Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 541
Filtrar
1.
Viruses ; 16(4)2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38675966

RESUMO

A devastating bluetongue (BT) epidemic caused by bluetongue virus serotype 3 (BTV-3) has spread throughout most of the Netherlands within two months since the first infection was officially confirmed in the beginning of September 2023. The epidemic comes with unusually strong suffering of infected cattle through severe lameness, often resulting in mortality or euthanisation for welfare reasons. In total, tens of thousands of sheep have died or had to be euthanised. By October 2023, more than 2200 locations with ruminant livestock were officially identified to be infected with BTV-3, and additionally, ruminants from 1300 locations were showing BTV-associated clinical symptoms (but not laboratory-confirmed BT). Here, we report on the spatial spread and dynamics of this BT epidemic. More specifically, we characterized the distance-dependent intensity of the between-holding transmission by estimating the spatial transmission kernel and by comparing it to transmission kernels estimated earlier for BTV-8 transmission in Northwestern Europe in 2006 and 2007. The 2023 BTV-3 kernel parameters are in line with those of the transmission kernel estimated previously for the between-holding spread of BTV-8 in Europe in 2007. The 2023 BTV-3 transmission kernel has a long-distance spatial range (across tens of kilometres), evidencing that in addition to short-distance dispersal of infected midges, other transmission routes such as livestock transports probably played an important role.


Assuntos
Vírus Bluetongue , Bluetongue , Epidemias , Sorogrupo , Animais , Bluetongue/epidemiologia , Bluetongue/transmissão , Bluetongue/virologia , Vírus Bluetongue/classificação , Países Baixos/epidemiologia , Ovinos , Bovinos , Doenças dos Bovinos/virologia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/transmissão
2.
J Virol ; 96(13): e0053122, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35727032

RESUMO

Segmented RNA viruses are a taxonomically diverse group that can infect plant, wildlife, livestock and human hosts. A shared feature of these viruses is the ability to exchange genome segments during coinfection of a host by a process termed "reassortment." Reassortment enables rapid evolutionary change, but where transmission involves a biological arthropod vector, this change is constrained by the selection pressures imposed by the requirement for replication in two evolutionarily distant hosts. In this study, we use an in vivo, host-arbovirus-vector model to investigate the impact of reassortment on two phenotypic traits, virus infection rate in the vector and virulence in the host. Bluetongue virus (BTV) (Reoviridae) is the causative agent of bluetongue (BT), an economically important disease of domestic and wild ruminants and deer. The genome of BTV comprises 10 linear segments of dsRNA, and the virus is transmitted between ruminants by Culicoides biting midges (Diptera: Ceratopogonidae). Five strains of BTV representing three serotypes (BTV-1, BTV-4, and BTV-8) were isolated from naturally infected ruminants in Europe and ancestral/reassortant lineage status assigned through full genome sequencing. Each strain was then assessed in parallel for the ability to replicate in vector Culicoides and to cause BT in sheep. Our results demonstrate that two reassortment strains, which themselves became established in the field, had obtained high replication ability in C. sonorensis from one of the ancestral virus strains, which allowed inferences of the genome segments conferring this phenotypic trait. IMPORTANCE Reassortment between virus strains can lead to major shifts in the transmission parameters and virulence of segmented RNA viruses, with consequences for spread, persistence, and impact. The ability of these pathogens to adapt rapidly to their environment through this mechanism presents a major challenge in defining the conditions under which emergence can occur. Utilizing a representative mammalian host-insect vector infection and transmission model, we provide direct evidence of this phenomenon in closely related ancestral and reassortant strains of BTV. Our results demonstrate that efficient infection of Culicoides observed for one of three ancestral BTV strains was also evident in two reassortant strains that had subsequently emerged in the same ecosystem.


Assuntos
Vetores Artrópodes , Vírus Bluetongue , Bluetongue , Ceratopogonidae , Doenças dos Ovinos , Animais , Vetores Artrópodes/virologia , Bluetongue/transmissão , Bluetongue/virologia , Vírus Bluetongue/classificação , Vírus Bluetongue/genética , Vírus Bluetongue/patogenicidade , Ceratopogonidae/virologia , Cervos , Fenótipo , Vírus Reordenados/metabolismo , Ovinos , Doenças dos Ovinos/transmissão , Doenças dos Ovinos/virologia , Replicação Viral
3.
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
4.
J Virol ; 96(1): e0167721, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34669428

RESUMO

Bluetongue virus (BTV), a member of the Orbivirus genus, is transmitted by biting midges (gnats, Culicoides sp.) and is one of the most widespread animal pathogens, causing serious outbreaks in domestic animals, particularly in sheep, with high economic impact. The non-enveloped BTV particle is a double-capsid structure of seven proteins and a genome of 10 double-stranded RNA segments. Although the outermost spike-like VP2 acts as the attachment protein during BTV entry, no specific host receptor has been identified for BTV. Recent high-resolution cryo-electron (cryoEM) structures and biological data have suggested that VP2 may interact with sialic acids (SAs). To confirm this, we have generated protein-based nanoparticles displaying multivalent VP2 and used them to probe glycan arrays. The data show that VP2 binds α2,3-linked SA with high affinity but also binds α2,6-linked SA. Further, Maackia amurensis lectin II (MAL II) and Sambucus nigra lectin (SNA), which specifically bind α2,3-linked and α2,6-linked SAs, respectively, inhibited BTV infection and virus growth in susceptible sheep cells while SNA alone inhibited virus growth in Culicoides-derived cells. A combination of hydrogen deuterium exchange mass spectrometry and site-directed mutagenesis allowed the identification of the specific SA binding residues of VP2. This study provides direct evidence that sialic acids act as key receptor for BTV and that the outer capsid protein VP2 specifically binds SA during BTV entry in both mammalian and insect cells. IMPORTANCE To date no receptor has been assigned for non-enveloped bluetongue virus. To determine if the outermost spike-like VP2 protein is responsible for host cell attachment via interaction with sialic acids, we first generated a protein-based VP2-nanoparticle, for the multivalent presentation of recombinant VP2 protein. Using nanoparticles displaying VP2 to probe a glycan array, we identified that VP2 binds both α2,3-linked and α2,6-linked sialic acids. Lectin inhibitors targeting both linkages of sialic acids showed strong inhibition to BTV infection and progeny virus production in mammalian cells; however the inhibition was only seen with the lectin targeting α2,6-linked sialic acid in insect vector cells. In addition, we identified the VP2 sialic acid binding sites in the exposed tip domain. Our data provides direct evidence that sialic acids act as key receptors for BTV attachment and entry in to both mammalian and insect cells.


Assuntos
Sítios de Ligação , Vírus Bluetongue/fisiologia , Bluetongue/virologia , Proteínas do Capsídeo/metabolismo , Internalização do Vírus , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Interações Hospedeiro-Patógeno , Lectinas/metabolismo , Espectrometria de Massas , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Receptores Virais/química , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo
5.
Viruses ; 13(11)2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34834993

RESUMO

Epizootic haemorragic disease (EHD) is an important disease of white-tailed deer and can cause a bluetongue-like illness in cattle. A definitive diagnosis of EHD relies on molecular assays such as real-time RT-qPCR or conventional PCR. Reverse transcription loop-mediated isothermal amplification (RT-LAMP) is a cost-effective, specific, and sensitive technique that provides an alternative to RT-qPCR. We designed two sets of specific primers targeting segment-9 of the EHD virus genome to enable the detection of western and eastern topotypes, and evaluated their performance in singleplex and multiplex formats using cell culture isolates (n = 43), field specimens (n = 20), and a proficiency panel (n = 10). The limit of detection of the eastern and western RT-LAMP assays was estimated as ~24.36 CT and as ~29.37 CT in relation to real-time RT-qPCR, respectively, indicating a greater sensitivity of the western topotype singleplex RT-LAMP. The sensitivity of the western topotype RT-LAMP assay, relative to the RT-qPCR assay, was 72.2%, indicating that it could be theoretically used to detect viraemic cervines and bovines. For the first time, an RT-LAMP assay was developed for the rapid detection of the EHD virus that could be used as either a field test or high throughput screening tool in established laboratories to control the spread of EHD.


Assuntos
Vírus da Doença Hemorrágica Epizoótica/isolamento & purificação , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Amplificação de Ácido Nucleico/métodos , Infecções por Reoviridae/diagnóstico , Infecções por Reoviridae/virologia , Animais , Bluetongue/virologia , Bovinos , Primers do DNA/genética , Cervos , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Reversa , Sensibilidade e Especificidade
6.
Viruses ; 13(11)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34835014

RESUMO

Bluetongue virus serotypes 1 to 24 are transmitted primarily by infected Culicoides midges, in which they also replicate. However, "atypical" BTV serotypes (BTV-25, -26, -27 and -28) have recently been identified that do not infect and replicate in adult Culicoides, or a Culicoides derived cell line (KC cells). These atypical viruses are transmitted horizontally by direct contact between infected and susceptible hosts (primarily small ruminants) causing only mild clinical signs, although the exact transmission mechanisms involved have yet to be determined. We used reverse genetics to generate a strain of BTV-1 (BTV-1 RGC7) which is less virulent, infecting IFNAR(-/-) mice without killing them. Reassortant viruses were also engineered, using the BTV-1 RGC7 genetic backbone, containing individual genome segments derived from BTV-26. These reassortant viruses were used to explore the genetic control of horizontal transmission (HT) in the IFNAR(-/-) mouse model. Previous studies showed that genome segments 1, 2 and 3 restrict infection of Culicoides cells, along with a minor role for segment 7. The current study demonstrates that genome segments 2, 5 and 10 of BTV-26 (coding for proteins VP2, NS1 and NS3/NS3a/NS5, respectively) are individually sufficient to promote HT.


Assuntos
Vírus Bluetongue/genética , Transmissão de Doença Infecciosa , Vírus Reordenados/genética , Animais , Bluetongue/virologia , Ceratopogonidae/virologia , Modelos Animais de Doenças , Engenharia Genética , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta , Sorogrupo
7.
Nat Microbiol ; 6(11): 1424-1432, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34702979

RESUMO

Bluetongue virus (BTV) is a non-enveloped virus and causes substantial morbidity and mortality in ruminants such as sheep. Fashioning a receptor-binding protein (VP2) and a membrane penetration protein (VP5) on the surface, BTV releases its genome-containing core (VP3 and VP7) into the host cell cytosol after perforation of the endosomal membrane. Unlike enveloped ones, the entry mechanisms of non-enveloped viruses into host cells remain poorly understood. Here we applied single-particle cryo-electron microscopy, cryo-electron tomography and structure-guided functional assays to characterize intermediate states of BTV cell entry in endosomes. Four structures of BTV at the resolution range of 3.4-3.9 Å show the different stages of structural rearrangement of capsid proteins on exposure to low pH, including conformational changes of VP5, stepwise detachment of VP2 and a small shift of VP7. In detail, sensing of the low-pH condition by the VP5 anchor domain triggers three major VP5 actions: projecting the hidden dagger domain, converting a surface loop to a protonated ß-hairpin that anchors VP5 to the core and stepwise refolding of the unfurling domains into a six-helix stalk. Cryo-electron tomography structures of BTV interacting with liposomes show a length decrease of the VP5 stalk from 19.5 to 15.5 nm after its insertion into the membrane. Our structures, functional assays and structure-guided mutagenesis experiments combined indicate that this stalk, along with dagger domain and the WHXL motif, creates a single pore through the endosomal membrane that enables the viral core to enter the cytosol. Our study unveils the detailed mechanisms of BTV membrane penetration and showcases general methods to study cell entry of other non-enveloped viruses.


Assuntos
Vírus Bluetongue/metabolismo , Bluetongue/virologia , Proteínas do Capsídeo/metabolismo , Endossomos/virologia , Animais , Vírus Bluetongue/química , Vírus Bluetongue/genética , Vírus Bluetongue/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Microscopia Crioeletrônica , Endossomos/química , Concentração de Íons de Hidrogênio , Modelos Moleculares , Ovinos , Doenças dos Ovinos/virologia , Internalização do Vírus
8.
Viruses ; 13(9)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34578322

RESUMO

In this article, we describe the development and evaluation of a double antigen sandwich enzyme-linked immunosorbent assay (ELISA) able to detect serotype 4-specific antibodies from BTV-4 infected or vaccinated animals using a recombinant BTV-4 VP2 protein. The coding sequence of VP2 was inserted into a pVote plasmid by recombination in the Gateway® cloning system. Vaccinia virus (VacV) was used as a vector for the expression of the recombinant VP2. After production in BSR cells, recombinant VP2 was purified by immunoprecipitation using a FLAG tag and then used both as the coated ELISA antigen and as the HRP-tagged conjugate. The performance of the ELISA was evaluated with 1186 samples collected from BTV negative, infected or vaccinated animals. The specificity and sensitivity of the BTV-4 ELISA were above the expected standards for the detection of anti-BTV-4 VP2 antibodies in animals reared in Europe or in the Mediterranean basin. Cross-reactions were observed with reference sera for serotypes 10 and 20, and to a lesser extent with serotypes 12, 17 and 24, due to their genetic proximity to serotype 4. Nevertheless, these serotypes have never been detected in Europe and the Mediterranean area. This ELISA, which requires only the production of a recombinant protein, can be used to detect BTV serotype 4-specific antibodies and is therefore an attractive alternative diagnostic method to serum neutralization.


Assuntos
Vírus Bluetongue/isolamento & purificação , Bluetongue/diagnóstico , Ensaio de Imunoadsorção Enzimática/métodos , Animais , Anticorpos Antivirais/sangue , Bluetongue/virologia , Europa (Continente) , Proteínas Recombinantes/genética , Sorogrupo , Ovinos , Vaccinia virus/imunologia
9.
Viruses ; 13(9)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34578362

RESUMO

Since the 2000s, the distribution of bluetongue virus (BTV) has changed, leading to numerous epidemics and economic losses in Europe. Previously, we found a BTV-4 field strain with a higher infection rate of a Culicoides vector than a BTV-1 field strain has. We reverse-engineered parental BTV-1 and BTV-4 strains and created BTV-1/BTV-4 reassortants to elucidate the influence of individual BTV segments on BTV replication in both C. sonorensis midges and in KC cells. Substitution of segment 2 (Seg-2) with Seg-2 from the rBTV-4 significantly increased vector infection rate in reassortant BTV-14S2 (30.4%) in comparison to reverse-engineered rBTV-1 (1.0%). Replacement of Seg-2, Seg-6 and Seg-7 with those from rBTV-1 in reassortant BTV-41S2S6S7 (2.9%) decreased vector infection rate in comparison to rBTV-4 (30.2%). However, triple-reassorted BTV-14S2S6S7 only replicated to comparatively low levels (3.0%), despite containing Seg-2, Seg-6 and Seg-7 from rBTV-4, indicating that vector infection rate is influenced by interactions of multiple segments and/or host-mediated amino acid substitutions within segments. Overall, these results demonstrated that we could utilize reverse-engineered viruses to identify the genetic basis influencing BTV replication within Culicoides vectors. However, BTV replication dynamics in KC cells were not suitable for predicting the replication ability of these virus strains in Culicoides midges.


Assuntos
Vírus Bluetongue/genética , Vírus Bluetongue/fisiologia , Ceratopogonidae/virologia , Insetos Vetores/virologia , Animais , Bluetongue/virologia , Linhagem Celular , Europa (Continente) , Vírus Reordenados/genética , Replicação Viral , Sequenciamento Completo do Genoma
10.
Parasit Vectors ; 14(1): 491, 2021 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-34563238

RESUMO

BACKGROUND: Bluetongue is a serious disease of ruminants caused by the bluetongue virus (BTV). BTV is transmitted by biting midges (Culicoides spp.). Serological evidence from livestock and the presence of at least one competent vector species of Culicoides suggests that transmission of BTV is possible and may have occurred in Kazakhstan. METHODS: We estimated the risk of transmission using a mathematical model of the reproduction number R0 for bluetongue. This model depends on livestock density and climatic factors which affect vector density. Data on climate and livestock numbers from the 2466 local communities were used. This, together with previously published model parameters, was used to estimate R0 for each month of the year. We plotted the results on isopleth maps of Kazakhstan using interpolation to smooth the irregular data. We also mapped the estimated proportion of the population requiring vaccination to prevent outbreaks of bluetongue. RESULTS: The results suggest that transmission of bluetongue in Kazakhstan is not possible in the winter from October to March. Assuming there are vector-competent species of Culicoides endemic in Kazakhstan, then low levels of risk first appear in the south of Kazakhstan in April before spreading north and intensifying, reaching maximum levels in northern Kazakhstan in July. The risk declined in September and had disappeared by October. CONCLUSION: These results should aid in surveillance efforts for the detection and control of bluetongue in Kazakhstan by indicating where and when outbreaks of bluetongue are most likely to occur. The results also indicate where vaccination efforts should be focussed to prevent outbreaks of disease.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/epidemiologia , Bluetongue/transmissão , Animais , Bluetongue/virologia , Clima , Insetos Vetores/fisiologia , Insetos Vetores/virologia , Gado/virologia , Modelos Teóricos , Estações do Ano
11.
Viruses ; 13(8)2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34452303

RESUMO

Statin derivatives can inhibit the replication of a range of viruses, including hepatitis C virus (HCV, Hepacivirus), dengue virus (Flavivirus), African swine fever virus (Asfarviridae) and poliovirus (Picornaviridae). We assess the antiviral effect of fluvastatin in cells infected with orbiviruses (bluetongue virus (BTV) and Great Island virus (GIV)). The synthesis of orbivirus outer-capsid protein VP2 (detected by confocal immunofluorescence imaging) was used to assess levels of virus replication, showing a reduction in fluvastatin-treated cells. A reduction in virus titres of ~1.7 log (98%) in fluvastatin-treated cells was detected by a plaque assay. We have previously identified a fourth non-structural protein (NS4) of BTV and GIV, showing that it interacts with lipid droplets in infected cells. Fluvastatin, which inhibits 3-hydroxy 3-methyl glutaryl CoA reductase in the mevalonic acid pathway, disrupts these NS4 interactions. These findings highlight the role of the lipid pathways in orbivirus replication and suggest a greater role for the membrane-enveloped orbivirus particles than previously recognised. Chemical intermediates of the mevalonic acid pathway were used to assess their potential to rescue orbivirus replication. Pre-treatment of IFNAR(-/-) mice with fluvastatin promoted their survival upon challenge with live BTV, although only limited protection was observed.


Assuntos
Antivirais/farmacologia , Vírus Bluetongue/efeitos dos fármacos , Fluvastatina/farmacologia , Ácido Mevalônico/metabolismo , Orbivirus/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Bluetongue/tratamento farmacológico , Bluetongue/virologia , Vírus Bluetongue/fisiologia , Linhagem Celular , Ceratopogonidae/enzimologia , Ceratopogonidae/virologia , Fluvastatina/uso terapêutico , Humanos , Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Redes e Vias Metabólicas , Camundongos , Orbivirus/fisiologia , Receptor de Interferon alfa e beta/genética , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/fisiologia
12.
Viruses ; 13(8)2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34452321

RESUMO

Bluetongue (BT) is a severe and economically important disease of ruminants that is widely distributed around the world, caused by the bluetongue virus (BTV). More than 28 different BTV serotypes have been identified in serum neutralisation tests (SNT), which, along with geographic variants (topotypes) within each serotype, reflect differences in BTV outer-capsid protein VP2. VP2 is the primary target for neutralising antibodies, although the basis for cross-reactions and serological variations between and within BTV serotypes is poorly understood. Recombinant BTV VP2 proteins (rVP2) were expressed in Nicotiana benthamiana, based on sequence data for isolates of thirteen BTV serotypes (primarily from Europe), including three 'novel' serotypes (BTV-25, -26 and -27) and alternative topotypes of four serotypes. Cross-reactions within and between these viruses were explored using rabbit anti-rVP2 sera and post BTV-infection sheep reference-antisera, in I-ELISA (with rVP2 target antigens) and SNT (with reference strains of BTV-1 to -24, -26 and -27). Strong reactions were generally detected with homologous rVP2 proteins or virus strains/serotypes. The sheep antisera were largely serotype-specific in SNT, but more cross-reactive by ELISA. Rabbit antisera were more cross-reactive in SNT, and showed widespread, high titre cross-reactions against homologous and heterologous rVP2 proteins in ELISA. Results were analysed and visualised by antigenic cartography, showing closer relationships in some, but not all cases, between VP2 topotypes within the same serotype, and between serotypes belonging to the same 'VP2 nucleotype'.


Assuntos
Vírus Bluetongue/classificação , Vírus Bluetongue/genética , Proteínas do Capsídeo/classificação , Proteínas do Capsídeo/genética , Reações Cruzadas/imunologia , Sorogrupo , Animais , Antígenos Virais/imunologia , Bluetongue/imunologia , Bluetongue/virologia , Vírus Bluetongue/imunologia , Proteínas do Capsídeo/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Coelhos/imunologia , Ruminantes/imunologia , Sorotipagem , Ovinos/imunologia , Nicotiana/genética
13.
Viruses ; 13(8)2021 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-34452376

RESUMO

Viral infections have long provided a platform to understand the workings of immunity. For instance, great strides towards defining basic immunology concepts, such as MHC restriction of antigen presentation or T-cell memory development and maintenance, have been achieved thanks to the study of lymphocytic choriomeningitis virus (LCMV) infections. These studies have also shaped our understanding of antiviral immunity, and in particular T-cell responses. In the present review, we discuss how bluetongue virus (BTV), an economically important arbovirus from the Reoviridae family that affects ruminants, affects adaptive immunity in the natural hosts. During the initial stages of infection, BTV triggers leucopenia in the hosts. The host then mounts an adaptive immune response that controls the disease. In this work, we discuss how BTV triggers CD8+ T-cell expansion and neutralizing antibody responses, yet in some individuals viremia remains detectable after these adaptive immune mechanisms are active. We present some unpublished data showing that BTV infection also affects other T cell populations such as CD4+ T-cells or γδ T-cells, as well as B-cell numbers in the periphery. This review also discusses how BTV evades these adaptive immune mechanisms so that it can be transmitted back to the arthropod host. Understanding the interaction of BTV with immunity could ultimately define the correlates of protection with immune mechanisms that would improve our knowledge of ruminant immunology.


Assuntos
Imunidade Adaptativa , Anticorpos Antivirais/sangue , Vírus Bluetongue/imunologia , Bluetongue/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Neutralizantes/sangue , Apresentação de Antígeno , Bluetongue/virologia , Ruminantes/imunologia , Ovinos/imunologia , Linfócitos T/classificação
14.
Parasit Vectors ; 14(1): 382, 2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34330315

RESUMO

The transmission of vector-borne diseases is governed by complex factors including pathogen characteristics, vector-host interactions, and environmental conditions. Temperature is a major driver for many vector-borne diseases including Bluetongue viral (BTV) disease, a midge-borne febrile disease of ruminants, notably livestock, whose etiology ranges from mild or asymptomatic to rapidly fatal, thus threatening animal agriculture and the economy of affected countries. Using modeling tools, we seek to predict where the transmission can occur based on suitable temperatures for BTV. We fit thermal performance curves to temperature-sensitive midge life-history traits, using a Bayesian approach. We incorporate these curves into S(T), a transmission suitability metric derived from the disease's basic reproductive number, [Formula: see text] This suitability metric encompasses all components that are known to be temperature-dependent. We use trait responses for two species of key midge vectors, Culicoides sonorensis and Culicoides variipennis present in North America. Our results show that outbreaks of BTV are more likely between 15[Formula: see text] C and [Formula: see text], with predicted peak transmission risk at 26 [Formula: see text] C. The greatest uncertainty in S(T) is associated with the following: the uncertainty in mortality and fecundity of midges near optimal temperature for transmission; midges' probability of becoming infectious post-infection at the lower edge of the thermal range; and the biting rate together with vector competence at the higher edge of the thermal range. We compare three model formulations and show that incorporating thermal curves into all three leads to similar BTV risk predictions. To demonstrate the utility of this modeling approach, we created global suitability maps indicating the areas at high and long-term risk of BTV transmission, to assess risk and to anticipate potential locations of disease establishment.


Assuntos
Bluetongue/transmissão , Ceratopogonidae/virologia , Insetos Vetores/virologia , Gado , Temperatura , Animais , Teorema de Bayes , Bluetongue/epidemiologia , Bluetongue/prevenção & controle , Bluetongue/virologia , Ceratopogonidae/crescimento & desenvolvimento , Surtos de Doenças/veterinária , Feminino , Insetos Vetores/crescimento & desenvolvimento , Gravidez , Fatores de Risco , Ovinos , Vacinas Virais/normas
15.
Viruses ; 13(5)2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-34067226

RESUMO

The bluetongue virus (BTV) is transmitted by Culicoides biting midges and causes bluetongue (BT), an OIE-notifiable disease of ruminants. At least 29 BTV serotypes are described as determined by the outer shell proteins VP2 and VP5. Vaccination is the most effective control measure. Inactivated and live-attenuated vaccines (LAVs) are currently available. These vaccines have their specific pros and cons, and both are not DIVA vaccines. The BT Disabled Infectious Single Animal (DISA) vaccine platform is based on LAV without nonessential NS3/NS3a expression and is applicable for many serotypes by the exchange of outer shell proteins. The DISA vaccine is effective and completely safe. Further, transmission of the DISA vaccine by midges is blocked (DISA principle). Finally, the DISA vaccine enables DIVA because of a lack of antibodies against the immunogenic NS3/NS3a protein (DIVA principle). The deletion of 72 amino acids (72aa) in NS3/NS3a is sufficient to block virus propagation in midges. Here, we show that a prototype DISA vaccine based on LAV with the 72aa deletion enables DIVA, is completely safe and induces a long-lasting serotype-specific protection in cattle. In conclusion, the in-frame deletion of 72-aa codons in the BT DISA/DIVA vaccine platform is sufficient to fulfil all the criteria for modern veterinary vaccines.


Assuntos
Vírus Bluetongue/genética , Vírus Bluetongue/imunologia , Doenças dos Bovinos/prevenção & controle , Vacinas Atenuadas/imunologia , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Bluetongue/prevenção & controle , Bluetongue/virologia , Bovinos , Genoma Viral , Imunização , Sorogrupo , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos
16.
Viruses ; 13(6)2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34071483

RESUMO

Bluetongue virus (BTV) is a segmented RNA virus transmitted by Culicoides midges. Climatic factors, animal movement, vector species, and viral mutation and reassortment may all play a role in the occurrence of BTV outbreaks among susceptible ruminants. We used two enzootic strains of BTV (BTV-2 and BTV-10) to explore the potential for Culicoides sonorensis, a key North American vector, to be infected with these viruses, and identify the impact of temperature variations on virogenesis during infection. While BTV-10 replicated readily in C. sonorensis following an infectious blood meal, BTV-2 was less likely to result in productive infection at biologically relevant exposure levels. Moreover, when C. sonorensis were co-exposed to both viruses, we did not detect reassortment between the two viruses, despite previous in vitro findings indicating that BTV-2 and BTV-10 are able to reassort successfully. These results highlight that numerous factors, including vector species and exposure dose, may impact the in vivo replication of varying BTV strains, and underscore the complexities of BTV ecology in North America.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/virologia , Dípteros/virologia , Temperatura , Animais , Técnicas de Cultura de Células , Linhagem Celular , Suscetibilidade a Doenças , Genótipo , Insetos Vetores/virologia , Vírus Reordenados , Ensaio de Placa Viral , Replicação Viral
17.
Virus Genes ; 57(4): 369-379, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34120252

RESUMO

The smallest polycistronic dsRNA segment-10 (S10) of bluetongue virus (BTV) encodes NS3/3A and putative NS5. The S10 sequence data of 46 Indian BTV field isolates obtained between 1985 and 2011 were determined and compared with the cognate sequences of global BTV strains. The largest ORF on S10 encodes NS3 (229 aa) and an amino-terminal truncated form of the protein (NS3A) and a putative NS5 (50-59 aa) due to alternate translation initiation site. The overall mean distance of the global NS3 was 0.1106 and 0.0269 at nt and deduced aa sequence, respectively. The global BTV strains formed four major clusters. The major cluster of Indian BTV strains was closely related to the viruses reported from Australia and China. A minor sub-cluster of Indian BTV strains were closely related to the USA strains and a few of the Indian strains were similar to the South African reference and vaccine strains. The global trait association of phylogenetic structure indicates the evolution of the global BTV S10 was not homogenous but rather represents a moderate level of geographical divergence. There was no evidence of an association between the virus and the host species, suggesting a random spread of the viruses. Conflicting selection pressure on the alternate coding sequences of the S10 was evident where NS3/3A might have evolved through strong purifying (negative) selection and NS5 through a positive selection. The presence of multiple positively selected codons on the putative NS5 may be advantageous for adaptation of the virus though their precise role is unknown.


Assuntos
Vírus Bluetongue/genética , Bluetongue/genética , RNA de Cadeia Dupla/genética , Proteínas não Estruturais Virais/genética , Animais , Austrália/epidemiologia , Bluetongue/patologia , Bluetongue/virologia , Vírus Bluetongue/classificação , China/epidemiologia , Vírus de RNA de Cadeia Dupla/classificação , Vírus de RNA de Cadeia Dupla/genética , Variação Genética/genética , Humanos , Índia/epidemiologia , Filogenia , Análise de Sequência de DNA , Ovinos/virologia
18.
Viruses ; 13(4)2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33918924

RESUMO

Transmission of bluetongue (BT) virus serotype 8 (BTV-8) via artificial insemination of contaminated frozen semen from naturally infected bulls was investigated in two independent experiments. Healthy, BT negative heifers were hormonally synchronized and artificially inseminated at oestrus. In total, six groups of three heifers received semen from four batches derived from three bulls naturally infected with BTV-8. Each experiment included one control heifer that was not inseminated and that remained BT negative throughout. BTV viraemia and seroconversion were determined in 8 out of 18 inseminated heifers, and BTV was isolated from five of these animals. These eight heifers only displayed mild clinical signs of BT, if any at all, but six of them experienced pregnancy loss between weeks four and eight of gestation, and five of them became BT PCR and antibody positive. The other two infected heifers gave birth at term to two healthy and BT negative calves. The BT viral load varied among the semen batches used and this had a significant impact on the infection rate, the time of onset of viraemia post artificial insemination, and the gestational stage at which pregnancy loss occurred. These results, which confirm unusual features of BTV-8 infection, should not be extrapolated to infection with other BTV strains without thorough evaluation. This study also adds weight to the hypothesis that the re-emergence of BTV-8 in France in 2015 may be attributable to the use of contaminated bovine semen.


Assuntos
Vírus Bluetongue/fisiologia , Bluetongue/transmissão , Doenças dos Bovinos/transmissão , Doenças dos Bovinos/virologia , Inseminação Artificial/veterinária , Preservação do Sêmen/veterinária , Sêmen/virologia , Aborto Animal/virologia , Animais , Bluetongue/virologia , Vírus Bluetongue/classificação , Vírus Bluetongue/imunologia , Vírus Bluetongue/isolamento & purificação , Bovinos , Feminino , França , Inseminação Artificial/efeitos adversos , Masculino , Gravidez , Preservação do Sêmen/efeitos adversos , Sorogrupo
19.
Viruses ; 13(5)2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33919269

RESUMO

We identified a putative novel atypical BTV serotype '36' in Swiss goat flocks. In the initial flock clinical signs consisting of multifocal purulent dermatitis, facial oedema and fever were observed. Following BTV detection by RT-qPCR, serotyping identified BTV-25 and also a putative novel BTV serotype in several of the affected goats. We successfully propagated the so-called "BTV-36-CH2019" strain in cell culture, developed a specific RT-qPCR targeting Segment 2, and generated the full genome by high-throughput sequencing. Furthermore, we experimentally infected goats with BTV-36-CH2019. Regularly, EDTA blood, serum and diverse swab samples were collected. Throughout the experiment, neither fever nor clinical disease was observed in any of the inoculated goats. Four goats developed BTV viremia, whereas one inoculated goat and the two contact animals remained negative. No viral RNA was detected in the swab samples collected from nose, mouth, eye, and rectum, and thus the experimental infection of goats using this novel BTV serotype delivered no indications for any clinical symptoms or vector-free virus transmission pathways. The subclinical infection of the four goats is in accordance with the reports for other atypical BTVs. However, the clinical signs of the initial goat flock did most likely not result from infection with the novel BTV-36-CH0219.


Assuntos
Vírus Bluetongue/classificação , Bluetongue/epidemiologia , Bluetongue/virologia , Ruminantes/virologia , Animais , Bluetongue/diagnóstico , Vírus Bluetongue/genética , Feminino , Doenças das Cabras/diagnóstico , Doenças das Cabras/epidemiologia , Doenças das Cabras/virologia , Cabras/virologia , Masculino , Filogenia , RNA Viral , Sorogrupo , Suíça/epidemiologia
20.
Vet Microbiol ; 256: 109046, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33780805

RESUMO

Lumpy Skin Disease (LSD) and Bluetongue (BT) are the main ruminants viral vector-borne diseases. LSD is endemic in Africa and has recently emerged in Europe and central Asia as a major threat to cattle industry. BT caused great economic damage in Europe during the last decade with a continuous spread to other countries. To control these diseases, vaccination is the only economically viable tool. For LSD, only live-attenuated vaccines (LAVs) are commercially available, whilst for BT both LAVs and inactivated vaccines are available with a limited number of serotypes. In this study, we developed an inactivated, oil adjuvanted bivalent vaccine against both diseases based on LSDV Neethling strain and BTV4. The vaccine was tested for safety and immunogenicity on cattle during a one-year period. Post-vaccination monitoring was carried out by VNT and ELISA. The vaccine was completely safe and elicited high neutralizing antibodies starting from the first week following the second injection up to one year. Furthermore, a significant correlation (R = 0.9040) was observed when comparing VNT and competitive ELISA in BTV4 serological response. Following BTV4 challenge, none of vaccinated and unvaccinated cattle were registered clinical signs, however vaccinated cattle showed full protection from viraemia. In summary, this study highlights the effectiveness of this combined vaccine as a promising solution for both LSD and BT control. It also puts an emphasis on the need for the development of other multivalent inactivated vaccines, which could be greatly beneficial for improving vaccination coverage in endemic countries and prophylaxis of vector-borne diseases.


Assuntos
Vírus Bluetongue/imunologia , Bluetongue/prevenção & controle , Doença Nodular Cutânea/prevenção & controle , Vírus da Doença Nodular Cutânea/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos , Animais , Bluetongue/virologia , Bovinos , Ensaio de Imunoadsorção Enzimática/veterinária , Feminino , Doença Nodular Cutânea/virologia , Masculino , Ovinos , Vacinação/veterinária , Vacinas Atenuadas/imunologia , Vacinas Combinadas/imunologia , Vacinas de Produtos Inativados/imunologia , Viremia/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA