Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Immunology ; 164(2): 266-278, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34003490

RESUMEN

Foot-and-mouth disease (FMD) is a highly contagious, economically devastating disease of cloven-hooved animals. The development of long-lasting effective FMD vaccines would greatly benefit the global FMD control programme. Deep analysis of adaptive immunity in cattle vaccinated against FMD is technically challenging due to the lack of species-specific tools. In this study, we aimed to identify CD4+ T-cell epitopes in the FMD virus (FMDV) capsid and to phenotype the CD4+ T cells that recognize them using bovine major histocompatibility complex (BoLA) class II tetramer. A BoLA class II tetramer based on the DRA/DRB3*020:02 allele and FMDV antigen-stimulated PBMCs from bovine vaccinates were used to successfully identify four epitopes in the FMDV capsid, three of which have not been previously reported; two epitopes were identified in the structural protein VP1, one in VP3 and one in VP4. Specificity of the three novel epitopes was confirmed by proliferation assay. All epitope-expanded T-cell populations produced IFN-γ in vitro, indicating a long-lasting Th1 cell phenotype after FMD vaccination. VP3-specific CD4+ T cells exhibited the highest frequency amongst the identified epitopes, comprising >0·004% of the CD4+ T-cell population. CD45RO+ CCR7+ defined central memory CD4+ T-cell subpopulations were present in higher frequency in FMDV-specific CD4+ T-cell populations from FMD-vaccinated cattle ex vivo. This indicates an important role in maintaining cell adaptive immunity after FMD vaccination. Notably, FMDV epitope-loaded tetramers detected the presence of FMDV-specific CD4+ T cells in bovine PBMC more than four years after vaccination. This work contributes to our understanding of vaccine efficacy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/virología , Proteínas de la Cápside/inmunología , Bovinos , Células Cultivadas , Epítopos de Linfocito T/inmunología , Fiebre Aftosa/virología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Serogrupo , Vacunación/métodos
2.
J Virol ; 95(9)2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33328306

RESUMEN

Classical swine fever virus (CSFV) is the causative agent of classical swine fever, a notifiable disease of economic importance that causes severe leukopenia, fever and haemorrhagic disease in domesticated pigs and wild boar across the globe. CSFV has been shown to antagonise the induction of type I IFN, partly through a function of its N-terminal protease (Npro) which binds IRF3 and targets it for proteasomal degradation. Additionally, Npro has been shown to antagonise apoptosis triggered by the dsRNA-homolog poly(I:C), however the exact mechanism by which this is achieved has not been fully elucidated. In this study we confirm the ability of Npro to inhibit dsRNA-mediated apoptosis and show that Npro is also able to antagonise Sendai virus-mediated apoptosis in PK-15 cells. Gene edited PK-15 cell lines were used to show the dsRNA-sensing pathogen recognition receptors (PRRs) TLR3 and RIG-I specifically respond to poly(I:C) and SeV respectively, subsequently triggering apoptosis through pathways that converge on IRF3 and culminate in the cleavage of caspase-3. Importantly, this IRF3-mediated apoptosis was found to be dependent on transcription-independent functions of IRF3 and also on Bax, a pro-apoptotic Bcl-2 family protein, through a direct interaction between the two proteins. Deletion of IRF3, stable expression of Npro and infection with wild-type CSFV were found to antagonise the mitochondrial localisation of Bax, a key hallmark of the intrinsic, mitochondrial pathway of apoptosis. Together, these findings show that Npro's putative interaction with IRF3 is involved not only in its antagonism of type I IFN, but also dsRNA-mediated mitochondrial apoptosis.Importance Responsible for severe haemorrhagic disease in domestic pigs and wild boar, classical swine fever is recognised by the World Organisation for Animal Health (OIE) and European Union as a notifiable disease of economic importance. Persistent infection, immunotolerance and early dissemination of the virus at local sites of infection have been linked to the antagonism of type I IFN induction by Npro This protein may further contribute to these phenomena by antagonising the induction of dsRNA-mediated apoptosis. Ultimately, apoptosis is an important innate mechanism by which cells counter viruses at local sites of infection, thus preventing wider spread and dissemination within the host, potentially also contributing to the onset of persistence. Elucidation of the mechanism by which Npro antagonises the apoptotic response will help inform the development of rationally-designed live-attenuated vaccines and antivirals for control of outbreaks in typically CSFV-free countries.

3.
J Virol ; 93(15)2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31092573

RESUMEN

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.


Asunto(s)
Búfalos , Portador Sano/veterinaria , Evolución Molecular , Virus de la Fiebre Aftosa/crecimiento & desarrollo , Fiebre Aftosa/inmunología , Fiebre Aftosa/virología , Evasión Inmune , Animales , Proteínas de la Cápside/genética , Portador Sano/inmunología , Portador Sano/virología , Epitelio/virología , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/inmunología , Inestabilidad Genómica , Centro Germinal/virología , Mutación , Orofaringe/virología , Análisis de Secuencia de ADN
4.
J Virol ; 91(10)2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28298597

RESUMEN

Foot-and-mouth disease virus (FMDV), particularly strains of the O and SAT serotypes, is notoriously unstable. Consequently, vaccines derived from heat-labile SAT viruses have been linked to the induction of immunity with a poor duration and hence require more frequent vaccinations to ensure protection. In silico calculations predicted residue substitutions that would increase interactions at the interpentamer interface, supporting increased stability. We assessed the stability of the 18 recombinant mutant viruses in regard to their growth kinetics, antigenicity, plaque morphology, genetic stability, and temperature, ionic, and pH stability by using Thermofluor and inactivation assays in order to evaluate potential SAT2 vaccine candidates with improved stability. The most stable mutant for temperature and pH stability was the S2093Y single mutant, while other promising mutants were the E3198A, L2094V, and S2093H single mutants and the F2062Y-H2087M-H3143V triple mutant. Although the S2093Y mutant had the greatest stability, it exhibited smaller plaques, a reduced growth rate, a change in monoclonal antibody footprint, and poor genetic stability properties compared to those of the wild-type virus. However, these factors affecting production can be overcome. The addition of 1 M NaCl was found to further increase the stability of the SAT2 panel of viruses. The S2093Y and S2093H mutants were selected for future use in stabilizing SAT2 vaccines.IMPORTANCE Foot-and-mouth disease virus (FMDV) causes a highly contagious acute vesicular disease in cloven-hoofed livestock and wildlife. The control of the disease by vaccination is essential, especially at livestock-wildlife interfaces. The instability of some serotypes, such as SAT2, affects the quality of vaccines and therefore the duration of immunity. We have shown that we can improve the stability of SAT2 viruses by mutating residues at the capsid interface through predictive modeling. This is an important finding for the potential use of such mutants in improving the stability of SAT2 vaccines in countries where FMD is endemic, which rely heavily on the maintenance of the cold chain, with potential improvement to the duration of immune responses.


Asunto(s)
Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/fisiología , Vacunas Virales/genética , Sustitución de Aminoácidos , Animales , Virus de la Fiebre Aftosa/inmunología , Inestabilidad Genómica , Concentración de Iones de Hidrógeno , Inmunogenicidad Vacunal , Iones , Cinética , Mutación , Serogrupo , Cloruro de Sodio/farmacología , Temperatura , Potencia de la Vacuna , Vacunas Virales/química
5.
J Immunol ; 196(10): 4214-26, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053760

RESUMEN

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.


Asunto(s)
Diarrea Mucosa Bovina Viral/inmunología , Células Dendríticas/inmunología , Virus de la Diarrea Viral Bovina/inmunología , Inmunidad Celular , Interferón-alfa/inmunología , Interleucinas/inmunología , Aciltransferasas/inmunología , Animales , Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Bovinos , Línea Celular , Proliferación Celular , Humanos , Tolerancia Inmunológica , Interferón-alfa/sangre , Interferones , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Monocitos/inmunología , Proteínas Recombinantes/inmunología , Sus scrofa
6.
J Virol ; 90(10): 5132-5140, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26962214

RESUMEN

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.


Asunto(s)
Búfalos/virología , Portador Sano/veterinaria , Virus de la Fiebre Aftosa/fisiología , Virus de la Fiebre Aftosa/patogenicidad , Fiebre Aftosa/virología , África/epidemiología , Animales , Animales Salvajes/virología , Anticuerpos Antivirales/sangre , Portador Sano/virología , Coinfección/epidemiología , Coinfección/veterinaria , Coinfección/virología , Ensayo de Inmunoadsorción Enzimática , Fiebre Aftosa/epidemiología , Fiebre Aftosa/inmunología , Fiebre Aftosa/transmisión , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/inmunología , Genoma Viral , Tonsila Palatina/virología , Serogrupo , Virulencia , Replicación Viral
7.
J Gen Virol ; 97(7): 1557-1565, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27002540

RESUMEN

Foot-and-mouth disease (FMD) has a major economic impact throughout the world and is a considerable threat to food security. Current FMD virus (FMDV) vaccines are made from chemically inactivated virus and need to contain intact viral capsids to maximize efficacy. FMDV exists as seven serotypes, each made up by a number of constantly evolving subtypes. A lack of immunological cross-reactivity between serotypes and between some strains within a serotype greatly complicates efforts to control FMD by vaccination. Thus, vaccines for one serotype do not afford protection against the others, and multiple-serotype-specific vaccines are required for effective control. The FMDV serotypes exhibit variation in their thermostability, and the capsids of inactivated preparations of the O, C and SAT serotypes are particularly susceptible to dissociation at elevated temperature. Methods to quantify capsid stability are currently limited, lack sensitivity and cannot accurately reflect differences in thermostability. Thus, new, more sensitive approaches to quantify capsid stability would be of great value for the production of more stable vaccines and to assess the effect of production conditions on vaccine preparations. Here we have investigated the application of a novel methodology (termed PaSTRy) that utilizes an RNA-binding fluorescent dye and a quantitative (q)PCR machine to monitor viral genome release and hence dissociation of the FMDV capsid during a slow incremental increase in temperature. PaSTRy was used to characterize capsid stability of all FMDV serotypes. Furthermore, we have used this approach to identify stabilizing factors for the most labile FMDV serotypes.


Asunto(s)
Proteínas de la Cápside/inmunología , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/prevención & control , Vacunas de Productos Inactivados/inmunología , Vacunas Virales/inmunología , Animales , Cápside/inmunología , Línea Celular , Cricetinae , Fiebre Aftosa/inmunología , Fiebre Aftosa/virología , Genoma Viral/genética , Cabras/virología , Calor , Reacción en Cadena de la Polimerasa , Serogrupo , Vacunación
8.
J Gen Virol ; 95(Pt 11): 2329-2345, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25000962

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/inmunología , Fiebre Aftosa/prevención & control , Vacunas Virales/uso terapéutico , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Bovinos , Fiebre Aftosa/virología , Inmunidad Celular , Inmunidad Humoral , Inmunidad Innata , Ratones , Modelos Animales , Sus scrofa , Linfocitos T/inmunología
9.
J Virol ; 87(15): 8735-44, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740982

RESUMEN

Field isolates of foot-and-mouth disease virus (FMDV) have a restricted cell tropism which is limited by the need for certain RGD-dependent integrin receptors. In contrast, cell culture-adapted viruses use heparan sulfate (HS) or other unidentified molecules as receptors to initiate infection. Here, we report several novel findings resulting from cell culture adaptation of FMDV. In cell culture, a virus with the capsid of the A/Turkey/2/2006 field isolate gained the ability to infect CHO and HS-deficient CHO cells as a result of a single glutamine (Q)-to-lysine (K) substitution at VP1-110 (VP1-(Q)110(K)). Using site-directed mutagenesis, the introduction of lysine at this same site also resulted in an acquired ability to infect CHO cells by type O and Asia-1 FMDV. However, this ability appeared to require a second positively charged residue at VP1-109. CHO cells express two RGD-binding integrins (α5ß1 and αvß5) that, although not used by FMDV, have the potential to be used as receptors; however, viruses with the VP1-(Q)110(K) substitution did not use these integrins. In contrast, the VP1-(Q)110(K) substitution appeared to result in enhanced interactions with αvß6, which allowed a virus with KGE in place of the normal RGD integrin-binding motif to use αvß6 as a receptor. Thus, our results confirmed the existence of nonintegrin, non-HS receptors for FMDV on CHO cells and revealed a novel, non-RGD-dependent use of αvß6 as a receptor. The introduction of lysine at VP1-110 may allow for cell culture adaptation of FMDV by design, which may prove useful for vaccine manufacture when cell culture adaptation proves intractable.


Asunto(s)
Adaptación Biológica , Virus de la Fiebre Aftosa/fisiología , Receptores Virales/metabolismo , Pase Seriado , Tropismo Viral , Animales , Células CHO , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Cricetinae , Análisis Mutacional de ADN , Virus de la Fiebre Aftosa/genética , Mutagénesis Sitio-Dirigida
10.
J Gen Virol ; 94(Pt 7): 1517-1527, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23559477

RESUMEN

Foot-and-mouth disease virus (FMDV) is one of the most extensively studied animal pathogens because it remains a major threat to livestock economies worldwide. However, the dynamics of FMDV infection are still poorly understood. The application of reverse genetics provides the opportunity to generate molecular tools to further dissect the FMDV life cycle. Here, we have used reverse genetics to determine the capsid packaging limitations for a selected insertion site in the FMDV genome. We show that exogenous RNA up to a defined length can be stably introduced into the FMDV genome, whereas larger insertions are excised by recombination events. This led us to construct a recombinant FMDV expressing the fluorescent marker protein, termed iLOV. Characterization of infectious iLOV-FMDV showed the virus has a plaque morphology and rate of growth similar to the parental virus. In addition, we show that cells infected with iLOV-FMDV are easily differentiated by flow cytometry using the inherent fluorescence of iLOV and that cells infected with iLOV-FMDV can be monitored in real-time with fluorescence microscopy. iLOV-FMDV therefore offers a unique tool to characterize FMDV infection in vitro, and its applications for in vivo studies are discussed.


Asunto(s)
Epitelio/virología , Virus de la Fiebre Aftosa/patogenicidad , Proteínas Fluorescentes Verdes/metabolismo , Recombinación Genética , Animales , Células Cultivadas , Efecto Citopatogénico Viral , Citometría de Flujo , Fiebre Aftosa/virología , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/fisiología , Cabras , Proteínas Fluorescentes Verdes/genética
11.
Viruses ; 15(12)2023 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-38140685

RESUMEN

Porcine reproductive and respiratory syndrome viruses (PRRSV-1 and -2) are the causative agents of one of the most important infectious diseases affecting the global pig industry. Previous studies, largely focused on PRRSV-2, have shown that non-structural protein-1α (NSP1α) and NSP1ß modulate host cell responses; however, the underlying molecular mechanisms remain to be fully elucidated. Therefore, we aimed to identify novel PRRSV-1 NSP1-host protein interactions to improve our knowledge of NSP1-mediated immunomodulation. NSP1α and NSP1ß from a representative western European PRRSV-1 subtype 1 field strain (215-06) were used to screen a cDNA library generated from porcine alveolar macrophages (PAMs), the primary target cell of PRRSV, using the yeast-2-hybrid system. This identified 60 putative binding partners for NSP1α and 115 putative binding partners for NSP1ß. Of those taken forward for further investigation, 3 interactions with NSP1α and 27 with NSP1ß were confirmed. These proteins are involved in the immune response, ubiquitination, nuclear transport, or protein expression. Increasing the stringency of the system revealed NSP1α interacts more strongly with PIAS1 than PIAS2, whereas NSP1ß interacts more weakly with TAB3 and CPSF4. Our study has increased our knowledge of the PRRSV-1 NSP1α and NSP1ß interactomes, further investigation of which could provide detailed insight into PRRSV immunomodulation and aid vaccine development.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Animales , Porcinos , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Línea Celular , Macrófagos Alveolares/metabolismo , Ubiquitinación , Técnicas del Sistema de Dos Híbridos , Proteínas no Estructurales Virales/metabolismo
12.
Vaccine ; 41(44): 6572-6578, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37679279

RESUMEN

Foot-and-mouth disease (FMD) is a highly contagious viral disease of livestock which is prevalent across Africa, the Middle East, Asia, and South America where it has a severe economic impact on the agriculture industry. Vaccination with inactivated viral vaccines is used as the main control measure in these endemic regions of the world, however the presence of multiple serotypes, subtypes, and the continual emergence of new, antigenically divergent strains limits its effectiveness. East Africa (EA) has been identified as a region that would particularly benefit from updated FMD vaccines, since those currently in use contain older strains which do not provide good protection against contemporary strains. Four serotypes are currently circulating in EA, necessitating the development of a quadrivalent vaccine containing representative strains of each serotype. A key consideration in the selection of vaccine strains is the stability of the virus particle, since the capsids readily dissociate on exposure to elevated temperatures, but only intact capsids induce protective immunity to FMD. Therefore, with a view to producing a more stable, updated quadrivalent vaccine for EA, we recently screened a panel of foot-and-mouth disease virus (FMDV) isolates from the region to select strains with naturally higher thermostabilities and confirmed their immunogenicity in cattle. Herein we describe the formulation and serological assessment of wild-type and recombinant quadrivalent vaccine candidates comprising these stable strains, and demonstrate that both vaccines generate high neutralising antibody titres against the homologous strains and also to heterologous strains from EA. Importantly, the vaccine passed the criteria set by the AgResults vaccine challenge project and offers good cross-protection against a panel of regional FMDV strains.

13.
J Gen Virol ; 93(Pt 11): 2371-2381, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22815275

RESUMEN

Foot-and-mouth disease (FMD) is a highly contagious and economically devastating disease of cloven-hoofed animals with an almost-worldwide distribution. Conventional FMD vaccines consisting of chemically inactivated viruses have aided in the eradication of FMD from Europe and remain the main tool for control in endemic countries. Although significant steps have been made to improve the quality of vaccines, such as improved methods of antigen concentration and purification, manufacturing processes are technically demanding and expensive. Consequently, there is large variation in the quality of vaccines distributed in FMD-endemic countries compared with those manufactured for emergency use in FMD-free countries. Here, we have used reverse genetics to introduce haemagglutinin (HA) and FLAG tags into the foot-and-mouth disease virus (FMDV) capsid. HA- and FLAG-tagged FMDVs were infectious, with a plaque morphology similar to the non-tagged parental infectious copy virus and the field virus. The tagged viruses utilized integrin-mediated cell entry and retained the tag epitopes over serial passages. In addition, infectious HA- and FLAG-tagged FMDVs were readily purified from small-scale cultures using commercial antibodies. Tagged FMDV offers a feasible alternative to the current methods of vaccine concentration and purification, a potential to develop FMD vaccine conjugates and a unique tool for FMDV research.


Asunto(s)
Proteínas de la Cápside/metabolismo , Epítopos , Virus de la Fiebre Aftosa/clasificación , Fiebre Aftosa/prevención & control , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Fiebre Aftosa/virología , Modelos Moleculares , Conformación Proteica , ARN Viral , Coloración y Etiquetado , Vacunas de ADN , Vacunas de Productos Inactivados
14.
J Virol ; 85(9): 4297-308, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307187

RESUMEN

Type I interferons (alpha/beta interferons [IFN-α/ß]) are the main innate cytokines that are able to induce a cellular antiviral state, thereby limiting viral replication and disease pathology. Plasmacytoid dendritic cells (pDCs) play a crucial role in the control of viral infections, especially in response to viruses that have evolved mechanisms to block the type I IFN signal transduction pathway. Using density gradient separation and cell sorting, we have highly enriched a population of bovine cells capable of producing high levels of biologically active type I IFN. These cells represented less than 0.1% of the total lymphocyte population in blood, pseudoafferent lymph, and lymph nodes. Phenotypic analysis identified these cells as bovine pDCs (CD3(-) CD14(-) CD21(-) CD11c(-) NK(-) TCRδ(-) CD4(+) MHC II(+) CD45RB(+) CD172a(+) CD32(+)). High levels of type I IFN were generated by these cells in vitro in response to Toll-like receptor 9 (TLR-9) agonist CpG and foot-and-mouth disease virus (FMDV) immune complexes. In contrast, immune complexes formed with UV-inactivated FMDV or FMDV empty capsids failed to elicit a type I IFN response. Depletion of CD4 cells in vivo resulted in levels of type I IFN in serum early during FMDV infection that were significantly lower than those for control animals. In conclusion, pDCs interacting with immune-complexed virus are the major source of type I interferon production during acute FMDV infection in cattle.


Asunto(s)
Enfermedades de los Bovinos/inmunología , Células Dendríticas/inmunología , Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/inmunología , Interferón Tipo I/metabolismo , Animales , Antígenos CD/análisis , Bovinos , Centrifugación por Gradiente de Densidad , Células Dendríticas/química , Citometría de Flujo
15.
Viruses ; 14(6)2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35746633

RESUMEN

Foot-and-mouth disease is an economically devastating disease of livestock caused by foot-and-mouth disease virus (FMDV). Vaccination is the most effective control measure in place to limit the spread of the disease; however, the success of vaccination campaigns is hampered by the antigenic diversity of FMDV and the rapid rate at which new strains emerge that escape pre-existing immunity. FMDV has seven distinct serotypes, and within each serotype are multiple strains that often induce little cross-protective immunity. The diversity of FMDV is a consequence of the high error rate of the RNA-dependent RNA polymerase, accompanied by extensive recombination between genomes during co-infection. Since multiple serotypes and strains co-circulate in regions where FMDV is endemic, co-infection is common, providing the conditions for recombination, and also for other events such as trans-encapsidation in which the genome of one virus is packaged into the capsid of the co-infecting virus. Here, we demonstrate that the co-infection of cells with two FMDVs of different serotypes results in trans-encapsidation of both viral genomes. Crucially, this facilitates the infection of new cells in the presence of neutralizing antibodies that recognize the capsid that is encoded by the packaged genome.


Asunto(s)
Coinfección , Virus de la Fiebre Aftosa , Fiebre Aftosa , Animales , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales , Proteínas de la Cápside/genética , Virus de la Fiebre Aftosa/genética , Serogrupo
16.
Viruses ; 14(3)2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35337028

RESUMEN

Foot-and-mouth disease (FMD) is endemic in large parts of sub-Saharan Africa, Asia and South America, where outbreaks in cloven-hooved livestock threaten food security and have severe economic impacts. Vaccination in endemic regions remains the most effective control strategy. Current FMD vaccines are produced from chemically inactivated foot-and-mouth disease virus (FMDV) grown in suspension cultures of baby hamster kidney 21 cells (BHK-21). Strain diversity means vaccines produced from one subtype may not fully protect against circulating disparate subtypes, necessitating the development of new vaccine strains that "antigenically match". However, some viruses have proven difficult to adapt to cell culture, slowing the manufacturing process, reducing vaccine yield and limiting the availability of effective vaccines, as well as potentiating the selection of undesired antigenic changes. To circumvent the need to cell culture adapt FMDV, we have used a systematic approach to develop recombinant suspension BHK-21 that stably express the key FMDV receptor integrin αvß6. We show that αvß6 expression is retained at consistently high levels as a mixed cell population and as a clonal cell line. Following exposure to field strains of FMDV, these recombinant BHK-21 facilitated higher virus yields compared to both parental and control BHK-21, whilst demonstrating comparable growth kinetics. The presented data supports the application of these recombinant αvß6-expressing BHK-21 in future FMD vaccine production.


Asunto(s)
Virus de la Fiebre Aftosa , Fiebre Aftosa , Vacunas Virales , Animales , Línea Celular , Virus de la Fiebre Aftosa/genética , Vacunación , Vacunas Virales/genética
17.
Viruses ; 13(12)2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34960702

RESUMEN

Foot-and-mouth disease, caused by foot-and-mouth disease virus (FMDV), is an economically devastating disease affecting several important livestock species. FMDV is antigenically diverse and exists as seven serotypes comprised of many strains which are poorly cross-neutralised by antibodies induced by infection or vaccination. Co-infection and recombination are important drivers of antigenic diversity, especially in regions where several serotypes co-circulate at high prevalence, and therefore experimental systems to study these events in vitro would be beneficial. Here we have utilised recombinant FMDVs containing an HA or a FLAG epitope tag within the VP1 capsid protein to investigate the products of co-infection in vitro. Co-infection with viruses from the same and from different serotypes was demonstrated by immunofluorescence microscopy and flow cytometry using anti-tag antibodies. FLAG-tagged VP1 and HA-tagged VP1 could be co-immunoprecipitated from co-infected cells, suggesting that newly synthesised capsids may contain VP1 proteins from both co-infecting viruses. Furthermore, we provide the first demonstration of trans-encapsidation of an FMDV genome into capsids comprised of proteins encoded by a co-infecting heterologous virus. This system provides a useful tool for investigating co-infection dynamics in vitro, particularly between closely related strains, and has the advantage that it does not depend upon the availability of strain-specific FMDV antibodies.


Asunto(s)
Cápside/metabolismo , Virus de la Fiebre Aftosa/fisiología , Fiebre Aftosa/virología , ARN Viral/metabolismo , Empaquetamiento del Genoma Viral , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Coinfección , Epítopos , Virus de la Fiebre Aftosa/genética , Virus de la Fiebre Aftosa/inmunología , Genoma Viral , Hemaglutininas Virales/genética , Hemaglutininas Virales/inmunología , ARN Viral/genética , Serogrupo
18.
Vaccine ; 39(35): 5015-5024, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34303562

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Aftosa , Fiebre Aftosa , Vacunas Virales , Animales , Proteínas de la Cápside/genética , Fiebre Aftosa/prevención & control , Serogrupo
19.
Viruses ; 13(4)2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807175

RESUMEN

Type I interferons (IFNs) are produced by most cells in response to virus infection and stimulate a program of anti-viral gene expression in neighboring cells to suppress virus replication. Type III IFNs have similar properties, however their effects are limited to epithelial cells at mucosal surfaces due to restricted expression of the type III IFN receptor. Rotavirus (RV) replicates in intestinal epithelial cells that respond predominantly to type III IFNs, and it has been shown that type III rather than type I IFNs are important for controlling RV infections in vivo. The RV NSP1 protein antagonizes the host type I IFN response by targeting IRF-3, IRF-5, IRF-7, or ß-TrCP for proteasome-mediated degradation in a strain-specific manner. Here we provide the first demonstration that NSP1 proteins from several human and animal RV strains antagonize type III as well as type I IFN induction. We also show that NSP1 is a potent inhibitor of IRF-1, a previously undescribed property of NSP1 which is conserved among human and animal RVs. Interestingly, all NSP1 proteins were substantially more effective inhibitors of IRF-1 than either IRF-3 or IRF-7 which has significance for evasion of basal anti-viral immunity and type III IFN induction in the intestinal epithelium.


Asunto(s)
Células Epiteliales/virología , Interferón Tipo I/antagonistas & inhibidores , Interferones/antagonistas & inhibidores , Rotavirus/inmunología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología , Animales , Células Epiteliales/inmunología , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Interferones/inmunología , Intestinos/citología , Rotavirus/química , Rotavirus/aislamiento & purificación
20.
J Gen Virol ; 91(Pt 3): 721-6, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19906943

RESUMEN

Classical swine fever is a notifiable disease of pigs. The causative agent, classical swine fever virus (CSFV), is highly contagious and causes mild to severe haemorrhagic disease depending on the virulence of the strain. The RNA genome of CSFV is translated as a single polyprotein that is processed to yield 12 proteins. Like other pestiviruses, the first protein to be translated is the N-terminal autoprotease termed N(pro). A novel pestiviral protein with no known cellular homologues, N(pro) antagonizes type I interferon (IFN) induction by binding and targeting the transcription factor IFN regulatory factor 3 (IRF-3) for ubiquitin-dependent proteasomal degradation. In this study, CSFV-infected PK-15 cells and stable cell lines were used to show that N(pro) is itself an unstable protein that is targeted for proteasomal degradation in a ubiquitin-dependent manner. In addition, N(pro) is not degraded as a direct consequence of its ability to interact with IRF-3 or to target IRF-3 for proteasomal degradation.


Asunto(s)
Virus de la Fiebre Porcina Clásica/inmunología , Endopeptidasas/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Porcinos , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA