Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(3): 105779, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38395305

RESUMEN

The newly discovered zoonotic coronavirus swine acute diarrhea syndrome coronavirus (SADS-CoV) causes acute diarrhea, vomiting, dehydration, and high mortality rates in newborn piglets. Although SADS-CoV uses different strategies to evade the host's innate immune system, the specific mechanism(s) by which it blocks the interferon (IFN) response remains unidentified. In this study, the potential of SADS-CoV nonstructural proteins (nsp) to inhibit the IFN response was detected. The results determined that nsp1 was a potent antagonist of IFN response. SADS-CoV nsp1 efficiently inhibited signal transducer and activator of transcription 1 (STAT1) phosphorylation by inducing Janus kinase 1 (JAK1) degradation. Subsequent research revealed that nsp1 induced JAK1 polyubiquitination through K11 and K48 linkages, leading to JAK1 degradation via the ubiquitin-proteasome pathway. Furthermore, SADS-CoV nsp1 induced CREB-binding protein degradation to inhibit IFN-stimulated gene production and STAT1 acetylation, thereby inhibiting STAT1 dephosphorylation and blocking STAT1 transport out of the nucleus to receive antiviral signaling. In summary, the results revealed the novel mechanisms by which SADS-CoV nsp1 blocks the JAK-STAT signaling pathway via the ubiquitin-proteasome pathway. This study yielded valuable findings on the specific mechanism of coronavirus nsp1 in inhibiting the JAK-STAT signaling pathway and the strategies of SADS-CoV in evading the host's innate immune system.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Complejo de la Endopetidasa Proteasomal , Enfermedades de los Porcinos , Proteínas no Estructurales Virales , Animales , Acetilación , Alphacoronavirus/fisiología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Porcinos , Ubiquitinas/metabolismo , Enfermedades de los Porcinos/metabolismo , Enfermedades de los Porcinos/virología , Células HEK293 , Células Vero , Humanos , Chlorocebus aethiops , Proteínas no Estructurales Virales/metabolismo
2.
J Virol ; 97(6): e0068923, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37289083

RESUMEN

Goblet cells and their secreted mucus are important elements of the intestinal mucosal barrier, which allows host cells to resist invasion by intestinal pathogens. Porcine deltacoronavirus (PDCoV) is an emerging swine enteric virus that causes severe diarrhea in pigs and causes large economic losses to pork producers worldwide. To date, the molecular mechanisms by which PDCoV regulates the function and differentiation of goblet cells and disrupts the intestinal mucosal barrier remain to be determined. Here, we report that in newborn piglets, PDCoV infection disrupts the intestinal barrier: specifically, there is intestinal villus atrophy, crypt depth increases, and tight junctions are disrupted. There is also a significant reduction in the number of goblet cells and the expression of MUC-2. In vitro, using intestinal monolayer organoids, we found that PDCoV infection activates the Notch signaling pathway, resulting in upregulated expression of HES-1 and downregulated expression of ATOH-1 and thereby inhibiting the differentiation of intestinal stem cells into goblet cells. Our study shows that PDCoV infection activates the Notch signaling pathway to inhibit the differentiation of goblet cells and their mucus secretion, resulting in disruption of the intestinal mucosal barrier. IMPORTANCE The intestinal mucosal barrier, mainly secreted by the intestinal goblet cells, is a crucial first line of defense against pathogenic microorganisms. PDCoV regulates the function and differentiation of goblet cells, thereby disrupting the mucosal barrier; however, the mechanism by which PDCoV disrupts the barrier is not known. Here, we report that in vivo, PDCoV infection decreases villus length, increases crypt depth, and disrupts tight junctions. Moreover, PDCoV activates the Notch signaling pathway, inhibiting goblet cell differentiation and mucus secretion in vivo and in vitro. Thus, our results provide a novel insight into the mechanism underlying intestinal mucosal barrier dysfunction caused by coronavirus infection.


Asunto(s)
Infecciones por Coronavirus , Células Caliciformes , Receptores Notch , Enfermedades de los Porcinos , Animales , Coronavirus , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/veterinaria , Células Caliciformes/citología , Transducción de Señal , Porcinos , Enfermedades de los Porcinos/patología , Enfermedades de los Porcinos/virología , Células Madre/citología , Diferenciación Celular , Receptores Notch/metabolismo
3.
J Med Virol ; 95(12): e29302, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38084773

RESUMEN

Alphavirus is a type of arbovirus that can infect both humans and animals. The amino acid sequence of the 6K protein, being one of the structural proteins of the alphavirus, is not conserved. Deletion of this protein will result in varying effects on different alphaviruses. Our study focuses on the function of the Getah virus (GETV) 6K protein in infected cells and mice. We successfully constructed infectious clone plasmids and created resulting viruses (rGETV and rGETV-Δ6K). Our comprehensive microscopic analysis revealed that the 6 K protein mainly stays in the endoplasmic reticulum. In addition, rGETV-Δ6K has lower thermal stability and sensitivity to temperature than GETV. Although the deletion of the 6K protein does not reduce virion production in ST cells, it affects the release of virions from host cells by inhibiting the process of E2 protein transportation to the plasma membrane. Subsequent in vivo testing demonstrated that neonatal mice infected with rGETV-Δ6K had a lower virus content, less significant pathological changes in tissue slices, and milder disease than those infected with the wild-type virus. Our results indicate that the 6K protein effectively reduces the viral titer by influencing the release of viral particles. Furthermore, the 6K protein play a role in the clinical manifestation of GETV disease.


Asunto(s)
Alphavirus , Humanos , Animales , Ratones , Alphavirus/metabolismo , Virulencia , Proteínas Virales/metabolismo , Replicación Viral , Secuencia de Aminoácidos
4.
J Med Virol ; 95(9): e29104, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37721411

RESUMEN

Swine acute diarrhea syndrome (SADS) is first reported in January 2017 in Southern China. It subsequently causes widespread outbreaks in multiple pig farms, leading to economic losses. Therefore, it is an urgent to understand the molecular mechanisms underlying the pathogenesis and immune evasion of Swine acute diarrhea syndrome coronavirus (SADS-CoV). Our research discovered that SADS-CoV inhibited the production of interferon-ß (IFN-ß) during viral infection. The nonstructural protein 1 (nsp1) prevented the phosphorylation of TBK1 by obstructing the interaction between TBK1 and Ub protein. Moreover, nsp1 induced the degradation of CREB-binding protein (CBP) through the proteasome-dependent pathway, thereby disrupting the IFN-ß enhancer and inhibiting IFN transcription. Finally, we identified nsp1-Phe39 as the critical amino acid that downregulated IFN production. In conclusion, our findings described two mechanisms in nsp1 that inhibited IFN production and provided new insights into the evasion strategy adopted by SADS-CoV to evade host antiviral immunity.


Asunto(s)
Alphacoronavirus , Proteína de Unión a CREB , Animales , Porcinos , Fosforilación , Aminoácidos , Interferón beta/genética
5.
Cell Commun Signal ; 21(1): 361, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38110975

RESUMEN

Getah virus (GETV) was becoming more serious and posing a potential threat to animal safety and public health. Currently, there is limited comprehension regarding the pathogenesis and immune evasion mechanisms employed by GETV. Our study reveals that GETV infection exhibits the capacity for interferon antagonism. Specifically, the nonstructural protein nsP2 of GETV plays a crucial role in evading the host immune response. GETV nsP2 effectively inhibits the induction of IFN-ß by blocking the phosphorylation and nuclear translocation of IRF3. Additionally, GETV nsP2 hinders the phosphorylation of STAT1 and its nuclear accumulation, leading to significantly impaired JAK-STAT signaling. Furthermore, the amino acids K648 and R649, situated in the C-terminal region of GETV nsP2, play a crucial role in facilitating nuclear localization. Not only do they affect the interference of nsP2 with the innate immune response, but they also exert an influence on the pathogenicity of GETV in mice. In summary, our study reveals novel mechanisms by which GETV evades the immune system, thereby offering a foundation for comprehending the pathogenic nature of GETV. Video Abstract.


Asunto(s)
Alphavirus , Interferones , Animales , Ratones , Línea Celular , Inmunidad Innata , Evasión Inmune
6.
Int J Mol Sci ; 24(18)2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37762237

RESUMEN

Porcine deltacoronavirus (PDCoV) is an emerging virus that poses a significant threat to the global swine industry. Its membrane (M) protein is crucial for virion assembly and virus-host interactions. We selected the hydrophilic region of M protein for prokaryotic expression, purification, and recombinant protein production. Utilizing hybridoma technology, we prepared the monoclonal antibody (mAb) 24-A6 against M protein. The mAb 24-A6 was shown to be suitable for use in immunofluorescence assays, western blotting, and immunoprecipitation, with specificity for PDCoV and no cross-reactivity with other five porcine viruses. The M protein was observed to be expressed as early as 3 h after PDCoV infection, increasing its expression over the duration of infection. Notably, the antigenic epitope of the M protein identified as 103SPESRL108 recognized by mAb 24-A6 was found within a conserved structural domain (SWWSFNPETNNL) of the coronavirus M protein, indicating a crucial overlap between a functionally important viral assembly region and a region recognized by the immune system. Our findings provide valuable insights into mAb 24-A6 targeting the antigenic epitope of M protein and may contribute to the development of diagnostic tools for PDCoV infection and fundamental research into the function of PDCoV M protein.


Asunto(s)
Anticuerpos Monoclonales , Proteínas de la Membrana , Animales , Porcinos , Deltacoronavirus , Epítopos
7.
Dig Dis Sci ; 67(9): 4410-4421, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34797444

RESUMEN

BACKGROUND: Slow transit constipation (STC) is caused by intestinal peristalsis dysfunction and is closely associated with disturbance of the intestinal microecological balance. Bacillus subtilis plays a positive role in the treatment of STC, but its mechanism needs to be further explored. AIMS: The purpose of the present study was to explore the effects and mechanism of B. subtilis on the pathophysiology of STC. METHODS: A STC mouse model was established with compound diphenoxylate, following which B. subtilis was used to treat STC. The effects and possible mechanism of B. subtilis on STC were investigated by assessing intestinal motility, histology of the colon, release of 5-HT in enterochromaffin cells (ECs) and the TGR5/TRPA1 pathway. Moreover, LC-MS targeted metabolomics was used to analyze the regulation of Bacillus subtilis on bile acid metabolisms in STC mice. RESULTS: Bacillus subtilis significantly increased 24 h defecations, fecal moisture and intestinal transport rate of STC mice, improved pathological damage of the colon and showed protective effects on the intestinal tract. The release of 5-HT from ECs and the bile acid receptor TGR5/TRPA1 pathway were significantly increased in STC mice treated with B. subtilis. In addition, the metabolomics results showed that the bile acid contents of STC mice were significantly decreased, and B. subtilis could increase the bile acid composition and content of STC mice. CONCLUSION: Bacillus subtilis regulates intestinal peristalsis of STC by promoting the release of 5-HT from ECs through bile acid metabolism and its receptor TGR5 pathway and plays a positive role in the treatment of STC.


Asunto(s)
Ácidos y Sales Biliares , Peristaltismo , Animales , Bacillus subtilis/metabolismo , Estreñimiento , Ratones , Serotonina
8.
Appl Microbiol Biotechnol ; 105(6): 2385-2397, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33660038

RESUMEN

Senecavirus A (SVA) is an emerging picornavirus that has been associated with vesicular disease and neonatal mortality in swine. The construction of SVA virus carrying foreign reporter gene provides a powerful tool in virus research. However, it is often fraught with rescuing a recombinant picornavirus harboring a foreign gene or maintaining the stability of foreign gene in the virus genome. Here, we successfully generated recombinant SVA GD05/2017 viruses (V-GD05-clone) expressing the green fluorescent protein (iLOV), red fluorescent protein (RFP), or NanoLuc luciferase (Nluc). These recombinant viruses have comparable growth kinetics to the parental virus. Genetic stability analysis indicated that V-GD05-iLOV was highly stable in retaining iLOV gene for more than 10 passages, while V-GD05-RFP and V-GD05-Nluc lost the foreign genes in five passages. In addition, high-intensity fluorescent signals were found in the V-GD05-RFP- and V-GD05-iLOV-infected cells by fluorescence observation and flow cytometry analysis, and the luciferase activity assay could quantitatively monitor the replication of V-GD05-Nluc. In order to identify the porcine cell receptor for SVA, anthrax toxin receptor 1 (ANTXR1) was knocked out or overexpressed in the ST-R cells. The ANTXR1 knock-out cells lost the ability for SVA infection, while overexpression of ANTXR1 significantly increased the cell permissivity. These results confirmed that ANTXR1 was the receptor for SVA to invade porcine cells as reported in the human cells. Overall, this study suggests that these SVA reporter viruses will be useful tools in elucidating virus pathogenesis and developing control measures. KEY POINTS: • We successfully generated SVA viruses expressing the iLOV, RFP, or Nluc. • The iLOV was genetically stable in the V-GD05-iLOV genome over ten passages. • ANTXR1 was the receptor for SVA to invade porcine cells.


Asunto(s)
Picornaviridae , Animales , Genes Reporteros , Genoma Viral , Picornaviridae/genética , Proteínas Recombinantes , Porcinos
9.
Microb Pathog ; 147: 104432, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32771656

RESUMEN

Senecavirus A (SVA)-associated vesicular disease (SAVD) has extensively been present in the swine industry during the past years. The mechanisms of SVA-host interactions at the molecular level, subsequent to SVA infection, are unclear. We studied the gene expression profiles of LLC-PK1 cells, with or without SVA infection, for 6 h and 12 h using an RNA-seq technology. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed on differentially expressed genes (DEGs). Immune-related genes and pathways were significantly modified after SVA infection. To confirm the RNA-seq data, 28 important DEGs were selected for RT-qPCR assays. All DEGs exhibited expression patterns consistent with the RNA-seq results. Among them, type I IFNs (including IFN-α and IFN-ß) showed the largest upregulation, followed by RSAD2, DDX58, MX1 and the 17 other DEGs. In contrary, ID2 and another 5 DEGs were down-regulated or unchanged. These results indicated that type I IFNs play a critical role in host immune responses against SVA infection at early stage, while other immune-regulated genes directly or indirectly participate in the host immune responses.


Asunto(s)
Interferón Tipo I , Picornaviridae , Animales , Antivirales , Perfilación de la Expresión Génica , Interferón Tipo I/genética , Picornaviridae/genética , Porcinos , Transcriptoma
10.
Rev Med Virol ; 29(1): e2018, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30411827

RESUMEN

Emerging porcine pestivirus diseases frequently challenge prevention and control strategies in the swine industry. Over the past decade, a few novel pestiviruses have been identified in pigs. This article focuses on the recently emerging atypical porcine pestivirus (APPV) that potentially threatens global swine herd health security. The virus was first identified in 2016, in the United States and thereafter, accumulated evidence shows that it is currently distributed in three continents. The clinical presentation of APPV-infected pigs is characterized by congenital tremor (CT) type A-II in piglets, while adult pigs may become persistent carriers and shedders. Here, a literature review is conducted to summarize the published findings in the virus genomic biology, transmission, epidemiology, pathogenesis, and diagnosis, which would shed light on acceleration of development of anti-APPV strategies.


Asunto(s)
Enfermedades Transmisibles Emergentes/veterinaria , Infecciones por Pestivirus/veterinaria , Pestivirus/clasificación , Pestivirus/aislamiento & purificación , Enfermedades de los Porcinos/epidemiología , Enfermedades de los Porcinos/virología , Crianza de Animales Domésticos , Animales , Portador Sano/epidemiología , Portador Sano/veterinaria , Portador Sano/virología , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/patología , Enfermedades Transmisibles Emergentes/virología , Pruebas Diagnósticas de Rutina , Transmisión de Enfermedad Infecciosa , Salud Global , Pestivirus/genética , Infecciones por Pestivirus/epidemiología , Infecciones por Pestivirus/patología , Infecciones por Pestivirus/virología , Porcinos , Enfermedades de los Porcinos/patología , Esparcimiento de Virus
11.
Int J Mol Sci ; 21(21)2020 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-33114322

RESUMEN

Transmissible gastroenteritis virus (TGEV) is a coronavirus associated with diarrhea and high mortality in piglets. To gain insight into the evolution and adaptation of TGEV, a comprehensive analysis of phylogeny and codon usage bias was performed. The phylogenetic analyses of maximum likelihood and Bayesian inference displayed two distinct genotypes: genotypes I and II, and genotype I was classified into subtypes Ia and Ib. The compositional properties revealed that the coding sequence contained a higher number of A/U nucleotides than G/C nucleotides, and that the synonymous codon third position was A/U-enriched. The principal component analysis based on the values of relative synonymous codon usage (RSCU) showed the genotype-specific codon usage patterns. The effective number of codons (ENC) indicated moderate codon usage bias in the TGEV genome. Dinucleotide analysis showed that CpA and UpG were over-represented and CpG was under-represented in the coding sequence of the TGEV genome. The analyses of Parity Rule 2 plot, ENC-plot, and neutrality plot displayed that natural selection was the dominant evolutionary driving force in shaping codon usage preference in genotypes Ia and II. In addition, natural selection played a major role, while mutation pressure had a minor role in driving the codon usage bias in genotype Ib. The codon adaptation index (CAI), relative codon deoptimization index (RCDI), and similarity index (SiD) analyses suggested that genotype I might be more adaptive to pigs than genotype II. Current findings contribute to understanding the evolution and adaptation of TGEV.


Asunto(s)
Uso de Codones , Evolución Molecular , Virus de la Gastroenteritis Transmisible/genética , Islas de CpG , Genoma Viral , Selección Genética
12.
Microb Pathog ; 136: 103695, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31449854

RESUMEN

Seneca Valley virus (SVV) has been identified as the causative agent of SVV-associated vesicular disease (SAVD). To investigate the pathogenicity of two newly isolated SVV strains (GD-S5/2018 and GD04/2017) in China, experimental infections of pigs were performed. In pig experiments, both SVV strains successfully infected all animals, evidenced by presence of virus shedding and robust protective antibody responses. SVV GD-S5/2018 infection resulted in characteristic clinical signs, and ulcerative lesions on the tongue and gums. However, SVV GD04/2017 did not cause any clinical symptoms except depression in pigs during the experiment. Taken together, these results demonstrate that SVV GD-S5/2018 is a virulent strain for pigs, whereas SVV GD04/2017 is nearly avirulent. The established animal models for SVV infection will be utilized to dissect the immunity and pathogenesis, and develop vaccines and antivirals.


Asunto(s)
Infecciones por Picornaviridae/veterinaria , Picornaviridae/patogenicidad , Enfermedad Vesicular Porcina/patología , Enfermedad Vesicular Porcina/virología , Animales , China , Picornaviridae/aislamiento & purificación , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Porcinos , Virulencia
13.
Rev Med Virol ; 28(2)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29316047

RESUMEN

Parainfluenza virus 5 (PIV5), known as canine parainfluenza virus in the veterinary field, is a negative-sense, nonsegmented, single-stranded RNA virus belonging to the Paramyxoviridae family. Parainfluenza virus 5 is an excellent viral vector and has been used as a live vaccine for kennel cough for many years in dogs without any safety concern. It can grow to high titers in many cell types, and its genome is stable even in the presence of foreign gene insertions. So far, PIV5 has been used to develop vaccines against influenza virus, respiratory syncytial virus, rabies virus, and Mycobacterium tuberculosis, demonstrating its ability to elicit robust and protective immune responses in preclinical animal models. Parainfluenza virus 5-based vaccines can be administered intranasally, intramuscularly, or orally. Interestingly, prior exposure of PIV5 does not prevent a PIV5-vectored vaccine from generating robust immunity, indicating that the vector can be used more than once. Here, these encouraging results are reviewed together along with discussion of the desirable advantages of the PIV5 vaccine vector to aid future vaccine design and to accelerate progression of PIV5-based vaccines into clinical trials.


Asunto(s)
Enfermedades de los Animales/prevención & control , Control de Enfermedades Transmisibles , Enfermedades Transmisibles/inmunología , Vectores Genéticos , Virus de la Parainfluenza 5 , Vacunas/inmunología , Enfermedades de los Animales/etiología , Animales , Control de Enfermedades Transmisibles/métodos , Expresión Génica , Ingeniería Genética , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Inmunidad , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/inmunología , Vacunas/administración & dosificación , Estructuras Virales , Replicación Viral
14.
J Virol ; 91(19)2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28747497

RESUMEN

Human respiratory syncytial virus (RSV) is the leading etiologic agent of lower respiratory tract infections in children, but no licensed vaccine exists. Previously, we developed two parainfluenza virus 5 (PIV5)-based RSV vaccine candidates that protect mice against RSV challenge. PIV5 was engineered to express either the RSV fusion protein (F) or the RSV major attachment glycoprotein (G) between the hemagglutinin-neuraminidase (HN) and RNA-dependent RNA polymerase (L) genes of the PIV5 genome [PIV5-RSV-F (HN-L) and PIV5-RSV-G (HN-L), respectively]. To investigate the stability of the vaccine candidates in vitro, they were passaged in Vero cells at high and low multiplicities of infection (MOIs) for 11 generations and the genome sequences, growth kinetics, and protein expression of the resulting viruses were compared with those of the parent viruses. Sporadic mutations were detected in the consensus sequences of the viruses after high-MOI passages, and mutation rates increased under low-MOI-passage conditions. None of the mutations abolished antigen expression. Increased numbers of mutations correlated with increased growth rates in vitro, indicating that the viruses evolved through the course of serial passages. We also examined the in vivo stability of the vaccine candidates after a single passage in African green monkeys. No mutations were detected in the consensus sequences of viruses collected from the bronchoalveolar lavage (BAL) fluid of the animals. In vivo, mutations in RSV G and PIV5 L were found in individual isolates of PIV5-RSV-G (HN-L), but plaque isolates of PIV5-RSV-F (HN-L) had no mutations. To improve upon the PIV5-RSV-F (HN-L) candidate, additional vaccine candidates were generated in which the gene for RSV F was inserted into earlier positions in the PIV5 genome. These insertions did not negatively impact the sequence stability of the vaccine candidates. The results suggest that the RSV F and G gene insertions are stable in the PIV5 genome. However, the function of the foreign gene insertion may need to be considered when designing PIV5-based vaccines.IMPORTANCE The genetic stability of live viral vaccines is important for safety and efficacy. PIV5 is a promising live viral vector and has been used to develop vaccines. In this work, we examined the genetic stability of a PIV5-based RSV vaccine in vitro and in vivo We found that insertions of foreign genes, such as the RSV F and G genes, were stably maintained in the PIV5 genome and there was no mutation that abolished the expression of RSV F or G. Interestingly, the function of the inserted gene may have an impact on PIV5 genome stability.


Asunto(s)
Glicoproteínas/genética , Proteína HN/genética , Virus de la Parainfluenza 5/genética , ARN Polimerasa Dependiente del ARN/genética , Virus Sincitial Respiratorio Humano/genética , Proteínas Virales de Fusión/genética , Animales , Líquido del Lavado Bronquioalveolar/virología , Línea Celular , Chlorocebus aethiops , Cricetinae , Inestabilidad Genómica/genética , Glicoproteínas/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Células Vero , Proteínas Virales de Fusión/inmunología
15.
Microb Pathog ; 117: 247-254, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29408315

RESUMEN

PEDV remains one of the most important swine diseases that infects pigs of all ages. It causes devastating viral enteric disease in piglets with a high mortality rate, leading to significant threats and huge economic loss to the pork industry. In this study, a transcriptomic shotgun sequencing (RNA-Seq) procedure was used to study gene responses against PEDV infection. Genome-wide analysis of differentially expressed genes (DEGs) was performed in Vero E6 cells post-PEDV infection. mTOR signaling pathway activator-MHY1485, and inhibitor-PP242 were used to study the antiviral function. Results revealed that the IRF3 was significantly up-regulated post-PEDV infection. Although most of the IFN-regulatory and -related genes evaluated in this study were either down-regulated or remained unchanged, IL11 behaved significantly up-regulated, with the peak at 16 hpi. Nearly 90% of PEDV infections were suppressed in the PP242 pretreated cells whereas the reverse effect was observed in the MYH1485 pretreated cells. Results indicated that the mTOR signaling pathway played a vital role in the PEDV antiviral regulation in the Vero E6 cells. Future studies will contribute to better understand the cellular antiviral mechanism against PEDV.


Asunto(s)
Infecciones por Coronavirus/patología , Expresión Génica/genética , Virus de la Diarrea Epidémica Porcina/fisiología , Proteoma/metabolismo , Células Vero/metabolismo , Células Vero/virología , Animales , Antivirales/farmacología , Chlorocebus aethiops , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Regulación hacia Abajo , Perfilación de la Expresión Génica , Indoles/antagonistas & inhibidores , Interleucina-11/metabolismo , Morfolinas/farmacología , Virus de la Diarrea Epidémica Porcina/efectos de los fármacos , Virus de la Diarrea Epidémica Porcina/patogenicidad , Proteómica/métodos , Purinas/antagonistas & inhibidores , Transducción de Señal , Porcinos/virología , Enfermedades de los Porcinos/virología , Transcriptoma , Triazinas/farmacología , Células Vero/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
J Gen Virol ; 97(2): 281-286, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26581410

RESUMEN

Porcine parainfluenza virus 1 (PPIV1) was first identified in 2013 in slaughterhouse pigs in Hong Kong, China. Here, two near-complete genomes were assembled from swine exhibiting acute respiratory disease that were 90.0-95.3% identical to Chinese PPIV1. Analysis of the HN gene from ten additional PPIV1-positive samples found 85.0-95.5% identity, suggesting genetic diversity between strains. Molecular analysis identified 17 out of 279 (6.1%) positive samples from pigs with respiratory disease. Eleven nursery pigs from a naturally infected herd were asymptomatic; however, nasal swabs from six pigs and the lungs of a single pig were quantitative reverse transcriptase (qRT)-PCR positive. Histopathology identified PPIV1 RNA in the nasal respiratory epithelium and trachea. Two serological assays demonstrated seroconversion of infected pigs and further analysis of 59 swine serum samples found 52.5% and 66.1% seropositivity, respectively. Taken together, the results confirm the widespread presence of PPIV1 in the US swine herd.


Asunto(s)
Virus de la Parainfluenza 1 Humana/aislamiento & purificación , Infecciones por Paramyxoviridae/veterinaria , Infecciones del Sistema Respiratorio/veterinaria , Enfermedades de los Porcinos/epidemiología , Enfermedades de los Porcinos/virología , Animales , Análisis por Conglomerados , Genoma Viral , Histocitoquímica , Datos de Secuencia Molecular , Mucosa Nasal/patología , Mucosa Nasal/virología , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/virología , Filogenia , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Análisis de Secuencia de ADN , Homología de Secuencia , Porcinos , Tráquea/patología , Tráquea/virología , Estados Unidos/epidemiología , Virología/métodos
17.
J Virol ; 89(6): 3427-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552723

RESUMEN

Rabies remains a major public health threat around the world. Once symptoms appear, there is no effective treatment to prevent death. In this work, we tested a recombinant parainfluenza virus 5 (PIV5) strain expressing the glycoprotein (G) of rabies (PIV5-G) as a therapy for rabies virus infection: we have found that PIV5-G protected mice as late as 6 days after rabies virus infection. PIV5-G is a promising vaccine for prevention and treatment of rabies virus infection.


Asunto(s)
Vectores Genéticos/genética , Virus de la Parainfluenza 5/genética , Vacunas Antirrábicas/administración & dosificación , Virus de la Rabia/inmunología , Rabia/prevención & control , Proteínas del Envoltorio Viral/administración & dosificación , Animales , Anticuerpos Antivirales/inmunología , Expresión Génica , Vectores Genéticos/metabolismo , Humanos , Ratones , Virus de la Parainfluenza 5/metabolismo , Rabia/inmunología , Rabia/virología , Vacunas Antirrábicas/genética , Vacunas Antirrábicas/inmunología , Virus de la Rabia/genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
18.
J Gen Virol ; 96(10): 2994-2998, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26219947

RESUMEN

Pestiviruses are some of the most significant pathogens affecting ruminants and swine. Here, we assembled a 11 276 bp contig encoding a predicted 3635 aa polyprotein from porcine serum with 68 % pairwise identity to that of a recently partially characterized Rhinolophus affinis pestivirus (RaPV) and approximately 25-28 % pairwise identity to those of other pestiviruses. The virus was provisionally named atypical porcine pestivirus (APPV). Metagenomic sequencing of 182 serum samples identified four additional APPV-positive samples. Positive samples originated from five states and ELISAs using recombinant APPV Erns found cross-reactive antibodies in 94 % of a collection of porcine serum samples, suggesting widespread distribution of APPV in the US swine herd. The molecular and serological results suggest that APPV is a novel, highly divergent porcine pestivirus widely distributed in US pigs.


Asunto(s)
Infecciones por Pestivirus/veterinaria , Pestivirus/clasificación , Pestivirus/aislamiento & purificación , Enfermedades de los Porcinos/virología , Animales , Anticuerpos Antivirales/sangre , Análisis por Conglomerados , Reacciones Cruzadas , Datos de Secuencia Molecular , Pestivirus/genética , Infecciones por Pestivirus/virología , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Suero/virología , Porcinos , Estados Unidos
19.
J Virol ; 88(5): 2600-10, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24352450

RESUMEN

Mumps is a highly contagious human disease, characterized by lateral or bilateral nonsuppurative swelling of the parotid glands and neurological complications that can result in aseptic meningitis or encephalitis. A mumps vaccination program implemented since the 1960s reduced mumps incidence by more than 99% and kept the mumps case numbers as low as hundreds of cases per year in the United States before 2006. However, a large mumps outbreak occurred in vaccinated populations in 2006 and again in 2009 in the United States, raising concerns about the efficacy of the vaccination program. Previously, we have shown that clinical isolate-based recombinant mumps viruses lacking expression of either the V protein (rMuVΔV) or the SH protein (rMuVΔSH) are attenuated in a neurovirulence test using newborn rat brains (P. Xu et al., Virology 417:126-136, 2011, http://dx.doi.org/10.1016/j.virol.2011.05.003; P. Xu et al., J. Virol. 86:1768-1776, 2012, http://dx.doi.org/10.1128/JVI.06019-11) and may be good candidates for vaccine development. In this study, we examined immunity induced by rMuVΔSH and rMuVΔV in mice. Furthermore, we generated recombinant mumps viruses lacking expression of both the V protein and the SH protein (rMuVΔSHΔV). Analysis of rMuVΔSHΔV indicated that it was stable in tissue culture cell lines. Importantly, rMuVΔSHΔV was immunogenic in mice, indicating that it is a promising candidate for mumps vaccine development.


Asunto(s)
Vacuna contra la Parotiditis/genética , Vacuna contra la Parotiditis/inmunología , Animales , Anticuerpos Antivirales/inmunología , Secuencia de Bases , Técnicas de Cultivo de Célula , Línea Celular , Chlorocebus aethiops , Femenino , Humanos , Inmunización , Masculino , Ratones , Paperas/inmunología , Paperas/prevención & control , Vacuna contra la Parotiditis/administración & dosificación , Virus de la Parotiditis/genética , Virus de la Parotiditis/inmunología , Mutación , Ratas , Recombinación Genética , Eliminación de Secuencia , Linfocitos T/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Células Vero , Proteínas Virales/genética , Proteínas Virales/inmunología
20.
J Virol ; 87(23): 12990-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067956

RESUMEN

J paramyxovirus (JPV) was first isolated from moribund mice with hemorrhagic lung lesions in Australia in the 1970s. Recent sequencing of JPV (JPV-LW) confirms that JPV is a paramyxovirus with several unique features. However, neither JPV-LW nor a recombinant JPV based on its sequence (rJPV-LW) caused obvious illness in mice. In this work, we analyzed a different JPV isolate (JPV-BH), which behaved differently from JPV-LW; JPV-BH grew more slowly in Vero cells and had less of a cytopathic effect on tissue culture cells but caused severe disease in mice. We have determined the whole genome sequence of JPV-BH. There were several nucleotide sequence differences between JPV-BH and JPV-LW, one in the leader sequence, one in the GX gene, and three in the L gene. The high sequence identity between JPV-BH and JPV-LW suggests that JPV-BH and JPV-LW are the same virus strain but were obtained at different passages from different laboratories. To understand the roles of these nucleotide sequence differences in pathogenicity in mice, we generated a recombinant JPV-BH strain (rJPV-BH) and hybrid rJPV-BH strains with sequences from the leader sequence (rJPV-BH-Le-LW), the GX gene (rJPV-BH-GX-LW), and the L gene (rJPV-BH-L-LW) of JPV-LW and compared their pathogenicities in mice. We have found that rJPV-BH-L-LW was attenuated in mice, indicating that nucleotide sequence differences in the L gene play a critical role in pathogenesis.


Asunto(s)
Infecciones por Paramyxoviridae/veterinaria , Paramyxovirinae/metabolismo , Paramyxovirinae/patogenicidad , Enfermedades de los Roedores/virología , Proteínas Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/genética , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA