Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(7): e0196422, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37358450

RESUMEN

Porcine epidemic diarrhea virus is a swine pathogen that has been responsible for significant animal and economic losses worldwide in recent years. In this manuscript, we report the generation of a reverse genetics system C(RGS) for the highly virulent US PEDV strain Minnesota (PEDV-MN; GenBank accession number KF468752), which was based on the assembly and cloning of synthetic DNA, using vaccinia virus as a cloning vector. Viral rescue was only possible following the substitution of 2 nucleotides within the 5'UTR and 2 additional nucleotides within the spike gene, based on the sequence of the cell culture-adapted strains. Besides displaying a highly pathogenic phenotype in newborn piglets, in comparison with the parental virus, the rescued recombinant PEDV-MN was used to confirm that the PEDV spike gene has an important role in PEDV virulence and that the impact of an intact PEDV ORF3 on viral pathogenicity is modest. Moreover, a chimeric virus with a TGEV spike gene in the PEDV backbone generated with RGS was able to replicate efficiently in vivo and could be readily transmitted between piglets. Although this chimeric virus did not cause severe disease upon the initial infection of piglets, there was evidence of increasing pathogenicity upon transmission to contact piglets. The RGS described in this study constitutes a powerful tool with which to study PEDV pathogenesis and can be used to generate vaccines against porcine enteric coronaviruses. IMPORTANCE PEDV is a swine pathogen that is responsible for significant animal and economic losses worldwide. Highly pathogenic variants can lead to a mortality rate of up to 100% in newborn piglets. The generation of a reverse genetics system for a highly virulent PEDV strain originating from the United States is an important step in phenotypically characterizing PEDV. The synthetic PEDV mirrored the authentic isolate and displayed a highly pathogenic phenotype in newborn piglets. With this system, it was possible to characterize potential viral virulence factors. Our data revealed that an accessory gene (ORF3) has a limited impact on pathogenicity. However, as it is also now known for many coronaviruses, the PEDV spike gene is one of the main determinants of pathogenicity. Finally, we show that the spike gene of another porcine coronavirus, namely, TGEV, can be accommodated in the PEDV genome background, suggesting that similar viruses can emerge in the field via recombination.


Asunto(s)
Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Animales , Estados Unidos , Porcinos , Virulencia/genética , Virus de la Diarrea Epidémica Porcina/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Genética Inversa , Infecciones por Coronavirus/prevención & control , Nucleótidos , Diarrea
2.
PLoS Pathog ; 18(8): e1010522, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36006954

RESUMEN

African Swine Fever virus (ASFV) is a large double-enveloped DNA virus of the Asfarviridae family that causes a lethal hemorrhagic disease in domestic pigs and wild boars. Since 2007, a highly virulent genotype II strain has emerged and spread in Europe and South-East Asia, where millions of animals succumbed to the disease. Field- and laboratory-attenuated strains of ASFV cause highly variable clinical disease severity and survival, and mechanisms remain unclear. We hypothesized that the immunological and hygienic status of pigs is a determinant of ASF disease course. Here we compared the immunological profile at baseline and in response to ASFV infection in specific pathogen-free (SPF) and farm-raised Large White domestic pigs. At steady state, SPF pigs showed lower white blood cell counts and a lower basal inflammatory and antiviral transcriptomic profile compared to farm pigs, associated with profound differences in gut microbiome composition. After inoculation with a highly virulent ASFV genotype II strain (Armenia 2008), severe clinical signs, viremia and pro-inflammatory cytokines appeared sooner in SPF pigs, indicating a reduced capacity to control early virus replication. In contrast, during infection with an attenuated field isolate (Estonia 2014), SPF pigs presented a milder and shorter clinical disease with full recovery, whereas farm pigs presented severe protracted disease with 50% lethality. Interestingly, farm pigs showed higher production of inflammatory cytokines, whereas SPF pigs produced more anti-inflammatory IL-1ra early after infection and presented a stronger expansion of leukocytes in the recovery phase. Altogether, our data indicate that the hygiene-dependent innate immune status has a double-edge sword impact on immune responses in ASF pathogenesis. While the higher baseline innate immune activity helps the host in reducing initial virus replication, it promotes immunopathological cytokine responses, and delays lymphocyte proliferation after infection with an attenuated strain. Such effects should be considered for live vaccine development and vigilance.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Virus de la Fiebre Porcina Africana/genética , Animales , Citocinas , Higiene , Índice de Severidad de la Enfermedad , Sus scrofa , Porcinos
3.
J Virol ; 96(14): e0043822, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35758667

RESUMEN

In this study, we assessed the potential synergistic effect of the Erns RNase activity and the poly-U insertion in the 3' untranslated region (UTR) of the low-virulence classical swine fever virus (CSFV) isolate Pinar de Rio (PdR) in innate and adaptive immunity regulation and its relationship with classical swine fever (CSF) pathogenesis in pigs. We knocked out the Erns RNase activity of PdR and replaced the long polyuridine sequence of the 3' UTR with 5 uridines found typically at this position, resulting in a double mutant, vPdR-H30K-5U. This mutant induced severe CSF in 5-day-old piglets and 3-week-old pigs, with higher lethality in the newborn (89.5%) than in the older (33.3%) pigs. However, the viremia and viral excretion were surprisingly low, while the virus load was high in the tonsils. Only alpha interferon (IFN-α) and interleukin 12 (IL-12) were highly and consistently elevated in the two groups. Additionally, high IL-8 levels were found in the newborn but not in the older pigs. This points toward a role of these cytokines in the CSF outcome, with age-related differences. The disproportional activation of innate immunity might limit systemic viral spread from the tonsils and increase virus clearance, inducing strong cytokine-mediated symptoms. Infection with vPdR-H30K-5U resulted in poor neutralizing antibody responses compared with results obtained previously with the parent and RNase knockout PdR. This study shows for the first time the synergistic effect of the 3' UTR and the Erns RNase function in regulating innate immunity against CSFV, favoring virus replication in target tissue and thus contributing to disease severity. IMPORTANCE CSF is one of the most relevant viral epizootic diseases of swine, with high economic and sanitary impact. Systematic stamping out of infected herds with and without vaccination has permitted regional virus eradication. However, the causative agent, CSFV, persists in certain areas of the world, leading to disease reemergence. Nowadays, low- and moderate-virulence strains that could induce unapparent CSF forms are prevalent, posing a challenge for disease eradication. Here, we show for the first time the synergistic role of lacking the Erns RNase activity and the 3' UTR polyuridine insertion from a low-virulence CSFV isolate in innate immunity disproportional activation. This might limit systemic viral spread to the tonsils and increase virus clearance, inducing strong cytokine-mediated symptoms, thus contributing to disease severity. These results highlight the role played by the Erns RNase activity and the 3' UTR in CSFV pathogenesis, providing new perspectives for novel diagnostic tools and vaccine strategies.


Asunto(s)
Virus de la Fiebre Porcina Clásica , Peste Porcina Clásica , Síndrome de Liberación de Citoquinas , Regiones no Traducidas 3'/genética , Inmunidad Adaptativa/genética , Animales , Peste Porcina Clásica/inmunología , Peste Porcina Clásica/patología , Peste Porcina Clásica/virología , Virus de la Fiebre Porcina Clásica/enzimología , Virus de la Fiebre Porcina Clásica/genética , Virus de la Fiebre Porcina Clásica/inmunología , Virus de la Fiebre Porcina Clásica/patogenicidad , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Citocinas , Inmunidad Innata/genética , Interferón-alfa/inmunología , Interleucina-12/inmunología , Ribonucleasas/genética , Ribonucleasas/metabolismo , Porcinos , Vacunas Virales , Virulencia/genética
4.
PLoS Pathog ; 17(4): e1009529, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33909707

RESUMEN

The human respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections in infants, possibly due to the properties of the immature neonatal pulmonary immune system. Using the newborn lamb, a classical model of human lung development and a translational model of RSV infection, we aimed to explore the role of cell-mediated immunity in RSV disease during early life. Remarkably, in healthy conditions, the developing T cell compartment of the neonatal lung showed major differences to that seen in the mature adult lung. The most striking observation being a high baseline frequency of bronchoalveolar IL-4-producing CD4+ and CD8+ T cells, which declined progressively over developmental age. RSV infection exacerbated this pro-type 2 environment in the bronchoalveolar space, rather than inducing a type 2 response per se. Moreover, regulatory T cell suppressive functions occurred very early to dampen this pro-type 2 environment, rather than shutting them down afterwards, while γδ T cells dropped and failed to produce IL-17. Importantly, RSV disease severity was related to the magnitude of those unconventional bronchoalveolar T cell responses. These findings provide novel insights in the mechanisms of RSV immunopathogenesis in early life, and constitute a major step for the understanding of RSV disease severity.


Asunto(s)
Pulmón/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones del Sistema Respiratorio/inmunología , Linfocitos T/patología , Animales , Animales Recién Nacidos , Diferenciación Celular/inmunología , Células Cultivadas , Preescolar , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Pulmón/crecimiento & desarrollo , Pulmón/patología , Pulmón/virología , Infecciones por Virus Sincitial Respiratorio/congénito , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones del Sistema Respiratorio/congénito , Infecciones del Sistema Respiratorio/patología , Ovinos/crecimiento & desarrollo , Ovinos/inmunología , Linfocitos T/inmunología , Linfocitos T/fisiología
5.
PLoS Pathog ; 17(7): e1009789, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34320038

RESUMEN

Lung-resident (LR) mesenchymal stem and stromal cells (MSCs) are key elements of the alveolar niche and fundamental regulators of homeostasis and regeneration. We interrogated their function during virus-induced lung injury using the highly prevalent respiratory syncytial virus (RSV) which causes severe outcomes in infants. We applied complementary approaches with primary pediatric LR-MSCs and a state-of-the-art model of human RSV infection in lamb. Remarkably, RSV-infection of pediatric LR-MSCs led to a robust activation, characterized by a strong antiviral and pro-inflammatory phenotype combined with mediators related to T cell function. In line with this, following in vivo infection, RSV invades and activates LR-MSCs, resulting in the expansion of the pulmonary MSC pool. Moreover, the global transcriptional response of LR-MSCs appears to follow RSV disease, switching from an early antiviral signature to repair mechanisms including differentiation, tissue remodeling, and angiogenesis. These findings demonstrate the involvement of LR-MSCs during virus-mediated acute lung injury and may have therapeutic implications.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/virología , Pulmón/inmunología , Células Madre Mesenquimatosas/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Animales , Humanos , Pulmón/citología , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/inmunología , Ovinos
6.
Nanomedicine ; 49: 102655, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36681171

RESUMEN

Herein, we provide the first description of a synthetic delivery method for self-replicating replicon RNAs (RepRNA) derived from classical swine fever virus (CSFV) using a Coatsome-replicon vehicle based on Coatsome® SS technologies. This results in an unprecedented efficacy when compared to well-established polyplexes, with up to ∼65 fold-increase of the synthesis of RepRNA-encoded gene of interest (GOI). We demonstrated the efficacy of such Coatsome-replicon vehicles for RepRNA-mediated induction of CD8 T-cell responses in mice. Moreover, we provide new insights on physical properties of the RepRNA, showing that the removal of all CSFV structural protein genes has a positive effect on the translation of the GOI. Finally, we successfully engineered RepRNA constructs encoding a porcine reproductive and respiratory syndrome virus (PRRSV) antigen, providing an example of antigen expression with potential application to combat viral diseases. The versatility and simplicity of modifying and manufacturing these Coatsome-replicon vehicle formulations represents a major asset to tackle foreseeable emerging pandemics.


Asunto(s)
Enfermedades Transmisibles , ARN , Porcinos , Ratones , Animales , ARN/genética , Antígenos , Enfermedades Transmisibles/genética , Replicón/genética
7.
Emerg Infect Dis ; 27(7): 1811-1820, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34152956

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread globally, and the number of worldwide cases continues to rise. The zoonotic origins of SARS-CoV-2 and its intermediate and potential spillback host reservoirs, besides humans, remain largely unknown. Because of ethical and experimental constraints and more important, to reduce and refine animal experimentation, we used our repository of well-differentiated airway epithelial cell (AEC) cultures from various domesticated and wildlife animal species to assess their susceptibility to SARS-CoV-2. We observed that SARS-CoV-2 replicated efficiently only in monkey and cat AEC culture models. Whole-genome sequencing of progeny viruses revealed no obvious signs of nucleotide transitions required for SARS-CoV-2 to productively infect monkey and cat AEC cultures. Our findings, together with previous reports of human-to-animal spillover events, warrant close surveillance to determine the potential role of cats, monkeys, and closely related species as spillback reservoirs for SARS-CoV-2.


Asunto(s)
Animales Salvajes , COVID-19 , Animales , Células Epiteliales , Humanos , Sistema Respiratorio , SARS-CoV-2
8.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31645448

RESUMEN

Low-virulence classical swine fever virus (CSFV) strains make CSF eradication particularly difficult. Few data are available on the molecular determinants of CSFV virulence. The aim of the present study was to assess a possible role for CSFV virulence of a unique, uninterrupted 36-uridine (poly-U) sequence found in the 3' untranslated region (3' UTR) of the low-virulence CSFV isolate Pinar de Rio (PdR). To this end, a pair of cDNA-derived viruses based on the PdR backbone were generated, one carrying the long poly-U insertion in the 3' UTR (vPdR-36U) and the other harboring the standard 5 uridines at this position (vPdR-5U). Two groups of 20 5-day-old piglets were infected with vPdR-36U and vPdR-5U. Ten contact piglets were added to each group. Disease progression, virus replication, and immune responses were monitored for 5 weeks. The vPdR-5U virus was significantly more virulent than the vPdR-36U virus, with more severe disease, higher mortality, and significantly higher viral loads in serum and body secretions, despite similar replication characteristics in cell culture. The two viruses were transmitted to all contact piglets. Ninety percent of the piglets infected with vPdR-36U seroconverted, while only one vPdR-5U-infected piglet developed antibodies. The vPdR-5U-infected piglets showed only transient alpha interferon (IFN-α) responses in serum after 1 week of infection, while the vPdR-36U-infected piglets showed sustained IFN-α levels during the first 2 weeks. Taken together, these data show that the 3' UTR poly-U insertion acquired by the PdR isolate reduces viral virulence and activates the innate and humoral immune responses without affecting viral transmission.IMPORTANCE Classical swine fever (CSF), a highly contagious viral disease of pigs, is still endemic in some countries of Asia and Central and South America. Considering that the 3' untranslated region (3' UTR) plays an important role in flavivirus replication, the present study showed for the first time that a long polyuridine sequence acquired in the 3' UTR by an endemic CSFV isolate can activate immunity, control viral replication, and modulate disease in piglets. Our findings provide new avenues for the development of novel vaccines against infections with CSF virus and other flaviviruses. Knowledge of molecular virulence determinants is also relevant for future development of rapid and efficient diagnostic tools for the prediction of the virulence of field isolates and for efficient CSF control.


Asunto(s)
Regiones no Traducidas 3'/inmunología , Virus de la Fiebre Porcina Clásica , Peste Porcina Clásica , Mutagénesis Insercional , Poli U , ARN Viral , Animales , Peste Porcina Clásica/genética , Peste Porcina Clásica/inmunología , Peste Porcina Clásica/patología , Virus de la Fiebre Porcina Clásica/genética , Virus de la Fiebre Porcina Clásica/inmunología , Virus de la Fiebre Porcina Clásica/patogenicidad , Humanos , Interferón-alfa/inmunología , Poli U/genética , Poli U/inmunología , ARN Viral/genética , ARN Viral/inmunología , Porcinos
9.
J Gen Virol ; 101(2): 156-167, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31922948

RESUMEN

Several pigeon paramyxovirus-1 (PPMV-1) outbreaks in feral pigeons were described recently in Switzerland. The potential of PPMV-1 to induce the notifiable Newcastle disease in chickens is discussed controversially. Therefore, in order to study epidemiologically relevant parameters such as the kinetics of PPMV-1 replication and shedding as well as seroconversion after infection, chickens were infected experimentally with a Swiss PPMV-1 isolate. This generated also defined sample material for the comparison of diagnostic tests. The infectivity of the Swiss PPMV-1 isolate for chickens was demonstrated successfully by virus shedding after experimental inoculation. Our data suggest that long-lasting shedding for up to 60 days can occur in chickens infected with PPMV-1. The isolate used here was of low pathogenicity for chickens. Different quantitative reverse transcription PCR assays were evaluated with a set of Swiss PPMV-1 isolates, and various samples from experimentally infected chickens were analysed with respect to their suitability for viral RNA detection. At 14 days post-infection, virus genome was detected mainly in spleen, caecal tonsils, heart, cloacal swabs, liver, proventriculus, duodenum and kidney tissue samples. Overall, the level of virus replication was low. Not all assays used routinely in diagnostics were capable of detecting viral genome from the isolates tested. Possible explanations are the genetic divergence of PPMV-1 and the low level of viral RNA in the samples. In contrast, two methods that are not used routinely proved more suitable for virus-genome detection. Importantly, the collection of material from various different organs is recommended, in addition to the kidney and brain analysed routinely. In conclusion, this study shows that there is a need to reconsider the type of samples and the protocols used for the detection of PPMV-1 RNA in chickens.


Asunto(s)
Infecciones por Avulavirus/diagnóstico , Avulavirus , Enfermedad de Newcastle/diagnóstico , Animales , Avulavirus/genética , Avulavirus/crecimiento & desarrollo , Avulavirus/aislamiento & purificación , Avulavirus/patogenicidad , Infecciones por Avulavirus/patología , Pollos , Columbidae/virología , Genoma Viral , Enfermedad de Newcastle/patología , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/genética , Virus de la Enfermedad de Newcastle/crecimiento & desarrollo , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Virus de la Enfermedad de Newcastle/patogenicidad , Enfermedades de las Aves de Corral/virología , Suiza , Virosis/veterinaria , Replicación Viral , Esparcimiento de Virus
10.
J Immunol ; 197(12): 4791-4806, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27837108

RESUMEN

Porcine dendritic cells (DCs) are relatively well characterized, but a clear-cut identification of all DC subsets combined with full transcriptional profiling was lacking, preventing an unbiased insight into the functional specializations of DC subsets. Using a large panel of Abs in multicolor flow cytometry, cell sorting, and RNA sequencing we identified and characterized the porcine equivalent of conventional DCs (cDC) 1 and cDC2 as well as plasmacytoid DCs (pDCs) in the peripheral blood of pigs. We demonstrate that cDC1 are CD135+CD14-CD172alowCADM1+wCD11R1+ cells, cDC2 are CD135+CD14-CD172a+CADM1+CD115+wCD11R1+CD1+ cells and pDCs are CD4+CD135+CD172a+CD123+CD303+ cells. As described in other species, only cDC1 express BATF3 and XCR1, cDC2 express FCER1A and FCGR2B, and only pDCs express TCF4 and NRP1 Nevertheless, despite these cross-species conserved subset-specific transcripts, porcine pDCs differed from the species described so far in many expressed genes and transcriptomic profiling clustered pDCs more distantly from cDCs than monocytes. The response of porcine DC subsets to TLR ligands revealed that pDCs are by far the most important source of TNF-α, IL-12p40, and of course IFN-α, whereas cDCs are most efficient in MHC and costimulatory molecule expression. Nevertheless, upregulation of CD40 and CD86 in cDCs was critically influenced or even dependent on the presence of pDCs, particularly for TLR 7 and 9 ligands. Our data demonstrate that extrapolation of data on DC biology from one species to another has to be done with care, and it shows how functional details have evolved differentially in different species.


Asunto(s)
Células Sanguíneas/fisiología , Células Dendríticas/fisiología , Especificidad de la Especie , Porcinos/inmunología , Transcriptoma , Animales , Antígenos CD/metabolismo , Diferenciación Celular , Separación Celular , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica , Interferón gamma/metabolismo , Subunidad p40 de la Interleucina-12/metabolismo , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Virol ; 90(17): 7740-7, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27334592

RESUMEN

UNLABELLED: Interferon regulatory factor 3 (IRF3) is a transcription factor involved in the activation of type I alpha/beta interferon (IFN-α/ß) in response to viral infection. Upon viral infection, the IRF3 monomer is activated into a phosphorylated dimer, which induces the transcription of interferon genes in the nucleus. Viruses have evolved several ways to target IRF3 in order to subvert the innate immune response. Pestiviruses, such as classical swine fever virus (CSFV), target IRF3 for ubiquitination and subsequent proteasomal degradation. This is mediated by the viral protein N(pro) that interacts with IRF3, but the molecular details for this interaction are largely unknown. We used recombinant N(pro) and IRF3 proteins and show that N(pro) interacts with IRF3 directly without additional proteins and forms a soluble 1:1 complex. The full-length IRF3 but not merely either of the individual domains is required for this interaction. The interaction between N(pro) and IRF3 is not dependent on the activation state of IRF3, since N(pro) binds to a constitutively active form of IRF3 in the presence of its transcriptional coactivator, CREB-binding protein (CBP). The results indicate that the N(pro)-binding site on IRF3 encompasses a region that is unperturbed by the phosphorylation and subsequent activation of IRF3 and thus excludes the dimer interface and CBP-binding site. IMPORTANCE: The pestivirus N-terminal protease, N(pro), is essential for evading the host's immune system by facilitating the degradation of interferon regulatory factor 3 (IRF3). However, the nature of the N(pro) interaction with IRF3, including the IRF3 species (inactive monomer versus activated dimer) that N(pro) targets for degradation, is largely unknown. We show that classical swine fever virus N(pro) and porcine IRF3 directly interact in solution and that full-length IRF3 is required for interaction with N(pro) Additionally, N(pro) interacts with a constitutively active form of IRF3 bound to its transcriptional cofactor, the CREB-binding protein. This is the first study to demonstrate that N(pro) is able to bind both inactive IRF3 monomer and activated IRF3 dimer and thus likely targets both IRF3 species for ubiquitination and proteasomal degradation.


Asunto(s)
Virus de la Fiebre Porcina Clásica/enzimología , Virus de la Fiebre Porcina Clásica/fisiología , Endopeptidasas/metabolismo , Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón/metabolismo , Proteínas Virales/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas
12.
Vet Res ; 47(1): 117, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27871316

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is an arterivirus responsible for a widespread contagious disease of domestic pigs with high economic impact. Switzerland is one of the rare PRRSV-free countries in Europe, although sporadic outbreaks have occurred in the past. The PRRSV isolate IVI-1173 from the short outbreak in Switzerland in 2012 was entirely sequenced, and a functional full-length cDNA clone was constructed. Genetic and antigenic characterization of IVI-1173 revealed the importance of amino acid 90 of the nucleocapsid protein N as part of a conformational epitope. IVI-1173 was not detected by SDOW17, a monoclonal antibody against N widely used to detect PRRSV-infected cells. Substitution of alanine at position 90 of N [N(A90)] with a threonine [N(T90)] restored reactivity of vIVI1173-N(T90) to SDOW17 completely. The relevance of this amino acid for the conformational SDOW17 epitope of PRRSV N was further confirmed by the opposite substitution in a functional cDNA clone of the genotype 2 isolate RVB-581. Finally, N proteins from ten genotype 1 strains differing from threonine at position 90 were analysed for reactivity with SDOW17. N(A90) totally disrupted or severely affected the epitope in 7 out of 8 strains tested. Based on these findings, 225 genotype 1 strains were screened for the prevalence of N(A90). N(A90) is rare in classical subtype 1 and in subtype 3 strains, but is frequent in Russian subtype 1 (70%) and in subtype 2 (45%) isolates. In conclusion, this study highlights the variable antigenic properties of N among genotype 1 PRRSV strains.


Asunto(s)
Antígenos Virales/inmunología , Nucleocápside/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/genética , Brotes de Enfermedades/veterinaria , Epítopos/genética , Epítopos/inmunología , Femenino , Citometría de Flujo/veterinaria , Técnica del Anticuerpo Fluorescente/veterinaria , Masculino , Nucleocápside/genética , Filogenia , Síndrome Respiratorio y de la Reproducción Porcina/epidemiología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Alineación de Secuencia , Análisis de Secuencia de ADN/veterinaria , Porcinos , Suiza/epidemiología
13.
Vet Res ; 47: 33, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26895704

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is the causative agent of one of the most devastating and economically significant viral disease of pigs worldwide. The vaccines currently available on the market elicit only limited protection. Recombinant vesicular stomatitis virus (VSV) replicon particles (VRP) have been used successfully to induce protection against influenza A virus (IAV) in chickens and bluetongue virus in sheep. In this study, VSV VRP expressing the PRRSV envelope proteins GP5, M, GP4, GP3, GP2 and the nucleocapsid protein N, individually or in combination, were generated and evaluated as a potential vector vaccine against PRRSV infection. High level expression of the recombinant PRRSV proteins was demonstrated in cell culture. However, none of the PRRSV antigens expressed from VRP, with the exception of the N protein, did induce any detectable antibody response in pigs before challenge infection with PRRSV. After challenge however, the antibody responses against GP5, GP4 and GP3 appeared in average 2 weeks earlier than in pigs vaccinated with the empty control VRP. No reduction of viremia was observed in the vaccinated group compared with the control group. When pigs were co-vaccinated with VRP expressing IAV antigens and VRP expressing PRRSV glycoproteins, only antibody responses to the IAV antigens were detectable. These data show that the VSV replicon vector can induce immune responses to heterologous proteins in pigs, but that the PRRSV envelope proteins expressed from VSV VRP are poorly immunogenic. Nevertheless, they prime the immune system for significantly earlier B-cell responses following PRRSV challenge infection.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/prevención & control , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Replicón/inmunología , Vacunas Virales/inmunología , Viremia/veterinaria , Animales , Glicoproteínas/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/virología , Porcinos , Vacunas Sintéticas/inmunología , Vesiculovirus/genética , Vesiculovirus/inmunología , Proteínas Virales/metabolismo , Viremia/inmunología , Viremia/prevención & control , Virión/inmunología
14.
Nanomedicine ; 12(3): 711-722, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26592962

RESUMEN

Self-amplifying replicon RNA (RepRNA) are large molecules (12-14 kb); their self-replication amplifies mRNA template numbers, affording several rounds of antigen production, effectively increasing vaccine antigen payloads. Their sensitivity to RNase-sensitivity and inefficient uptake by dendritic cells (DCs) - absolute requirements for vaccine design - were tackled by condensing RepRNA into synthetic, nanoparticulate, polyethylenimine (PEI)-polyplex delivery vehicles. Polyplex-delivery formulations for small RNA molecules cannot be transferred to RepRNA due to its greater size and complexity; the N:P charge ratio and impact of RepRNA folding would influence polyplex condensation, post-delivery decompaction and the cytosolic release essential for RepRNA translation. Polyplex-formulations proved successful for delivery of RepRNA encoding influenza virus hemagglutinin and nucleocapsid to DCs. Cytosolic translocation was facilitated, leading to RepRNA translation. This efficacy was confirmed in vivo, inducing both humoral and cellular immune responses. Accordingly, this paper describes the first PEI-polyplexes providing efficient delivery of the complex and large, self-amplifying RepRNA vaccines. FROM THE CLINICAL EDITOR: The use of self-amplifying replicon RNA (RepRNA) to increase vaccine antigen payloads can potentially be useful in effective vaccine design. Nonetheless, its use is limited by the degradation during the uptake process. Here, the authors attempted to solve this problem by packaging RepRNA using polyethylenimine (PEI)-polyplex delivery vehicles. The efficacy was confirmed in vivo by the appropriate humoral and cellular immune responses. This novel delivery method may prove to be very useful for future vaccine design.


Asunto(s)
Antígenos/genética , Polietileneimina/química , ARN/administración & dosificación , ARN/genética , Replicón , Vacunas/administración & dosificación , Vacunas/genética , Animales , Antígenos/inmunología , Línea Celular , Células Dendríticas/inmunología , Inmunidad Celular , Inmunidad Humoral , Ratones Endogámicos BALB C , Biosíntesis de Proteínas , ARN/inmunología , ARN/farmacocinética , Porcinos , Vacunas/inmunología , Vacunas/farmacocinética
15.
J Gen Virol ; 96(9): 2623-2635, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26018962

RESUMEN

Classical swine fever virus (CSFV) causes a highly contagious disease in pigs that can range from a severe haemorrhagic fever to a nearly unapparent disease, depending on the virulence of the virus strain. Little is known about the viral molecular determinants of CSFV virulence. The nonstructural protein NS4B is essential for viral replication. However, the roles of CSFV NS4B in viral genome replication and pathogenesis have not yet been elucidated. NS4B of the GPE- vaccine strain and of the highly virulent Eystrup strain differ by a total of seven amino acid residues, two of which are located in the predicted trans-membrane domains of NS4B and were described previously to relate to virulence, and five residues clustering in the N-terminal part. In the present study, we examined the potential role of these five amino acids in modulating genome replication and determining pathogenicity in pigs. A chimeric low virulent GPE- -derived virus carrying the complete Eystrup NS4B showed enhanced pathogenicity in pigs. The in vitro replication efficiency of the NS4B chimeric GPE- replicon was significantly higher than that of the replicon carrying only the two Eystrup-specific amino acids in NS4B. In silico and in vitro data suggest that the N-terminal part of NS4B forms an amphipathic α-helix structure. The N-terminal NS4B with these five amino acid residues is associated with the intracellular membranes. Taken together, this is the first gain-of-function study showing that the N-terminal domain of NS4B can determine CSFV genome replication in cell culture and viral pathogenicity in pigs.


Asunto(s)
Virus de la Fiebre Porcina Clásica/fisiología , Virus de la Fiebre Porcina Clásica/patogenicidad , Peste Porcina Clásica/virología , Membranas Intracelulares/virología , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Secuencias de Aminoácidos , Animales , Virus de la Fiebre Porcina Clásica/química , Virus de la Fiebre Porcina Clásica/genética , Estructura Secundaria de Proteína , Transporte de Proteínas , Porcinos , Proteínas no Estructurales Virales/genética , Virulencia
16.
J Gen Virol ; 96(Pt 7): 1746-56, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25809915

RESUMEN

The viral protein Npro is unique to the genus Pestivirus within the family Flaviviridae. After autocatalytic cleavage from the nascent polyprotein, Npro suppresses type I IFN (IFN-α/ß) induction by mediating proteasomal degradation of IFN regulatory factor 3 (IRF-3). Previous studies found that the Npro-mediated IRF-3 degradation was dependent of a TRASH domain in the C-terminal half of Npro coordinating zinc by means of the amino acid residues C112, C134, D136 and C138. Interestingly, four classical swine fever virus (CSFV) isolates obtained from diseased pigs in Thailand in 1993 and 1998 did not suppress IFN-α/ß induction despite the presence of an intact TRASH domain. Through systematic analyses, it was found that an amino acid mutation at position 40 or mutations at positions 17 and 61 in the N-terminal half of Npro of these four isolates were related to the lack of IRF-3-degrading activity. Restoring a histidine at position 40 or both a proline at position 17 and a lysine at position 61 based on the sequence of a functional Npro contributed to higher stability of the reconstructed Npro compared with the Npro from the Thai isolate. This led to enhanced interaction of Npro with IRF-3 along with its degradation by the proteasome. The results of the present study revealed that amino acid residues in the N-terminal domain of Npro are involved in the stability of Npro, in interaction of Npro with IRF-3 and subsequent degradation of IRF-3, leading to downregulation of IFN-α/ß production.


Asunto(s)
Virus de la Fiebre Porcina Clásica/inmunología , Endopeptidasas/química , Endopeptidasas/inmunología , Interacciones Huésped-Patógeno , Factores Reguladores del Interferón/antagonistas & inhibidores , Interferón Tipo I/antagonistas & inhibidores , Proteínas Virales/química , Proteínas Virales/inmunología , Sustitución de Aminoácidos , Animales , Peste Porcina Clásica/virología , Virus de la Fiebre Porcina Clásica/genética , Virus de la Fiebre Porcina Clásica/aislamiento & purificación , Análisis Mutacional de ADN , Regulación hacia Abajo , Endopeptidasas/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Mutación Missense , Unión Proteica , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Estructura Terciaria de Proteína , Porcinos , Tailandia , Proteínas Virales/genética
18.
PLoS Pathog ; 9(6): e1003412, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785283

RESUMEN

Plasmacytoid dendritic cells (pDC) have been shown to efficiently sense HCV- or HIV-infected cells, using a virion-free pathway. Here, we demonstrate for classical swine fever virus, a member of the Flaviviridae, that this process is much more efficient in terms of interferon-alpha induction when compared to direct stimulation by virus particles. By employment of virus replicon particles or infectious RNA which can replicate but not form de novo virions, we exclude a transfer of virus from the donor cell to the pDC. pDC activation by infected cells was mediated by a contact-dependent RNA transfer to pDC, which was sensitive to a TLR7 inhibitor. This was inhibited by drugs affecting the cytoskeleton and membrane cholesterol. We further demonstrate that a unique viral protein with ribonuclease activity, the viral E(rns) protein of pestiviruses, efficiently prevented this process. This required intact ribonuclease function in intracellular compartments. We propose that this pathway of activation could be of particular importance for viruses which tend to be mostly cell-associated, cause persistent infection, and are non-cytopathogenic.


Asunto(s)
Virus de la Fiebre Porcina Clásica/inmunología , Peste Porcina Clásica/inmunología , Células Dendríticas/inmunología , Endorribonucleasas/inmunología , Células Plasmáticas/inmunología , Proteínas Virales/inmunología , Animales , Línea Celular , Peste Porcina Clásica/patología , Células Plasmáticas/patología , ARN Viral/inmunología , Porcinos , Receptor Toll-Like 7/inmunología
19.
PLoS Pathog ; 9(10): e1003704, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24146623

RESUMEN

Pestiviruses express their genome as a single polypeptide that is subsequently cleaved into individual proteins by host- and virus-encoded proteases. The pestivirus N-terminal protease (N(pro)) is a cysteine autoprotease that cleaves between its own C-terminus and the N-terminus of the core protein. Due to its unique sequence and catalytic site, it forms its own cysteine protease family C53. After self-cleavage, N(pro) is no longer active as a protease. The released N(pro) suppresses the induction of the host's type-I interferon-α/ß (IFN-α/ß) response. N(pro) binds interferon regulatory factor-3 (IRF3), the key transcriptional activator of IFN-α/ß genes, and promotes degradation of IRF3 by the proteasome, thus preventing induction of the IFN-α/ß response to pestivirus infection. Here we report the crystal structures of pestivirus N(pro). N(pro) is structurally distinct from other known cysteine proteases and has a novel "clam shell" fold consisting of a protease domain and a zinc-binding domain. The unique fold of N(pro) allows auto-catalysis at its C-terminus and subsequently conceals the cleavage site in the active site of the protease. Although many viruses interfere with type I IFN induction by targeting the IRF3 pathway, little information is available regarding structure or mechanism of action of viral proteins that interact with IRF3. The distribution of amino acids on the surface of N(pro) involved in targeting IRF3 for proteasomal degradation provides insight into the nature of N(pro)'s interaction with IRF3. The structures thus establish the mechanism of auto-catalysis and subsequent auto-inhibition of trans-activity of N(pro), and its role in subversion of host immune response.


Asunto(s)
Virus de la Fiebre Porcina Clásica/enzimología , Proteasas de Cisteína/química , Interferón Tipo I , Pliegue de Proteína , Animales , Catálisis , Dominio Catalítico , Virus de la Fiebre Porcina Clásica/genética , Cristalografía por Rayos X , Proteasas de Cisteína/genética , Proteasas de Cisteína/metabolismo , Relación Estructura-Actividad , Porcinos
20.
Vet Res ; 46: 78, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26159607

RESUMEN

Classical swine fever (CSF) causes major losses in pig farming, with various degrees of disease severity. Efficient live attenuated vaccines against classical swine fever virus (CSFV) are used routinely in endemic countries. However, despite intensive vaccination programs in these areas for more than 20 years, CSF has not been eradicated. Molecular epidemiology studies in these regions suggests that the virus circulating in the field has evolved under the positive selection pressure exerted by the immune response to the vaccine, leading to new attenuated viral variants. Recent work by our group demonstrated that a high proportion of persistently infected piglets can be generated by early postnatal infection with low and moderately virulent CSFV strains. Here, we studied the immune response to a hog cholera lapinised virus vaccine (HCLV), C-strain, in six-week-old persistently infected pigs following post-natal infection. CSFV-negative pigs were vaccinated as controls. The humoral and interferon gamma responses as well as the CSFV RNA loads were monitored for 21 days post-vaccination. No vaccine viral RNA was detected in the serum samples and tonsils from CSFV postnatally persistently infected pigs for 21 days post-vaccination. Furthermore, no E2-specific antibody response or neutralising antibody titres were shown in CSFV persistently infected vaccinated animals. Likewise, no of IFN-gamma producing cell response against CSFV or PHA was observed. To our knowledge, this is the first report demonstrating the absence of a response to vaccination in CSFV persistently infected pigs.


Asunto(s)
Virus de la Fiebre Porcina Clásica/inmunología , Peste Porcina Clásica/inmunología , Inmunidad Celular , Inmunidad Humoral , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Animales , Peste Porcina Clásica/virología , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA