polimerasa II/química , Proteínas no Estructurales Virales/química , Animales , Línea Celular Transformada , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestructura , Plexo Coroideo/citología , Plexo Coroideo/metabolismo , Plexo Coroideo/virología , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Células Ependimogliales/metabolismo , Células Ependimogliales/ultraestructura , Regulación de la Expresión Génica , Células HeLa , Humanos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Orthobunyavirus/genética , Orthobunyavirus/metabolismo , Señales de Clasificación de Proteína , Transporte de Proteínas , Proteolisis , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Ovinos , Transducción de Señal , Transcripción Genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
11.
BMC Biol ; 14: 69, 2016 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27538435

RESUMEN

BACKGROUND: After viral infection and the stimulation of some pattern-recognition receptors, TANK-binding kinase I (TBK1) is activated by K63-linked polyubiquitination followed by trans-autophosphorylation. While the activated TBK1 induces type I interferon production by phosphorylating the transcription factor IRF3, the precise molecular mechanisms underlying TBK1 activation remain unclear. RESULTS: We report here the localization of the ubiquitinated and phosphorylated active form of TBK1 to the Golgi apparatus after the stimulation of RIG-I-like receptors (RLRs) or Toll-like receptor-3 (TLR3), due to TBK1 K63-linked ubiquitination on lysine residues 30 and 401. The ubiquitin-binding protein optineurin (OPTN) recruits ubiquitinated TBK1 to the Golgi apparatus, leading to the formation of complexes in which TBK1 is activated by trans-autophosphorylation. Indeed, OPTN deficiency in various cell lines and primary cells impairs TBK1 targeting to the Golgi apparatus and its activation following RLR or TLR3 stimulation. Interestingly, the Bluetongue virus NS3 protein binds OPTN at the Golgi apparatus, neutralizing its activity and thereby decreasing TBK1 activation and downstream signaling. CONCLUSIONS: Our results highlight an unexpected role of the Golgi apparatus in innate immunity as a key subcellular gateway for TBK1 activation after RNA virus infection.


Asunto(s)
Aparato de Golgi/virología , Inmunidad Innata , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones por Virus ARN/inmunología , Proteínas de Ciclo Celular , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Aparato de Golgi/metabolismo , Células HEK293 , Células HeLa , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Proteínas de Transporte de Membrana , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Virus ARN , Receptores Inmunológicos , Transducción de Señal , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Factor de Transcripción TFIIIA/genética , Factor de Transcripción TFIIIA/metabolismo , Transfección , Ubiquitinación , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
14.
J Gen Virol ; 97(9): 2073-2083, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27435041

RESUMEN

During the compulsory vaccination programme against bluetongue virus serotype 1 (BTV-1) in Corsica (France) in 2014, a BTV strain belonging to a previously uncharacterized serotype (BTV-27) was isolated from asymptomatic goats. The present study describes the detection and molecular characterization of two additional distinct BTV-27 variants found in goats in Corsica in 2014 and 2015. The full coding genome of these two novel BTV-27 variants show high homology (90-93 % nucleotide/93-95 % amino acid) with the originally described BTV-27 isolate from Corsican goats in 2014. These three variants constitute the novel serotype BTV-27 ('BTV-27/FRA2014/v01 to v03'). Phylogenetic analyses with the 26 other established BTV serotypes revealed the closest relationship to BTV-25 (SWI2008/01) (80 % nucleotide/86 % amino acid) and to BTV-26 (KUW2010/02) (73-74 % nucleotide/80-81 % amino acid). However, highest sequence homologies between individual segments of BTV-27/FRA2014/v01-v03 with BTV-25 and BTV-26 vary. All three variants share the same segment 2 nucleotype with BTV-25. Neutralization assays of anti-BTV27/FRA2014/v01-v03 sera with a reassortant virus containing the outer capsid proteins of BTV-25 (BTV1VP2/VP5 BTV25) further confirmed that BTV-27 represents a distinct BTV serotype. Relationships between the variants and with BTV-25 and BTV-26, hypotheses about their origin, reassortment events and evolution are discussed.


Asunto(s)
Virus de la Lengua Azul/clasificación , Virus de la Lengua Azul/aislamiento & purificación , Lengua Azul/virología , Serogrupo , Animales , Enfermedades Asintomáticas , Análisis por Conglomerados , Francia , Genoma Viral , Cabras , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia
15.
J Virol ; 89(20): 10467-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246581

RESUMEN

UNLABELLED: Bluetongue virus (BTV) is an arbovirus transmitted to livestock by midges of the Culicoides family and is the etiological agent of a hemorrhagic disease in sheep and other ruminants. In mammalian cells, BTV particles are released primarily by virus-induced cell lysis, while in insect cells they bud from the plasma membrane and establish a persistent infection. BTV possesses a ten-segmented double-stranded RNA genome, and NS3 proteins are encoded by segment 10 (Seg-10). The viral nonstructural protein 3 (NS3) plays a key role in mediating BTV egress as well as in impeding the in vitro synthesis of type I interferon in mammalian cells. In this study, we asked whether genetically distant NS3 proteins can alter BTV-host interactions. Using a reverse genetics approach, we showed that, depending on the NS3 considered, BTV replication kinetics varied in mammals but not in insects. In particular, one of the NS3 proteins analyzed harbored a proline at position 24 that leads to its rapid intracellular decay in ovine but not in Culicoides cells and to the attenuation of BTV virulence in a mouse model of disease. Overall, our data reveal that the genetic variability of Seg-10/NS3 differentially modulates BTV replication kinetics in a host-specific manner and highlight the role of the host-specific variation in NS3 protein turnover rate. IMPORTANCE: BTV is the causative agent of a severe disease transmitted between ruminants by biting midges of Culicoides species. NS3, encoded by Seg-10 of the BTV genome, fulfills key roles in BTV infection. As Seg-10 sequences from various BTV strains display genetic variability, we assessed the impact of different Seg-10 and NS3 proteins on BTV infection and host interactions. In this study, we revealed that various Seg-10/NS3 proteins alter BTV replication kinetics in mammals but not in insects. Notably, we found that NS3 protein turnover may vary in ovine but not in Culicoides cells due to a single amino acid residue that, most likely, leads to rapid and host-dependent protein degradation. Overall, this study highlights that genetically distant BTV Seg-10/NS3 influence BTV biological properties in a host-specific manner and increases our understanding of how NS3 proteins contribute to the outcome of BTV infection.


Asunto(s)
Virus de la Lengua Azul/genética , Células Endoteliales/virología , Regulación Viral de la Expresión Génica , Genoma Viral , Proteínas no Estructurales Virales/genética , Replicación Viral/genética , Secuencia de Aminoácidos , Animales , Aorta/metabolismo , Aorta/patología , Aorta/virología , Virus de la Lengua Azul/química , Virus de la Lengua Azul/metabolismo , Línea Celular Transformada , Ceratopogonidae , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Plexo Coroideo/virología , Cricetulus , Células Endoteliales/metabolismo , Células Endoteliales/patología , Especificidad del Huésped , Ratones , Datos de Secuencia Molecular , Cultivo Primario de Células , Estabilidad Proteica , Proteolisis , Genética Inversa , Ovinos , Transducción de Señal , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Liberación del Virus/genética
16.
J Virol ; 88(18): 10792-802, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008919

RESUMEN

UNLABELLED: Bluetongue virus (BTV) is a double-stranded RNA (dsRNA) virus that causes an economically important disease in ruminants. BTV infection is a strong inducer of type I interferon (IFN-I) in multiple cell types. It has been shown recently that BTV and, more specifically, the nonstructural protein NS3 of BTV are able to modulate the IFN-I synthesis pathway. However, nothing is known about the ability of BTV to counteract IFN-I signaling. Here, we investigated the effect of BTV on the IFN-I response pathway and, more particularly, the Janus tyrosine kinase (JAK)/signal transducer and activator of transcription protein (STAT) signaling pathway. We found that BTV infection triggered the expression of IFN-stimulated genes (ISGs) in A549 cells. However, when BTV-infected cells were stimulated with external IFN-I, we showed that activation of the IFN-stimulated response element (ISRE) promoter and expression of ISGs were inhibited. We found that this inhibition involved two different mechanisms that were dependent on the time of infection. After overnight infection, BTV blocked specifically the phosphorylation and nuclear translocation of STAT1. This inhibition correlated with the redistribution of STAT1 in regions adjacent to the nucleus. At a later time point of infection, BTV was found to interfere with the activation of other key components of the JAK/STAT pathway and to induce the downregulation of JAK1 and TYK2 protein expression. Overall, our study indicates for the first time that BTV is able to interfere with the JAK/STAT pathway to modulate the IFN-I response. IMPORTANCE: Bluetongue virus (BTV) causes a severe disease in ruminants and has an important impact on the livestock economy in areas of endemicity such as Africa. The emergence of strains, such as serotype 8 in Europe in 2006, can lead to important economic losses due to commercial restrictions and prophylactic measures. It has been known for many years that BTV is a strong inducer of type I interferon (IFN-I) in vitro and in vivo in multiple cell types. However, the ability of BTV to interact with the IFN-I system remains unclear. Here, we report that BTV is able to modulate the IFN-I response by interfering with the Janus tyrosine kinase (JAK)/signal transducer and activator of transcription protein (STAT) signaling pathway. These findings contribute to knowledge of how BTV infection interferes with the host's innate immune response and becomes pathogenic. This will also be important for the design of efficacious vaccine candidates.


Asunto(s)
Virus de la Lengua Azul/fisiología , Lengua Azul/metabolismo , Interferón Tipo I/metabolismo , Animales , Lengua Azul/genética , Lengua Azul/virología , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/genética , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
17.
Virologie (Montrouge) ; 19(4): 178-186, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33065904

RESUMEN

Upon viral infection, infected cells mount an antiviral response that culminates with the production of type I IFN (IFN-α/ß) and other pro-inflammatory cytokines that control the infection. Production of type I IFN occurs both in vivo and in vitro in response to Bluetongue virus (BTV), an arthropod-borne virus, but the underlying mechanisms responsible for this event remained unknown until recently. This review describes the recent advances in the identification of cellular sensors and signalling pathways involved in this process. In non-hematopoietic cells, expression of IFN-ß in response to BTV infection depends on the activation of the RNA helicases retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). In contrast, induction of IFN-α/ß synthesis in sheep primary plasmacytoid dendritic cells (pDCs) required the MyD88 adaptor independently of the Toll-like receptor 7 (TLR7), as well as the kinases dsRNA-activated protein kinase (PKR) and stress-activated protein kinase (SAPK)/Jun N-terminal protein kinase (JNK). In order to counteract this antiviral response, most of viruses have elaborated mechanisms to hinder its action. This review also describes the ability of BTV to interfere with the IFN pathway and the recent findings describing the non-structural viral protein NS3 as a powerful antagonist of the host cellular response.

18.
Emerg Infect Dis ; 20(12): 2123-5, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25418049

RESUMEN

During 2000-2013, 4 genotypes of bluetongue virus (BTV) were detected in Corsica, France. At the end of 2013, a compulsory BTV-1 vaccination campaign was initiated among domestic ruminants; biological samples from goats were tested as part of a corresponding monitoring program. A BTV strain with nucleotide sequences suggestive of a novel serotype was detected.


Asunto(s)
Virus de la Lengua Azul/clasificación , Lengua Azul/epidemiología , Lengua Azul/virología , Cabras/virología , Animales , Virus de la Lengua Azul/genética , Francia/epidemiología , Genotipo , Filogenia , Vigilancia en Salud Pública , ARN Viral
19.
J Virol ; 87(14): 8241-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23658442

RESUMEN

Upon infection with Bluetongue virus (BTV), an arthropod-borne virus, type I interferon (IFN-I) is produced in vivo and in vitro. IFN-I is essential for the establishment of an antiviral cellular response, and most if not all viruses have elaborated strategies to counteract its action. In this study, we assessed the ability of BTV to interfere with IFN-I synthesis and identified the nonstructural viral protein NS3 as an antagonist of the IFN-I system.


Asunto(s)
Virus de la Lengua Azul/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/antagonistas & inhibidores , Transducción de Señal/inmunología , Proteínas no Estructurales Virales/metabolismo , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón Tipo I/biosíntesis , Luciferasas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteínas no Estructurales Virales/farmacología
20.
J Virol ; 87(16): 9333-43, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23785206

RESUMEN

Human and animal hemorrhagic viruses initially target dendritic cells (DCs). It has been proposed, but not documented, that both plasmacytoid DCs (pDCs) and conventional DCs (cDCs) may participate in the cytokine storm encountered in these infections. In order to evaluate the contribution of DCs in hemorrhagic virus pathogenesis, we performed a genome-wide expression analysis during infection by Bluetongue virus (BTV), a double-stranded RNA virus that induces hemorrhagic fever in sheep and initially infects cDCs. Both pDCs and cDCs accumulated in regional lymph nodes and spleen during BTV infection. The gene response profiles were performed at the onset of the disease and markedly differed with the DC subtypes and their lymphoid organ location. An integrative knowledge-based analysis revealed that blood pDCs displayed a gene signature related to activation of systemic inflammation and permeability of vasculature. In contrast, the gene profile of pDCs and cDCs in lymph nodes was oriented to inhibition of inflammation, whereas spleen cDCs did not show a clear functional orientation. These analyses indicate that tissue location and DC subtype affect the functional gene expression program induced by BTV and suggest the involvement of blood pDCs in the inflammation and plasma leakage/hemorrhage during BTV infection in the real natural host of the virus. These findings open the avenue to target DCs for therapeutic interventions in viral hemorrhagic diseases.


Asunto(s)
Sangre/inmunología , Virus de la Lengua Azul/inmunología , Lengua Azul/inmunología , Células Dendríticas/inmunología , Perfilación de la Expresión Génica , Ganglios Linfáticos/inmunología , Animales , Células Cultivadas , Masculino , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA