Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 373
Filtrar
2.
Prev Vet Med ; 232: 106328, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39191049

RESUMO

Bluetongue virus (BT) is a vector-borne virus that causes a disease, called bluetongue, which results in significant economic loss and morbidity in sheep, cattle, goats and wild ungulates across all continents of the world except Antarctica. Despite the geographical breadth of its impact, most BT epidemiological models are informed by parameters derived from the 2006-2009 BTV-8 European outbreak. The aim of this study was to develop a highly adaptable model for BT which could be used elsewhere in the world, as well as to identify the parameters which most influence outbreak dynamics, so that policy makers can be properly informed with the most current information to aid in disease planning. To provide a framework for future outbreak modelling and an updated parameterisation that reflects natural variation in infections, a newly developed and parameterised two-host, two-vector species ordinary differential equation model was formulated and analysed. The model was designed to be adaptable to be implemented in any region of the world and able to model both epidemic and endemic scenarios. It was parameterised using a systematic literature review of host-to-vector and vector-to-host transmission rates, host latent periods, host infectious periods, and vaccine protection factors. The model was demonstrated using the updated parameters, with South Africa as a setting based on the Western Cape's known cattle and sheep populations, local environmental parameters, and Culicoides spp. presence data. The sensitivity analysis identified that the duration of the infectious period for sheep and cows had the greatest impact on the outbreak length and number of animals infected at the peak of the outbreak. Transmission rates from cows and sheep to C. imicola midges greatly influenced the day on which the peak of the outbreak occurred, along with the duration of incubation period, and infectious period for cows. Finally, the protection factor of the vaccine had the greatest influence on the total number of animals infected. This knowledge could aid in the development of control measures. Due to gradual climate and anthropological change resulting in alterations in vector habitat suitability, BT outbreaks are likely to continue to increase in range and frequency. Therefore, this research provides an updated BT modelling framework for future outbreaks around the world to explore transmission, outbreak dynamics and control measures.


Assuntos
Vírus Bluetongue , Bluetongue , Doenças dos Bovinos , Surtos de Doenças , Animais , Bovinos , Bluetongue/epidemiologia , Bluetongue/transmissão , Bluetongue/virologia , Bluetongue/prevenção & controle , Vírus Bluetongue/patogenicidade , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , Doenças dos Bovinos/transmissão , Ceratopogonidae/virologia , Surtos de Doenças/prevenção & controle , Surtos de Doenças/veterinária , Cabras/virologia , Insetos Vetores/virologia , Ovinos/virologia , África do Sul/epidemiologia , Modelos Epidemiológicos
5.
Front Immunol ; 15: 1424307, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39011043

RESUMO

Introduction: Bluetongue (BT) poses a significant threat to the livestock industry, affecting various animal species and resulting in substantial economic losses. The existence of numerous BT virus (BTV) serotypes has hindered control efforts, highlighting the need for broad-spectrum vaccines. Methodology: In this study, we evaluated the conserved amino acid sequences within key non-structural (NS) proteins of BTV and identified numerous highly conserved murine- and bovine-specific MHC class I-restricted (MHC-I) CD8+ and MHC-II-restricted CD4+ epitopes. We then screened these conserved epitopes for antigenicity, allergenicity, toxicity, and solubility. Using these epitopes, we developed in silico-based broad-spectrum multiepitope vaccines with Toll-like receptor (TLR-4) agonists. The predicted proinflammatory cytokine response was assessed in silico using the C-IMMSIM server. Structural modeling and refinement were achieved using Robetta and GalaxyWEB servers. Finally, we assessed the stability of the docking complexes through extensive 100-nanosecond molecular dynamics simulations before considering the vaccines for codon optimization and in silico cloning. Results: We found many epitopes that meet these criteria within NS1 and NS2 proteins and developed in silico broad-spectrum vaccines. The immune simulation studies revealed that these vaccines induce high levels of IFN-γ and IL-2 in the vaccinated groups. Protein-protein docking analysis demonstrated promising epitopes with strong binding affinities to TLR-4. The docked complexes were stable, with minimal Root Mean Square Deviation and Root Mean Square Fluctuation values. Finally, the in silico-cloned plasmids have high % of GC content with > 0.8 codon adaptation index, suggesting they are suitable for expressing the protein vaccines in prokaryotic system. Discussion: These next-generation vaccine designs are promising and warrant further investigation in wet lab experiments to assess their immunogenicity, safety, and efficacy for practical application in livestock. Our findings offer a robust framework for developing a comprehensive, broad-spectrum vaccine, potentially revolutionizing BT control and prevention strategies in the livestock industry.


Assuntos
Vírus Bluetongue , Biologia Computacional , Epitopos de Linfócito T , Proteínas não Estruturais Virais , Vacinas Virais , Animais , Vírus Bluetongue/imunologia , Epitopos de Linfócito T/imunologia , Vacinas Virais/imunologia , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/genética , Camundongos , Biologia Computacional/métodos , Sorogrupo , Bovinos , Bluetongue/prevenção & controle , Bluetongue/imunologia , Bluetongue/virologia , Sequência Conservada
6.
Front Immunol ; 15: 1440407, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39072326

RESUMO

Introduction: Bluetongue (BT), caused by bluetongue virus (BTV), is an important arthropod-borne livestock disease listed by the World Organization for Animal Health. Live-attenuated and inactivated vaccines have permitted to control BT but they do not simultaneously protect against the myriad of BTV serotypes. Recently, we identified the highly conserved BTV nonstructural protein NS1 and the N-terminal region of NS2 as antigens capable of conferring multiserotype protection against BTV. Methods: Here, we designed Modified Vaccinia Ankara (MVA) viral vectors that expressed BTV-4 proteins VP2 or VP7 along with NS1 and NS2-Nt as well as MVAs that expressed proteins VP2, VP7 or NS1 and NS2-Nt. Results: Immunization of IFNAR(-/-) mice with two doses of MVA-NS1-2A-NS2-Nt protected mice from BTV-4M infection by the induction of an antigen-specific T cell immune response. Despite rMVA expressing VP7 alone were not protective in the IFNAR(-/-) mouse model, inclusion of VP7 in the vaccine formulation amplified the cell-mediated response induced by NS1 and NS2-Nt. Expression of VP2 elicited protective non-cross-reactive neutralizing antibodies (nAbs) in immunized animals and improved the protection observed in the MVA-NS1-2A-NS2-Nt immunized mice when these three BTV antigens were co-expressed. Moreover, vaccines candidates co-expressing VP2 or VP7 along with NS1 and NS2-Nt provided multiserotype protection. We assessed protective efficacy of both vaccine candidates in sheep against virulent challenge with BTV-4M. Discussion: Immunization with MVA-VP7-NS1-2A-NS2-Nt partially dumped viral replication and clinical disease whereas administration of MVA-VP2-NS1-2A-NS2-Nt promoted a complete protection, preventing viraemia and the pathology produced by BTV infection.


Assuntos
Vírus Bluetongue , Bluetongue , Proteínas do Capsídeo , Vetores Genéticos , Receptor de Interferon alfa e beta , Vaccinia virus , Proteínas não Estruturais Virais , Vacinas Virais , Animais , Vírus Bluetongue/imunologia , Vírus Bluetongue/genética , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/genética , Bluetongue/prevenção & controle , Bluetongue/imunologia , Bluetongue/virologia , Camundongos , Vacinas Virais/imunologia , Vacinas Virais/genética , Vaccinia virus/genética , Vaccinia virus/imunologia , Receptor de Interferon alfa e beta/genética , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/genética , Camundongos Knockout , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/sangue , Feminino
9.
Front Immunol ; 15: 1328820, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38357545

RESUMO

Introduction: Bluetongue virus (BTV) is an arthropod-borne Orbivirus that is almost solely transmitted by Culicoides biting midges and causes a globally important haemorrhagic disease, bluetongue (BT), in susceptible ruminants. Infection with BTV is characterised by immunosuppression and substantial lymphopenia at peak viraemia in the host. Methods: In this study, the role of cell-mediated immunity and specific T-cell subsets in BTV pathogenesis, clinical outcome, viral dynamics, immune protection, and onwards transmission to a susceptible Culicoides vector is defined in unprecedented detail for the first time, using an in vivo arboviral infection model system that closely mirrors natural infection and transmission of BTV. Individual circulating CD4+, CD8+, or WC1+ γδ T-cell subsets in sheep were depleted through the administration of specific monoclonal antibodies. Results: The absence of cytotoxic CD8+ T cells was consistently associated with less severe clinical signs of BT, whilst the absence of CD4+ and WC1+ γδ T cells both resulted in an increased clinical severity. The absence of CD4+ T cells also impaired both a timely protective neutralising antibody response and the production of IgG antibodies targeting BTV non-structural protein, NS2, highlighting that the CD4+ T-cell subset is important for a timely protective immune response. T cells did not influence viral replication characteristics, including onset/dynamics of viraemia, shedding, or onwards transmission of BTV to Culicoides. We also highlight differences in T-cell dependency for the generation of immunoglobulin subclasses targeting BTV NS2 and the structural protein, VP7. Discussion: This study identifies a diverse repertoire of T-cell functions during BTV infection in sheep, particularly in inducing specific anti-viral immune responses and disease manifestation, and will support more effective vaccination strategies.


Assuntos
Arbovírus , Vírus Bluetongue , Bluetongue , Ceratopogonidae , Ovinos , Animais , Gado , Viremia , Linfócitos T CD8-Positivos , Ruminantes , Subpopulações de Linfócitos T , Bluetongue/prevenção & controle , Ceratopogonidae/fisiologia
10.
Vet Rec ; 194(1): 49, 2024 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-38180194
11.
13.
Bull Math Biol ; 85(7): 69, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37318632

RESUMO

Bluetongue (BT) is a well-known vector-borne disease that infects ruminants such as sheep, cattle, and deer with high mortality rates. Recent outbreaks in Europe highlight the importance of understanding vector-host dynamics and potential courses of action to mitigate the damage that can be done by BT. We present an agent-based model, entitled 'MidgePy', that focuses on the movement of individual Culicoides spp. biting midges and their interactions with ruminants to understand their role as vectors in BT outbreaks, especially in regions that do not regularly experience outbreaks. The results of our sensitivity analysis suggest that midge survival rate has a significant impact on the probability of a BTV outbreak as well as its severity. Using midge flight activity as a proxy for temperature, we found that an increase in environmental temperature corresponded with an increased probability of outbreak after identifying parameter regions where outbreaks are more likely to occur. This suggests that future methods to control BT spread could combine large-scale vaccination programs with biting midge population control measures such as the use of pesticides. Spatial heterogeneity in the environment is also explored to give insight on optimal farm layouts to reduce the potential for BT outbreaks.


Assuntos
Vírus Bluetongue , Bluetongue , Ceratopogonidae , Cervos , Bovinos , Ovinos , Animais , Bluetongue/epidemiologia , Bluetongue/prevenção & controle , Modelos Biológicos , Insetos Vetores , Conceitos Matemáticos , Ruminantes , Surtos de Doenças/veterinária
14.
Arch Virol ; 168(7): 179, 2023 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-37310539

RESUMO

Bluetongue virus (BTV) causes bluetongue disease in ruminants and sheep. The current live attenuated and inactivated vaccines available for prevention pose several risks, and there is thus a need for vaccines that are safer, economically viable, and effective against multiple circulating serotypes. This work describes the development of recombinant virus-like particle (VLP) vaccine candidates in plants, which are assembled by co-expression of the four BTV serotype 8 major structural proteins. We show that substitution of a neutralising tip domain of BTV8 VP2 with that of BTV1 VP2 resulted in the assembly of VLPs that stimulated serotype-specific antibodies as well as virus-specific neutralising antibodies.


Assuntos
Vírus Bluetongue , Bluetongue , Vacinas de Partículas Semelhantes a Vírus , Animais , Ovinos , Vírus Bluetongue/genética , Anticorpos , Bluetongue/prevenção & controle , Sorogrupo , Vacinas de Partículas Semelhantes a Vírus/genética
15.
J Infect Dev Ctries ; 17(2): 147-156, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36897898

RESUMO

Bluetongue (BT), once considered a disease of sheep confined to the southern African region, has spread all over the world. BT is a viral disease caused by the bluetongue virus (BTV). BT is regarded as an economically important disease in ruminants of compulsory notification to OIE. BTV is transmitted by the bite of Culicoides species. Research over the years has led to a better understanding of the disease, the nature of the virus life cycle between ruminants and Culicoides species, and its distribution in different geographical regions. Advances have also been made in understanding the molecular structure and function of the virus, the biology of the Culicoides species, its ability to transmit the disease, and the persistence of the virus inside the Culicoides and the mammalian hosts. Global climate change has enabled the colonization of new habitats and the spread of the virus into additional species of the Culicoides vector. This review highlights some of the current findings on the status of BT in the world based on the latest research on disease aspects, virus-host-vector interactions, and the different diagnostic approaches and control strategies available for BTV.


Assuntos
Vírus Bluetongue , Bluetongue , Ceratopogonidae , Animais , Ovinos , Insetos Vetores , Ruminantes , Bluetongue/prevenção & controle
16.
Risk Anal ; 43(6): 1124-1136, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35994609

RESUMO

Bluetongue (BT) causes an economic loss of $3 billion every year in the world. After two serious occurrences of BT (bluetongue virus [BTV] occurrence in 2006 and 2015), France has been controlling for decades, but it has not been eradicated. As the largest live cattle export market in the world, France is also one of the major exporters of breeding animals and genetic materials in the world. The biosafety of its exported cattle and products has always been a concern. The scenario tree quantitative model was used to analyze the risk of BTV release from French exported live cattle and bovine semen. The results showed that with the increase in vaccination coverage rates, the risk decreased. If the vaccine coverage is 0%, the areas with the highest average risk probability of BTV-4 and BTV-8 release from exported live cattle were Haute-Savoie and Puy-de-Dôme, and the risk was 2.96 × 10-4 and 4.25 × 10-4 , respectively. When the vaccine coverage was 90%, the risk probability of BTV-4 and BTV-8 release from exported live cattle was 2.96 × 10-5 and 4.24 × 10-5 , respectively. The average probability of BTV-8 release from bovine semen was 1.09 × 10-10 . Sensitivity analysis showed that the probability of false negative polymerase chain reaction (PCR) test and the probability of BT infection in the bull breeding station had an impact on the model. The identification of high-risk areas and the discovery of key control measures provide a reference for decision makers to assess the risk of French exports of live cattle and bovine semen.


Assuntos
Vírus Bluetongue , Bluetongue , Doenças dos Bovinos , Ovinos/genética , Animais , Bovinos , Masculino , Sorogrupo , Vírus Bluetongue/genética , França/epidemiologia , Bluetongue/epidemiologia , Bluetongue/prevenção & controle , Reação em Cadeia da Polimerase , Doenças dos Bovinos/epidemiologia
17.
Trop Biomed ; 39(1): 47-54, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-35507924

RESUMO

There are many infectious animal diseases in T urkey and generally, vaccination is the primarly control strategy to combat them. However, it is difficult to apply all vaccines in a definite period in the field due to limitations of the labor and finance. Rapid vaccination and effective use of labor can be possible with the help of simultaneous vaccine administrations. The study aims to show the effects of simultaneous foot-and-mouth disease (FMD), peste des petits ruminants (PPR), sheep pox and goat pox (SGP), and bluetongue (BT) vaccine administration on the antibody response of sheep. For this aim, 30 sheep were divided into one experiment and 5 control groups. Blood samples were collected in each group at 0, 30 and 60 days post-vaccination (DPV). Immune response was measured with virus neutralization test (VNT) and, liquid phase blocking ELISA (LPBE) for FMDV; VNT for BTV and PPR. A live virus challenge study was performed to determine the immune response of SGP vaccine. As a result, antibody titers for each vaccine agent decreased on 60 DPV with the simultaneous vaccination except FMD. The difference between means of antibody titer decrease with single and simultaneous vaccinations is significant especially for BTV and PPR vaccines at 60DPV (p<0.05). Briefly, this decreasing immune response of three live vaccines can be explained with the development of the interference, administration of these vaccines from the same injection site, the effect of cytokines, especially IL-10 effect of SGP vaccine. It was concluded that four vaccines can not be used simultaneously in sheep.


Assuntos
Bluetongue , Febre Aftosa , Doenças das Cabras , Peste dos Pequenos Ruminantes , Vírus da Peste dos Pequenos Ruminantes , Doenças dos Ovinos , Animais , Anticorpos Antivirais , Formação de Anticorpos , Bluetongue/prevenção & controle , Febre Aftosa/prevenção & controle , Doenças das Cabras/prevenção & controle , Cabras , Peste dos Pequenos Ruminantes/prevenção & controle , Ovinos , Doenças dos Ovinos/prevenção & controle , Vacinação/veterinária , Vacinas Atenuadas
18.
Methods Mol Biol ; 2465: 177-193, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35118622

RESUMO

Modified vaccinia virus Ankara (MVA) is employed widely as an experimental vaccine vector for its abortive replication in mammalian cells and high expression level of foreign/heterologous genes. Recombinant MVAs (rMVAs) are used as platforms for protein production as well as vectors to generate vaccines against a wide range of infectious diseases and other pathologies. The portrait of the virus combines desirable elements such as high-level biological safety, the ability to activate appropriate innate immune mediators upon vaccination , and the capacity to deliver substantial amounts of heterologous antigens. rMVAs encoding proteins of Bluetongue virus (BTV), an orbivirus that infects domestic and wild ruminants through transmission by biting midges of the Culicoides species, are excellent vaccine candidates against this virus. In this chapter, we describe the methods for the generation of rMVAs encoding VP2, NS1, and VP7 proteins of BTV . The included protocols cover the cloning of VP2, NS1, and VP7 BTV-4 genes in a transfer plasmid, the construction of rMVAs, the titration of virus working stocks, and the protein expression analysis by immunofluorescence and radiolabeling of rMVA infected cells as well as virus purification procedure.


Assuntos
Vírus Bluetongue , Bluetongue , Vacinas Virais , Animais , Bluetongue/prevenção & controle , Vírus Bluetongue/genética , Proteínas do Capsídeo/genética , Mamíferos , Ovinos , Vaccinia virus/genética
19.
J Virol ; 96(3): e0161421, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34787454

RESUMO

Bluetongue, caused by bluetongue virus (BTV), is a widespread arthropod-borne disease of ruminants that entails a recurrent threat to the primary sector of developed and developing countries. In this work, we report modified vaccinia virus Ankara (MVA) and ChAdOx1-vectored vaccines designed to simultaneously express the immunogenic NS1 protein and/or NS2-Nt, the N-terminal half of protein NS2 (NS21-180). A single dose of MVA or ChAdOx1 expressing NS1-NS2-Nt improved the protection conferred by NS1 alone in IFNAR(-/-) mice. Moreover, mice immunized with ChAdOx1/MVA-NS1, ChAdOx1/MVA-NS2-Nt, or ChAdOx1/MVA-NS1-NS2-Nt developed strong cytotoxic CD8+ T-cell responses against NS1, NS2-Nt, or both proteins and were fully protected against a lethal infection with BTV serotypes 1, 4, and 8. Furthermore, although a single immunization with ChAdOx1-NS1-NS2-Nt partially protected sheep against BTV-4, the administration of a booster dose of MVA-NS1-NS2-Nt promoted a faster viral clearance, reduction of the period and level of viremia and also protected from the pathology produced by BTV infection. IMPORTANCE Current BTV vaccines are effective but they do not allow to distinguish between vaccinated and infected animals (DIVA strategy) and are serotype specific. In this work we have develop a DIVA multiserotype vaccination strategy based on adenoviral (ChAdOx1) and MVA vaccine vectors, the most widely used in current phase I and II clinical trials, and the conserved nonstructural BTV proteins NS1 and NS2. This immunization strategy solves the major drawbacks of the current marketed vaccines.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/prevenção & controle , Vetores Genéticos/genética , Vaccinia virus/genética , Proteínas não Estruturais Virais/genética , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus Bluetongue/classificação , Vetores Genéticos/imunologia , Imunidade Celular , Imunização , Imunogenicidade da Vacina , Sorogrupo , Ovinos , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vaccinia virus/imunologia , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
20.
J Virol Methods ; 298: 114288, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34536487

RESUMO

Previously, we reported the detection of two novel bluetongue virus (BTV) strains (SPvvvv/02 and SPvvvv/03), possibly representing new BTV genotypes, in a batch of sheeppox vaccine. We developed type-specific RT-qPCR assays (targeting genome segment 2) for these two new BTV strains. The limit of detection of both assays was 10 genome copies/µl and no cross-reactivity with other BTV genotypes was observed. The performance of three other BTV group-specific diagnostic assays was also tested against the putative novel genotypes. RT-qPCR assays targeting BTV segment 9 and 10 detected both strains (SPvvvv/02 and SPvvvv/03) whereas a BTV segment 1 RT-qPCR assay was unable to detect either BTV strain. The work presented here expands upon the current repertoire of RT-qPCR assays for BTV genotype determination.


Assuntos
Vírus Bluetongue , Bluetongue , Vacinas , Animais , Bluetongue/diagnóstico , Bluetongue/prevenção & controle , Vírus Bluetongue/genética , Genótipo , Reação em Cadeia da Polimerase em Tempo Real , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA