Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Monoclon Antib Immunodiagn Immunother ; 42(5): 178-181, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37855908

RESUMO

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.


Assuntos
Vírus da Gastroenterite Transmissível , Animais , Suínos , Camundongos , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/metabolismo , Metiltransferases , Exonucleases , Anticorpos Monoclonais , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/química , Éxons/genética
2.
J Virol ; 96(18): e0096222, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36073923

RESUMO

Intestinal stem cells (ISCs) play an important role in tissue repair after injury. A recent report delineates the effect of transmissible gastroenteritis virus (TGEV) infection on the small intestine of recovered pigs. However, the mechanism behind the epithelium regeneration upon TGEV infection remains unclear. To address this, we established a TGEV infection model based on the porcine intestinal organoid monolayer. The results illustrated that the porcine intestinal organoid monolayer was susceptible to TGEV. In addition, the TGEV infection initiated the interferon and inflammatory responses following the loss of absorptive enterocytes and goblet cells. However, TGEV infection did not disturb epithelial integrity but induced the proliferation of ISCs. Furthermore, TGEV infection activated the Wnt/ß-catenin pathway by upregulating the accumulation and nuclear translocation of ß-catenin, as well as promoting the expression of Wnt target genes, such as C-myc, Cyclin D1, Mmp7, Lgr5, and Sox9, which were associated with the self-renewal of ISCs. Collectively, these data demonstrated that the TGEV infection activated the Wnt/ß-catenin pathway to promote the self-renewal of ISCs and resulted in intestinal epithelium regeneration. IMPORTANCE The intestinal epithelium is a physical barrier to enteric viruses and commensal bacteria. It plays an essential role in maintaining the balance between the host and intestinal microenvironment. In addition, intestinal stem cells (ISCs) are responsible for tissue repair after injury. Therefore, prompt self-renewal of intestinal epithelium will facilitate the rebuilding of the physical barrier and maintain gut health. In the manuscript, we found that the transmissible gastroenteritis virus (TGEV) infection did not disturb epithelial integrity but induced the proliferation of ISCs and facilitated epithelium regeneration. Detailed mechanism investigations revealed that the TGEV infection activated the Wnt/ß-catenin pathway to promote the self-renewal of ISCs and resulted in intestinal epithelium regeneration. These findings will contribute to understanding the mechanism of intestinal epithelial regeneration and reparation upon viral infection.


Assuntos
Células-Tronco , Vírus da Gastroenterite Transmissível , Animais , Ciclina D1/metabolismo , Interferons/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/virologia , Metaloproteinase 7 da Matriz , Células-Tronco/citologia , Células-Tronco/virologia , Suínos , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
3.
Vet Microbiol ; 272: 109497, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35785658

RESUMO

Transmissible gastroenteritis virus (TGEV) infection can lead to mitochondrial damage in porcine intestinal epithelial cells-jejunum 2 (IPEC-J2) cell line. The abnormal opening of mitochondrial permeability transition pore (mPTP) is the most important factor for mitochondrial damage. We previously demonstrated that circEZH2 could inhibit the abnormal opening of mPTP by binding miR-22. However, circEZH2 binding to miR-22 cannot completely enable mPTP opening to recover to normal level compared with the control group. So, we assume that circEZH2 also regulates the mPTP opening in other ways. To prove it, we identified the differentially expressed proteins (DEPs) caused by circEZH2 and circEZH2-interacting proteins by liquid chromatography-tandem mass spectrometry (LC-MS/MS). It turns out there are 123 DEPs (0.83 ≤ fold change ≥ 1.2) upon overexpression circEZH2 and 200 proteins interacted with circEZH2. The kyoto encyclopedia of genes and genomes (KEGG) analysis, gene ontology (GO) analysis, subcellular localization analysis, and protein interaction network results show that the DEPs and circEZH2-interacting proteins may involve in the regulation of mPTP opening. RNA immunoprecipitation (RIP) assay and flow cytometry (FCM) results indicate that circEZH2 can inhibit the opening of mPTP by interacting with Pi carrier (PiC, also named SLC25A3). Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, and FCM results reveal that circEZH2 can inhibit mPTP opening by promoting the expression of radical s-adenosyl methionine domain-containing protein 2 (RSAD2). In addition, PiC can promote RSAD2 expression. The data indicate that circEZH2 inhibits TGEV-induced mPTP opening by interacting with PiC and upregulating RSAD2.


Assuntos
MicroRNAs , Vírus da Gastroenterite Transmissível , Animais , Cromatografia Líquida/veterinária , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Suínos , Espectrometria de Massas em Tandem/veterinária , Vírus da Gastroenterite Transmissível/metabolismo
4.
Cells ; 11(11)2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35681454

RESUMO

Effective airborne transmission of coronaviruses via liquid microdroplets requires a virion structure that must withstand harsh environmental conditions. Due to the demanding biosafety requirements for the study of human respiratory viruses, it is important to develop surrogate models to facilitate their investigation. Here we explore the mechanical properties and nanostructure of transmissible gastroenteritis virus (TGEV) virions in liquid milieu and their response to different chemical agents commonly used as biocides. Our data provide two-fold results on virus stability: First, while particles with larger size and lower packing fraction kept their morphology intact after successive mechanical aggressions, smaller viruses with higher packing fraction showed conspicuous evidence of structural damage and content release. Second, monitoring the structure of single TGEV particles in the presence of detergent and alcohol in real time revealed the stages of gradual degradation of the virus structure in situ. These data suggest that detergent is three orders of magnitude more efficient than alcohol in destabilizing TGEV virus particles, paving the way for optimizing hygienic protocols for viruses with similar structure, such as SARS-CoV-2.


Assuntos
COVID-19 , Vírus da Gastroenterite Transmissível , Detergentes/farmacologia , Humanos , SARS-CoV-2 , Vírus da Gastroenterite Transmissível/metabolismo , Vírion/metabolismo
5.
Vet Microbiol ; 261: 109189, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34375914

RESUMO

Transmissible gastroenteritis (TGE) is an acute viral disease and characterized as severe acute inflammation response that leads to diarrhea, vomiting, and high lethality of piglets. Transmissible gastroenteritis virus (TGEV), a member of coronavirus, is the pathogen of TGE. We previously found NF-κB pathway was activated and 65 miRNAs were changed in response to inflammation caused by TGEV in cell line porcine intestinal epithelial cells-jejunum 2 (IPEC-J2). Bioinformatics results showed that these altered miRNAs were relevant to inflammation. In this study, the candidate targets of differentially expressed (DE) miRNAs were predicted and analyzed using Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Based on the results of KEGG analysis, miR-885-3p might participate in regulating activation of NF-κB pathway and TNF pathway. To study the function of miR-885-3p, miR-885-3p mimics and inhibitors were artificially synthesized and respectively used for overexpression and silence of miR-885-3p in cells. Our results showed that miR-885-3p inhibited NF-κB signaling pathway and tumor necrosis factor-α (TNF-α) production. B-cell CLL/lymphoma 10 (Bcl-10) was identified as the target of miR-885-3p, and promoted NF-κB pathway activation and TNF-α production. It was found that TGEV open reading frame 3b (TGEV-ORF3b) suppressed Bcl-10 expression, activation of NF-κB pathway, and TNF-α production by uniquely up-regulated miR-885-3p expression. Overall, the results indicated that TGEV-ORF3b counteracted NF-κB pathway and TNF-α via regulating miR-885-3p and Bcl-10.


Assuntos
Proteína 10 de Linfoma CCL de Células B/metabolismo , Gastroenterite Suína Transmissível/virologia , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteína 10 de Linfoma CCL de Células B/genética , Linhagem Celular , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação da Expressão Gênica , Mucosa Intestinal/citologia , MicroRNAs/genética , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Suínos , Regulação para Cima , Proteínas Virais
6.
Biomed Res Int ; 2021: 6689471, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34307666

RESUMO

This article is aimed at analyzing the structure and function of the spike (S) proteins of porcine enteric coronaviruses, including transmissible gastroenteritis virus (TGEV), porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus (PDCoV), and swine acute diarrhea syndrome coronavirus (SADS-CoV) by applying bioinformatics methods. The physical and chemical properties, hydrophilicity and hydrophobicity, transmembrane region, signal peptide, phosphorylation and glycosylation sites, epitope, functional domains, and motifs of S proteins of porcine enteric coronaviruses were predicted and analyzed through online software. The results showed that S proteins of TGEV, PEDV, SADS-CoV, and PDCoV all contained transmembrane regions and signal peptide. TGEV S protein contained 139 phosphorylation sites, 24 glycosylation sites, and 53 epitopes. PEDV S protein had 143 phosphorylation sites, 22 glycosylation sites, and 51 epitopes. SADS-CoV S protein had 109 phosphorylation sites, 20 glycosylation sites, and 43 epitopes. PDCoV S protein had 124 phosphorylation sites, 18 glycosylation sites, and 52 epitopes. Moreover, TGEV, PEDV, and PDCoV S proteins all contained two functional domains and two motifs, spike_rec_binding and corona_S2. The corona_S2 consisted of S2 subunit heptad repeat 1 (HR1) and S2 subunit heptad repeat 2 (HR2) region profiles. Additionally, SADS-CoV S protein was predicted to contain only one functional domain, the corona_S2. This analysis of the biological functions of porcine enteric coronavirus spike proteins can provide a theoretical basis for the design of antiviral drugs.


Assuntos
Infecções por Coronavirus/epidemiologia , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/ultraestrutura , Alphacoronavirus/metabolismo , Alphacoronavirus/patogenicidade , Animais , Biologia Computacional/métodos , Coronavirus/imunologia , Coronavirus/ultraestrutura , Bases de Dados Genéticas , Deltacoronavirus/metabolismo , Deltacoronavirus/patogenicidade , Epitopos/imunologia , Vírus da Diarreia Epidêmica Suína/metabolismo , Vírus da Diarreia Epidêmica Suína/patogenicidade , Glicoproteína da Espícula de Coronavírus/metabolismo , Suínos/virologia , Doenças dos Suínos/virologia , Vírus da Gastroenterite Transmissível/metabolismo , Vírus da Gastroenterite Transmissível/patogenicidade
7.
mBio ; 13(1): e0360021, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35100869

RESUMO

Pyroptosis, a programmed cell death, functions as an innate immune effector mechanism and plays a crucial role against microbial invasion. Gasdermin D (GSDMD), as the main pyroptosis effector, mediates pyroptosis and promotes releasing proinflammatory molecules into the extracellular environment through pore-forming activity, modifying inflammation and immune responses. While the substantial importance of GSDMD in microbial infection and cancer has been widely investigated, the role of GSDMD in virus infection, including coronaviruses, remains unclear. Enteric coronavirus transmissible gastroenteritis virus (TGEV) and porcine deltacoronavirus (PDCoV) are the major agents for lethal watery diarrhea in neonatal pigs and pose the potential for spillover from pigs to humans. In this study, we found that alphacoronavirus TGEV upregulated and activated GSDMD, resulting in pyroptosis after infection. Furthermore, the fragment of swine GSDMD from amino acids 242 to 279 (242-279 fragment) was required to induce pyroptosis. Notably, GSDMD strongly inhibited both TGEV and PDCoV infection. Mechanistically, the antiviral activity of GSDMD was mediated through promoting the nonclassical release of antiviral beta interferon (IFN-ß) and then enhancing the interferon-stimulated gene (ISG) responses. These findings showed that GSDMD dampens coronavirus infection by an uncovered GSDMD-mediated IFN secretion, which may present a novel target of coronavirus antiviral therapeutics. IMPORTANCE Coronaviruses, primarily targeting respiratory and gastrointestinal epithelia in vivo, have a serious impact on humans and animals. GSDMD, a main executioner of pyroptosis, is highly expressed in epithelial cells and involves viral infection pathogenesis. While the functions and importance of GSDMD as a critical regulator of inflammasome activities in response to intracellular bacterial infection have been extensively investigated, the roles of GSDMD during coronavirus infection remain unclear. We here show that alphacoronavirus TGEV triggered pyroptosis and upregulated GSDMD expression, while GSDMD broadly suppressed the infection of enteric coronavirus TGEV and PDCoV by its pore-forming activity via promoting unconventional release of IFN-ß. Our study highlights the importance of GSDMD as a regulator of innate immunity and may open new avenues for treating coronavirus infection.


Assuntos
Infecções por Coronavirus , Coronavirus , Vírus da Gastroenterite Transmissível , Suínos , Animais , Humanos , Interferon beta/metabolismo , Gasderminas , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/metabolismo , Antivirais/metabolismo
8.
Viruses ; 12(4)2020 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-32260595

RESUMO

Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) have been reported to use aminopeptidase N (APN) as a cellular receptor. Recently, the role of APN as a receptor for PEDV has been questioned. In our study, the role of APN in PEDV and TGEV infections was studied in primary porcine enterocytes. After seven days of cultivation, 89% of enterocytes presented microvilli and showed a two- to five-fold higher susceptibility to PEDV and TGEV. A significant increase of PEDV and TGEV infection was correlated with a higher expression of APN, which was indicative that APN plays an important role in porcine coronavirus infections. However, PEDV and TGEV infected both APN positive and negative enterocytes. PEDV and TGEV Miller showed a higher infectivity in APN positive cells than in APN negative cells. In contrast, TGEV Purdue replicated better in APN negative cells. These results show that an additional receptor exists, different from APN for porcine coronaviruses. Subsequently, treatment of enterocytes with neuraminidase (NA) had no effect on infection efficiency of TGEV, implying that terminal cellular sialic acids (SAs) are no receptor determinants for TGEV. Treatment of TGEV with NA significantly enhanced the infection which shows that TGEV is masked by SAs.


Assuntos
Antígenos CD13/metabolismo , Gastroenterite Suína Transmissível/patologia , Vírus da Diarreia Epidêmica Suína/metabolismo , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Infecções por Coronavirus/patologia , Infecções por Coronavirus/veterinária , Enterócitos/virologia , Hidrocortisona/farmacologia , Insulina/farmacologia , Mucosa Respiratória/virologia , Espermidina/farmacologia , Suínos , Células Vero , Ligação Viral , Replicação Viral/efeitos dos fármacos
9.
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
10.
J Virol ; 92(12)2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29618640

RESUMO

Identification of cellular receptors used by coronavirus (CoV) entry into the host cells is critical to an understanding of pathogenesis and to development of intervention strategies. The fourth CoV genus, Deltacoronavirus, evolutionarily related to the Gammacoronavirus, has just been defined recently. In the current study, we demonstrate that porcine aminopeptidase N (pAPN) acts as a cross-genus CoV functional receptor for both enteropathogenic porcine deltacoronovirus (PDCoV) and alphacoronovirus (AlphaCoV) (transmissible gastroenteritis virus [TGEV]) based upon three lines of evidence. First, the soluble S1 protein of PDCoV bound to the surface of target porcine cell lines known to express pAPN as efficiently as TGEV-S1, which could be blocked by soluble pAPN pretreatment. Second, both PDCoV-S1 and TGEV-S1 physically recognized and interacted with pAPN by coimmunoprecipitation in pAPN cDNA-transfected cells and by dot blot hybridization assay. Finally, exogenous expression of pAPN in refractory cells conferred susceptibility to PDCoV-S1 binding and to PDCoV entry and productive infection. PDCoV-S1 appeared to have a lower pAPN-binding affinity and likely consequent lower infection efficiency in pAPN-expressing refractory cells than TGEV-S1, suggesting that there may be differences between these two viruses in the virus-binding regions in pAPN. This study paves the way for dissecting the molecular mechanisms of PDCoV-host interactions and pathogenesis as well as facilitates future vaccine development and intervention strategies against PDCoV infection.IMPORTANCE The emergence of new human and animal coronaviruses is believed to have occurred through interspecies transmission that is mainly mediated by a species-specific receptor of the host. Among the four genera of the Coronavirinae, a couple of functional receptors for the representative members in the genera Alphacoronavirus and Betacoronavirus have been identified, whereas receptors for Gammacoronavirus and Deltacoronavirus, which are believed to originate from birds, are still unknown. Porcine coronaviruses, including the newly discovered porcine deltacoronavirus (PDCoV) associated with diarrhea in newborn piglets, have posed a serious threat to the pork industry in Asia and North America. Here, we report that PDCoV employs the alphacoronavirus TGEV functional receptor porcine aminopeptidase N (pAPN) for cellular entry, demonstrating the usage of pAPN as a cross-genus CoV functional receptor. The identification of the PDCoV receptor provides another example of the expanded host range of CoV and paves the way for further investigation of PDCoV-host interaction and pathogenesis.


Assuntos
Antígenos CD13/metabolismo , Coronavirus/metabolismo , Receptores Virais/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Ligação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Cricetinae , Especificidade de Hospedeiro/genética , Receptores de Coronavírus , Receptores Virais/genética , Suínos , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Células Vero , Internalização do Vírus
11.
Acta Virol ; 62(1): 41-49, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29521102

RESUMO

Transmissible gastroenteritis virus (TGEV) is an important pathogen in swine that is responsible for substantial economic losses. Previous studies suggest that the TGEV non-structural protein 7 (nsp7) plays an important role in the viral assembly process. However, the subcellular localization and other functions of the TGEV nsp7 protein are still unclear. In this study we have examined the subcellular localization and other functions of TGEV nsp7 protein through analysis of its effects on cell growth, cell cycle progression, interleukin 8 (IL-8) expression, and NF-κB activation. Our results showed that the nsp7 protein is localized in the cytoplasm and has no effect on intestinal epithelial cells (IECs) growth, cell cycle, and cyclin A expression. Further studies showed that TGEV nsp7 protein had no effect on GRP78 expression, could not induce endoplasmic reticulum (ER) stress and activate NF-κB activity. Interestingly, the IECs expressing nsp7 protein secreted lower levels of IL-8 than control cells. This is the first report to demonstrate the subcellular localization and novel functions of TGEV nsp7 protein. These findings provide novel information about the function of the poorly characterized TGEV non-structural protein 7.


Assuntos
Células Epiteliais/virologia , Regulação da Expressão Gênica/fisiologia , Interleucina-8/metabolismo , Suínos , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Proliferação de Células/fisiologia , Células Cultivadas , Regulação para Baixo , Células Epiteliais/fisiologia , Interleucina-8/genética , Mucosa Intestinal/citologia , Transporte Proteico , Proteínas não Estruturais Virais/genética
12.
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
13.
Virology ; 464-465: 397-405, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25113909

RESUMO

The spike protein S of coronaviruses contains a highly conserved cytoplasmic cysteine-rich motif adjacent to the transmembrane region. This motif is palmitoylated in the Betacoronaviruses MHV and SARS-CoV. Here, we demonstrate by metabolic labeling with [(3)H]-palmitic acid that the S protein of transmissible gastroenteritis coronavirus (TGEV), an Alphacoronavirus, is palmitoylated as well. This is relevant for TGEV replication as virus growth was compromised by the general palmitoylation inhibitor 2-bromopalmitate. Mutation of individual cysteine clusters in the cysteine-rich motif of S revealed that all cysteines must be replaced to abolish acylation and incorporation of S into virus-like particles (VLP). Conversely, the interaction of S with the M protein, essential for VLP incorporation of S, was not impaired by lack of palmitoylation. Thus, palmitoylation of the S protein of Alphacoronaviruses is dispensable for S-M interaction, but required for the generation of progeny virions.


Assuntos
Proteína S/química , Proteína S/metabolismo , Doenças dos Suínos/virologia , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas da Matriz Viral/metabolismo , Vírion/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Lipoilação , Dados de Sequência Molecular , Ligação Proteica , Proteína S/genética , Estrutura Terciária de Proteína , Alinhamento de Sequência , Suínos , Vírus da Gastroenterite Transmissível/química , Vírus da Gastroenterite Transmissível/genética , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética , Vírion/química , Vírion/genética
14.
Virology ; 464-465: 274-286, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25108114

RESUMO

Coronaviruses (CoVs) are positive-stranded RNA viruses with potential as immunization vectors, expressing high levels of heterologous genes and eliciting both secretory and systemic immune responses. Nevertheless, its high recombination rate may result in the loss of the full-length foreign gene, limiting their use as vectors. Transmissible gastroenteritis virus (TGEV) was engineered to express porcine reproductive and respiratory syndrome virus (PRRSV) small protein domains, as a strategy to improve heterologous gene stability. After serial passage in tissue cultures, stable expression of small PRRSV protein antigenic domains was achieved. Therefore, size reduction of the heterologous genes inserted in CoV-derived vectors led to the stable expression of antigenic domains. Immunization of piglets with these TGEV vectors led to partial protection against a challenge with a virulent PRRSV strain, as immunized animals showed reduced clinical signs and lung damage. Further improvement of TGEV-derived vectors will require the engineering of vectors with decreased recombination rate.


Assuntos
Antígenos Virais/genética , Antígenos Virais/imunologia , Expressão Gênica , Vetores Genéticos/genética , Síndrome Respiratória e Reprodutiva Suína/virologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Vírus da Gastroenterite Transmissível/genética , Animais , Anticorpos Antivirais/imunologia , Antígenos Virais/administração & dosagem , Antígenos Virais/química , Vetores Genéticos/metabolismo , Imunização , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vírus da Síndrome Respiratória e Reprodutiva Suína/química , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Suínos , Vírus da Gastroenterite Transmissível/metabolismo , Vacinas Virais/administração & dosagem , Vacinas Virais/química , Vacinas Virais/genética , Vacinas Virais/imunologia
15.
Biochem Biophys Res Commun ; 442(1-2): 33-7, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24225120

RESUMO

Transmissible gastroenteritis virus (TGEV), an enteropathogenic coronavirus, causes severe lethal watery diarrhea and dehydration in piglets. Previous studies indicate that TGEV infection induces cell apoptosis in host cells. In this study, we investigated the roles and regulation of reactive oxygen species (ROS) in TGEV-activated apoptotic signaling. The results showed that TGEV infection induced ROS accumulation, whereas UV-irradiated TGEV did not promote ROS accumulation. In addition, TGEV infection lowered mitochondrial transmembrane potential in PK-15 cell line, which could be inhibited by ROS scavengers, pyrrolidinedithiocarbamic (PDTC) and N-acetyl-l-cysteine (NAC). Furthermore, the two scavengers significantly inhibited the activation of p38 MAPK and p53 and further blocked apoptosis occurrence through suppressing the TGEV-induced Bcl-2 reduction, Bax redistribution, cytochrome c release and caspase-3 activation. These results suggest that oxidative stress pathway might be a key element in TGEV-induced apoptosis and TGEV pathogenesis.


Assuntos
Apoptose , Gastroenterite Suína Transmissível/metabolismo , Gastroenterite Suína Transmissível/patologia , Espécies Reativas de Oxigênio/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Acetilcisteína/farmacologia , Animais , Linhagem Celular , Sequestradores de Radicais Livres/farmacologia , Gastroenterite Suína Transmissível/virologia , Potencial da Membrana Mitocondrial , Estresse Oxidativo , Pirrolidinas/farmacologia , Transdução de Sinais , Suínos , Tiocarbamatos/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Monoclon Antib Immunodiagn Immunother ; 32(5): 326-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24111863

RESUMO

The purified C subunit of the recombinant porcine aminopeptidase N (rpAPN-C) protein was used as an immobilized target to screen potential ligands against rpAPN-C from a 12-mer phage display random peptide library. After five rounds of biopanning, five phage clones showed specific binding affinities to rpAPN-C. In 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assays, the phage clone PM1, which contained the HDAISWTHYHPW peptide sequence, had a protective effect against TGEV infection in swine testis cells. Therefore, the HDAISWTHYHPW peptide sequence has a potential use as a small molecular therapeutic agent against TGEV infection.


Assuntos
Antígenos CD13/genética , Gastroenterite Suína Transmissível/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Subunidades Proteicas/metabolismo , Testículo/virologia , Vírus da Gastroenterite Transmissível/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Técnicas de Visualização da Superfície Celular , Ensaio de Imunoadsorção Enzimática/veterinária , Masculino , Dados de Sequência Molecular , Oligopeptídeos/genética , Biblioteca de Peptídeos , Subunidades Proteicas/genética , Suínos , Sais de Tetrazólio , Tiazóis , Vírus da Gastroenterite Transmissível/fisiologia , Internalização do Vírus
17.
Antiviral Res ; 99(3): 383-90, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23830854

RESUMO

The membrane (M) protein is one of the major structural proteins of coronavirus particles. In this study, the M protein of transmissible gastroenteritis virus (TGEV) was used to biopan a 12-mer phage display random peptide library. Three phages expressing TGEV-M-binding peptides were identified and characterized in more depth. A phage-based immunosorbent assay (phage-ELISA) capable of differentiating TGEV from other coronaviruses was developed using one phage, phTGEV-M7, as antigen. When the phage-ELISA was compared to conventional antibody-based ELISA for detecting infections, phage-ELISA exhibited greater sensitivity. A chemically synthesized, TGEV-M7 peptide (pepTGEV-M7; HALTPIKYIPPG) was evaluated for antiviral activity. Plaque-reduction assays revealed that pepTGEV-M7 was able to prevent TGEV infection in vitro (p<0.01) following pretreatment of the virus with the peptide. Indirect immunofluorescence and real-time RT-PCR confirmed the inhibitory effects of the peptide. These results indicate that pepTGEV-M7 might be utilized for virus-specific diagnostics and treatment.


Assuntos
Gastroenterite Suína Transmissível/virologia , Peptídeos/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas da Matriz Viral/metabolismo , Sequência de Aminoácidos , Animais , Antivirais/química , Antivirais/farmacologia , Proteínas M de Coronavírus , Gastroenterite Suína Transmissível/diagnóstico , Gastroenterite Suína Transmissível/tratamento farmacológico , Ligantes , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/genética , Suínos , Vírus da Gastroenterite Transmissível/efeitos dos fármacos , Vírus da Gastroenterite Transmissível/genética , Proteínas da Matriz Viral/genética
18.
Virology ; 442(1): 74-81, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23628137

RESUMO

Coronavirus spike (S) protein assembles into virions via its carboxy-terminus, which is composed of a transmembrane domain and an endodomain. Here, the carboxy-terminal charge-rich motif in the endodomain was verified to be critical for the specificity of S assembly into mouse hepatitis virus (MHV). Recombinant MHVs exhibited a range of abilities to accommodate the homologous S endodomains from the betacoronaviruses bovine coronavirus and human SARS-associated coronavirus, the alphacoronavirus porcine transmissible gastroenteritis virus (TGEV), and the gammacoronavirus avian infectious bronchitis virus respectively. Interestingly, in TGEV endodomain chimeras the reverting mutations resulted in stronger S incorporation into virions, and a net gain of negatively charged residues in the charge-rich motif accounted for the improvement. Additionally, MHV S assembly could also be rescued by the acidic carboxy-terminal domain of the nucleocapsid protein. These results indicate an important role for negatively charged endodomain residues in the incorporation of MHV S protein into assembled virions.


Assuntos
Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Vírus da Hepatite Murina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Montagem de Vírus , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Coronavirus Bovino/genética , Coronavirus Bovino/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Vírus da Hepatite Murina/química , Vírus da Hepatite Murina/genética , Mutação , Estrutura Terciária de Proteína/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Glicoproteína da Espícula de Coronavírus , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas do Envelope Viral/genética
19.
Trop Anim Health Prod ; 45(5): 1087-91, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23224864

RESUMO

A multiplex PCR assay was developed and evaluated for its ability to simultaneously detect three viral infections of swine. Specific primers were carefully selected from articles published for each of the following three viruses: porcine circovirus type II (PCV2), porcine teschovirus (PTV) and porcine transmissible gastroenteritis virus (TGEV). Each target produced a specific amplicon with a size of 353 bp (PCV2), 168 bp (PTV) and 499 bp (TGEV). The sensitivity of the multiplex PCR using purified plasmid constructs containing the specific viral target fragments was 6.60 × 10(2), 8.43 × 10(2) and 7.30 × 10(2) copies for PCV2, PTV and TGEV, respectively. Among 127 samples which were collected from Heilongjiang, Jilin, Henan and Guangxi provinces, the single infection of PCV2, PTV and TGEV was 99.21, 46.88 and 65.35 %, respectively, and co-infection of the three viruses was 26.77 %. In conclusion, the multiplex PCR has the potential to be useful for routine molecular diagnosis and epidemiology.


Assuntos
Infecções por Circoviridae/veterinária , Coinfecção/veterinária , Reação em Cadeia da Polimerase Multiplex/métodos , Infecções por Picornaviridae/veterinária , Doenças dos Suínos/diagnóstico , Animais , China , Infecções por Circoviridae/diagnóstico , Infecções por Circoviridae/virologia , Circovirus/genética , Circovirus/isolamento & purificação , Circovirus/metabolismo , Coinfecção/diagnóstico , Coinfecção/virologia , DNA Viral/genética , DNA Viral/metabolismo , Gastroenterite Suína Transmissível/diagnóstico , Gastroenterite Suína Transmissível/virologia , Reação em Cadeia da Polimerase Multiplex/veterinária , Infecções por Picornaviridae/diagnóstico , Infecções por Picornaviridae/virologia , RNA Viral/genética , RNA Viral/metabolismo , Suínos , Doenças dos Suínos/virologia , Teschovirus/genética , Teschovirus/isolamento & purificação , Teschovirus/metabolismo , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/isolamento & purificação , Vírus da Gastroenterite Transmissível/metabolismo
20.
Virol J ; 8: 435, 2011 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-21910859

RESUMO

BACKGROUND: Transmissible gastroenteritis virus (TGEV) has a sialic acid binding activity that is believed to be important for enteropathogenicity, but that has so far appeared to be dispensable for infection of cultured cells. The aims of this study were to determine the effect of sialic acid binding for the infection of cultured cells under unfavorable conditions, and comparison of TGEV strains and mutants, as well as the avian coronavirus IBV concerning their dependence on the sialic acid binding activity. METHODS: The infectivity of different viruses was analyzed by a plaque assay after adsorption times of 5, 20, and 60 min. Prior to infection, cultured cells were either treated with neuraminidase to deplete sialic acids from the cell surface, or mock-treated. In a second approach, pre-treatment of the virus with porcine intestinal mucin was performed, followed by the plaque assay after a 5 min adsorption time. A student's t-test was used to verify the significance of the results. RESULTS: Desialylation of cells only had a minor effect on the infection by TGEV strain Purdue 46 when an adsorption period of 60 min was allowed for initiation of infection. However, when the adsorption time was reduced to 5 min the infectivity on desialylated cells decreased by more than 60%. A TGEV PUR46 mutant (HAD3) deficient in sialic acid binding showed a 77% lower titer than the parental virus after a 5 min adsorption time. After an adsorption time of 60 min the titer of HAD3 was 58% lower than that of TGEV PUR46. Another TGEV strain, TGEV Miller, and IBV Beaudette showed a reduction in infectivity after neuraminidase treatment of the cultured cells irrespective of the virion adsorption time. CONCLUSIONS: Our results suggest that the sialic acid binding activity facilitates the infection by TGEV under unfavorable environmental conditions. The dependence on the sialic acid binding activity for an efficient infection differs in the analyzed TGEV strains.


Assuntos
Membrana Celular/metabolismo , Gastroenterite Suína Transmissível/virologia , Glicoproteínas de Membrana/metabolismo , Neuraminidase/farmacologia , Ácidos Siálicos/metabolismo , Testículo/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Aves , Membrana Celular/efeitos dos fármacos , Membrana Celular/virologia , Células Cultivadas , Chlorocebus aethiops , Gastroenterite Suína Transmissível/metabolismo , Vírus da Bronquite Infecciosa/metabolismo , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/química , Mucinas/farmacologia , Mutação , Neuraminidase/metabolismo , Ligação Proteica/efeitos dos fármacos , Ácidos Siálicos/antagonistas & inibidores , Glicoproteína da Espícula de Coronavírus , Suínos , Testículo/citologia , Testículo/efeitos dos fármacos , Testículo/virologia , Vírus da Gastroenterite Transmissível/efeitos dos fármacos , Vírus da Gastroenterite Transmissível/genética , Vírus da Gastroenterite Transmissível/patogenicidade , Células Vero , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/química , Ensaio de Placa Viral , Vírion/efeitos dos fármacos , Vírion/metabolismo , Ligação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA