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











Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
NPJ Vaccines ; 5(1): 1, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31908850

RESUMO

Foot and mouth disease (FMD) is a highly contagious viral disease with high economic impact, representing a major threat for cloven-hooved mammals worldwide. Vaccines based on adjuvanted inactivated virus (iFMDV) induce effective protective immunity implicating antibody (Ab) responses. To reduce the biosafety constraints of the manufacturing process, a non-replicative human adenovirus type 5 vector encoding FMDV antigens (Ad5-FMDV) has been developed. Here we compared the immunogenicity of iFMDV and Ad5-FMDV with and without the ISA206VG emulsion-type adjuvant in sheep. Contrasted Ab responses were obtained: iFMDV induced the highest Ab levels, Ad5-FMDV the lowest ones, and ISA206VG increased the Ad5-FMDV-induced Ab responses to protective levels. Each vaccine generated heterogeneous Ab responses, with high and low responders, the latter being considered as obstacles to vaccine effectiveness. A transcriptomic study on total blood responses at 24 h post-vaccination revealed several blood gene module activities correlating with long-term Ab responses. Downmodulation of T cell modules' activities correlated with high responses to iFMDV and to Ad5-FMDV+ISA206VG vaccines as also found in other systems vaccinology studies in humans and sheep. The impact of cell cycle activity depended on the vaccine types, as it positively correlated with higher responses to iFMDV but negatively to non-adjuvanted Ad5-FMDV. Finally an elevated B cell activity at 24 h correlated with high Ab responses to the Ad5-FMDV+ISA206VG vaccine. This study provides insights into the early mechanisms driving the Ab response induced by different vaccine regimens including Ad5 vectors and points to T cell modules as early biomarker candidates of different vaccine-type efficacy across species.

4.
Transbound Emerg Dis ; 67(1): 133-148, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31419374

RESUMO

Foot-and-mouth disease virus (FMDV) causes a highly contagious vesicular disease in livestock, with serious consequences for international trade. The virus persists in the nasopharynx of cattle and this slows down the process to obtain an FMDV-free status after an outbreak. To study biological mechanisms, or to identify molecules that can be targeted to diagnose or interfere with persistence, we developed a model of persistent FMDV infection in bovine dorsal soft palate (DSP). Primary DSP cells were isolated after commercial slaughter and were cultured in multilayers at the air-liquid interface. After 5 weeks of culture without further passage, the cells were infected with FMDV strain O/FRA/1/2001. Approximately, 20% of cells still had a polygonal morphology and displayed tight junctions as in stratified squamous epithelia. Subsets of cells expressed cytokeratin and most or all cells expressed vimentin. In contrast to monolayers in medium, multilayers in air demonstrated only a limited cytopathic effect. Integrin αV ß6 expression was observed in mono- but not in multilayers. FMDV antigen, FMDV RNA and live virus were detected from day 1 to 28, with peaks at day 1 and 2. The proportion of infected cells was highest at 24 hr (3% and 36% of cells at an MOI of 0.01 and 1, respectively). At day 28 after infection, at a time when animals that still harbour FMDV are considered carriers, FMDV antigen was detected in 0.2%-2.1% of cells, in all layers, and live virus was isolated from supernatants of 6/8 cultures. On the consensus level, the viral genome did not change within the first 24 hr after infection. Only a few minor single nucleotide variants were detected, giving no indication of the presence of a viral quasispecies. The air-liquid interface model of DSP brings new possibilities to investigate FMDV persistence in a controlled manner.


Assuntos
Antígenos Virais/imunologia , Doenças dos Bovinos/virologia , Vírus da Febre Aftosa/imunologia , Febre Aftosa/virologia , Genoma Viral/genética , Animais , Bovinos , Linhagem Celular , Células Cultivadas , Células Epiteliais/virologia , Feminino , Vírus da Febre Aftosa/isolamento & purificação , Imuno-Histoquímica/veterinária , Masculino , Palato Mole/virologia , RNA Viral/análise , Suínos
5.
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
6.
Viruses ; 11(1)2019 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-30642035

RESUMO

Foot-and-mouth disease (FMD) is the most devastating disease of cloven-hoofed livestock, with a crippling economic burden in endemic areas and immense costs associated with outbreaks in free countries. Foot-and-mouth disease virus (FMDV), a picornavirus, will spread rapidly in naïve populations, reaching morbidity rates of up to 100% in cattle. Even after recovery, over 50% of cattle remain subclinically infected and infectious virus can be recovered from the nasopharynx. The pathogen and host factors that contribute to FMDV persistence are currently not understood. Using for the first time primary bovine soft palate multilayers in combination with proteogenomics, we analyzed the transcriptional responses during acute and persistent FMDV infection. During the acute phase viral RNA and protein was detectable in large quantities and in response hundreds of interferon-stimulated genes (ISG) were overexpressed, mediating antiviral activity and apoptosis. Although the number of pro-apoptotic ISGs and the extent of their regulation decreased during persistence, some ISGs with antiviral activity were still highly expressed at that stage. This indicates a long-lasting but ultimately ineffective stimulation of ISGs during FMDV persistence. Furthermore, downregulation of relevant genes suggests an interference with the extracellular matrix that may contribute to the skewed virus-host equilibrium in soft palate epithelial cells.


Assuntos
Doenças dos Bovinos/imunologia , Células Epiteliais/virologia , Febre Aftosa/imunologia , Interações Hospedeiro-Patógeno , Palato Mole/citologia , Proteogenômica , Animais , Apoptose , Bovinos , Doenças dos Bovinos/virologia , Células Cultivadas , Biologia Computacional , Regulação para Baixo , Vírus da Febre Aftosa , Expressão Gênica , Perfilação da Expressão Gênica , Imunidade Inata , Interferons/genética , Palato Mole/virologia , RNA Viral/genética
7.
Transbound Emerg Dis ; 66(1): 537-545, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30394662

RESUMO

Competitive-ELISA (c-ELISA) is the most widely used serological test for the detection of Bluetongue virus (BTV) viral protein 7 (VP7) antibodies (Ab). However, these BTV c-ELISAs cannot to distinguish between IgG and IgM. IgM Ab are generated shortly after the primary immune response against an infectious agent, indicating a recent infection or exposure to antigens, such as after vaccination. Because the BTV genome or anti-VP7 Ab can be detected in ruminant blood months after infection, BTV diagnostic tools cannot discriminate between recent and old infections. In this study, we evaluated an IgM-capture ELISA prototype to detect ruminant anti-BTV VP7 IgM on 1,650 serum samples from cattle, sheep, or goats. Animals were BTV-naive, infected, or/and vaccinated with BTV-1, -2, -4, -8, -9, -16, or -27, and we also included 30 sera from cattle infected with the Epizootic haemorrhagic disease virus (EHDV) serotype 6. Results demonstrated that this ELISA kit is specific and can detect the presence of IgM with satisfactory diagnostic specificity and sensitivity from 1 to 5 weeks after BTV infection in domestic ruminants (for goats and cattle; for sheep, at least up to 24 days). The peak of anti-VP7 IgM was reached when the level of infectious viruses and BTV RNA in blood were the highest. The possibility of detecting BTV-RNA in IgM-positive sera allows the amplification and sequencing of the partial RNA segment 2 (encoding the serotype specific to VP2) to determine the causative BTV serotype/strain. Therefore, BTV IgM ELISA can detect the introduction of BTV (or EHDV) in an area with BTV-seropositive domestic animals regardless of their serological BTV status. This approach may also be of particular interest for retrospective epidemiological studies on frozen serum samples.


Assuntos
Animais Domésticos/virologia , Anticorpos Antivirais/sangue , Vírus Bluetongue/imunologia , Bluetongue/diagnóstico , Ensaio de Imunoadsorção Enzimática/veterinária , Imunoglobulina M/sangue , Proteínas do Core Viral/imunologia , Animais , Bluetongue/imunologia , Bluetongue/virologia , Bovinos , Doenças dos Bovinos/diagnóstico , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/virologia , Diagnóstico Precoce , Doenças das Cabras/diagnóstico , Doenças das Cabras/imunologia , Doenças das Cabras/virologia , Cabras , Estudos Retrospectivos , Ruminantes , Sorogrupo , Testes Sorológicos/métodos , Ovinos , Doenças dos Ovinos/diagnóstico , Doenças dos Ovinos/imunologia , Doenças dos Ovinos/virologia
8.
Transbound Emerg Dis ; 66(1): 83-90, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30070433

RESUMO

The laboratory diagnosis of African horse sickness (AHS) is important for: (a) demonstrating freedom from infection in a population, animals or products for trade (b) assessing the efficiency of eradication policies; (c) laboratory confirmation of clinical diagnosis; (d) estimating the prevalence of AHS infection; and (e) assessing postvaccination immune status of individual animals or populations. Although serological techniques play a secondary role in the confirmation of clinical cases, their use is very important for all the other purposes due to their high throughput, ease of use and good cost-benefit ratio. The main objective of this study was to support the validation of AHS VP7 Blocking ELISA up to the Stage 3 of the World Animal Health Organization (OIE) assay validation pathway. To achieve this, a collaborative ring trial, which included all OIE Reference Laboratories and other AHS-specialist diagnostic centres, was conducted in order to assess the diagnostic performance characteristics of the VP7 Blocking ELISA. In this trial, a panel of sera of different epidemiological origin and infection status was used. Through this comprehensive evaluation we can conclude that the VP7 Blocking ELISA satisfies the OIE requirements of reproducibility. The VP7 Blocking ELISA, in its commercial version is ready to enter Stage 4 of the validation pathway (Programme Implementation). Specifically, this will require testing the diagnostic performance of the assay using contemporary serum samples collected during control campaigns in endemic countries.


Assuntos
Vírus da Doença Equina Africana/isolamento & purificação , Doença Equina Africana/diagnóstico , Testes Diagnósticos de Rotina/veterinária , Ensaio de Imunoadsorção Enzimática/veterinária , Doenças dos Cavalos/diagnóstico , Animais , Antígenos Virais/sangue , Testes Diagnósticos de Rotina/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Cavalos , Reprodutibilidade dos Testes , Proteínas do Core Viral/sangue
9.
Vaccine ; 36(16): 2193-2198, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29544690

RESUMO

Vaccination is a key element in the control of foot-and-mouth disease (FMD). The majority of the antigenic sites that induce protective immune responses are localized on the FMD virus (FMDV) capsid that is formed by four virus-encoded structural proteins, VP1 to VP4. In the present study, recombinant canine adenovirus type 2 (CAV2)-based FMD vaccines, Cav-P1/3C R° and Cav-VP1 R°, respectively expressing the structural P1 precursor protein along with the non-structural 3C protein or expressing the structural VP1 protein of the FMDV strain O/FRA/1/2001, were evaluated as novel vaccines against FMD. A strong humoral immune response was elicited in guinea pigs (GP) following immunization with Cav-P1/3C R°, while administration of Cav-VP1 R° did not induce a satisfying antibody response in GP or mice. GP were then used as an experimental model for the determination of the protection afforded by the Cav-P1/3C R° vaccine against challenge with the FMDV strain O1 Manisa/Turkey/1969. The Cav-P1/3C R° vaccine protected GP from generalized FMD to a similar extent as a high potency double-oil emulsion O1 Manisa vaccine. The results of the present study show that CAV2-based vector vaccines can express immunogenic FMDV antigens and offer protection against generalized FMD in GP. This suggest that Cav-P1/3C R° FMDV vaccine may protect natural host species from FMD. In combination with an appropriate diagnostic test, the Cav-P1/3C R° FMDV vaccine may also serve as a marker vaccine to differentiate vaccinated from infected animals.


Assuntos
Adenovirus Caninos/genética , Adenovirus Caninos/imunologia , Reações Cruzadas/imunologia , Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Cães , Feminino , Cobaias , Imunização , Imunogenicidade da Vacina , Masculino , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
10.
Virologie (Montrouge) ; 19(4): 178-186, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-33065904

RESUMO

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.

11.
PLoS One ; 9(11): e111605, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25364822

RESUMO

Bluetongue virus (BTV) is an economically important Orbivirus transmitted by biting midges to domestic and wild ruminants. The need for new vaccines has been highlighted by the occurrence of repeated outbreaks caused by different BTV serotypes since 1998. The major group-reactive antigen of BTV, VP7, is conserved in the 26 serotypes described so far, and its role in the induction of protective immunity has been proposed. Viral-based vectors as antigen delivery systems display considerable promise as veterinary vaccine candidates. In this paper we have evaluated the capacity of the BTV-2 serotype VP7 core protein expressed by either a non-replicative canine adenovirus type 2 (Cav-VP7 R0) or a leporipoxvirus (SG33-VP7), to induce immune responses in sheep. Humoral responses were elicited against VP7 in almost all animals that received the recombinant vectors. Both Cav-VP7 R0 and SG33-VP7 stimulated an antigen-specific CD4+ response and Cav-VP7 R0 stimulated substantial proliferation of antigen-specific CD8+ lymphocytes. Encouraged by the results obtained with the Cav-VP7 R0 vaccine vector, immunized animals were challenged with either the homologous BTV-2 or the heterologous BTV-8 serotype and viral burden in plasma was followed by real-time RT-PCR. The immune responses triggered by Cav-VP7 R0 were insufficient to afford protective immunity against BTV infection, despite partial protection obtained against homologous challenge. This work underscores the need to further characterize the role of BTV proteins in cross-protective immunity.


Assuntos
Antígenos Virais/genética , Vírus Bluetongue/genética , Bluetongue/imunologia , Expressão Gênica , Vetores Genéticos/genética , Proteínas do Core Viral/genética , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Bluetongue/prevenção & controle , Bluetongue/virologia , Vírus Bluetongue/imunologia , Linhagem Celular , Cricetinae , Reações Cruzadas/imunologia , Cães , Feminino , Imunidade Celular , Imunização , Masculino , Coelhos , Ovinos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas do Core Viral/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
12.
Virus Res ; 182: 59-70, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24211608

RESUMO

The innate immune response is the first line of defence against viruses, involving the production of type I IFN (IFN-α/ß) and other pro-inflammatory cytokines that control the infection. It also shapes the adaptive immune response generated by both T and B cells. Production of type I IFN occurs both in vivo and in vitro in response to Bluetongue virus (BTV), an arthropod-borne virus. However, the mechanisms responsible for the production of IFN-ß in response to BTV remained unknown until recently and are still not completely understood. In this review, we describe the recent advances in the identification of cellular sensors and signalling pathways involved in this process. The RNA helicases retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) were shown to be involved in the expression of IFN-ß as well as in the control of BTV infection in non-haematopoietic cells. 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). As type I IFN is essential for the establishment of an antiviral cellular response, most of viruses have elaborated counteracting mechanisms to hinder its action. This review also addresses the ability of BTV to interfere with IFN-ß synthesis and the recent findings describing the non-structural viral protein NS3 as a powerful antagonist of the host cellular response.


Assuntos
Vírus Bluetongue/imunologia , Evasão da Resposta Imune , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Ovinos/imunologia , Ovinos/virologia , Animais , Interferon Tipo I/antagonistas & inibidores , Receptores Imunológicos , Transdução de Sinais
13.
J Virol ; 87(1): 543-57, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23097432

RESUMO

Coinfection of a cell by two different strains of a segmented virus can give rise to a "reassortant" with phenotypic characteristics that might differ from those of the parental strains. Bluetongue virus (BTV) is a double-stranded RNA (dsRNA) segmented virus and the cause of bluetongue, a major infectious disease of livestock. BTV exists as at least 26 different serotypes (BTV-1 to BTV-26). Prompted by the isolation of a field reassortant between BTV-1 and BTV-8, we systematically characterized the process of BTV reassortment. Using a reverse genetics approach, our study clearly indicates that any BTV-1 or BTV-8 genome segment can be rescued in the heterologous "backbone." To assess phenotypic variation as a result of reassortment, we examined viral growth kinetics and plaque sizes in in vitro experiments and virulence in an experimental mouse model of bluetongue disease. The monoreassortants generated had phenotypes that were very similar to those of the parental wild-type strains both in vitro and in vivo. Using a forward genetics approach in cells coinfected with BTV-1 and BTV-8, we have shown that reassortants between BTV-1 and BTV-8 are generated very readily. After only four passages in cell culture, we could not detect wild-type BTV-1 or BTV-8 in any of 140 isolated viral plaques. In addition, most of the isolated reassortants contained heterologous VP2 and VP5 structural proteins, while only 17% had homologous VP2 and VP5 proteins. Our study has shown that reassortment in BTV is very flexible, and there is no fundamental barrier to the reassortment of any genome segment. Given the propensity of BTV to reassort, it is increasingly important to have an alternative classification system for orbiviruses.


Assuntos
Vírus Bluetongue/genética , Genoma Viral , RNA Viral/genética , Vírus Reordenados/genética , Recombinação Genética , Animais , Vírus Bluetongue/crescimento & desenvolvimento , Genótipo , Camundongos , Dados de Sequência Molecular , Fenótipo , Genética Reversa , Análise de Sequência de DNA , Ensaio de Placa Viral , Proteínas Estruturais Virais/genética
14.
J Virol ; 86(21): 11789-99, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22915805

RESUMO

Bluetongue virus (BTV), an arthropod-borne member of the Reoviridae family, is a double-stranded RNA virus that causes an economically important livestock disease that has spread across Europe in recent decades. Production of type I interferon (alpha/beta interferon [IFN-α/ß]) has been reported in vivo and in vitro upon BTV infection. However, the cellular sensors and signaling pathways involved in this process remain unknown. Here we studied the mechanisms responsible for the production of IFN-ß in response to BTV serotype 8. Upon BTV infection of A549 cells, expression of IFN-ß and other proinflammatory cytokines was strongly induced at both the protein and mRNA levels. This response appeared to be dependent on virus replication, since exposure to UV-inactivated virus failed to induce IFN-ß. We also demonstrated that BTV infection activated the transcription factors IFN regulatory factor 3 and nuclear factor κB. We investigated the role of several pattern recognition receptors in this response and showed that expression of IFN-ß was greatly reduced after small-interfering-RNA-mediated knockdown of the RNA helicase encoded by retinoic acid-inducible gene I (RIG-I) or melanoma differentiation-associated gene 5 (MDA5). In contrast, silencing of MyD88, Toll-like receptor 3, or the recently described DexD/H-box helicase DDX1 sensor had no or a weak effect on IFN-ß induction, suggesting that the RIG-I-like receptor pathway is specifically engaged for BTV sensing. Moreover, we also showed that overexpression of either RIG-I or MDA5 impaired BTV expression in infected A549 cells. Overall, this indicates that RIG-I and MDA5 can both contribute to the recognition and control of BTV infection.


Assuntos
Vírus Bluetongue/imunologia , RNA Helicases DEAD-box/metabolismo , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Interferon beta/biossíntese , Animais , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Perfilação da Expressão Gênica , Inativação Gênica , Humanos , Helicase IFIH1 Induzida por Interferon , Interferon beta/genética , Receptores Imunológicos
15.
J Virol ; 86(10): 5817-28, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22438548

RESUMO

Dendritic cells (DCs), especially plasmacytoid DCs (pDCs), produce large amounts of alpha/beta interferon (IFN-α/ß) upon infection with DNA or RNA viruses, which has impacts on the physiopathology of the viral infections and on the quality of the adaptive immunity. However, little is known about the IFN-α/ß production by DCs during infections by double-stranded RNA (dsRNA) viruses. We present here novel information about the production of IFN-α/ß induced by bluetongue virus (BTV), a vector-borne dsRNA Orbivirus of ruminants, in sheep primary DCs. We found that BTV induced IFN-α/ß in skin lymph and in blood in vivo. Although BTV replicated in a substantial fraction of the conventional DCs (cDCs) and pDCs in vitro, only pDCs responded to BTV by producing a significant amount of IFN-α/ß. BTV replication in pDCs was not mandatory for IFN-α/ß production since it was still induced by UV-inactivated BTV (UV-BTV). Other inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and IL-12p40, were also induced by UV-BTV in primary pDCs. The induction of IFN-α/ß required endo-/lysosomal acidification and maturation. However, despite being an RNA virus, UV-BTV did not signal through Toll-like receptor 7 (TLR7) for IFN-α/ß induction. In contrast, pathways involving the MyD88 adaptor and kinases dsRNA-activated protein kinase (PKR) and stress-activated protein kinase (SAPK)/Jun N-terminal protein kinase (JNK) were implicated. This work highlights the importance of pDCs for the production of innate immunity cytokines induced by a dsRNA virus, and it shows that a dsRNA virus can induce IFN-α/ß in pDCs via a novel TLR-independent and Myd88-dependent pathway. These findings have implications for the design of efficient vaccines against dsRNA viruses.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/imunologia , Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Animais , Bluetongue/genética , Bluetongue/virologia , Vírus Bluetongue/genética , Vírus Bluetongue/fisiologia , Células Cultivadas , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/virologia , Feminino , Imunidade Inata , Interferon Tipo I/genética , Glicoproteínas de Membrana , Fator 88 de Diferenciação Mieloide/genética , Receptores de Interleucina-1 , Ovinos/imunologia , Ovinos/virologia , Transdução de Sinais , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética
16.
PLoS One ; 7(12): e52513, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300693

RESUMO

Gene expression profiling of the blood cell response induced early after vaccination has previously been demonstrated to predict the immunogenicity of vaccines. In this study, we evaluated whether the analysis of the gene expression profile of skin-migrated dendritic cells (DCs) could be informative for the in vitro prediction of immunogenicity of vaccine, using canine adenovirus serotype 2 (CAV2) as vaccine vector. CAV2 has been shown to induce immunity to transgenes in several species including sheep and is an interesting alternative to human adenovirus-based vectors, based on the safety records of the parental strain in dogs and the lack of pre-existing immunity in non-host species. Skin-migrated DCs were collected from pseudo-afferent lymph in sheep. Both the CD11b(+) -type and CD103(+) -type skin-migrated DCs were transduced by CAV2. An analysis of the global gene response to CAV2 in the two skin DC subsets showed that the gene response in CD11b(+) -type DCs was far higher and broader than in the CD103(+) -type DCs. A newly released integrative analytic tool from Ingenuity systems revealed that the CAV2-modulated genes in the CD11b(+) -type DCs clustered in several activated immunogenicity-related functions, such as immune response, immune cell trafficking and inflammation. Thus gene profiling in skin-migrated DC in vitro indicates that the CD11b(+) DC type is more responsive to CAV2 than the CD103(+) DC type, and provides valuable information to help in evaluating and possibly improving viral vector vaccine effectiveness.


Assuntos
Adenovirus Caninos/genética , Movimento Celular/imunologia , DNA Recombinante/genética , Células Dendríticas/imunologia , Vetores Genéticos/genética , Pele/imunologia , Transcriptoma/imunologia , Animais , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Cães , Feminino , Humanos , Cadeias alfa de Integrinas/metabolismo , Ovinos , Vacinação
17.
Vaccine ; 29(6): 1304-10, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21134446

RESUMO

Safe and efficient vaccination is important for rabies prevention in domestic animals. Replicative vectors expressing the rabies virus glycoprotein, derived from canine adenovirus have been reported to be promising vaccines in various animal models. In this paper we compare the potential of a replicative and a non-replicative vector, both based on canine adenovirus type 2 and expressing the rabies glycoprotein. Upon inoculation in sheep, immune responses against the rabies virus protein elicited by recombinant vectors were monitored. All immunised sheep produced a rapid and potent neutralizing antibody response against rabies virus after a single inoculation of either replicative or non-replicative recombinant canine adenovirus type 2. In addition, the non-replicative vector expressing the rabies glycoprotein stimulated antigen-specific CD4(+) and CD8(+) lymphocyte proliferation as well as IFN-γ production. These results suggest that vectors derived from canine adenovirus 2 could be considered for the development of promising vaccines in the ruminant species.


Assuntos
Adenovirus Caninos/genética , Portadores de Fármacos , Vetores Genéticos , Vacina Antirrábica/imunologia , Vírus da Raiva/imunologia , Raiva/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Interferon gama/metabolismo , Masculino , Vacina Antirrábica/genética , Vírus da Raiva/genética , Ovinos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
18.
Antiviral Res ; 81(3): 217-25, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19103226

RESUMO

To evaluate the feasibility of using pseudorabies virus (PrV) glycoprotein B (gB) as a carrier of foot and mouth disease virus (FMDV) antigens in DNA immunization, FMDV B- and T-cell epitopes were inserted either between the two B-cell epitopes of the N-term subunit of PrV-gB (BT-PrV-gB-N-term construct) or within the B-cell epitope of the C-term subunit of PrV-gB (BT-PrV-gB-C-term construct). Two animal experiments were performed, each with three injections of plasmids 2 weeks apart, followed by a booster inoculation of peptides corresponding to the FMDV epitopes. Control groups of pigs were injected with plasmids encoding either PrV-gB or FMDV-BT, or with empty-pcDNA3. The results of both assays were combined. Significant titers of FMDV neutralizing antibodies were detected after the peptides boost in groups injected with the BT-PrV-gB-C-term construct. Insignificant amounts were detected in groups injected with the BT-PrV-gB-N-term and FMDV-BT constructs. PBMCs from the BT-PrV-gB-N-term groups, isolated after the peptide boost injection, produced IFN-gamma and IL-4 mRNAs in vitro when stimulated with FMDV peptides. This was not observed with the other groups. These results imply that PrV-gB can be used to carry FMDV antigens in a DNA vaccine.


Assuntos
Antígenos Virais/genética , Vírus da Febre Aftosa/genética , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/genética , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Vírus da Febre Aftosa/imunologia , Imunização Secundária , Interferon gama/biossíntese , Interleucina-4/biossíntese , Leucócitos Mononucleares/imunologia , Testes de Neutralização , Suínos , Proteínas Virais/genética , Proteínas Virais/imunologia
19.
J Gen Virol ; 88(Pt 11): 3078-3088, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17947533

RESUMO

The complete nucleotide sequence of Middelburg virus (MIDV) was determined for strain MIDV-857 from Zimbabwe. The isolation of this virus in 1993 from a horse that died showing severe clinical signs represents the first indication that MIDV can cause severe disease in equids. Full-length cDNA copies of the viral genome were successfully synthesized by an innovative RT-PCR amplification approach using an 'anchor primer' combined with the SMART methodology described previously for the synthesis of full-length cDNA copies from genome segments of dsRNA viruses. The MIDV-857 genome is 11,674 nt, excluding the 5'-terminal cap structure and poly(A) tail (which varies in length from approximately 180 to approximately 220 residues). The organization of the genome is like that of other alphaviruses, including a read-through stop codon between the nsP3 and nsP4 genes. However, phylogenetic analyses of the structural protein amino acid sequences suggested that the MIDV E1 gene was generated by recombination with a Semliki Forest virus-like virus. This hypothesis was supported by bootscanning analysis using a recombination-detection program. The 3' untranslated region of MIDV-857 also contains a 112 nt duplication. This study reports the first full-length sequence of MIDV, which was obtained from a single RT-PCR product.


Assuntos
Alphavirus/genética , Genoma Viral , Regiões 3' não Traduzidas/genética , Alphavirus/isolamento & purificação , Infecções por Alphavirus/veterinária , Animais , Sequência de Bases , Códon de Terminação/genética , Doenças dos Cavalos/virologia , Cavalos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Filogenia , Capuzes de RNA/genética , RNA Mensageiro/genética , Recombinação Genética , Vírus da Floresta de Semliki/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Baço/virologia , Proteínas não Estruturais Virais/genética , Proteínas Estruturais Virais/genética , Zimbábue
20.
Vaccine ; 25(37-38): 6774-83, 2007 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-17669563

RESUMO

The development of recombinant capripoxviruses for protective immunization of ruminants against bluetongue virus (BTV) infection is described. Sheep (n=11) and goats (n=4) were immunized with BTV recombinant capripoxviruses (BTV-Cpox) individually expressing four different genes encoding two capsid proteins (VP2 and VP7) and two non-structural proteins (NS1, NS3) of BTV serotype 2 (BTV-2). Seroconversion was observed against NS3, VP7 and VP2 in both species and a lymphoproliferation specific to BTV antigens was also demonstrated in goats. Finally, partial protection of sheep challenged 3 weeks after BTV-Cpox administration with a virulent strain of BTV-2, was observed.


Assuntos
Vírus Bluetongue/imunologia , Vírus Bluetongue/metabolismo , Capripoxvirus/imunologia , Capripoxvirus/metabolismo , Expressão Gênica , Proteínas Virais/imunologia , Proteínas Virais/metabolismo , Animais , Vírus Bluetongue/genética , Vírus Bluetongue/patogenicidade , Capripoxvirus/genética , Células Cultivadas , Chlorocebus aethiops , Vetores Genéticos/genética , Cabras , Ovinos , Taxa de Sobrevida , Proteínas Virais/genética , Vacinas Virais/efeitos adversos , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA