Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Emerg Infect Dis ; 30(4): 738-751, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38478379

RESUMEN

Highly pathogenic avian influenza (HPAI) viruses have potential to cross species barriers and cause pandemics. Since 2022, HPAI A(H5N1) belonging to the goose/Guangdong 2.3.4.4b hemagglutinin phylogenetic clade have infected poultry, wild birds, and mammals across North America. Continued circulation in birds and infection of multiple mammalian species with strains possessing adaptation mutations increase the risk for infection and subsequent reassortment with influenza A viruses endemic in swine. We assessed the susceptibility of swine to avian and mammalian HPAI H5N1 clade 2.3.4.4b strains using a pathogenesis and transmission model. All strains replicated in the lung of pigs and caused lesions consistent with influenza A infection. However, viral replication in the nasal cavity and transmission was only observed with mammalian isolates. Mammalian adaptation and reassortment may increase the risk for incursion and transmission of HPAI viruses in feral, backyard, or commercial swine.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Animales , Aves , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar , Mamíferos , Filogenia , Aves de Corral , Porcinos
2.
PLoS Pathog ; 18(3): e1010197, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35312736

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19) in humans, has a broad host range, and is able to infect domestic and wild animal species. Notably, white-tailed deer (WTD, Odocoileus virginianus), the most widely distributed cervid species in the Americas, were shown to be highly susceptible to SARS-CoV-2 in challenge studies and reported natural infection/exposure rates approaching 30-40% in free-ranging WTD in the U.S. Thus, understanding the infection and transmission dynamics of SARS-CoV-2 in WTD is critical to prevent future zoonotic transmission to humans, at the human-WTD interface during hunting or venison farming, and for implementation of effective disease control measures. Here, we demonstrated that following intranasal inoculation with SARS-CoV-2 B.1 lineage, WTD fawns (~8-month-old) shed infectious virus up to day 5 post-inoculation (pi), with high viral loads shed in nasal and oral secretions. This resulted in efficient deer-to-deer transmission on day 3 pi. Consistent a with lack of infectious SARS-CoV-2 shedding after day 5 pi, no transmission was observed to contact animals added on days 6 and 9 pi. We have also investigated the tropism and sites of SARS-CoV-2 replication in adult WTD (3-4 years of age). Infectious virus was detected up to day 6 pi in nasal secretions, and from various respiratory-, lymphoid-, and central nervous system tissues, indicating broad tissue tropism and multiple sites of virus replication. The study provides important insights on the infection and transmission dynamics of SARS-CoV-2 in WTD, a wild animal species that is highly susceptible to infection and with the potential to become a reservoir for the virus in the field.


Asunto(s)
COVID-19 , Ciervos , Animales , COVID-19/veterinaria , SARS-CoV-2 , Tropismo
3.
J Virol ; 95(11)2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692203

RESUMEN

The origin of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus causing the global coronavirus disease 19 (COVID-19) pandemic, remains a mystery. Current evidence suggests a likely spillover into humans from an animal reservoir. Understanding the host range and identifying animal species that are susceptible to SARS-CoV-2 infection may help to elucidate the origin of the virus and the mechanisms underlying cross-species transmission to humans. Here we demonstrated that white-tailed deer (Odocoileus virginianus), an animal species in which the angiotensin converting enzyme 2 (ACE2) - the SARS-CoV-2 receptor - shares a high degree of similarity to humans, are highly susceptible to infection. Intranasal inoculation of deer fawns with SARS-CoV-2 resulted in established subclinical viral infection and shedding of infectious virus in nasal secretions. Notably, infected animals transmitted the virus to non-inoculated contact deer. Viral RNA was detected in multiple tissues 21 days post-inoculation (pi). All inoculated and indirect contact animals seroconverted and developed neutralizing antibodies as early as day 7 pi. The work provides important insights into the animal host range of SARS-CoV-2 and identifies white-tailed deer as a susceptible wild animal species to the virus.IMPORTANCEGiven the presumed zoonotic origin of SARS-CoV-2, the human-animal-environment interface of COVID-19 pandemic is an area of great scientific and public- and animal-health interest. Identification of animal species that are susceptible to infection by SARS-CoV-2 may help to elucidate the potential origin of the virus, identify potential reservoirs or intermediate hosts, and define the mechanisms underlying cross-species transmission to humans. Additionally, it may also provide information and help to prevent potential reverse zoonosis that could lead to the establishment of a new wildlife hosts. Our data show that upon intranasal inoculation, white-tailed deer became subclinically infected and shed infectious SARS-CoV-2 in nasal secretions and feces. Importantly, indirect contact animals were infected and shed infectious virus, indicating efficient SARS-CoV-2 transmission from inoculated animals. These findings support the inclusion of wild cervid species in investigations conducted to assess potential reservoirs or sources of SARS-CoV-2 of infection.

4.
J Virol ; 94(17)2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32554697

RESUMEN

Coronaviruses (CoVs) have repeatedly emerged from wildlife hosts and infected humans and livestock animals to cause epidemics with significant morbidity and mortality. CoVs infect various organs, including respiratory and enteric systems, as exemplified by newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The constellation of viral factors that contribute to developing enteric disease remains elusive. Here, we investigated CoV interferon antagonists for their contribution to enteric pathogenesis. Using an infectious clone of an enteric CoV, porcine epidemic diarrhea virus (icPEDV), we generated viruses with inactive versions of interferon antagonist nonstructural protein 1 (nsp1), nsp15, and nsp16 individually or combined into one virus designated icPEDV-mut4. Interferon-responsive PK1 cells were infected with these viruses and produced higher levels of interferon responses than were seen with wild-type icPEDV infection. icPEDV-mut4 elicited robust interferon responses and was severely impaired for replication in PK1 cells. To evaluate viral pathogenesis, piglets were infected with either icPEDV or icPEDV-mut4. While the icPEDV-infected piglets exhibited clinical disease, the icPEDV-mut4-infected piglets showed no clinical symptoms and exhibited normal intestinal pathology at day 2 postinfection. icPEDV-mut4 replicated in the intestinal tract, as revealed by detection of viral RNA in fecal swabs, with sequence analysis documenting genetic stability of the input strain. Importantly, icPEDV-mut4 infection elicited IgG and neutralizing antibody responses to PEDV. These results identify nsp1, nsp15, and nsp16 as virulence factors that contribute to the development of PEDV-induced diarrhea in swine. Inactivation of these CoV interferon antagonists is a rational approach for generating candidate vaccines to prevent disease and spread of enteric CoVs, including SARS-CoV-2.IMPORTANCE Emerging coronaviruses, including SARS-CoV-2 and porcine CoVs, can infect enterocytes, cause diarrhea, and be shed in the feces. New approaches are needed to understand enteric pathogenesis and to develop vaccines and therapeutics to prevent the spread of these viruses. Here, we exploited a reverse genetic system for an enteric CoV, porcine epidemic diarrhea virus (PEDV), and outline an approach of genetically inactivating highly conserved viral factors known to limit the host innate immune response to infection. Our report reveals that generating PEDV with inactive versions of three viral interferon antagonists, nonstructural proteins 1, 15, and 16, results in a highly attenuated virus that does not cause diarrhea in animals and elicits a neutralizing antibody response in virus-infected animals. This strategy may be useful for generating live attenuated vaccine candidates that prevent disease and fecal spread of enteric CoVs, including SARS-CoV-2.


Asunto(s)
Infecciones por Coronavirus/inmunología , Coronavirus/inmunología , Interferones/inmunología , Virus de la Diarrea Epidémica Porcina/inmunología , Vacunas Atenuadas/inmunología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Betacoronavirus/inmunología , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/prevención & control , Diarrea/patología , Diarrea/virología , Modelos Animales de Enfermedad , Endorribonucleasas/antagonistas & inhibidores , Heces/virología , Íleon/patología , Inmunidad Innata , Yeyuno/patología , Pandemias , Neumonía Viral/inmunología , Virus de la Diarrea Epidémica Porcina/genética , ARN Viral , ARN Polimerasa Dependiente del ARN , SARS-CoV-2 , Porcinos , Enfermedades de los Porcinos/virología , Células Vero , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/inmunología
5.
Respiration ; 100(8): 811-815, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34044399

RESUMEN

The radiological findings of COVID-19 are well-described, including its evolution. In an earlier report of admission chest radiographs of patients with COVID-19, we anecdotally noted relative sparing of the left upper zone (LUZ). We subsequently aimed to describe the main chest radiograph findings in another cohort, focusing on zonal predominance. The admission chest radiographs of 111 patients with CO-VID-19 pneumonia requiring intensive care admission were reviewed by 2 thoracic radiologists and categorized according to the predominant pattern into either ground-glass opacities (GGOs), alveolar infiltrates and/or consolidation, or reticular and/or nodular infiltrates or an equal combination of both, and the extent of disease involvement of each of the zones using a modified Radiologic Assessment of Lung Edema (RALE) score. Parenchymal changes were detected in all. In total, 106 radiographs showed GGOs, alveolar infiltrates, and/or consolidation, and 5 had a combination of reticular/nodular infiltrates as well as GGOs, alveolar infiltrates, and/or consolidation. The LUZ had a significant lower grading score than the right upper zone: 1 versus 2 (p < 0.001). Likewise, the upper zones had a significant lower score than the mid and lower zones (p < 0.001). Our findings confirmed the relative sparing of the LUZ in severe COVID-19 pneumonia.


Asunto(s)
COVID-19/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Radiografía Torácica
6.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728254

RESUMEN

Identifying viral antagonists of innate immunity and determining if they contribute to pathogenesis are critical for developing effective strategies to control emerging viruses. Previously, we reported that an endoribonuclease (EndoU) encoded by murine coronavirus plays a pivotal role in evasion of host innate immune defenses in macrophages. Here, we asked if the EndoU activity of porcine epidemic diarrhea coronavirus (PEDV), which causes acute diarrhea in swine, plays a role in antagonizing the innate response in porcine epithelial cells and macrophages, the sites of viral replication. We constructed an infectious clone of PEDV-Colorado strain (icPEDV-wt) and an EndoU-mutant PEDV (icPEDV-EnUmt) by changing the codon for a catalytic histidine residue of EndoU to alanine (His226Ala). We found that both icPEDV-wt and icPEDV-EnUmt propagated efficiently in interferon (IFN)-deficient Vero cells. In contrast, the propagation of icPEDV-EnUmt was impaired in porcine epithelial cells (LLC-PK1), where we detected an early and robust transcriptional activation of type I and type III IFNs. Infection of piglets with the parental Colorado strain, icPEDV-wt, or icPEDV-EnUmt revealed that all viruses replicated in the gut and induced diarrhea; however, there was reduced viral shedding and mortality in the icPEDV-EnUmt-infected animals. These results demonstrate that EndoU activity is not required for PEDV replication in immortalized, IFN-deficient Vero cells, but is important for suppressing the IFN response in epithelial cells and macrophages, which facilitates replication, shedding, and pathogenesis in vivo We conclude that PEDV EndoU activity is a key virulence factor that suppresses both type I and type III IFN responses.IMPORTANCE Coronaviruses (CoVs) can emerge from an animal reservoir into a naive host species to cause pandemic respiratory or gastrointestinal diseases with significant mortality in humans or domestic animals. Porcine epidemic diarrhea virus (PEDV), an alphacoronavirus (alpha-CoV), infects gut epithelial cells and macrophages, inducing diarrhea and resulting in high mortality in piglets. How PEDV suppresses the innate immune response was unknown. We found that mutating a viral endoribonuclease, EndoU, results in a virus that activates both the type I interferon response and the type III interferon response in macrophages and epithelial cells. This activation of interferon resulted in limited viral replication in epithelial cell cultures and was associated with reduced virus shedding and mortality in piglets. This study reveals a role for EndoU activity as a virulence factor in PEDV infection and provides an approach for generating live-attenuated vaccine candidates for emerging coronaviruses.


Asunto(s)
Infecciones por Coronavirus , Endorribonucleasas , Interferón Tipo I/inmunología , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Proteínas Virales , Animales , Línea Celular , Infecciones por Coronavirus/enzimología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/veterinaria , Endorribonucleasas/genética , Endorribonucleasas/inmunología , Interferón Tipo I/genética , Virus de la Diarrea Epidémica Porcina/enzimología , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/inmunología , Porcinos , Enfermedades de los Porcinos/enzimología , Enfermedades de los Porcinos/genética , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Proteínas Virales/genética , Proteínas Virales/inmunología , Esparcimiento de Virus/inmunología
7.
BMC Bioinformatics ; 20(1): 265, 2019 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-31132991

RESUMEN

BACKGROUND: In standard high throughput sequencing analysis, genetic variants are not assigned to a homologous chromosome of origin. This process, called haplotype phasing, can reveal information important for understanding the relationship between genetic variants and biological phenotypes. For example, in genes that carry multiple heterozygous missense variants, phasing resolves whether one or both gene copies are altered. Here, we present a novel approach to phasing variants that takes advantage of unique properties of paired tumor:normal sequencing data from cancer studies. RESULTS: VAF phasing uses changes in variant allele frequency (VAF) between tumor and normal samples in regions of somatic chromosomal gain or loss to phase germline variants. We apply VAF phasing to 6180 samples from the Cancer Genome Atlas (TCGA) and demonstrate that our method is highly concordant with other standard phasing methods, and can phase an average of 33% more variants than other read-backed phasing methods. Using variant annotation tools designed to score gene haplotypes, we find a suggestive association between carrying multiple missense variants in a single copy of a cancer predisposition gene and earlier age of cancer diagnosis. CONCLUSIONS: VAF phasing exploits unique properties of tumor genomes to increase the number of germline variants that can be phased over standard read-backed methods in paired tumor:normal samples. Our phase-informed association testing results call attention to the need to develop more tools for assessing the joint effect of multiple genetic variants.


Asunto(s)
Variación Genética , Neoplasias/genética , Análisis de Secuencia de ADN , Secuencia de Bases , Variaciones en el Número de Copia de ADN/genética , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad , Haplotipos/genética , Humanos
8.
BMC Vet Res ; 14(1): 352, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30453952

RESUMEN

BACKGROUND: Senecavirus A, commonly known as Seneca Valley virus (SVV), is a picornavirus that has been infrequently associated with porcine idiopathic vesicular disease (PIVD). In late 2014 there were multiple PIVD outbreaks in several states in Brazil and samples from those cases tested positive for SVV. Beginning in July of 2015, multiple cases of PIVD were reported in the United States in which a genetically similar SVV was also detected. These events suggested SVV could induce vesicular disease, which was recently demonstrated with contemporary US isolates that produced mild disease in pigs. It was hypothesized that stressful conditions may exacerbate the expression of clinical disease and the following experiment was performed. Two groups of 9-week-old pigs were given an intranasal SVV challenge with one group receiving an immunosuppressive dose of dexamethasone prior to challenge. After challenge animals were observed for the development of clinical signs and serum and swabs were collected to study viral shedding and antibody production. In addition, pigs were euthanized 2, 4, 6, 8, and 12 days post inoculation (dpi) to demonstrate tissue distribution of virus during acute infection. RESULTS: Vesicular disease was experimentally induced in both groups with the duration and magnitude of clinical signs similar between groups. During acute infection [0-14 days post infection (dpi)], SVV was detected by PCR in serum, nasal swabs, rectal swabs, various tissues, and in swabs from ruptured vesicles. From 15 to 30 dpi, virus was less consistently detected in nasal and rectal swabs, and absent from most serum samples. Virus neutralizing antibody was detected by 5 dpi and lasted until the end of the study. CONCLUSION: Treatment with an immunosuppressive dose of dexamethasone did not drastically alter the clinical disease course of SVV in experimentally infected nursery aged swine. A greater understanding of SVV pathogenesis and factors that could exacerbate disease can help the swine industry with control and prevention strategies directed against this virus.


Asunto(s)
Dexametasona/farmacología , Inmunosupresores/farmacología , Infecciones por Picornaviridae/veterinaria , Picornaviridae , Enfermedades de los Porcinos/virología , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/sangre , Porcinos , Enfermedad Vesicular Porcina/virología , Esparcimiento de Virus/efectos de los fármacos
9.
PLoS Genet ; 11(6): e1005347, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26125563

RESUMEN

The childhood epileptic encephalopathies (EE's) are seizure disorders that broadly impact development including cognitive, sensory and motor progress with severe consequences and comorbidities. Recently, mutations in DNM1 (dynamin 1) have been implicated in two EE syndromes, Lennox-Gastaut Syndrome and Infantile Spasms. Dnm1 encodes dynamin 1, a large multimeric GTPase necessary for activity-dependent membrane recycling in neurons, including synaptic vesicle endocytosis. Dnm1Ftfl or "fitful" mice carry a spontaneous mutation in the mouse ortholog of DNM1 and recapitulate many of the disease features associated with human DNM1 patients, providing a relevant disease model of human EE's. In order to examine the cellular etiology of seizures and behavioral and neurological comorbidities, we engineered a conditional Dnm1Ftfl mouse model of DNM1 EE. Observations of Dnm1Ftfl/flox mice in combination with various neuronal subpopulation specific cre strains demonstrate unique seizure phenotypes and clear separation of major neurobehavioral comorbidities from severe seizures associated with the germline model. This demonstration of pleiotropy suggests that treating seizures per se may not prevent severe comorbidity observed in EE associated with dynamin-1 mutations, and is likely to have implications for other genetic forms of EE.


Asunto(s)
Dinamina I/genética , Epilepsia/genética , Animales , Conducta Animal , Modelos Animales de Enfermedad , Dinamina I/metabolismo , Electroencefalografía , Epilepsia/epidemiología , Epilepsia/mortalidad , Epilepsia/patología , Femenino , Eliminación de Gen , Humanos , Lactante , Síndrome de Lennox-Gastaut/epidemiología , Síndrome de Lennox-Gastaut/genética , Masculino , Ratones Mutantes , Neuronas/patología , Fenotipo , Prosencéfalo/metabolismo , Prosencéfalo/fisiopatología , Espasmos Infantiles/epidemiología , Espasmos Infantiles/genética , Transmisión Sináptica
10.
BMC Genomics ; 18(1): 458, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28606096

RESUMEN

BACKGROUND: Cancer research to date has largely focused on somatically acquired genetic aberrations. In contrast, the degree to which germline, or inherited, variation contributes to tumorigenesis remains unclear, possibly due to a lack of accessible germline variant data. Here we called germline variants on 9618 cases from The Cancer Genome Atlas (TCGA) database representing 31 cancer types. RESULTS: We identified batch effects affecting loss of function (LOF) variant calls that can be traced back to differences in the way the sequence data were generated both within and across cancer types. Overall, LOF indel calls were more sensitive to technical artifacts than LOF Single Nucleotide Variant (SNV) calls. In particular, whole genome amplification of DNA prior to sequencing led to an artificially increased burden of LOF indel calls, which confounded association analyses relating germline variants to tumor type despite stringent indel filtering strategies. The samples affected by these technical artifacts include all acute myeloid leukemia and practically all ovarian cancer samples. CONCLUSIONS: We demonstrate how technical artifacts induced by whole genome amplification of DNA can lead to false positive germline-tumor type associations and suggest TCGA whole genome amplified samples be used with caution. This study draws attention to the need to be sensitive to problems associated with a lack of uniformity in data generation in TCGA data.


Asunto(s)
Artefactos , Bases de Datos Genéticas , Genómica , Mutación de Línea Germinal , Neoplasias/genética , Genoma Humano/genética , Humanos , Mutación con Pérdida de Función
11.
Emerg Infect Dis ; 22(7): 1246-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27315363

RESUMEN

Senecavirus A has been infrequently associated with vesicular disease in swine since 1988. However, clinical disease has not been reproduced after experimental infection with this virus. We report vesicular disease in 9-week-old pigs after Sencavirus A infection by the intranasal route under experimental conditions.


Asunto(s)
Infecciones por Picornaviridae/veterinaria , Picornaviridae , Enfermedades de los Porcinos/virología , Animales , Enfermedades del Pie/patología , Enfermedades del Pie/veterinaria , Enfermedades del Pie/virología , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Porcinos , Enfermedades de los Porcinos/patología
12.
Sci Rep ; 14(1): 11171, 2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750049

RESUMEN

White-tailed deer (Odocoileus virginianus) have emerged as a reservoir host for SARS-CoV-2 given their susceptibility to infection and demonstrated high rates of seroprevalence and infection across the United States. As SARS-CoV-2 circulates within free-ranging white-tailed deer populations, there is the risk of transmission to other wildlife species and even back to the human population. The goal of this study was to determine the susceptibility, shedding, and immune response of North American elk (Cervus elaphus canadensis) to experimental infection with SARS-CoV-2, to determine if another wide-ranging cervid species could potentially serve as a reservoir host for the virus. Here we demonstrate that while North American elk do not develop clinical signs of disease, they do develop a neutralizing antibody response to infection, suggesting the virus is capable of replicating in this mammalian host. Additionally, we demonstrate SARS-CoV-2 RNA presence in the medial retropharyngeal lymph nodes of infected elk three weeks after experimental infection. Consistent with previous observations in humans, these data may highlight a mechanism of viral persistence for SARS-CoV-2 in elk.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19 , Ciervos , ARN Viral , SARS-CoV-2 , Animales , Ciervos/virología , SARS-CoV-2/genética , SARS-CoV-2/fisiología , COVID-19/virología , ARN Viral/genética , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Esparcimiento de Virus , Reservorios de Enfermedades/virología , Femenino
13.
Vet Microbiol ; 289: 109958, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38181600

RESUMEN

Neonatal mortality has been increasingly reported on swine breeding farms experiencing swine idiopathic vesicular disease (SIVD) outbreaks, which can be accompanied by lethargy, diarrhea, and neurologic signs in neonates. Seneca Valley Virus (SVV), or Senecavirus A, has been detected in clinical samples taken from pigs with SIVD. Experimental SVV inoculation has caused vesicular disease in pigs, particularly during the stages from weaning to finishing. However, it remains crucial to investigate whether SVV directly contributes to the increase in neonatal mortality rates. The following study was conducted to chronicle the pathogenesis of SVV infection in sows and their offspring. Ten sows were intranasally inoculated with 4.75 × 107 plaque-forming units of the virus per sow either late in gestation (n = 5) or within fourteen days of farrowing (n = 5). Each sow replicated SVV following intranasal inoculation, but only one out of ten sows developed a vesicular lesion on the snout. Evidence of transplacental infection was observed in two litters, and an additional two litters became infected following parturition out of five litters from sows inoculated in late gestation. No clinical signs were observed in the infected neonates. Likewise, no clinical signs were observed in the other five litters inoculated after farrowing, although each piglet did replicate the challenge virus. In this study, the experimental challenge of SVV did not result in neonatal mortality in contrast to observations in the field; however, it has shed light on the pathogenesis of the virus, the transmission of SVV between sows and their offspring, and host immune response that can help shape control measures in the field.


Asunto(s)
Infecciones por Picornaviridae , Picornaviridae , Enfermedades de los Porcinos , Porcinos , Animales , Femenino , Embarazo , Infecciones por Picornaviridae/veterinaria , Brotes de Enfermedades/veterinaria
14.
Front Genet ; 14: 1292671, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38075681

RESUMEN

Pseudorabies virus (PRV)-the causative agent of Aujeszky's disease-was eliminated from commercial pig production herds in the United States (US) in 2004; however, PRV remains endemic among invasive feral swine (Sus scrofa). The circulation of PRV among abundant, widespread feral swine populations poses a sustained risk for disease spillover to production herds. Risk-based surveillance has been successfully implemented for PRV in feral swine populations in the US. However, understanding the role of host genetics in infection status may offer new insights into the epidemiology and disease dynamics of PRV that can be applied to management strategies. Genetic mechanisms underlying host susceptibility to PRV are relatively unknown; therefore, we sought to identify genomic regions associated with PRV infection status among naturally infected feral swine using genome-wide association studies (GWAS) and gene set enrichment analysis of single nucleotide polymorphism data (GSEA-SNP). Paired serological and genotypic data were collected from 6,081 feral swine distributed across the invaded range within the contiguous US. Three complementary study populations were developed for GWAS: 1) comprehensive population consisting of feral swine throughout the invaded range within the contiguous US; 2) population of feral swine under high, but temporally variable PRV infection pressure; and 3) population of feral swine under temporally stable, high PRV infection pressure. We identified one intronic SNP associated with PRV infection status within candidate gene AKAP6 on autosome 7. Various gene sets linked to metabolic pathways were enriched in the GSEA-SNP. Ultimately, improving disease surveillance efforts in feral swine will be critical to further understanding of the role host genetics play in PRV infection status, helping secure the health of commercial pork production.

15.
Viruses ; 15(8)2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37632109

RESUMEN

Atypical porcine pestivirus (APPV) was found to be associated with pigs demonstrating congenital tremors (CT), and clinical signs in pigs have been reproduced after experimental challenge. Subsequently, APPV has been identified in both symptomatic and asymptomatic swine of all ages globally. The objective of this research was to perform a longitudinal study following two cohorts of pigs, those born in litters with pigs exhibiting CT and those born in litters without CT, to analyze the virus and antibody dynamics of APPV infection in serum from birth to market. There was a wide range in the percentage of affected pigs (8-75%) within CT-positive litters. After co-mingling with CT-positive litters at weaning, pigs from CT-negative litters developed viremia that was cleared after approximately 2 months, with the majority seroconverting by the end of the study. In contrast, a greater percentage of pigs exhibiting CT remained PCR positive throughout the growing phase, with less than one-third of these animals seroconverting. APPV RNA was present in multiple tissues from pigs in both groups at the time of marketing. This study improved our understanding of the infection dynamics of APPV in swine and the impact that the immune status and timing of infection have on the persistence of APPV in serum and tissues.


Asunto(s)
Anticuerpos , Pestivirus , Animales , Porcinos , Estudios Longitudinales , Pestivirus/genética , Reacción en Cadena de la Polimerasa , Temblor/veterinaria
16.
PLoS One ; 17(4): e0267145, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35486625

RESUMEN

Foot-and-mouth disease virus (FMDV) is a picornavirus that produces a highly transmissible vesicular disease that can devastate meat and dairy production to such an extent that FMDV-free countries commit significant economic resources to maintain their FMDV-free status. Senecavirus A (SVA), also a picornavirus, causes vesicular disease in swine that is indistinguishable from FMDV. Since 2015, SVA outbreaks have been reported around the world requiring FMDV-free countries to investigate these cases to rule out FMDV. Understanding the pathogenesis of the SVA and its ability to transmit to naïve populations is critical to formulating control and prevention measures, which could reduce FMDV investigations. The primary objective of this study was to determine the infectious dose of SVA in market weight and neonatal pigs. A 2011 SVA isolate was serially hundred-fold diluted to create four challenge inoculums ranging from 106.5 to 100.5 TCID50/ml. Four market weight pigs individually housed were intranasally inoculated with 5 mL of each dose (n = 16). Serial ten-fold dilutions were used to create 6 challenge inoculums ranging from 105.5 to 100.5 TCID50/ml for neonatal pigs. Again, four animals in individual housing were challenged orally with 2 mL of each dose (n = 24). Detection of SVA by PCR in collected samples and/or neutralizing antibody response was utilized to classify an animal as infected. The minimum infectious dose for this study in market weight animals was 1,260 TCID50/ml (103.1 TCID50/ml) and for neonates it was 316 TCID50/ml (102.5 TCID50/ml). Knowledge of the infectious dose of SVA can guide biosecurity and disinfection measures to control the spread of SVA.


Asunto(s)
Virus de la Fiebre Aftosa , Picornaviridae , Enfermedades de los Porcinos , Animales , Anticuerpos Neutralizantes , Porcinos
17.
Vaccine ; 40(12): 1747-1754, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35183385

RESUMEN

Senecavirus A (SVA), commonly known as Seneca Valley virus (SVV) is a causative agent for vesicular disease in swine. It has been found across the globe including the United States, Brazil, and China. Clinical disease caused by this virus is identical to foot-and-mouth disease virus (FMDV). Since FMDV has the potential to cause severe economic consequences in FMDV-free countries, those countries are on high alert for signs of vesicles in swine and an investigation is performed to rule out the presence of FMDV if observed. In countries where SVA cases have continued to occur, investigations and testing can cause a burden on personnel and resources. The objectives of this study were to test the efficacy of a whole-virus inactivated SVA vaccine against challenge in nursery-aged pigs, mature sows, and to assess the protection of passive maternal immunity generated by immunized dams. Animals were given two doses of the vaccine intramuscularly three weeks apart and challenged intranasally two weeks after the second dose. Non-vaccinated animals challenged with SVA developed clinical signs of disease, replicated virus, and developed a neutralizing antibody response. Vaccinated animals had robust neutralizing titers after two doses; and after challenge, did not develop vesicular disease and had limited rectal shedding. Piglets suckling immunized dams and challenged with SVA at 3-6 days-of-age had neutralizing titers prior to challenge and did not replicate or shed virus. An efficacious vaccine could improve swine welfare and reduce the economic consequences of continued foreign animal disease investigations.


Asunto(s)
Infecciones por Picornaviridae , Enfermedades de los Porcinos , Vacunas Virales , Animales , Anticuerpos Antivirales , Femenino , Picornaviridae , Infecciones por Picornaviridae/veterinaria , Porcinos , Enfermedades de los Porcinos/prevención & control , Vacunas de Productos Inactivados
18.
Front Vet Sci ; 9: 827082, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35296061

RESUMEN

Streptococcus equi subspecies zooepidemicus (SEZ) is a zoonotic pathogen capable of causing severe disease in many mammalian species. Historically, SEZ has not been a common cause of disease in pigs in North America; however, in 2019, SEZ caused mortality events leading to severe illness and 30-50% mortality in exposed animal groups. Because of the rapid progression of disease, it is important to investigate intervention strategies to prevent disease development. In this study, pigs were divided into four groups: (1) vaccinated with an inactivated SEZ vaccine generated from a highly mucoid 2019 mortality event isolate; (2) vaccinated with an inactivated SEZ vaccine generated from a genetically similar, non-mucoid isolate from a guinea pig; (3) and (4) sham vaccinated. Following boost vaccination, groups 1-3 were challenged with a 2019 mortality event isolate and group 4 were non-challenged controls. Antibody titers were higher for SEZ vaccinated animals than sham vaccinated animals; however, no anamnestic response was observed, and titers were lower than typically seen following the use of inactivated vaccines. Vaccination did not provide protection from disease development or mortality following challenge, which could be associated with the comparatively low antibody titers generated by vaccination. Surviving pigs also remained colonized and transmitted SEZ to naïve contact pigs 3 weeks following challenge, indicating that healthy animals can act as a source of SEZ exposure. Future investigation should evaluate different vaccine formulations, such as increased antigen load or an alternative adjuvant, that could induce a more robust adaptive immune response.

19.
Viruses ; 14(12)2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36560813

RESUMEN

Senecavirus A (SVA) is a causative agent for vesicular disease in swine, which is clinically indistinguishable from other vesicular diseases of swine including foot-and-mouth disease (FMD). Recently, it was reported that buffalo in Guangdong, China were experiencing clinical symptoms similar to FMD including mouth ulcers and lameness tested positive for SVA. The objective of this study was to determine the susceptibility of cattle (Bos taurus) to SVA infection. Initial in vitro work using the PrimeFlow assay demonstrated that bovine cell lines and peripheral blood mononuclear cells from cattle were susceptible to SVA infection. Subsequently, six colostrum-deprived Holstein calves were challenged with SVA intranasally. No vesicular lesions were observed after challenge. Serum, oral, nasal, and rectal swabs tested for SVA nucleic acid did not support significant viral replication and there was no evidence of seroconversion. Therefore, demonstrating cattle from this study were not susceptible to experimental SVA infection.


Asunto(s)
Fiebre Aftosa , Infecciones por Picornaviridae , Picornaviridae , Enfermedades de los Porcinos , Femenino , Embarazo , Bovinos , Animales , Porcinos , Calostro , Leucocitos Mononucleares , Línea Celular
20.
Front Vet Sci ; 9: 923878, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35812884

RESUMEN

Vesicular disease caused by Senecavirus A (SVA) is clinically indistinguishable from foot-and-mouth disease (FMD) and other vesicular diseases of swine. When a vesicle is observed in FMD-free countries, a costly and time-consuming foreign animal disease investigation (FADI) is performed to rule out FMD. Recently, there has been an increase in the number of FADIs and SVA positive samples at slaughter plants in the U.S. The objectives of this investigation were to: (1) describe the environmental burden of SVA in sow slaughter plants; (2) determine whether there was a correlation between PCR diagnostics, virus isolation (VI), and swine bioassay results; and (3) phylogenetically characterize the genetic diversity of contemporary SVA isolates. Environmental swabs were collected from three sow slaughter plants (Plants 1-3) and one market-weight slaughter plant (Plant 4) between June to December 2020. Of the 426 samples taken from Plants 1-3, 304 samples were PCR positive and 107 were VI positive. There was no detection of SVA by PCR or VI at Plant 4. SVA positive samples were most frequently found in the summer (78.3% June-September, vs. 59.4% October-December), with a peak at 85% in August. Eighteen PCR positive environmental samples with a range of Ct values were selected for a swine bioassay: a single sample infected piglets (n = 2). A random subset of the PCR positive samples was sequenced; and phylogenetic analysis demonstrated co-circulation and divergence of two genetically distinct groups of SVA. These data demonstrate that SVA was frequently found in the environment of sow slaughter plants, but environmental persistence and diagnostic detection was not indicative of whether a sampled was infectious to swine. Consequently, a more detailed understanding of the epidemiology of SVA and its environmental persistence in the marketing chain is necessary to reduce the number of FADIs and aide in the development of control measures to reduce the spread of SVA.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA