Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 872
Filtrar
1.
Microb Pathog ; 191: 106646, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631414

RESUMEN

Porcine viral diarrhea is a common ailment in clinical settings, causing significant economic losses to the swine industry. Notable culprits behind porcine viral diarrhea encompass transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus (PDCoV), and porcine rotavirus-A (PoRVA). Co-infections involving the viruses are a common occurrence in clinical settings, thereby amplifying the complexities associated with differential diagnosis. As a consequence, it is therefore necessary to develop a method that can detect and differentiate all four porcine diarrhea viruses (TGEV, PEDV, PDCoV, and PoRVA) with a high sensitivity and specificity. Presently, polymerase chain reaction (PCR) is the go-to method for pathogen detection. In comparison to conventional PCR, TaqMan real-time PCR offers heightened sensitivity, superior specificity, and enhanced accuracy. This study aimed to develop a quadruplex real-time RT-qPCR assay, utilizing TaqMan probes, for the distinctive detection of TGEV, PEDV, PDCoV, and PoRVA. The quadruplex real-time RT-qPCR assay, as devised in this study, exhibited the capacity to avoid the detection of unrelated pathogens and demonstrated commendable specificity, sensitivity, repeatability, and reproducibility, boasting a limit of detection (LOD) of 27 copies/µL. In a comparative analysis involving 5483 clinical samples, the results from the commercial RT-qPCR kit and the quadruplex RT-qPCR for TGEV, PEDV, PDCoV, and PoRVA detection were entirely consistent. Following sample collection from October to March in Guangxi Zhuang Autonomous Region, we assessed the prevalence of TGEV, PEDV, PDCoV, and PoRVA in piglet diarrhea samples, revealing positive detection rates of 0.2 % (11/5483), 8.82 % (485/5483), 1.22 % (67/5483), and 4.94 % (271/5483), respectively. The co-infection rates of PEDV/PoRVA, PEDV/PDCoV, TGEV/PED/PoRVA, and PDCoV/PoRVA were 0.39 %, 0.11 %, 0.01 %, and 0.03 %, respectively, with no detection of other co-infections, as determined by the quadruplex real-time RT-qPCR. This research not only established a valuable tool for the simultaneous differentiation of TGEV, PEDV, PDCoV, and PoRVA in practical applications but also provided crucial insights into the prevalence of these viral pathogens causing diarrhea in Guangxi.


Asunto(s)
Virus de la Diarrea Epidémica Porcina , Reacción en Cadena en Tiempo Real de la Polimerasa , Rotavirus , Sensibilidad y Especificidad , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/aislamiento & purificación , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/aislamiento & purificación , Virus de la Diarrea Epidémica Porcina/clasificación , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/diagnóstico , Rotavirus/genética , Rotavirus/aislamiento & purificación , Rotavirus/clasificación , Gastroenteritis Porcina Transmisible/diagnóstico , Gastroenteritis Porcina Transmisible/virología , Deltacoronavirus/genética , Deltacoronavirus/aislamiento & purificación , Diarrea/virología , Diarrea/veterinaria , Diarrea/diagnóstico , Coronavirus/genética , Coronavirus/aislamiento & purificación , Coronavirus/clasificación , Heces/virología , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología
2.
Virology ; 595: 110072, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38599031

RESUMEN

Porcine respiratory coronavirus (PRCV) was initially detected in Europe, and later in the United States of America (US), in the 1980s. In this study we obtained and compared PRCV sequences from Europe and the US, and investigated how these are related to transmissible gastroenteritis virus (TGEV) sequences. The whole genome sequences of Danish (1/90-DK), Italian (PRCV15087/12 III NPTV Parma), and Belgian PRCV (91V44) strains are presented. These sequences were aligned with nine other PRCV sequences from Europe and the US, and 43 TGEV sequences. Following alignment of the PRCV sequences, it was apparent that multiple amino acid variations in the structural proteins were distinct between the European and US strains. The alignments were used to build phylogenetic trees to infer the evolutionary relationships between the strains. In these trees, the European PRCV strains clustered as a separate group, whereas the US strains of PRCV all clustered with TGEVs.


Asunto(s)
Genoma Viral , Filogenia , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/clasificación , Europa (Continente) , Enfermedades de los Porcinos/virología , Estados Unidos , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/veterinaria , Coronavirus/genética , Coronavirus/clasificación , Gastroenteritis Porcina Transmisible/virología
3.
Talanta ; 274: 126024, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38583330

RESUMEN

The detection of transmissible gastroenteritis virus (TGEV) is of great significance to reduce the loss of pig industry. A LAMP-visualization/PFC self-powered dual-mode output sensor platform was constructed to detect TGEV by combining a simple and intuitive photoelectrochromic material with a highly sensitive PFC self-powered sensing platform without external power supply. The PFC sensing substrate was constructed using CdS nanoparticles modified ZnO NRs (CdS/ZnO NRs) as the photoanode, which exhibited high photoactivity, and Prussian blue (PB) as the cathode. After LAMP reaction on the optical anode, visual signals caused by PB discolorimetry can be detected semi-quantitatively, or PFC power density electrical signals collected by electrochemical workstation can be used. The output power density value is logarithm of TGEV concentration. The linear relationship was good within the detection range of 0.075 fg/µL-7.5 ng/µL, with a detection limit of 0.025 fg/µL (S/N = 3). This multi-signal output sensing platform provides more choices for quantifying TGEV detection results, and the two methods can be mutually verified, which meets the needs of different scenarios and improves the reliability of detection. It has a good effect in the actual sample detection, without the use of expensive and complex instruments, and has a broad application prospect.


Asunto(s)
Polímeros de Fluorocarbono , Técnicas de Diagnóstico Molecular , Técnicas de Amplificación de Ácido Nucleico , Virus de la Gastroenteritis Transmisible , Óxido de Zinc , Virus de la Gastroenteritis Transmisible/aislamiento & purificación , Óxido de Zinc/química , Animales , Porcinos , Límite de Detección , Compuestos de Cadmio/química , Técnicas Electroquímicas/métodos , Técnicas Electroquímicas/instrumentación , Nanopartículas/química , Sulfuros/química
4.
Emerg Microbes Infect ; 13(1): 2332653, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38517703

RESUMEN

Coinfection with multiple viruses is a common phenomenon in clinical settings and is a crucial driver of viral evolution. Although numerous studies have demonstrated viral recombination arising from coinfections of different strains of a specific species, the role of coinfections of different species or genera during viral evolution is rarely investigated. Here, we analyzed coinfections of and recombination events between four different swine enteric coronaviruses that infect the jejunum and ileum in pigs, including porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), and swine acute diarrhea syndrome coronavirus (SADS-CoV), and a deltacoronavirus, porcine deltacoronavirus (PDCoV). Various coinfection patterns were observed in 4,468 fecal and intestinal tissue samples collected from pigs in a 4-year survey. PEDV/PDCoV was the most frequent coinfection. However, recombination analyses have only detected events involving PEDV/TGEV and SADS-CoV/TGEV, indicating that inter-species recombination among coronaviruses is most likely to occur within the same genus. We also analyzed recombination events within the newly identified genus Deltacoronavirus and found that sparrows have played a unique host role in the recombination history of the deltacoronaviruses. The emerging virus PDCoV, which can infect humans, has a different recombination history. In summary, our study demonstrates that swine enteric coronaviruses are a valuable model for investigating the relationship between viral coinfection and recombination, which provide new insights into both inter- and intraspecies recombination events among swine enteric coronaviruses, and extend our understanding of the relationship between coronavirus coinfection and recombination.


Asunto(s)
Alphacoronavirus , Coinfección , Infecciones por Coronavirus , Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Humanos , Porcinos , Animales , Coinfección/veterinaria , Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Gastroenteritis Transmisible/genética , Recombinación Genética
5.
Vet Microbiol ; 292: 110055, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38513523

RESUMEN

Transmissible gastroenteritis virus (TGEV) is characterized by watery diarrhea, vomiting, and dehydration and is associated with high mortality especially in newborn piglets, causing significant economic losses to the global pig industry. Hypoxia inducible factor-1α (HIF-1α) has been identified as a key regulator of TGEV-induced inflammation, but understanding of the effect of HIF-1α on TGEV infection remains limited. This study found that TGEV infection was associated with a marked increase in HIF-1α expression in ST cells and an intestinal organoid epithelial monolayer. Furthermore, HIF-1α was shown to facilitate TGEV infection by targeting viral replication, which was achieved by restraining type I and type III interferon (IFN) production. In vivo experiments in piglets demonstrated that the HIF-1α inhibitor BAY87-2243 significantly reduced HIF-1α expression and inhibited TGEV replication and pathogenesis by activating IFN production. In summary, we unveiled that HIF-1α facilitates TGEV replication by restraining type I and type III IFN production in vitro, ex vivo, and in vivo. The findings from this study suggest that HIF-1α could be a novel antiviral target and candidate drug against TGEV infection.


Asunto(s)
Gastroenteritis Porcina Transmisible , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Interferón lambda , Intestinos , Replicación Viral , Hipoxia/veterinaria
6.
BMC Genomics ; 25(1): 239, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38438836

RESUMEN

BACKGROUND: Acute diarrhea, dehydration and death in piglets are all symptoms of transmissible gastroenteritis virus (TGEV), which results in significant financial losses in the pig industry. It is important to understand the pathogenesis and identify new antiviral targets by revealing the metabolic interactions between TGEV and host cells. RESULTS: We performed metabolomic and transcriptomic analyses of swine testicular cells infected with TGEV. A total of 1339 differential metabolites and 206 differentially expressed genes were detected post TEGV infection. The differentially expressed genes were significantly enriched in the HIF-1 signaling pathway and PI3K-Akt signaling. Integrated analysis of differentially expressed genes and differential metabolites indicated that they were significantly enriched in the metabolic processes such as nucleotide metabolism, biosynthesis of cofactors and purine metabolism. In addition, the results showed that most of the detected metabolites involved in the bile secretion was downregulated during TGEV infection. Furthermore, exogenous addition of key metabolite deoxycholic acid (DCA) significantly enhanced TGEV replication by NF-κB and STAT3 signal pathways. CONCLUSIONS: We identified a significant metabolite, DCA, related to TGEV replication. It added TGEV replication in host cells by inhibiting phosphorylation of NF-κB and STAT3. This study provided novel insights into the metabolomic and transcriptomic alterations related to TGEV infection and revealed potential molecular and metabolic targets for the regulation of TGEV infection.


Asunto(s)
FN-kappa B , Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Fosforilación , Fosfatidilinositol 3-Quinasas , Perfilación de la Expresión Génica , Transcriptoma , Ácido Desoxicólico/farmacología
7.
Vet J ; 304: 106083, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38365083

RESUMEN

Transmissible gastroenteritis virus (TGEV) is an important pathogen capable of altering the expression profile of cellular miRNA. In this study, the potential of Polygonum cillinerve polysaccharide (PCP) to treat TGEV-infected piglets was evaluated through in vivo experiments. High-throughput sequencing technology was employed to identify 9 up-regulated and 17 down-regulated miRNAs during PCP-mediated inhibition of TGEV infection in PK15 cells. Additionally, miR-181 was found to be associated with target genes of key proteins in the apoptosis pathway. PK15 cells were treated with various concentrations of PCP following transfection with miR-181 mimic or inhibitor. Real-time PCR assessed the impact on TGEV replication, while electron microscopy (TEM) and Hoechst fluorescence staining evaluated cellular functionality. Western blot analysis was utilized to assess the expression of key signaling factors-cytochrome C (cyt C), caspase 9, and P53-in the apoptotic signaling pathway. The results showed that compared with the control group, 250 µg/mL PCP significantly inhibited TGEV gRNA replication and gene N expression (P < 0.01). Microscopic examination revealed uniform cell morphology and fewer floating cells in PCP-treated groups (250 and 125 µg/mL). TEM analysis showed no typical virus structure in the 250 µg/mL PCP group, and apoptosis staining indicated a significant reduction in apoptotic cells at this concentration. Furthermore, PCP may inhibit TGEV-induced apoptosis via the Caspase-dependent mitochondrial pathway following miR-181 transfection. These findings provide a theoretical basis for further exploration into the mechanism of PCP's anti-TGEV properties.


Asunto(s)
MicroARNs , Polygonum , Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Polygonum/genética , ARN Guía de Sistemas CRISPR-Cas , Transducción de Señal , MicroARNs/genética
8.
J Virol ; 98(2): e0137723, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38197629

RESUMEN

Gut microbiota-derived metabolites are important for the replication and pathogenesis of many viruses. However, the roles of bacterial metabolites in swine enteric coronavirus (SECoV) infection remain poorly understood. Recent studies show that SECoVs infection in vivo significantly alters the composition of short-chain fatty acids (SCFAs)-producing gut microbiota. This prompted us to investigate whether and how SCFAs impact SECoV infection. Employing alphacoronavirus transmissible gastroenteritis virus (TGEV), a major cause of diarrhea in piglets, as a model, we found that SCFAs, particularly butyrate, enhanced TGEV infection both in porcine intestinal epithelial cells and swine testicular (ST) cells at the late stage of viral infection. This effect depended on the inhibited productions of virus-induced type I interferon (IFN) and downstream antiviral IFN-stimulated genes (ISGs) by butyrate. Mechanistically, butyrate suppressed the expression of retinoic acid-inducible gene I (RIG-I), a key viral RNA sensor, and downstream mitochondrial antiviral-signaling (MAVS) aggregation, thereby impairing type I IFN responses and increasing TGEV replication. Using pharmacological and genetic approaches, we showed that butyrate inhibited RIG-I-induced type I IFN signaling by suppressing class I histone deacetylase (HDAC). In summary, we identified a novel mechanism where butyrate enhances TGEV infection by suppressing RIG-I-mediated type I IFN responses. Our findings highlight that gut microbiota-derived metabolites like butyrate can be exploited by SECoV to dampen innate antiviral immunity and establish infection in the intestine.IMPORTANCESwine enteric coronaviruses (SECoVs) infection in vivo alters the composition of short-chain fatty acids (SCFAs)-producing gut microbiota, but whether microbiota-derived SCFAs impact coronavirus gastrointestinal infection is largely unknown. Here, we demonstrated that SCFAs, particularly butyrate, substantially increased alphacoronavirus TGEV infection at the late stage of infection, without affecting viral attachment or internalization. Furthermore, enhancement of TGEV by butyrate depended on impeding virus-induced type I interferon (IFN) responses. Mechanistically, butyrate suppressed the cytoplasmic viral RNA sensor RIG-I expression and downstream type I IFN signaling activation by inhibiting class I HDAC, thereby promoting TGEV infection. Our work reveals novel functions of gut microbiota-derived SCFAs in enhancing enteric coronavirus infection by impairing RIG-I-dependent type I IFN responses. This implies that bacterial metabolites could be therapeutic targets against SECoV infection by modulating antiviral immunity in the intestine.


Asunto(s)
Butiratos , Infecciones por Coronavirus , Coronavirus , Microbioma Gastrointestinal , Interferón Tipo I , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Animales , Butiratos/metabolismo , Coronavirus/fisiología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Interferón Tipo I/inmunología , ARN Viral , Porcinos , Virus de la Gastroenteritis Transmisible/fisiología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología
9.
Vet Microbiol ; 290: 109956, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38217919

RESUMEN

Swine enteric coronaviruses, such as porcine epidemic diarrhea virus (PEDV) or transmissible gastroenteritis virus (TGEV), have risen concern for the porcine industry and research community due to the increase in their virulence, their potential recombination capacity and the emergence of new variants. This in vivo study aims to compare the impact of three different strains of swine enteric coronaviruses [(two G1b (S-INDEL) PEDV strains and a recombinant TGEV-PEDV or Swine enteric coronavirus (SeCoV)] in the intestine of 3-weeks-old infected piglets, focusing on the pathology and main components of the intestinal barrier, including the number of goblet cells, and the expression of IgA as well as FoxP3, a regulatory T cell marker. Severity of lesions was evidenced in the three infected groups and was highly correlated with the viral load in feces and the frequency of viral antigen-positive cells. Furthermore, higher cellular death together with an increase in the expression of the FoxP3 marker was detected in the duodenum and jejunum of infected animals at 3 days post-infection. Our results highlight a recruitment of FoxP3+ cells in the small intestine of infected animals which may represent a response to the tissue damage caused by viral replication and cell death. Further studies should be addressed to determine the potential role of these cells during swine enteric coronavirus infections.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Porcinos , Animales , Coronavirus/genética , Infecciones por Coronavirus/veterinaria , Intestino Delgado , Virus de la Diarrea Epidémica Porcina/genética , Factores de Transcripción Forkhead/genética
10.
Biosens Bioelectron ; 246: 115900, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38056342

RESUMEN

The outbreak of transmissible gastroenteritis virus (TGEV) will cause huge economic losses to the whole pig industry. Hence, there is urgent need to develop a rapid and ultrasensitive method for detection of TGEV. As a nucleic acid detection technique, loop-mediated isothermal amplification (LAMP) can achieve quantitative detection of targeted nucleic acids with high sensitivity and selectivity. Nevertheless, the signal outputs of LAMP method must be acquired by complicated instruments. In this work, we firstly developed a LAMP photochromic sensing chip for porcine TGEV detection by combination of the photochromic sensing chip and nucleic acid amplification. The detection signal was based on color change of electrochromic material rather than electrical signal, and thus the detection signal can be obtained by visualization without relying on complicated instrument. The entire test was performed with small fluorinated indium tin oxide electrodes modified with zinc oxide (ZnO) (a photocatalytic material) and Prussian blue (PB) (an electrochromic material). When photoinduced electrons produced by ZnO were injected into PB under light, the PB was reduced to Prussian white. The higher the concentration of TGEV, the more double-stranded DNA was produced after amplification. The amplified product produced greater impedance, and fewer electron was transferred, which affect the corresponding color change of PB. The sensing chip also showed highly sensitive response to TGEV, with the minimum limit of detection was determined to be 2.5 fg/µL. The sensing chip developed herein will provide a new avenue for DNA amplification detection by visualization.


Asunto(s)
Técnicas Biosensibles , Ácidos Nucleicos , Virus de la Gastroenteritis Transmisible , Óxido de Zinc , Porcinos , Animales , Virus de la Gastroenteritis Transmisible/genética , Sensibilidad y Especificidad , Técnicas de Amplificación de Ácido Nucleico/métodos
11.
J Virol ; 98(1): e0123923, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38099687

RESUMEN

Coronaviruses (CoVs) pose a major threat to human and animal health worldwide, which complete viral replication by hijacking host factors. Identifying host factors essential for the viral life cycle can deepen our understanding of the mechanisms of virus-host interactions. Based on our previous genome-wide CRISPR screen of α-CoV transmissible gastroenteritis virus (TGEV), we identified the host factor dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), but not DYRK1B, as a critical factor in TGEV replication. Rescue assays and kinase inhibitor experiments revealed that the effect of DYRK1A on viral replication is independent of its kinase activity. Nuclear localization signal modification experiments showed that nuclear DYRK1A facilitated virus replication. Furthermore, DYRK1A knockout significantly downregulated the expression of the TGEV receptor aminopeptidase N (ANPEP) and inhibited viral entry. Notably, we also demonstrated that DYRK1A is essential for the early stage of TGEV replication. Transmission electron microscopy results indicated that DYRK1A contributes to the formation of double-membrane vesicles in a kinase-independent manner. Finally, we validated that DYRK1A is also a proviral factor for mouse hepatitis virus, porcine deltacoronavirus, and porcine sapelovirus. In conclusion, our work demonstrated that DYRK1A is an essential host factor for the replication of multiple viruses, providing new insights into the mechanism of virus-host interactions and facilitating the development of new broad-spectrum antiviral drugs.IMPORTANCECoronaviruses, like other positive-sense RNA viruses, can remodel the host membrane to form double-membrane vesicles (DMVs) as their replication organelles. Currently, host factors involved in DMV formation are not well defined. In this study, we used transmissible gastroenteritis virus (TGEV) as a virus model to investigate the regulatory mechanism of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) on coronavirus. Results showed that DYRK1A significantly inhibited TGEV replication in a kinase-independent manner. DYRK1A knockout (KO) can regulate the expression of receptor aminopeptidase N (ANPEP) and endocytic-related genes to inhibit virus entry. More importantly, our results revealed that DYRK1A KO notably inhibited the formation of DMV to regulate the virus replication. Further data proved that DYRK1A is also essential in the replication of mouse hepatitis virus, porcine deltacoronavirus, and porcine sapelovirus. Taken together, our findings demonstrated that DYRK1A is a conserved factor for positive-sense RNA viruses and provided new insights into its transcriptional regulation activity, revealing its potential as a candidate target for therapeutic design.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Quinasas DyrK , Animales , Humanos , Ratones , Antígenos CD13/genética , Coronavirus/clasificación , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Deltacoronavirus , Virus de la Hepatitis Murina/fisiología , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Tirosina , Replicación Viral/fisiología , Quinasas DyrK/metabolismo
12.
J Gen Virol ; 104(12)2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38116760

RESUMEN

Transmissible gastroenteritis virus (TGEV) is a coronavirus that infects piglets with severe diarrhoea, vomiting, dehydration, and even death, causing huge economic losses to the pig industry. The underlying pathogenesis of TGEV infection and the effects of TGEV infection on host metabolites remain poorly understood. To investigate the critical metabolites and regulatory factors during TGEV infection in intestinal porcine epithelial cells (IPEC-J2), we performed metabolomic and transcriptomic analyses of TGEV-infected IPEC-J2 cells by LC/MS and RNA-seq techniques. A total of 87 differential metabolites and 489 differentially expressed genes were detected. A series of metabolites and candidate genes from glutathione metabolism and AMPK signalling pathway were examined through combined analysis of metabolome and transcriptome. We found glutathione peroxidase 3 (GPX3) is markedly reduced after TGEV infection, and a significant negative correlation between AMPK signalling pathway and TGEV infection. Exogenous addition of the AMPK activator COH-SR4 significantly downregulates stearoyl coenzyme A (SCD1) mRNA and inhibits TGEV replication; while exogenous GSK-690693 significantly promotes TGEV infection by inhibiting AMPK signalling pathway. In summary, our study provides insights into the key metabolites and regulators for TGEV infection from the metabolome and transcriptome perspective, which will offer promising antiviral metabolic and molecular targets and enrich the understanding of the existence of a similar mechanism in the host.


Asunto(s)
Gastroenteritis Porcina Transmisible , Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Proteínas Quinasas Activadas por AMP , Línea Celular , Células Epiteliales , Perfilación de la Expresión Génica , Gastroenteritis Porcina Transmisible/genética
13.
Int J Mol Sci ; 24(21)2023 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-37958953

RESUMEN

Transmissible gastroenteritis virus (TGEV) is an important swine enteric coronavirus causing viral diarrhea in pigs of all ages. Currently, the development of antiviral agents targeting host proteins to combat viral infection has received great attention. The heat shock protein 90 (HSP90) is a critical host factor and has important regulatory effects on the infection of various viruses. However, its roles in porcine coronavirus infection remain unclear. In this study, the effect of HSP90 on TGEV infection was evaluated. In addition, the influence of its inhibitor VER-82576 on proinflammatory cytokine (IL-6, IL-12, TNF-α, CXCL10, and CXCL11) production induced by TGEV infection was further analyzed. The results showed that the knockdown of HSP90AB1 and HSP90 inhibitor VER-82576 treatment resulted in a reduction in TGEV M gene mRNA levels, the N protein level, and virus titers in a dose-dependent manner, while the knockdown of HSP90AA1 and KW-2478 treatment had no significant effect on TGEV infection. A time-of-addition assay indicated that the inhibitory effect of VER-82576 on TGEV infection mainly occurred at the early stage of viral replication. Moreover, the TGEV-induced upregulation of proinflammatory cytokine (IL-6, IL-12, TNF-α, CXCL10, and CXCL11) expression was significantly inhibited by VER-82576. In summary, these findings indicated that HSP90AB1 is a host factor enhancing TGEV infection, and the HSP90 inhibitor VER-82576 could reduce TGEV infection and proinflammatory cytokine production, providing a new perspective for TGEV antiviral drug target design.


Asunto(s)
Gastroenteritis Porcina Transmisible , Virus de la Gastroenteritis Transmisible , Porcinos , Animales , Virus de la Gastroenteritis Transmisible/genética , Gastroenteritis Porcina Transmisible/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Interleucina-6/farmacología , Citocinas/genética , Citocinas/farmacología , Interleucina-12/farmacología
14.
Front Immunol ; 14: 1251001, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37942329

RESUMEN

Introduction: Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are major intestinal coronaviruses that cause vomiting, diarrhea, dehydration, and mortality in piglets. These viruses coexist and lead to significant economic losses in the swine industry. Virus-like particles (VLPs) have emerged as promising alternatives to conventional inactivated vaccines due to their exceptional safety, efficacy, and ability to provide multi-disease protection with a single dose. Methods: Our study focused on specific antigenic epitopes from the PEDV S protein (SS2 and 2C10 regions) and the TGEV S protein (A and D sites) as target candidates. These epitopes were integrated into the ADDomer framework, and we successfully generated recombinant proteins AD, AD-P, AD-T, and AD-PT using the baculovirus expression vector system (BEVS). By meticulously optimizing conditions in High Five cells, we successfully expressed and purified the recombinant proteins. Subsequently, we developed the recombinant ADDomer-VLP vaccine and conducted a comprehensive evaluation of its efficacy in piglets. Results: Following ultrafiltration concentration and sucrose gradient centrifugation purification, the recombinant proteins self-assembled into VLPs as observed by transmission electron microscopy (TEM). Administration of the vaccine did not result in any adverse reactions in the immunized piglets. Additionally, no significant instances of fever were detected in any of the experimental groups, and there were no notable changes in average daily weight gain compared to the control group that received PBS. The recombinant ADDomer-VLP vaccines demonstrated strong immunogenicity, effectively stimulating the production of neutralizing antibodies against both PEDV and TGEV. Moreover, the recombinant ADDomer-VLP vaccine induced elevated levels of IFN-γ, IL-2, and IL-4, and enhanced cytotoxic T lymphocyte (CTL) activity in the peripheral blood of piglets. Discussion: These recombinant VLPs have demonstrated the ability to induce strong cellular and humoral immune responses in piglets, making them an incredibly promising platform for the rapid and simplified development of epitope vaccines.


Asunto(s)
Virus de la Gastroenteritis Transmisible , Vacunas de Partículas Similares a Virus , Animales , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Epítopos , Anticuerpos Antivirales , Vacunas Sintéticas , Inmunidad
15.
Monoclon Antib Immunodiagn Immunother ; 42(5): 178-181, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37855908

RESUMEN

Porcine transmissible gastroenteritis virus (TGEV) infection results in severe gastrointestinal disease manifesting vomiting, diarrhea in neonatal porcine, with extremely high mortality. Monoclonal antibody (MAb) specific to TGEV nonstructural protein (NSP)14 that contains two functional domains, exonuclease (ExoN) and methyltransferase (MTase) domains, may help elucidate the role of NSP14 in the viral life-cycle. In this study, we developed a murine MAb, designated 12F1, against TGEV NSP14 using traditional cell-fusion technique. It was shown the MAb can exclusively bind to viral NSP14, as evidenced by the results of indirect fluorescent assay and western blotting. Intriguingly, epitope screening assay shown that 12F1 targets a hinge region connecting ExoN and N7-MTase of NSP14.


Asunto(s)
Virus de la Gastroenteritis Transmisible , Animales , Porcinos , Ratones , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/metabolismo , Metiltransferasas , Exonucleasas , Anticuerpos Monoclonales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/química , Exones/genética
16.
J Virol ; 97(11): e0120923, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37843366

RESUMEN

IMPORTANCE: Porcine epidemic diarrhea caused by porcine coronaviruses remains a major threat to the global swine industry. Fatty acids are extensively involved in the whole life of the virus. In this study, we found that docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) significantly reduced the viral load of porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), and porcine delta coronavirus (PDCoV) and acted on the replication of the viruses rather than attachment and entry. We further confirmed that DHA and EPA inhibited PEDV replication by alleviating the endoplasmic reticulum stress. Meanwhile, DHA and EPA alleviate PEDV-induced inflammation and reactive oxygen species (ROS) levels and enhance the cellular antioxidant capacity. These data indicate that DHA and EPA have antiviral effects on porcine coronaviruses and provide a molecular basis for the development of new fatty acid-based therapies to control porcine coronavirus infection and transmission.


Asunto(s)
Infecciones por Coronavirus , Coronavirus , Ácidos Docosahexaenoicos , Ácido Eicosapentaenoico , Enfermedades de los Porcinos , Animales , Coronavirus/fisiología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/veterinaria , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/farmacología , Virus de la Diarrea Epidémica Porcina/fisiología , Porcinos , Enfermedades de los Porcinos/tratamiento farmacológico , Virus de la Gastroenteritis Transmisible/fisiología , Replicación Viral/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos
17.
Microb Pathog ; 183: 106320, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37625663

RESUMEN

BACKGROUND: Transmissible gastroenteritis virus (TGEV), which belongs to the coronaviruses (CoVs), causes diarrhea and high mortality rates in piglets and poses a huge threat and loss to the pig industry in China. METHOD: We estimated the prevalence of TGEV in Chinese pig animals from 1983 to 2022 by screening 36 papers on TGEV investigations in China from databases such as China Knowledge Network (CNKI), Wanfang Database, Science and Technology Journal Repository (VIP), PubMed, and ScienceDirect, excluding duplicate literature and other host studies according to the exclusion criteria we developed, and excluding literature with incomplete data to extract information from studies that could estimate the prevalence of TGEV infection in pigs in mainland China. RESULTS: A total of 36 studies (including data from 50,403 pigs) met our evaluation criteria. The overall estimated prevalence of TGEV infection in pigs in China is 10% (3887/50403), and the prevalence of TGEV in northeast China is 38% (2582/3078700) is significantly higher than the rest of China. The prevalence of TGEV infection was related to the sampling season and region. CONCLUSION: The results of the study show that the prevalence of TGEV is clearly seasonal and regional. Therefore, further research and monitoring of the prevalence of TGEV infection and the development of control programs based on different conditions are essential. In addition, effective and robust regulatory measures should be taken in colder regions to prevent the spread and transmission of TGEV in pigs.


Asunto(s)
Gastroenteritis Porcina Transmisible , Virus de la Gastroenteritis Transmisible , Animales , China/epidemiología , Diarrea , Gastroenteritis/epidemiología , Gastroenteritis/veterinaria , Prevalencia , Porcinos , Gastroenteritis Porcina Transmisible/epidemiología , Gastroenteritis Porcina Transmisible/virología
18.
J Virol ; 97(6): e0058923, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37255428

RESUMEN

The inflammasome pathway is a critical early response mechanism of the host that detects pathogens, initiates the production of inflammatory cytokines, and recruits effector cells to the infection site. Nonetheless, the mechanism of inflammasome activation in coronavirus infection and its biological functions in host defense remain unclear. Transmissible gastroenteritis virus (TGEV), a member of the genus Alphacoronavirus, is a significant pathogen that mainly infects piglets and causes intestinal inflammation and inflammatory cell infiltration. Here, we investigated the mechanism of inflammasome activation in intestinal epithelial cells (IECs) infected with TGEV. We observed a substantial increase in interleukin 1ß (IL-1ß) and IL-18 levels in both IECs and TGEV-infected porcine intestinal tissues. Furthermore, TGEV infection resulted in increased activation of caspase-1 and the NLRP1 (NOD-like receptor [NLR]-containing pyrin domain [PYD]) inflammasome. Our findings revealed that TGEV infection impeded the interaction between porcine NLRP1 (pNLRP1) and porcine dipeptidyl peptidases 9 (pDPP9), yet it did not reduce the expression of pDPP9. Importantly, the ZU5 domain, not the function-to-find domain (FIIND) reported in human NLRP1, was identified as the minimal domain of pNLRP1 for pDPP9 binding. In addition, the robust type I IFN expression induced by TGEV infection also upregulated pNLRP1 expression and pNLRP1 itself acts as an interferon-stimulated gene to counteract TGEV infection. Our data demonstrate that pNLRP1 has antiviral capabilities against coronavirus infection, which highlights its potential as a novel therapeutic target for coronavirus antiviral therapy. IMPORTANCE Coronavirus primarily targets the epithelial cells of the respiratory and gastrointestinal tracts, leading to damage in both humans and animals. NLRP1 is a direct sensor for RNA virus infection which is highly expressed in epithelial barrier tissues. However, until recently, the precise molecular mechanisms underlying its activation in coronavirus infection and subsequent downstream events remained unclear. In this study, we demonstrate that the alphacoronavirus TGEV induces the production of IL-1ß and IL-18 and upregulates the expression of pNLRP1. Furthermore, we found that pNLRP1 can serve as an interferon-stimulated gene (ISG) to inhibit the infection of enterovirus TGEV. Our research highlights the crucial role of NLRP1 as a regulator of innate immunity in TGEV infection and shows that it may serve as a potential therapeutic target for the treatment of coronavirus infection.


Asunto(s)
Gastroenteritis Porcina Transmisible , Inflamasomas , Proteínas NLR , Virus de la Gastroenteritis Transmisible , Animales , Inflamasomas/inmunología , Interferón Tipo I , Interleucina-18 , Proteínas NLR/inmunología , Porcinos , Gastroenteritis Porcina Transmisible/inmunología , Gastroenteritis Porcina Transmisible/transmisión
19.
J Virol ; 97(4): e0012823, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36975782

RESUMEN

Coronavirus membrane protein is a major component of the viral envelope and plays a central role in the viral life cycle. Studies of the coronavirus membrane protein (M) have mainly focused on its role in viral assembly and budding, but whether M protein is involved in the initial stage of viral replication remains unclear. In this study, eight proteins in transmissible gastroenteritis virus (TGEV)-infected cells coimmunoprecipitated with monoclonal antibodies (MAb) against M protein in PK-15 cells, heat shock cognate protein 70 (HSC70), and clathrin were identified by matrix-assisted laser desorption ionization-tandem time of flight mass spectrometry (MALDI-TOF MS). Further studies demonstrated that HSC70 and TGEV M colocalized on the cell surface in early stages of TGEV infection; specifically, HSC70 bound M protein through its substrate-binding domain (SBD) and preincubation of TGEV with anti-M serum to block the interaction of M and HSC70 reduced the internalization of TGEV, thus demonstrating that the M-HSC70 interaction mediates the internalization of TGEV. Remarkably, the process of internalization was dependent on clathrin-mediated endocytosis (CME) in PK-15 cells. Furthermore, inhibition of the ATPase activity of HSC70 reduced the efficiency of CME. Collectively, our results indicated that HSC70 is a newly identified host factor involved in TGEV infection. Taken together, our findings clearly illustrate a novel role for TGEV M protein in the viral life cycle and present a unique strategy used by HSC70 to promote TGEV infection in which the interaction with M protein directs viral internalization. These studies provide new insights into the life cycle of coronaviruses. IMPORTANCE TGEV is the causative agent of porcine diarrhea, a viral disease that economically affects the pig industry in many countries. However, the molecular mechanisms underlying viral replication remain incompletely understood. Here, we provide evidence of a previously undescribed role of M protein in viral replication during early stages. We also identified HSC70 as a new host factor affecting TGEV infection. We demonstrate that the interaction between M and HSC70 directs TGEV internalization in a manner dependent on CME, thus revealing a novel mechanism for TGEV replication. We believe that this study may change our understanding of the first steps of infection of cells with coronavirus. This study should facilitate the development of anti-TGEV therapeutic agents by targeting the host factors and may provide a new strategy for the control of porcine diarrhea.


Asunto(s)
Clatrina , Proteínas M de Coronavirus , Endocitosis , Proteínas del Choque Térmico HSC70 , Virus de la Gastroenteritis Transmisible , Internalización del Virus , Virus de la Gastroenteritis Transmisible/fisiología , Clatrina/metabolismo , Proteínas M de Coronavirus/metabolismo , Línea Celular , Humanos , Animales , Replicación Viral
20.
Virus Res ; 329: 199103, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36963723

RESUMEN

A variety of swine enteric coronaviruses (SECoVs) have emerged and are prevalent in pig populations, including porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine deltacoronavirus (PDCoV), and swine acute diarrhea syndrome (SADS)-CoV, a newly identified bat-origin CoV with zoonotic potential. Unfortunately, available traditional, inactivated and attenuated SECoV vaccines are of limited efficacy against the variants currently circulating in most pig populations. In this study, we evaluated the role of host factor heat shock protein 90 (Hsp90) as an antiviral target against SECoVs, exemplified by SADS-CoV. Pharmacological inhibition of Hsp90 diminished SADS-CoV replication significantly in porcine and human cell lines, and also decreased replication of SADS-CoV in a porcine intestinal enteroid model. Further mechanistic experiments revealed that both porcine and human isoforms of Hsp90 interact with the SADS-CoV nucleocapsid (N) protein, and inhibition of Hsp90 resulted in autophagic degradation of N protein. Moreover, we linked Hsp90 to virus-induced cellular pyroptosis, as SADS-CoV was found to trigger caspase-1/gasdermin-d-mediated pyroptotic cell death, which was mitigated by inhibition of Hsp90. Finally, we demonstrated that Hsp90 also associated with N proteins and was involved in propagation of PEDV, PDCoV and TGEV. This study thus extends our understanding of immune responses to SADS-CoV infection and offers a new potential therapeutic option against four SECoVs.


Asunto(s)
Alphacoronavirus , Infecciones por Coronavirus , Virus de la Diarrea Epidémica Porcina , Enfermedades de los Porcinos , Virus de la Gastroenteritis Transmisible , Animales , Humanos , Alphacoronavirus/genética , Antivirales/farmacología , Proteínas de Choque Térmico , Porcinos , Proteínas HSP90 de Choque Térmico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...