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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 18(5): e1009942, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35512014

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Aftosa , Fiebre Aftosa , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales/metabolismo , Proteínas de la Cápside/metabolismo , Bovinos , Células Dendríticas Foliculares/metabolismo , Virus de la Fiebre Aftosa/genética , Centro Germinal , Ratones , Receptores de Complemento/metabolismo
3.
PLoS Pathog ; 16(1): e1008235, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31905219

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/genética , Enfermedades de los Bovinos/virología , Epistasis Genética , Virus de la Fiebre Aftosa/genética , Fiebre Aftosa/virología , Animales , Búfalos , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Bovinos , Evolución Molecular , Virus de la Fiebre Aftosa/metabolismo , Genoma Viral , Filogenia , ARN Viral/genética , Recombinación Genética
4.
Vet Res ; 53(1): 63, 2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35927724

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Aftosa , Fiebre Aftosa , Animales , Anticuerpos Antivirales , Búfalos , Comercio , Fiebre/veterinaria , Virus de la Fiebre Aftosa/fisiología , Inmunidad , Internacionalidad
5.
Emerg Infect Dis ; 26(7): 1521-1525, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32568048

RESUMEN

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.


Asunto(s)
Animales Salvajes , Infecciones por Bunyaviridae , Animales , Animales Domésticos , Orthobunyavirus , Sudáfrica/epidemiología
6.
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
7.
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
8.
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
9.
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
10.
J Virol ; 87(10): 5447-60, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23468490

RESUMEN

We have previously shown that delivery of the porcine type I interferon gene (poIFN-α/ß) with a replication-defective human adenovirus vector (adenovirus 5 [Ad5]) can sterilely protect swine challenged with foot-and-mouth disease virus (FMDV) 1 day later. However, the need of relatively high doses of Ad5 limits the applicability of such a control strategy in the livestock industry. Venezuelan equine encephalitis virus (VEE) empty replicon particles (VRPs) can induce rapid protection of mice against either homologous or, in some cases, heterologous virus challenge. As an alternative approach to induce rapid protection against FMDV, we have examined the ability of VRPs containing either the gene for green fluorescent protein (VRP-GFP) or poIFN-α (VRP-poIFN-α) to block FMDV replication in vitro and in vivo. Pretreatment of swine or bovine cell lines with either VRP significantly inhibited subsequent infection with FMDV as early as 6 h after treatment and for at least 120 h posttreatment. Furthermore, mice pretreated with either 10(7) or 10(8) infectious units of VRP-GFP and challenged with a lethal dose of FMDV 24 h later were protected from death. Protection was induced as early as 6 h after treatment and lasted for at least 48 h and correlated with induction of an antiviral response and production of IFN-α. By 6 h after treatment several genes were upregulated, and the number of genes and the level of induction increased at 24 h. Finally, we demonstrated that the chemokine IP-10, which is induced by IFN-α and VRP-GFP, is directly involved in protection against FMDV.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/genética , Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/prevención & control , Terapia Genética/métodos , Vectores Genéticos , Interferón-alfa/genética , Interferón-alfa/inmunología , Animales , Modelos Animales de Enfermedad , Fiebre Aftosa/inmunología , Ratones , Ratones Endogámicos C57BL , Análisis de Supervivencia
11.
J Virol ; 86(3): 1316-27, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22114339

RESUMEN

Foot-and-mouth disease virus (FMDV) leader proteinase (L(pro)) cleaves itself from the viral polyprotein and cleaves the translation initiation factor eIF4G. As a result, host cell translation is inhibited, affecting the host innate immune response. We have demonstrated that L(pro) is also associated with degradation of nuclear factor κB (NF-κB), a process that requires L(pro) nuclear localization. Additionally, we reported that disruption of a conserved protein domain within the L(pro) coding sequence, SAP mutation, prevented L(pro) nuclear retention and degradation of NF-κB, resulting in in vitro attenuation. Here we report that inoculation of swine with this SAP-mutant virus does not cause clinical signs of disease, viremia, or virus shedding even when inoculated at doses 100-fold higher than those required to cause disease with wild-type (WT) virus. Remarkably, SAP-mutant virus-inoculated animals developed a strong neutralizing antibody response and were completely protected against challenge with WT FMDV as early as 2 days postinoculation and for at least 21 days postinoculation. Early protection correlated with a distinct pattern in the serum levels of proinflammatory cytokines in comparison to the levels detected in animals inoculated with WT FMDV that developed disease. In addition, animals inoculated with the FMDV SAP mutant displayed a memory T cell response that resembled infection with WT virus. Our results suggest that L(pro) plays a pivotal role in modulating several pathways of the immune response. Furthermore, manipulation of the L(pro) coding region may serve as a viable strategy to derive live attenuated strains with potential for development as effective vaccines against foot-and-mouth disease.


Asunto(s)
Virus de la Fiebre Aftosa/genética , Fiebre Aftosa/prevención & control , Mutación , Enfermedades de los Porcinos/prevención & control , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Antivirales/biosíntesis , Secuencia de Bases , Línea Celular , Cricetinae , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Fiebre Aftosa/inmunología , Virus de la Fiebre Aftosa/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Enfermedades de los Porcinos/inmunología , Linfocitos T/inmunología , Vacunas Virales/inmunología
12.
J Virol ; 86(8): 4477-87, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301155

RESUMEN

Interferons (IFNs) are the first line of defense against viral infections. Although type I and II IFNs have proven effective to inhibit foot-and-mouth disease virus (FMDV) replication in swine, a similar approach had only limited efficacy in cattle. Recently, a new family of IFNs, type III IFN or IFN-λ, has been identified in human, mouse, chicken, and swine. We have identified bovine IFN-λ3 (boIFN-λ3), also known as interleukin 28B (IL-28B), and demonstrated that expression of this molecule using a recombinant replication-defective human adenovirus type 5 (Ad5) vector, Ad5-boIFN-λ3, exhibited antiviral activity against FMDV in bovine cell culture. Furthermore, inoculation of cattle with Ad5-boIFN-λ3 induced systemic antiviral activity and upregulation of IFN-stimulated gene expression in the upper respiratory airways and skin. In the present study, we demonstrated that disease could be delayed for at least 6 days when cattle were inoculated with Ad5-boIFN-λ3 and challenged 24 h later by intradermolingual inoculation with FMDV. Furthermore, the delay in the appearance of disease was significantly prolonged when treated cattle were challenged by aerosolization of FMDV, using a method that resembles the natural route of infection. No clinical signs of FMD, viremia, or viral shedding in nasal swabs was found in the Ad5-boIFN-λ3-treated animals for at least 9 days postchallenge. Our results indicate that boIFN-λ3 plays a critical role in the innate immune response of cattle against FMDV. To this end, this work represents the most successful biotherapeutic strategy so far tested to control FMDV in cattle.


Asunto(s)
Antivirales , Enfermedades de los Bovinos/terapia , Fiebre Aftosa/terapia , Interferón gamma/genética , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Bovinos , Enfermedades de los Bovinos/inmunología , Línea Celular , Cricetinae , Fiebre Aftosa/genética , Fiebre Aftosa/inmunología , Virus de la Fiebre Aftosa/inmunología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Leucocitos Mononucleares/metabolismo , Resultado del Tratamiento
13.
Plants (Basel) ; 10(2)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671188

RESUMEN

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.

14.
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
15.
Science ; 374(6563): 104-109, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34591637

RESUMEN

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.


Asunto(s)
Búfalos/virología , Enfermedades Endémicas/veterinaria , Virus de la Fiebre Aftosa/patogenicidad , Fiebre Aftosa/transmisión , Fiebre Aftosa/virología , Animales , Virus de la Fiebre Aftosa/aislamiento & purificación , Población , Zoonosis/virología
16.
Vet Microbiol ; 137(1-2): 66-73, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19200664

RESUMEN

A truncated ORF2 capsid HEV antigen derived from a genotype 3 strain was developed in insect cells and insect larvae, and compared with the Sar55 antigen and a commercial ELISA. The antigen expressed in insect cells showed a better correlation with Sar55 (kappa value (k)=0.84) than the insect larvae antigen (k=0.69), and a better reproducibility as indicated by the intra and interplate variation coefficients. Commercial ELISA designed for human diagnosis but adapted to animal use using specific secondary antibodies demonstrated to have a very low sensitivity. The insect cell expressed antigen was used to develop an ELISA to detect anti-HEV-IgG in serum samples of different domestic animal and rodents. Seropositivity in the studied animal populations was 71.4% for pigs, 0.60% for goats, 1.92% for sheep, and 11.11% for cats. None of the 1170 cattle samples or 166 rodent samples analyzed was positive.


Asunto(s)
Animales Domésticos , Anticuerpos Antivirales/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Virus de la Hepatitis E/inmunología , Virus de la Hepatitis E/aislamiento & purificación , Roedores , Animales , Antígenos Virales , Línea Celular , Genotipo , Virus de la Hepatitis E/genética , Inmunoglobulina G/sangre , Insectos/virología , Larva/virología , Sensibilidad y Especificidad , España/epidemiología
17.
J Virol Methods ; 154(1-2): 167-74, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18773923

RESUMEN

The main aim of the present study was to describe new methods for the identification of antibodies against the PCV2 capsid (Cap) and replicase (Rep) proteins in pig sera. Specifically, two new indirect enzyme-linked immunosorbent assays (ELISA) were developed based on recombinant PCV2 Cap (rCap) and Rep/Rep' (rRep) proteins expressed in baculovirus and produced in Trichoplusia ni insect larvae. Both assays were validated by testing serum samples in a longitudinal study of 107 animals with different clinico-pathological features of PCV2 infection: pigs with postweaning multisystemic wasting syndrome (PMWS), wasted pigs without a diagnosis of PMWS and healthy animals. Longitudinal antibody profiles indicated that healthy animals had significantly higher anti-Cap and anti-Rep antibody levels than the rest of the animal groups at 11 weeks of age. Moreover, PMWS affected pigs could be distinguished from the rest of the pig groups by their lower anti-Rep antibody levels at 11 weeks of age and at necropsy. The results demonstrate the potential of these two ELISAs for large-scale serological studies. This study represents the first longitudinal study of the induction of anti-Cap and anti-Rep antibodies in farms affected by PMWS, from 1 week of age until the occurrence of disease.


Asunto(s)
Anticuerpos Antivirales/sangre , Antígenos Virales , Proteínas de la Cápside , Infecciones por Circoviridae/veterinaria , Circovirus/aislamiento & purificación , ADN Polimerasa Dirigida por ADN , Animales , Antígenos Virales/genética , Baculoviridae/genética , Proteínas de la Cápside/genética , Infecciones por Circoviridae/diagnóstico , Circovirus/inmunología , ADN Polimerasa Dirigida por ADN/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Larva/virología , Lepidópteros/virología , Sus scrofa
18.
Viruses ; 10(5)2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29693634

RESUMEN

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.


Asunto(s)
Virus de la Fiebre Aftosa/genética , Fiebre Aftosa/virología , Genoma Viral , Cuasiespecies , Recombinación Genética , Animales , Búfalos , Proteínas de la Cápside/genética , Bovinos , Enfermedades de los Bovinos/virología , Línea Celular , Cricetinae , Evolución Molecular , Polimorfismo de Nucleótido Simple/genética , ARN Viral/genética
19.
Sci Rep ; 8(1): 13654, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30209254

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/inmunología , Quimera/inmunología , Virus de la Fiebre Aftosa/inmunología , Fiebre Aftosa/prevención & control , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Cápside/inmunología , Línea Celular , Quimera/genética , Cricetinae , Fiebre Aftosa/inmunología , Virus de la Fiebre Aftosa/genética , Cabras , Porcinos
20.
J Virol Methods ; 146(1-2): 86-95, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17640741

RESUMEN

Porcine circovirus type 2 (PCV2) is detected in high amounts within the characteristic microscopic lesions of postweaning multisystemic wasting syndrome (PMWS) affected pigs. In spite of recent advances on disease pathogenesis, the precise cell types that support viral replication are still a major issue of scientific discussion. In this study, a new methodology to detect cell types that support PCV2 replication was designed. For this purpose, two in situ hybridisation (ISH) methods were developed and applied on tissues of PMWS naturally affected pigs using two probes designed from the ORF1 sequence of the virus. While the complementary probe (CP) detected ssDNA, ORF1 mRNA and replicative form (RF) of PCV2, the RF probe (RFP) exclusively hybridised with the RF of the virus, thus, only labelling cells where PCV2 replication is taking place. Both probes demonstrated to be specific and equally sensitive by an in vitro Southern blot hybridisation assay. ISH labelling with the CP was extensive in lymphoid tissues and of variable amount in other non-lymphoid tissues. With this probe, mainly macrophage-like cells were labelled but also other cell types such as hepatocytes and other epithelial cells. Tissues in which RFP labelling was found more frequently were lung, inguinal and mesenteric lymph nodes, tonsil and liver. Labelling with the RFP was always nuclear, and found in the same cell types as with the CP, although in a relatively low proportion of them; labelling of macrophage-like cells was infrequent. Therefore, the results indicate that at least a certain proportion of macrophages may support PCV2 replication, but main cells where PCV2 replicates are of epithelial/endothelial origin. In summary, the present study permitted the study of cell types that support PCV2 replication by the use of ISH on formalin-fixed, paraffin-embedded tissues of PMWS affected pigs.


Asunto(s)
Circovirus/fisiología , Hibridación in Situ , Macrófagos/virología , Enfermedades de los Porcinos/virología , Replicación Viral , Síndrome Debilitante/veterinaria , Animales , Infecciones por Circoviridae/veterinaria , Infecciones por Circoviridae/virología , Adhesión en Parafina , Porcinos/virología , Fijación del Tejido , Síndrome Debilitante/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA