Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Gen Virol ; 104(12)2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38116760

RESUMO

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.


Assuntos
Gastroenterite Suína Transmissível , Vírus da Gastroenterite Transmissível , Animais , Suínos , Vírus da Gastroenterite Transmissível/genética , Proteínas Quinases Ativadas por AMP , Linhagem Celular , Células Epiteliais , Perfilação da Expressão Gênica , Gastroenterite Suína Transmissível/genética
2.
Int J Mol Sci ; 24(21)2023 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-37958953

RESUMO

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.


Assuntos
Gastroenterite Suína Transmissível , Vírus da Gastroenterite Transmissível , Suínos , Animais , Vírus da Gastroenterite Transmissível/genética , Gastroenterite Suína Transmissível/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Interleucina-6/farmacologia , Citocinas/genética , Citocinas/farmacologia , Interleucina-12/farmacologia
3.
PLoS Pathog ; 17(12): e1010113, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34871328

RESUMO

Emerging coronaviruses (CoVs) pose a severe threat to human and animal health worldwide. To identify host factors required for CoV infection, we used α-CoV transmissible gastroenteritis virus (TGEV) as a model for genome-scale CRISPR knockout (KO) screening. Transmembrane protein 41B (TMEM41B) was found to be a bona fide host factor involved in infection by CoV and three additional virus families. We found that TMEM41B is critical for the internalization and early-stage replication of TGEV. Notably, our results also showed that cells lacking TMEM41B are unable to form the double-membrane vesicles necessary for TGEV replication, indicating that TMEM41B contributes to the formation of CoV replication organelles. Lastly, our data from a mouse infection model showed that the KO of this factor can strongly inhibit viral infection and delay the progression of a CoV disease. Our study revealed that targeting TMEM41B is a highly promising approach for the development of broad-spectrum anti-viral therapeutics.


Assuntos
Sistemas CRISPR-Cas , Gastroenterite Suína Transmissível/virologia , Interações Hospedeiro-Patógeno , Proteínas de Membrana/fisiologia , Organelas/virologia , Vírus da Gastroenterite Transmissível/fisiologia , Replicação Viral , Animais , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/transmissão , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Suínos
4.
Elife ; 92020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32876563

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) and transmissible gastroenteritis virus (TGEV) are two highly infectious and lethal viruses causing major economic losses to pig production. Here, we report generation of double-gene-knockout (DKO) pigs harboring edited knockout alleles for known receptor proteins CD163 and pAPN and show that DKO pigs are completely resistant to genotype 2 PRRSV and TGEV. We found no differences in meat-production or reproductive-performance traits between wild-type and DKO pigs, but detected increased iron in DKO muscle. Additional infection challenge experiments showed that DKO pigs exhibited decreased susceptibility to porcine deltacoronavirus (PDCoV), thus offering unprecedented in vivo evidence of pAPN as one of PDCoV receptors. Beyond showing that multiple gene edits can be combined in a livestock animal to achieve simultaneous resistance to two major viruses, our study introduces a valuable model for investigating infection mechanisms of porcine pathogenic viruses that exploit pAPN or CD163 for entry.


Pig epidemics are the biggest threat to the pork industry. In 2019 alone, hundreds of billions of dollars worldwide were lost due to various pig diseases, many of them caused by viruses. The porcine reproductive and respiratory virus (PRRS virus for short), for instance, leads to reproductive disorders such as stillbirths and premature labor. Two coronaviruses ­ the transmissible gastroenteritis virus (or TGEV) and the porcine delta coronavirus ­ cause deadly diarrhea and could potentially cross over into humans. Unfortunately, there are still no safe and effective methods to prevent or control these pig illnesses, but growing disease-resistant pigs could reduce both financial and animal losses. Traditionally, breeding pigs to have a particular trait is a slow process that can take many years. But with gene editing technology, it is possible to change or remove specific genes in a single generation of animals. When viruses infect a host, they use certain proteins on the surface of the host's cells to find their inside: the PRRS virus relies a protein called CD163, and TGEV uses pAPN. Xu, Zhou, Mu et al. used gene editing technology to delete the genes that encode the CD163 and pAPN proteins in pigs. When the animals were infected with PRRS virus or TGEV, the non-edited pigs got sick but the gene-edited animals remained healthy. Unexpectedly, pigs without CD163 and pAPN also coped better with porcine delta coronavirus infections, suggesting that CD163 and pAPN may also help this coronavirus infect cells. Finally, the gene-edited pigs reproduced and produced meat as well as the control pigs. These experiments show that gene editing can be a powerful technology for producing animals with desirable traits. The gene-edited pigs also provide new knowledge about how porcine viruses infect pigs, and may offer a starting point to breed disease-resistant animals on a larger scale.


Assuntos
Antígenos CD13/deficiência , Infecções por Coronavirus/prevenção & controle , Coronavirus/patogenicidade , Gastroenterite Suína Transmissível/prevenção & controle , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Receptores de Superfície Celular/deficiência , Vírus da Gastroenterite Transmissível/patogenicidade , Animais , Animais Geneticamente Modificados , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/imunologia , Composição Corporal , Antígenos CD13/genética , Antígenos CD13/imunologia , Coronavirus/imunologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Suscetibilidade a Doenças , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/virologia , Técnicas de Silenciamento de Genes , Interações entre Hospedeiro e Microrganismos , Indústria de Embalagem de Carne , Fenótipo , Síndrome Respiratória e Reprodutiva Suína/genética , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Sus scrofa/genética , Suínos , Vírus da Gastroenterite Transmissível/imunologia , Aumento de Peso
5.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32796075

RESUMO

The intestinal organoid culture system is a pathbreaking working model for investigating pathogen-host interactions in the intestines. However, due to the limitations of the first generation of intestinal organoids, basal-out structure and growth in Matrigel, most pathogens can rarely attach to the apical membrane directly and hardly initiate infection. In this study, we first developed a next-generation porcine intestinal organoid culture system, characterized by an apical membrane on the surface, called apical-out. To investigate the infectivity and antiviral immune responses of this apical-out porcine intestinal organoid, a swine enteric virus, transmissible gastroenteritis virus (TGEV), was employed to inoculate the culture system. Both reverse transcription-quantitative PCR (RT-qPCR) and immunofluorescence assay (IFA) analysis demonstrated that TGEV replicated in the apical-out porcine intestinal organoid culture system. Additionally, our results illustrated that TGEV infection significantly upregulated the expression levels of alpha interferon (IFN-α), IFN-λ1, interferon-stimulated gene 15 (ISG15), ISG58, tumor necrosis factor alpha (TNF-α), and interleukin 6 (IL-6) in this culture system. Hence, we successfully developed a porcine intestinal apical-out organoid culture system, which will facilitate the investigation of pathogen-host interactions in pig intestines.IMPORTANCE Intestinal organoids are a newly developed culture system for investigating pathogen-host interactions. Intestinal organoid models have been widely used since their development, because the results obtained from this type of culture model better represent physiological conditions than those from well-established cell lines. The three-dimensional (3D) porcine intestinal organoid model was reported in 2018 and 2019 for the investigation of intestinal pathogens. However, those organoid culture models were basal-out intestinal organoids, which are not suitable for porcine enteric virus research because they invade the intestines via the apical side of epithelial cells on villi. In this study, we developed a porcine apical-out intestinal organoid culture system and verified its infectivity, type I and type III interferon (IFN) antiviral responses, and inflammatory responses following infection by a swine enteric virus. Our results imply that this apical-out porcine intestinal organoid culture system is an ideal model for the investigation of interactions between swine enteric viruses and the intestines.


Assuntos
Células Epiteliais/imunologia , Gastroenterite Suína Transmissível/imunologia , Interações Hospedeiro-Patógeno/imunologia , Mucosa Intestinal/imunologia , Organoides/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Animais , Bioensaio , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferons/genética , Interferons/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Organoides/patologia , Organoides/virologia , Suínos , Vírus da Gastroenterite Transmissível/crescimento & desenvolvimento , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Ubiquitinas/genética , Ubiquitinas/imunologia , Replicação Viral
6.
J Microbiol Biotechnol ; 30(4): 515-525, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-31838830

RESUMO

Interferon (IFN)-λ plays an essential role in mucosal cells which exhibit strong antiviral activity. Lactobacillus plantarum (L. plantarum) has substantial application potential in the food and medical industries because of its probiotic properties. Alphacoronaviruses, especially porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV), cause high morbidity and mortality in piglets resulting in economic loss. Co-infection by these two viruses is becoming increasingly frequent. Therefore, it is particularly important to develop a new drug to prevent diarrhea infected with mixed viruses in piglets. In this study, we first constructed an anchored expression vector with CWA (C-terminal cell wall anchor) on L. plantarum. Second, we constructed two recombinant L. plantarum strains that anchored IFN-λ3 via pgsA (N-terminal transmembrane anchor) and CWA. Third, we demonstrated that both recombinant strains possess strong antiviral effects against coronavirus infection in the intestinal porcine epithelial cell line J2 (IPEC-J2). However, recombinant L. plantarum with the CWA anchor exhibited a more powerful antiviral effect than recombinant L. plantarum with pgsA. Consistent with this finding, Lb.plantarum-pSIP-409-IFN-λ3-CWA enhanced the expression levels of IFN-stimulated genes (ISGs) (ISG15, OASL, and Mx1) in IPEC-J2 cells more than did recombinant Lb.plantarum-pSIP-409-pgsA'-IFN-λ3. Our study verifies that recombinant L. plantarum inhibits PEDV and TGEV infection in IPEC-J2 cells, which may offer great potential for use as a novel oral antiviral agent in therapeutic applications for combating porcine epidemic diarrhea and transmissible gastroenteritis. This study is the first to show that recombinant L. plantarum suppresses PEDV and TGEV infection of IPEC-J2 cells.


Assuntos
Infecções por Coronavirus/veterinária , Gastroenterite Suína Transmissível/prevenção & controle , Interferons/administração & dosagem , Lactobacillus plantarum/genética , Doenças dos Suínos/prevenção & controle , Animais , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Feminino , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/virologia , Expressão Gênica , Interferons/genética , Interferons/imunologia , Lactobacillus plantarum/metabolismo , Masculino , Vírus da Diarreia Epidêmica Suína/fisiologia , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Vírus da Gastroenterite Transmissível/fisiologia
7.
BMC Genomics ; 20(1): 806, 2019 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-31684870

RESUMO

BACKGROUND: Transmissible gastroenteritis virus (TGEV) infection can cause acute inflammation. Long noncoding RNAs (lncRNAs) play important roles in a number of biological process including inflammation response. However, whether lncRNAs participate in TGEV-induced inflammation in porcine intestinal epithelial cells (IPECs) is largely unknown. RESULTS: In this study, the next-generation sequencing (NGS) technology was used to analyze the profiles of lncRNAs in Mock and TGEV-infected porcine intestinal epithelial cell-jejunum 2 (IPEC-J2) cell line. A total of 106 lncRNAs were differentially expressed. Many differentially expressed lncRNAs act as elements to competitively attach microRNAs (miRNAs) which target to messenger RNA (mRNAs) to mediate expression of genes that related to toll-like receptors (TLRs), NOD-like receptors (NLRs), tumor necrosis factor (TNF), and RIG-I-like receptors (RLRs) pathways. Functional analysis of the binding proteins and the up/down-stream genes of the differentially expressed lncRNAs revealed that lncRNAs were principally related to inflammatory response. Meanwhile, we found that the differentially expressed lncRNA TCONS_00058367 might lead to a reduction of phosphorylation of transcription factor p65 (p-p65) in TGEV-infected IPEC-J2 cells by negatively regulating its antisense gene promyelocytic leukemia (PML). CONCLUSIONS: The data showed that differentially expressed lncRNAs might be involved in inflammatory response induced by TGEV through acting as miRNA sponges, regulating their up/down-stream genes, or directly binding proteins.


Assuntos
Gastroenterite Suína Transmissível/genética , RNA Longo não Codificante/genética , Vírus da Gastroenterite Transmissível/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Sequenciamento de Nucleotídeos em Larga Escala , Inflamação/genética , MicroRNAs/genética , Suínos
8.
Sci Rep ; 9(1): 13186, 2019 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515498

RESUMO

Swine enteric diseases have caused significant economic loss and have been considered as the major threat to the global swine industry. Several coronaviruses, including transmissible gastroenteritis virus (TGEV) and porcine epidemic diarrhea virus (PEDV), have been identified as the causative agents of these diseases. Effective measures to control these diseases are lacking. The major host cells of transmissible gastroenteritis virus and porcine epidemic diarrhea virus have thought to be epithelial cells on small intestine villi. Aminopeptidase-N (APN) has been described as the putative receptor for entry of transmissible gastroenteritis virus and porcine epidemic diarrhea virus into cells in vitro. Recently, Whitworth et al. have reported that APN knockout pigs are resistant to TGEV but not PEDV after weaning. However, it remains unclear if APN-null neonatal pigs are protected from TGEV. Here we report the generation of APN-null pigs by using CRISPR/Cas9 technology followed by somatic cell nuclear transfer. APN-null pigs are produced with normal pregnancy rate and viability, indicating lack of APN is not embryonic lethal. After viral challenge, APN-null neonatal piglets are resistant to highly virulent transmissible gastroenteritis virus. Histopathological analyses indicate APN-null pigs exhibit normal small intestine villi, while wildtype pigs show typical lesions in small intestines. Immunochemistry analyses confirm that no transmissible gastroenteritis virus antigen is detected in target tissues in APN-null piglets. However, upon porcine epidemic diarrhea virus challenge, APN-null pigs are still susceptible with 100% mortality. Collectively, this report provides a viable tool for producing animals with enhanced resistance to TGEV and clarifies that APN is dispensable for the PEDV infection in pigs.


Assuntos
Animais Geneticamente Modificados , Antígenos CD13/deficiência , Infecções por Coronavirus , Gastroenterite Suína Transmissível , Vírus da Diarreia Epidêmica Suína/metabolismo , Suínos , Vírus da Gastroenterite Transmissível/metabolismo , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Animais Geneticamente Modificados/virologia , Antígenos CD13/metabolismo , Infecções por Coronavirus/enzimologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/virologia , Gastroenterite Suína Transmissível/enzimologia , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/prevenção & controle , Gastroenterite Suína Transmissível/virologia , Vírus da Diarreia Epidêmica Suína/genética , Suínos/genética , Suínos/metabolismo , Suínos/virologia , Vírus da Gastroenterite Transmissível/genética
9.
Mol Cell Proteomics ; 18(1): 51-64, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30257878

RESUMO

Transmissible gastroenteritis virus (TGEV) is a member of Coronaviridae family. Our previous research showed that TGEV infection could induce mitochondrial dysfunction and upregulate miR-222 level. Therefore, we presumed that miR-222 might be implicated in regulating mitochondrial dysfunction induced by TGEV infection. To verify the hypothesis, the effect of miR-222 on mitochondrial dysfunction was tested and we showed that miR-222 attenuated TGEV-induced mitochondrial dysfunction. To investigate the underlying molecular mechanism of miR-222 in TGEV-induced mitochondrial dysfunction, a quantitative proteomic analysis of PK-15 cells that were transfected with miR-222 mimics and infected with TGEV was performed. In total, 4151 proteins were quantified and 100 differentially expressed proteins were obtained (57 upregulated, 43 downregulated), among which thrombospondin-1 (THBS1) and cluster of differentiation 47 (CD47) were downregulated. THBS1 was identified as the target of miR-222. Knockdown of THBS1 and CD47 decreased mitochondrial Ca2+ level and increased mitochondrial membrane potential (MMP) level. Reversely, overexpression of THBS1 and CD47 elevated mitochondrial Ca2+ level and reduced mitochondrial membrane potential (MMP) level. Together, our data establish a significant role of miR-222 in regulating mitochondrial dysfunction in response to TGEV infection.


Assuntos
Antígeno CD47/metabolismo , Gastroenterite Suína Transmissível/metabolismo , MicroRNAs/genética , Mitocôndrias/metabolismo , Trombospondina 1/metabolismo , Vírus da Gastroenterite Transmissível/patogenicidade , Animais , Antígeno CD47/genética , Cálcio/metabolismo , Linhagem Celular , Gastroenterite Suína Transmissível/genética , Regulação da Expressão Gênica , Potencial da Membrana Mitocondrial , Mapas de Interação de Proteínas , Proteômica/métodos , Suínos , Trombospondina 1/genética , Transfecção
10.
Vet Res ; 49(1): 95, 2018 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-30236161

RESUMO

Transmissible gastroenteritis virus (TGEV) is a coronavirus that causes severe diarrhea in suckling piglets. TGEV primarily targets and infects porcine intestinal epithelial cells, which play an important role in nutrient absorption. However, the effects of TGEV infection on nutrient absorption in swine have not yet been investigated. In this study, we evaluated the impact of TGEV infection on arginine uptake using the porcine small intestinal epithelial cell line IPEC-J2 as a model system. High performance liquid chromatography (HPLC) analyses showed that TGEV infection leads to reduced arginine uptake at 48 hours post-infection (hpi). Expression of cationic amino acid transporter 1 (CAT-1) was attenuated as well. TGEV infection induced activation of phospho-protein kinase C α (p-PKC α), phospho-epidermal growth factor receptor (p-EGFR), and enhanced the expression of caveolin-1, all of which appear to be involved in down-regulating arginine uptake and CAT-1 expression. These results illuminate the relationship between TGEV infection and nutrient absorption, and further our understanding of the mechanisms of TGEV infection.


Assuntos
Arginina/metabolismo , Transportador 1 de Aminoácidos Catiônicos/genética , Regulação para Baixo , Gastroenterite Suína Transmissível/genética , Regulação da Expressão Gênica , Vírus da Gastroenterite Transmissível/fisiologia , Animais , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Linhagem Celular , Gastroenterite Suína Transmissível/fisiopatologia , Gastroenterite Suína Transmissível/virologia , Intestino Delgado/metabolismo , Intestino Delgado/virologia , Transdução de Sinais , Suínos
11.
Virology ; 507: 170-178, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28448848

RESUMO

Transmissible gastroenteritis virus (TGEV) is a porcine enteric coronavirus which causes lethal severe watery diarrhea in piglets. The pathogenesis of TGEV is strongly associated with inflammation. In this study, we found that TGEV infection activates transcription factors NF-κB, IRF3 and AP-1 in a time- and dose-dependent manner in porcine kidney cells. Treatment with the NF-κB-specific inhibitor BAY11-7082 significantly decreased TGEV-induced proinflammatory cytokine production, but did not affect virus replication. Phosphorylation of NF-κB subunit p65 and proinflammatory cytokine production were greatly decreased after knockdown of retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) or its adaptors MAVS and STING, while only slight reduction was observed in cells following silencing of Toll-like receptor adaptors, MyD88 and TRIF. Furthermore, TGEV infection significantly upregulated mRNA expression of RIG-I and MDA5. Taken together, our results indicate that the RLR signaling pathway is involved in TGEV-induced inflammatory responses.


Assuntos
Gastroenterite Suína Transmissível/imunologia , Gastroenterite Suína Transmissível/virologia , NF-kappa B/imunologia , Vírus da Gastroenterite Transmissível/fisiologia , Animais , Citocinas/genética , Citocinas/imunologia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Gastroenterite Suína Transmissível/genética , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , NF-kappa B/genética , Fosforilação , Transdução de Sinais , Suínos , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/imunologia , Vírus da Gastroenterite Transmissível/genética
12.
Virus Genes ; 53(2): 226-232, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27848068

RESUMO

The membrane (M) protein is the most abundant component of the porcine transmissible gastroenteritis virus (TGEV) particle. To exploit the possibility of using RNA interference (RNAi) as a strategy against TGEV infection, three plasmids (pRNAT-1, pRNAT-2, and pRNAT-3) expressing short hairpin RNAs were designed to target three different coding regions of the M gene of TGEV. The plasmids were constructed and transiently transfected into a porcine kidney cells, PK-15, to determine whether these constructs inhibited TGEV production. The analysis of cytopathic effects demonstrated that pRNAT-2 and pRNAT-3 could protect PK-15 cells against pathological changes specifically and efficiently. Additionally, indirect immunofluorescence and 50% tissue culture infectious dose (TCID50) assays showed that pRNAT-2 and pRNAT-3 inhibited the multiplication of the virus at the protein level effectively. Quantitative real-time PCR further confirmed that the amounts of viral RNAs in cell cultures pre-transfected with the three plasmids were reduced by 13, 68, and 70%, respectively. This is the first report showing that RNAi targeting of the M gene. Our results could promote studies of the specific function of viral genes associated with TGEV infection and might provide a theoretical basis for potential therapeutic applications.


Assuntos
Gastroenterite Suína Transmissível/genética , Proteínas de Membrana/genética , RNA Interferente Pequeno/genética , Vírus da Gastroenterite Transmissível/genética , Animais , Gastroenterite Suína Transmissível/terapia , Gastroenterite Suína Transmissível/virologia , Rim/patologia , Rim/virologia , Proteínas de Membrana/antagonistas & inibidores , Interferência de RNA , Suínos/genética , Suínos/virologia , Vírus da Gastroenterite Transmissível/patogenicidade , Replicação Viral/genética
13.
Sci Rep ; 6: 32154, 2016 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-27555521

RESUMO

It has been well characterized that the neonatal Fc receptor (FcRn) transports maternal IgG to a fetus or newborn and protects IgG from degradation. We previously reported that FcRn is expressed in a model of normal porcine intestinal epithelial cells (IPEC-J2). Transmissible gastroenteritis is an acute enteric disease of swine that is caused by transmissible gastroenteritis virus (TGEV). How porcine FcRn (pFcRn) expression is regulated by pathogenic infection remains unknown. Our research shows that IPEC-J2 cells infected with TGEV had up-regulated pFcRn expression. In addition, the NF-κB signaling pathway was activated in IPEC-J2 cells by TGEV infection. Furthermore, treatment of TGEV-infected IPEC-J2 cells with the NF-κB-specific inhibitor BAY 11-7082 resulted in down-regulation of pFcRn expression. Transient transfection of pFcRn promoter luciferase report plasmids with overexpression of NF-κB p65 transcription factor enhanced the activation of the luciferase report plasmids. We identified four NF-κB transcription factor binding sites in the promoter region of this gene using luciferase reporter system, chromatin immunoprecipitation, electromobility shift assay, and supershift analysis. Together, the data provide the first evidence that TGEV infection up-regulates pFcRn expression via activation of NF-κB signaling.


Assuntos
Gastroenterite Suína Transmissível/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , NF-kappa B/metabolismo , Receptores Fc/genética , Animais , Sítios de Ligação , Linhagem Celular , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/imunologia , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Nitrilas/farmacologia , Regiões Promotoras Genéticas , Receptores Fc/imunologia , Receptores Fc/metabolismo , Transdução de Sinais , Sulfonas/farmacologia , Suínos , Vírus da Gastroenterite Transmissível/patogenicidade
14.
Biomed Res Int ; 2016: 7590569, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27294133

RESUMO

Porcine kobuvirus (PKV) has circulated throughout China in recent years. Although many studies have detected it throughout the world, its molecular epidemiology has not been characterized in northwest China. To understand its prevalence, 203 fecal samples were collected from different regions of Gansu Province and tested with reverse transcription-polymerase chain reaction. In this study, we tested these samples for PKV, porcine epidemic diarrhea virus (PEDV), and sapovirus and analyzed the amplified 2C gene fragments of PKV. Overall, 126 (62.1%) samples were positive for PKV. Of the 74 piglets samples among the 203 fecal samples, 65 (87.8%) were positive for PKV. PKV infection was often accompanied by PEDV, but the relationship between the two viruses must be confirmed. A phylogenetic analysis indicated that the PKV strains isolated from the same regions clustered on the same branches. This investigation shows that PKV infections are highly prevalent in pigs in northwest China, especially in piglets with symptoms of diarrhea.


Assuntos
Coinfecção , Gastroenterite Suína Transmissível , Kobuvirus/genética , Vírus da Diarreia Epidêmica Suína , RNA Viral , Sapovirus/genética , Exantema Vesicular de Suínos , Animais , China/epidemiologia , Coinfecção/sangue , Coinfecção/epidemiologia , Coinfecção/genética , Coinfecção/virologia , Gastroenterite Suína Transmissível/sangue , Gastroenterite Suína Transmissível/epidemiologia , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/virologia , Epidemiologia Molecular , Infecções por Picornaviridae/sangue , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/virologia , RNA Viral/sangue , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Suínos , Exantema Vesicular de Suínos/sangue , Exantema Vesicular de Suínos/epidemiologia , Exantema Vesicular de Suínos/genética , Exantema Vesicular de Suínos/virologia
15.
Int J Biol Sci ; 11(8): 913-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26157346

RESUMO

Transmissible gastroenteritis virus (TGEV) is a member of Coronaviridae family. TGEV infection has emerged as a major cause of severe gastroenteritis and leads to alterations of many cellular processes. Meanwhile, the pathogenic mechanism of TGEV is still unclear. microRNAs (miRNAs) are a novel class of small non-coding RNAs which are involved in the regulation of numerous biological processes such as viral infection and cell apoptosis. Accumulating data show that miRNAs are involved in the process of coronavirus infection such as replication of severe acute respiratory syndrome coronavirus (SARS-CoV). However, the link between miRNAs and TGEV infection is unknown. In this study, we performed microRNA microarray assay and predicted targets of altered miRNAs. The results showed TGEV infection caused the change of miRNAs profile. Then we selected miR-4331 for further analysis and subsequently identified cell division cycle-associated protein 7 (CDCA7) as the target of miR-4331. Moreover, miR-4331 showed the ability to inhibit transcription of TGEV gene 7 (a non-structure gene) via directly targeting CDCA7. In conclusion, differentially expressed miR-4331 that is caused by TGEV infection can suppress transcription of TGEV gene 7 via targeting cellular CDCA7. Our key finding is that TGEV selectively manipulates the expression of some cellular miRNAs to regulate its subgenomic transcription.


Assuntos
Gastroenterite Suína Transmissível/genética , Genes Virais , MicroRNAs/genética , Transcrição Gênica/genética , Vírus da Gastroenterite Transmissível/genética , Animais , Reação em Cadeia da Polimerase em Tempo Real , Suínos
16.
Virus Res ; 200: 56-63, 2015 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-25533531

RESUMO

Nucleocapsid (N) protein of transmissible gastroenteritis virus (TGEV) packages viral RNA genome to form a ribonucleoprotein complex. In addition to its function as a structural protein, N protein is involved in cell apoptosis or cell-cycle regulation. N protein possibly interacts with host factors to modulate cellular functions. To identify cellular proteins that interacted with N protein of TGEV, methods of GST pull-down and Co-IP were utilized to precipitate cellular proteins of swine testicular (ST). Bound cellular proteins were resolved by SDS-PAGE. Analysis of interacting proteins by mass spectrometry allowed identification of 15 cellular protein bands representative of 12 cellular proteins including vimentin that bound to N protein. Furthermore, the function of vimentin cytoskeleton in ST cells during TGEV infection was examined. Vimentin cytoskeleton was required for virus replication. The present study thus provides protein-related information about interaction of TGEV N protein with host cell that should be useful for understanding host cell response to coronavirus pathogenesis infection and the underlying mechanism of coronavirus replication.


Assuntos
Gastroenterite Suína Transmissível/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Vimentina/metabolismo , Animais , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/virologia , Proteínas do Nucleocapsídeo/genética , Ligação Proteica , Suínos , Vírus da Gastroenterite Transmissível/genética , Vimentina/genética
17.
PLoS One ; 9(10): e110647, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25333634

RESUMO

The interactions occurring between a virus and a host cell during a viral infection are complex. The purpose of this paper was to analyze altered cellular protein levels in porcine transmissible gastroenteritis coronavirus (TGEV)-infected swine testicular (ST) cells in order to determine potential virus-host interactions. A proteomic approach using isobaric tags for relative and absolute quantitation (iTRAQ)-coupled two-dimensional liquid chromatography-tandem mass spectrometry identification was conducted on the TGEV-infected ST cells. The results showed that the 4-plex iTRAQ-based quantitative approach identified 4,112 proteins, 146 of which showed significant changes in expression 48 h after infection. At 64 h post infection, 219 of these proteins showed significant change, further indicating that a larger number of proteomic changes appear to occur during the later stages of infection. Gene ontology analysis of the altered proteins showed enrichment in multiple biological processes, including cell adhesion, response to stress, generation of precursor metabolites and energy, cell motility, protein complex assembly, growth, developmental maturation, immune system process, extracellular matrix organization, locomotion, cell-cell signaling, neurological system process, and cell junction organization. Changes in the expression levels of transforming growth factor beta 1 (TGF-ß1), caspase-8, and heat shock protein 90 alpha (HSP90α) were also verified by western blot analysis. To our knowledge, this study is the first time the response profile of ST host cells following TGEV infection has been analyzed using iTRAQ technology, and our description of the late proteomic changes that are occurring after the time of vigorous viral production are novel. Therefore, this study provides a solid foundation for further investigation, and will likely help us to better understand the mechanisms of TGEV infection and pathogenesis.


Assuntos
Gastroenterite Suína Transmissível/genética , Proteoma/genética , Testículo/metabolismo , Animais , Linhagem Celular , Cromatografia Líquida , Gastroenterite Suína Transmissível/metabolismo , Gastroenterite Suína Transmissível/patologia , Gastroenterite Suína Transmissível/virologia , Regulação Viral da Expressão Gênica , Masculino , Suínos , Testículo/patologia , Testículo/virologia , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/patogenicidade
18.
Biochem Biophys Res Commun ; 445(2): 497-503, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24548406

RESUMO

Our previous studies showed that TGEV infection could induce cell cycle arrest and apoptosis via activation of p53 signaling in cultured host cells. However, it is unclear which viral gene causes these effects. In this study, we investigated the effects of TGEV nucleocapsid (N) protein on PK-15 cells. We found that TGEV N protein suppressed cell proliferation by causing cell cycle arrest at the S and G2/M phases and apoptosis. Characterization of various cellular proteins that are involved in regulating cell cycle progression demonstrated that the expression of N gene resulted in an accumulation of p53 and p21, which suppressed cyclin B1, cdc2 and cdk2 expression. Moreover, the expression of TGEV N gene promoted translocation of Bax to mitochondria, which in turn caused the release of cytochrome c, followed by activation of caspase-3, resulting in cell apoptosis in the transfected PK-15 cells following cell cycle arrest. Further studies showed that p53 inhibitor attenuated TGEV N protein induced cell cycle arrest at S and G2/M phases and apoptosis through reversing the expression changes of cdc2, cdk2 and cyclin B1 and the translocation changes of Bax and cytochrome c induced by TGEV N protein. Taken together, these results demonstrated that TGEV N protein might play an important role in TGEV infection-induced p53 activation and cell cycle arrest at the S and G2/M phases and apoptosis occurrence.


Assuntos
Apoptose , Gastroenterite Suína Transmissível/metabolismo , Interações Hospedeiro-Patógeno , Proteínas do Nucleocapsídeo/metabolismo , Suínos/virologia , Vírus da Gastroenterite Transmissível/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/virologia , Expressão Gênica , Proteínas do Nucleocapsídeo/genética
19.
Hybridoma (Larchmt) ; 29(4): 345-50, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20715993

RESUMO

Transmissible gastroenteritis virus (TGEV) is a member of coronaviruses. The viral spike (S) protein mediates the interaction between TGEV and its susceptible cells. Here, we expressed a truncated gene encoding the N terminal half of TGEV S gene (designated S1 gene) in a prokaryotic system. The resulting S1 protein was used to immunize BALB/c mice followed by the generation of a monoclonal antibody (MAb). A generated MAb (7F9) was identified by ELISA and the chromosome number of the hybridoma cell was analyzed. The immunoreactivity of the MAb to TGEV S protein was confirmed by Western blot analysis. Moreover, immunofluorescence assays showed that the MAb is able to detect cell infection by TGEV. The MAb achieved in this study can be used as a specific diagnostic reagent for detecting TGEV S protein.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Gastroenterite Suína Transmissível/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas Recombinantes/imunologia , Vírus da Gastroenterite Transmissível/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Especificidade de Anticorpos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Gastroenterite Suína Transmissível/genética , Gastroenterite Suína Transmissível/metabolismo , Hibridomas , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Glicoproteína da Espícula de Coronavírus , Suínos , Testículo/citologia , Testículo/metabolismo , Vírus da Gastroenterite Transmissível/genética
20.
J Virol Methods ; 133(1): 27-33, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16300838

RESUMO

Porcine epidemic diarrhea virus (PEDV), a member of the family Coronaviridae, has caused a devastating enteric disease in the Korean swine industry. Previously, the differences between virulent field PEDV strains and a Vero cell culture adapted PEDV DR13 strain were determined using restriction fragment length polymorphism analysis (RFLP), and PEDV shedding patterns in pigs were reported. In an extension to these studies, an internal control was constructed and quantitative analysis of virus shedding after oral inoculation was established. A parent field PEDV and a cell culture adapted PEDV DR13 were inoculated orally to colostrum-deprived 1-day-old piglets, commercial 2-week-old pigs, and sows (1-5 ml dose, 10(5.8)-10(6.0) TCID(50)/0.1 ml). PEDV shedding was monitored every day and virus levels were measured using a quantitative reverse transcriptase polymerase chain reaction (RT-PCR) method. In fecal samples from experimentally-inoculated pigs, the level of virus excreted peaked at 2 days after oral inoculation and gradually decreased thereafter. In addition, PEDV from field specimens was quantified using the same RT-PCR assay to determine shedding viral load. This suggests that measurement of PEDV shedding viral load in pigs, by quantitative RT-PCR, may be a useful tool for estimating the transmission potential of PEDV in the swine population.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/isolamento & purificação , Gastroenterite Suína Transmissível/virologia , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Animais , Chlorocebus aethiops , Coronaviridae/genética , Infecções por Coronaviridae/diagnóstico , Infecções por Coronaviridae/virologia , DNA Complementar , Fezes/virologia , Gastroenterite Suína Transmissível/diagnóstico , Gastroenterite Suína Transmissível/genética , Polimorfismo de Fragmento de Restrição , RNA Viral/genética , Padrões de Referência , Suínos , Doenças dos Suínos/diagnóstico , Doenças dos Suínos/virologia , Células Vero , Carga Viral , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...