Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vet Res ; 53(1): 63, 2022 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-35927724

RESUMO

Foot-and-mouth disease (FMD) is one of the most important livestock diseases restricting international trade. While African buffalo (Syncerus caffer) act as the main wildlife reservoir, viral and immune response dynamics during FMD virus acute infection have not been described before in this species. We used experimental needle inoculation and contact infections with three Southern African Territories serotypes to assess clinical, virological and immunological dynamics for thirty days post infection. Clinical FMD in the needle inoculated buffalo was mild and characterised by pyrexia. Despite the absence of generalised vesicles, all contact animals were readily infected with their respective serotypes within the first two to nine days after being mixed with needle challenged buffalo. Irrespective of the route of infection or serotype, there were positive associations between the viral loads in blood and the induction of host innate pro-inflammatory cytokines and acute phase proteins. Viral loads in blood and tonsil swabs were tightly correlated during the acute phase of the infection, however, viraemia significantly declined after a peak at four days post-infection (dpi), which correlated with the presence of detectable neutralising antibodies. In contrast, infectious virus was isolated in the tonsil swabs until the last sampling point (30 dpi) in most animals. The pattern of virus detection in serum and tonsil swabs was similar for all three serotypes in the direct challenged and contact challenged animals. We have demonstrated for the first time that African buffalo are indeed systemically affected by FMD virus and clinical FMD in buffalo is characterized by a transient pyrexia. Despite the lack of FMD lesions, infection of African buffalo was characterised by high viral loads in blood and oropharynx, rapid and strong host innate and adaptive immune responses and high transmissibility.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Anticorpos Antivirais , Búfalos , Comércio , Febre/veterinária , Vírus da Febre Aftosa/fisiologia , Imunidade , Internacionalidade
2.
PLoS Pathog ; 18(5): e1009942, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35512014

RESUMO

Previous studies have shown after the resolution of acute infection and viraemia, foot-and-mouth disease virus (FMDV) capsid proteins and/or genome are localised in the light zone of germinal centres of lymphoid tissue in cattle and African buffalo. The pattern of staining for FMDV proteins was consistent with the virus binding to follicular dendritic cells (FDCs). We have now demonstrated a similar pattern of FMDV protein staining in mouse spleens after acute infection and showed FMDV proteins are colocalised with FDCs. Blocking antigen binding to complement receptor type 2 and 1 (CR2/CR1) prior to infection with FMDV significantly reduced the detection of viral proteins on FDCs and FMDV genomic RNA in spleen samples. Blocking the receptors prior to infection also significantly reduced neutralising antibody titres, through significant reduction in their avidity to the FMDV capsid. Therefore, the binding of FMDV to FDCs and sustained induction of neutralising antibody responses are dependent on FMDV binding to CR2/CR1 in mice.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/metabolismo , Proteínas do Capsídeo/metabolismo , Bovinos , Células Dendríticas Foliculares/metabolismo , Vírus da Febre Aftosa/genética , Centro Germinativo , Camundongos , Receptores de Complemento/metabolismo
3.
Science ; 374(6563): 104-109, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34591637

RESUMO

Extremely contagious pathogens are a global biosecurity threat because of their high burden of morbidity and mortality, as well as their capacity for fast-moving epidemics that are difficult to quell. Understanding the mechanisms enabling persistence of highly transmissible pathogens in host populations is thus a central problem in disease ecology. Through a combination of experimental and theoretical approaches, we investigated how highly contagious foot-and-mouth disease viruses persist in the African buffalo, which serves as their wildlife reservoir. We found that viral persistence through transmission among acutely infected hosts alone is unlikely. However, the inclusion of occasional transmission from persistently infected carriers reliably rescues the most infectious viral strain from fade-out. Additional mechanisms such as antigenic shift, loss of immunity, or spillover among host populations may be required for persistence of less transmissible strains.


Assuntos
Búfalos/virologia , Doenças Endêmicas/veterinária , Vírus da Febre Aftosa/patogenicidade , Febre Aftosa/transmissão , Febre Aftosa/virologia , Animais , Vírus da Febre Aftosa/isolamento & purificação , População , Zoonoses/virologia
4.
Vaccine ; 39(35): 5015-5024, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34303562

RESUMO

Foot-and-mouth disease (FMD) is a global burden on the livestock industry. The causative agent, FMD virus (FMDV), is highly infectious and exists in seven distinct serotypes. Vaccination remains the most effective control strategy in endemic regions and current FMD vaccines are made from inactivated preparations of whole virus. The inherent instability of FMDV and the emergence of new strains presents challenges to efficacious vaccine development. Currently, vaccines available in East Africa are comprised of relatively historic strains with unreported stabilities. As an initial step to produce an improved multivalent FMD vaccine we have identified naturally stable East African FMDV strains for each of the A, O, SAT1 and SAT2 serotypes and investigated their potential for protecting ruminants against strains that have recently circulated in East Africa. Interestingly, high diversity in stability between and within serotypes was observed, and in comparison to non-African A serotype viruses reported to date, the East African strains tested in this study are less stable. Candidate vaccine strains were adapted to propagation in BHK-21 cells with minimal capsid changes and used to generate vaccinate sera that effectively neutralised a panel of FMDV strains selected to improve FMD vaccines used in East Africa. This work highlights the importance of combining tools to predict and assess FMDV vaccine stability, with cell culture adaptation and serological tests in the development of FMD vaccines.


Assuntos
Vírus da Febre Aftosa , Febre Aftosa , Vacinas Virais , Animais , Proteínas do Capsídeo/genética , Febre Aftosa/prevenção & controle , Sorogrupo
5.
Plants (Basel) ; 10(2)2021 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671188

RESUMO

Current understanding of the effects of extreme temperature on alpine evergreens is very limited for ecosystems under Mediterranean climate (characterised by a drought period in summer), despite being exceptionally biodiverse systems and highly vulnerable under a global change scenario. We thus assessed (i) seasonal change and (ii) effect of ontogeny (young vs. mature leaves) on thermal sensitivity of Erysimum scoparium, a keystone evergreen of Teide mountain (Canary Islands). Mature leaves were comparatively much more vulnerable to moderately high leaf-temperature (≥+40 and <+50 °C) than other alpine species. Lowest LT50 occurred in autumn (-9.0 ± 1.6 °C as estimated with Rfd, and -12.9 ± 1.5 °C with Fv/Fm). Remarkably, young leaves showed stronger freezing tolerance than mature leaves in spring (LT50 -10.3 ± 2.1 °C vs. -5.6 ± 0.9 °C in mature leaves, as estimated with Rfd). Our data support the use of Rfd as a sensitive parameter to diagnose temperature-related damage in the leaves of mountain plants. On a global change scenario, E. scoparium appears as a well-prepared species for late-frost events, however rather vulnerable to moderately high temperatures.

7.
Emerg Infect Dis ; 26(7): 1521-1525, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32568048

RESUMO

We screened nonequine animals with unexplained neurologic signs or death in South Africa during 2010-2018 for Shuni virus (SHUV). SHUV was detected in 3.3% of wildlife, 1.1% of domestic, and 2.0% of avian species. Seropositivity was also demonstrated in wildlife. These results suggest a range of possible SHUV hosts in Africa.


Assuntos
Animais Selvagens , Infecções por Bunyaviridae , Animais , Animais Domésticos , Orthobunyavirus , África do Sul/epidemiologia
8.
PLoS Pathog ; 16(1): e1008235, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31905219

RESUMO

Although recombination is known to occur in foot-and-mouth disease virus (FMDV), it is considered only a minor determinant of virus sequence diversity. Analysis at phylogenetic scales shows inter-serotypic recombination events are rare, whereby recombination occurs almost exclusively in non-structural proteins. In this study we have estimated recombination rates within a natural host in an experimental setting. African buffaloes were inoculated with a SAT-1 FMDV strain containing two major viral sub-populations differing in their capsid sequence. This population structure enabled the detection of extensive within-host recombination in the genomic region coding for structural proteins and allowed recombination rates between the two sub-populations to be estimated. Quite surprisingly, the effective recombination rate in VP1 during the acute infection phase turns out to be about 0.1 per base per year, i.e. comparable to the mutation/substitution rate. Using a high-resolution map of effective within-host recombination in the capsid-coding region, we identified a linkage disequilibrium pattern in VP1 that is consistent with a mosaic structure with two main genetic blocks. Positive epistatic interactions between co-evolved variants appear to be present both within and between blocks. These interactions are due to intra-host selection both at the RNA and protein level. Overall our findings show that during FMDV co-infections by closely related strains, capsid-coding genes recombine within the host at a much higher rate than expected, despite the presence of strong constraints dictated by the capsid structure. Although these intra-host results are not immediately translatable to a phylogenetic setting, recombination and epistasis must play a major and so far underappreciated role in the molecular evolution of the virus at all scales.


Assuntos
Proteínas do Capsídeo/genética , Doenças dos Bovinos/virologia , Epistasia Genética , Vírus da Febre Aftosa/genética , Febre Aftosa/virologia , Animais , Búfalos , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Bovinos , Evolução Molecular , Vírus da Febre Aftosa/metabolismo , Genoma Viral , Filogenia , RNA Viral/genética , Recombinação Genética
9.
J Virol ; 93(15)2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31092573

RESUMO

African buffaloes (Syncerus caffer) are the principal "carrier" hosts of foot-and-mouth disease virus (FMDV). Currently, the epithelia and lymphoid germinal centers of the oropharynx have been identified as sites for FMDV persistence. We carried out studies in FMDV SAT1 persistently infected buffaloes to characterize the diversity of viruses in oropharyngeal epithelia, germinal centers, probang samples (oropharyngeal scrapings), and tonsil swabs to determine if sufficient virus variation is generated during persistence for immune escape. Most sequencing reads of the VP1 coding region of the SAT1 virus inoculum clustered around 2 subpopulations differing by 22 single-nucleotide variants of intermediate frequency. Similarly, most sequences from oropharynx tissue clustered into two subpopulations, albeit with different proportions, depending on the day postinfection (dpi). There was a significant difference between the populations of viruses in the inoculum and in lymphoid tissue taken at 35 dpi. Thereafter, until 400 dpi, no significant variation was detected in the viral populations in samples from individual animals, germinal centers, and epithelial tissues. Deep sequencing of virus from probang or tonsil swab samples harvested prior to postmortem showed less within-sample variability of VP1 than that of tissue sample sequences analyzed at the same time. Importantly, there was no significant difference in the ability of sera collected between 14 and 400 dpi to neutralize the inoculum or viruses isolated at later time points in the study from the same animal. Therefore, based on this study, there is no evidence of escape from antibody neutralization contributing to FMDV persistent infection in African buffalo.IMPORTANCE Foot-and-mouth disease virus (FMDV) is a highly contagious virus of cloven-hoofed animals and is recognized as the most important constraint to international trade in animals and animal products. African buffaloes (Syncerus caffer) are efficient carriers of FMDV, and it has been proposed that new virus variants are produced in buffalo during the prolonged carriage after acute infection, which may spread to cause disease in livestock populations. Here, we show that despite an accumulation of low-frequency sequence variants over time, there is no evidence of significant antigenic variation leading to immune escape. Therefore, carrier buffalo are unlikely to be a major source of new virus variants.


Assuntos
Búfalos , Portador Sadio/veterinária , Evolução Molecular , Vírus da Febre Aftosa/crescimento & desenvolvimento , Febre Aftosa/imunologia , Febre Aftosa/virologia , Evasão da Resposta Imune , Animais , Proteínas do Capsídeo/genética , Portador Sadio/imunologia , Portador Sadio/virologia , Epitélio/virologia , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/imunologia , Instabilidade Genômica , Centro Germinativo/virologia , Mutação , Orofaringe/virologia , Análise de Sequência de DNA
10.
Sci Rep ; 8(1): 13654, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30209254

RESUMO

Foot-and-mouth disease virus (FMDV) is highly contagious and infects cloven-hoofed domestic livestock leading to foot-and-mouth disease (FMD). FMD outbreaks have severe economic impact due to production losses and associated control measures. FMDV is found as seven distinct serotypes, but there are numerous subtypes within each serotype, and effective vaccines must match the subtypes circulating in the field. In addition, the O and Southern African Territories (SAT) serotypes, are relatively more thermolabile and their viral capsids readily dissociate into non-immunogenic pentameric subunits, which can compromise the effectiveness of FMD vaccines. Here we report the construction of a chimeric clone between the SAT2 and O serotypes, designed to have SAT2 antigenicity. Characterisation of the chimeric virus showed growth kinetics equal to that of the wild type SAT2 virus with better thermostability, attributable to changes in the VP4 structural protein. Sequence and structural analyses confirmed that no changes from SAT2 were present elsewhere in the capsid as a consequence of the VP4 changes. Following exposure to an elevated temperature the thermostable SAT2-O1K chimera induced higher neutralizing-antibody titres in comparison to wild type SAT2 virus.


Assuntos
Proteínas do Capsídeo/imunologia , Quimera/imunologia , Vírus da Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Capsídeo/imunologia , Linhagem Celular , Quimera/genética , Cricetinae , Febre Aftosa/imunologia , Vírus da Febre Aftosa/genética , Cabras , Suínos
11.
Viruses ; 10(5)2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29693634

RESUMO

Recombination is one of the determinants of genetic diversity in the foot-and-mouth disease virus (FMDV). FMDV sequences have a mosaic structure caused by extensive intra- and inter-serotype recombination, with the exception of the capsid-encoding region. While these genome-wide patterns of broad-scale recombination are well studied, not much is known about the patterns of recombination that may exist within infected hosts. In addition, detection of recombination among viruses evolving at the within-host level is challenging due to the similarity of the sequences and the limitations in differentiating recombination from point mutations. Here, we present the first analysis of recombination events between closely related FMDV sequences occurring within buffalo hosts. The detection of these events was made possible by the occurrence of co-infection of two viral swarms with about 1% nucleotide divergence. We found more than 15 recombination events, unequally distributed across eight samples from different animals. The distribution of these events along the FMDV genome was neither uniform nor related to the phylogenetic distribution of recombination breakpoints, suggesting a mismatch between within-host evolutionary pressures and long-term selection for infectivity and transmissibility.


Assuntos
Vírus da Febre Aftosa/genética , Febre Aftosa/virologia , Genoma Viral , Quase-Espécies , Recombinação Genética , Animais , Búfalos , Proteínas do Capsídeo/genética , Bovinos , Doenças dos Bovinos/virologia , Linhagem Celular , Cricetinae , Evolução Molecular , Polimorfismo de Nucleotídeo Único/genética , RNA Viral/genética
12.
J Virol Methods ; 247: 38-44, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28532601

RESUMO

Bioluminescence is a powerful tool in the study of viral infection both in vivo and in vitro. Foot-and-mouth disease virus (FMDV) has a small RNA genome with a limited tolerance to foreign RNA entities. There has been no success in making a reporter FMDV expressing a luciferase in infected cell culture supernatants. We report here for the first time a replication-competent FMDV encoding Nanoluciferase, named as Nano-FMDV. Nano-FMDV is genetically stable during serial passages in cells and exhibits growth kinetics and plaque morphology similar to its parental virus. There are applications for the use of Nano-FMDV such as real-time monitoring of FMDV replication in vitro and in vivo.


Assuntos
Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/fisiologia , Genes Reporter , Luciferases/biossíntese , Coloração e Rotulagem/métodos , Replicação Viral , Animais , Linhagem Celular , Instabilidade Genômica , Medições Luminescentes , Ensaio de Placa Viral , Cultura de Vírus , Viroses
13.
Front Immunol ; 8: 1944, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29375568

RESUMO

Detecting exposure to new or emerging pathogens is a critical challenge to protecting human, domestic animal, and wildlife health. Yet, current techniques to detect infections typically target known pathogens of humans or economically important animals. In the face of the current surge in infectious disease emergence, non-specific disease surveillance tools are urgently needed. Tracking common host immune responses indicative of recent infection may have potential as a non-specific diagnostic approach for disease surveillance. The challenge to immunologists is to identify the most promising markers, which ideally should be highly conserved across pathogens and host species, become upregulated rapidly and consistently in response to pathogen invasion, and remain elevated beyond clearance of infection. This study combined an infection experiment and a longitudinal observational study to evaluate the utility of non-specific markers of inflammation [NSMI; two acute phase proteins (haptoglobin and serum amyloid A), two pro-inflammatory cytokines (IFNγ and TNF-α)] as indicators of pathogen exposure in a wild mammalian species, African buffalo (Syncerus caffer). Specifically, in the experimental study, we asked (1) How quickly do buffalo mount NSMI responses upon challenge with an endemic pathogen, foot-and-mouth disease virus; (2) for how long do NSMI remain elevated after viral clearance and; (3) how pronounced is the difference between peak NSMI concentration and baseline NSMI concentration? In the longitudinal study, we asked (4) Are elevated NSMI associated with recent exposure to a suite of bacterial and viral respiratory pathogens in a wild population? Among the four NSMI that we tested, haptoglobin showed the strongest potential as a surveillance marker in African buffalo: concentrations quickly and consistently reached high levels in response to experimental infection, remaining elevated for almost a month. Moreover, elevated haptoglobin was indicative of recent exposure to two respiratory pathogens assessed in the longitudinal study. We hope this work motivates studies investigating suites of NSMI as indicators for pathogen exposure in a broader range of both pathogen and host species, potentially transforming how we track disease burden in natural populations.

14.
Virology ; 499: 340-349, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27743960

RESUMO

Foot-and-mouth-disease (FMD) remains the most infectious livestock disease worldwide. Although commercially available inactivated or adenovirus-vectored-vaccines (Ad5-FMD) are effective, they require 5-7 days to induce protection. Therefore, new control strategies that stimulate rapid immune responses are needed. Expression of bovine interferon λ3 using the Ad5-vector platform (Ad5-boIFNλ3) is able to delay disease in cattle, but clinical signs appear at 9 days after challenge. We hypothesized that combination of Ad5-boIFNλ3 and Ad5-FMD could induce immediate and lasting protection against FMD. Cattle were vaccinated with an Ad5-FMD, Ad5-boIFNλ3, or the combination of both, followed by challenge at three days post-immunization. All animals treated with Ad5-FMD combined with Ad5-boIFNλ3 were fully protected against FMD, despite the absence of systemic neutralizing antibodies or antiviral activity at the time of challenge. Induction of a strong cell-mediated immune response suggested that Ad5-boIFNλ3 is able to act as an adjuvant of Ad5-FMD vaccine in cattle.


Assuntos
Vírus da Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Imunidade Celular , Vacinas Virais/imunologia , Adenoviridae/genética , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Bovinos , Febre Aftosa/imunologia , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Vacinação , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
15.
J Immunol ; 196(10): 4214-26, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27053760

RESUMO

The pestivirus noncytopathic bovine viral diarrhea virus (BVDV) can suppress IFN production in the majority of cell types in vitro. However, IFN is detectable in serum during acute infection in vivo for ∼5-7 d, which correlates with a period of leucopoenia and immunosuppression. In this study, we demonstrate that a highly enriched population of bovine plasmacytoid dendritic cells (DCs) produced IFN in response to BVDV in vitro. We further show that the majority of the IFN produced in response to infection both in vitro and in vivo is type III IFN and acid labile. Further, we show IL-28B (IFN-λ3) mRNA is induced in this cell population in vitro. Supernatant from plasmacytoid DCs harvested postinfection with BVDV or recombinant bovine IFN-α or human IL-28B significantly reduced CD4(+) T cell proliferation induced by tubercle bacillus Ag 85-stimulated monocyte-derived DCs. Furthermore, these IFNs induced IFN-stimulated gene expression predominantly in monocyte-derived DCs. IFN-treated immature DCs derived from murine bone marrow also had a reduced capacity to stimulate T cell proliferative responses to tubercle bacillus Ag 85. Immature DCs derived from either source had a reduced capacity for Ag uptake following IFN treatment that is dose dependent. Immunosuppression is a feature of a number of pestivirus infections; our studies suggest type III IFN production plays a key role in the pathogenesis of this family of viruses. Overall, in a natural host, we have demonstrated a link between the induction of type I and III IFN after acute viral infection and transient immunosuppression.


Assuntos
Doença das Mucosas por Vírus da Diarreia Viral Bovina/imunologia , Células Dendríticas/imunologia , Vírus da Diarreia Viral Bovina/imunologia , Imunidade Celular , Interferon-alfa/imunologia , Interleucinas/imunologia , Aciltransferases/imunologia , Animais , Antígenos de Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Bovinos , Linhagem Celular , Proliferação de Células , Humanos , Tolerância Imunológica , Interferon-alfa/sangue , Interferons , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , Proteínas Recombinantes/imunologia , Sus scrofa
16.
J Virol ; 90(10): 5132-5140, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26962214

RESUMO

UNLABELLED: Foot-and-mouth disease (FMD) virus (FMDV) circulates as multiple serotypes and strains in many regions of endemicity. In particular, the three Southern African Territories (SAT) serotypes are maintained effectively in their wildlife reservoir, the African buffalo, and individuals may harbor multiple SAT serotypes for extended periods in the pharyngeal region. However, the exact site and mechanism for persistence remain unclear. FMD in buffaloes offers a unique opportunity to study FMDV persistence, as transmission from carrier ruminants has convincingly been demonstrated for only this species. Following coinfection of naive African buffaloes with isolates of three SAT serotypes from field buffaloes, palatine tonsil swabs were the sample of choice for recovering infectious FMDV up to 400 days postinfection (dpi). Postmortem examination identified infectious virus for up to 185 dpi and viral genomes for up to 400 dpi in lymphoid tissues of the head and neck, focused mainly in germinal centers. Interestingly, viral persistence in vivo was not homogenous, and the SAT-1 isolate persisted longer than the SAT-2 and SAT-3 isolates. Coinfection and passage of these SAT isolates in goat and buffalo cell lines demonstrated a direct correlation between persistence and cell-killing capacity. These data suggest that FMDV persistence occurs in the germinal centers of lymphoid tissue but that the duration of persistence is related to virus replication and cell-killing capacity. IMPORTANCE: Foot-and-mouth disease virus (FMDV) causes a highly contagious acute vesicular disease in domestic livestock and wildlife species. African buffaloes (Syncerus caffer) are the primary carrier hosts of FMDV in African savannah ecosystems, where the disease is endemic. We have shown that the virus persists for up to 400 days in buffaloes and that there is competition between viruses during mixed infections. There was similar competition in cell culture: viruses that killed cells quickly persisted more efficiently in passaged cell cultures. These results may provide a mechanism for the dominance of particular viruses in an ecosystem.


Assuntos
Búfalos/virologia , Portador Sadio/veterinária , Vírus da Febre Aftosa/fisiologia , Vírus da Febre Aftosa/patogenicidade , Febre Aftosa/virologia , África/epidemiologia , Animais , Animais Selvagens/virologia , Anticorpos Antivirais/sangue , Portador Sadio/virologia , Coinfecção/epidemiologia , Coinfecção/veterinária , Coinfecção/virologia , Ensaio de Imunoadsorção Enzimática , Febre Aftosa/epidemiologia , Febre Aftosa/imunologia , Febre Aftosa/transmissão , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/imunologia , Genoma Viral , Tonsila Palatina/virologia , Sorogrupo , Virulência , Replicação Viral
17.
Nat Struct Mol Biol ; 22(10): 788-94, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26389739

RESUMO

Virus capsids are primed for disassembly, yet capsid integrity is key to generating a protective immune response. Foot-and-mouth disease virus (FMDV) capsids comprise identical pentameric protein subunits held together by tenuous noncovalent interactions and are often unstable. Chemically inactivated or recombinant empty capsids, which could form the basis of future vaccines, are even less stable than live virus. Here we devised a computational method to assess the relative stability of protein-protein interfaces and used it to design improved candidate vaccines for two poorly stable, but globally important, serotypes of FMDV: O and SAT2. We used a restrained molecular dynamics strategy to rank mutations predicted to strengthen the pentamer interfaces and applied the results to produce stabilized capsids. Structural analyses and stability assays confirmed the predictions, and vaccinated animals generated improved neutralizing-antibody responses to stabilized particles compared to parental viruses and wild-type capsids.


Assuntos
Proteínas do Capsídeo/química , Vírus da Febre Aftosa/química , Febre Aftosa/prevenção & controle , Modelos Moleculares , Vacinas Virais/química , Animais , Anticorpos Neutralizantes/sangue , Sequência de Bases , Proteínas do Capsídeo/metabolismo , Biologia Computacional/métodos , Microscopia Crioeletrônica , Cristalografia por Raios X , Desenho de Fármacos , Ensaio de Imunoadsorção Enzimática , Febre Aftosa/imunologia , Vírus da Febre Aftosa/imunologia , Microscopia Eletrônica , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Vacinas Virais/imunologia
18.
Virology ; 468-470: 283-292, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25216089

RESUMO

Foot-and-mouth disease virus (FMDV) causes a highly contagious disease of cloven-hoofed animals. We have previously demonstrated that a replication-defective human adenovirus 5 vector carrying the FMDV capsid coding region of serotype A24 Cruzeiro (Ad5-CI-A24-2B) protects swine and cattle against FMDV challenge by 7 days post-vaccination. However, since relatively large amounts of Ad5-CI-A24-2B are required to induce protection this strategy could be costly for livestock production. Poly ICLC is a synthetic double stranded RNA that activates multiple innate and adaptive immune pathways. In this study, we have tested for the first time, the adjuvant effect of poly ICLC in combination with Ad5-CI-A24-2B in swine. We found that the combination resulted in a reduction of the vaccine protective dose by 80-fold. Interestingly, the lowest dose of Ad5-CI-A24-2B plus 1mg of poly ICLC protected animals against challenge even in the absence of detectable FMDV-specific neutralizing antibodies at the time of challenge.


Assuntos
Adenovírus Humanos , Carboximetilcelulose Sódica/análogos & derivados , Febre Aftosa/prevenção & controle , Vetores Genéticos , Poli I-C/farmacologia , Polilisina/análogos & derivados , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/sangue , Carboximetilcelulose Sódica/farmacologia , Células HEK293 , Humanos , Polilisina/farmacologia , Suínos , Doenças dos Suínos/prevenção & controle , Replicação Viral
19.
J Gen Virol ; 95(Pt 11): 2329-2345, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25000962

RESUMO

Laboratory animal models have provided valuable insight into foot-and-mouth disease virus (FMDV) pathogenesis in epidemiologically important target species. While not perfect, these models have delivered an accelerated time frame to characterize the immune responses in natural hosts and a platform to evaluate therapeutics and vaccine candidates at a reduced cost. Further expansion of these models in mice has allowed access to genetic mutations not available for target species, providing a powerful and versatile experimental system to interrogate the immune response to FMDV and to target more expensive studies in natural hosts. The purpose of this review is to describe commonly used FMDV infection models in laboratory animals and to cite examples of when these models have failed or successfully provided insight relevant for target species, with an emphasis on natural and vaccine-induced immunity.


Assuntos
Vírus da Febre Aftosa/imunologia , Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Vacinas Virais/uso terapêutico , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Bovinos , Febre Aftosa/virologia , Imunidade Celular , Imunidade Humoral , Imunidade Inata , Camundongos , Modelos Animais , Sus scrofa , Linfócitos T/imunologia
20.
J Interferon Cytokine Res ; 34(10): 810-21, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24786495

RESUMO

In recent years, we have developed novel strategies to control foot-and-mouth disease (FMD), including the use of biotherapeutics such as interferons (IFN) delivered by a replication-defective human adenovirus type 5 (Ad5). Swine can be sterilely protected after vaccination with an Ad5 that encodes porcine type I IFN (poIFN-α), and cattle can be similarly protected or develop significantly reduced disease when treated with an Ad5 delivering bovine type III IFN (boIFN-λ3). Here, we have evaluated the efficacy of porcine IFN-λ3 (poIFN-λ3) against FMD virus in vivo. Swine inoculated with different doses of Ad5-poIFN-λ3 were protected against disease in a dose-dependent manner. Despite the absence of systemic antiviral activity, 7 out of 10 Ad5-poIFN-λ3 inoculated animals did not develop disease or viremia, and the other 3 inoculated animals displayed delayed and milder disease by 7 days postchallenge as compared with control animals inoculated with an Ad5 control vector. While analysis of gene expression showed significant induction of IFN and IFN-stimulated genes in Ad5-poIFN-λ3-treated cultured porcine epithelial kidney cells, there was limited gene induction in peripheral blood monocytes isolated from treated swine. These results suggest that treatment with Ad5-poIFN-λ3 is an effective biotherapeutic strategy against FMD in swine.


Assuntos
Terapia Biológica/métodos , Febre Aftosa/prevenção & controle , Interleucinas/imunologia , Doenças dos Suínos/prevenção & controle , Adenoviridae , Animais , Bovinos , Células Cultivadas , Perfilação da Expressão Gênica , Vetores Genéticos , Humanos , Interleucinas/genética , Monócitos/imunologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...