Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Food Environ Virol ; 11(2): 184-192, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30903596

RESUMEN

An extremely affordable virus concentration method based on adsorption-elution to glass wool and subsequent reconcentration through polyethylene glycol 6000 (PEG) precipitation was optimized to recover not only non-enveloped viruses but also enveloped viruses. Hepatitis A virus (HAV) and transmissible gastroenteritis virus (TGEV) were employed as surrogates for naked and enveloped viruses, respectively, to set up the methodology. Initial experimentation in small-volume samples showed that both types of particles readily adsorbed to the positively charged glass wool but were poorly detached from it through standard elution with 0.05 M glycine with 3% of beef extract buffer, pH 9.5, with elution efficiencies of 7.2% and 2.6%, for HAV and TGEV, respectively. To improve the recovery of enveloped viruses, several modifications in the elution were assayed: increasing the elution pH, extending glass wool and eluent contact time, adding a detergent, or performing the elution by recirculation or under agitation. Considering practicability and performance, recircularization of the eluent at pH 11.0 for 20 min was the elution procedure of choice, with efficiencies of 25.7% and 18.8% for HAV and TGEV in 50 L of water. Additionally, employing 20% PEG instead of 10% for virus reconcentration improved recoveries up to 47% and 51%, respectively. The optimized procedure was applied to detect naturally occurring HAV and coronaviruses in surface water of Wadi Hanifa, Riyadh. HAV was detected in 38% of the samples, while one sample was positive for an alphacoronavirus. This cheap virus detection system enables the comprehensive surveillance of viruses present in water samples.


Asunto(s)
Agua Dulce/virología , Vidrio/química , Virus de la Hepatitis A/química , Virus de la Gastroenteritis Transmisible/química , Virología/métodos , Adsorción , Virus de la Hepatitis A/aislamiento & purificación , Virus de la Gastroenteritis Transmisible/aislamiento & purificación , Virología/instrumentación , Virus/química , Virus/aislamiento & purificación
2.
Virulence ; 9(1): 1685-1698, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30322331

RESUMEN

Transmissible gastroenteritis virus (TGEV) infection causes acute enteritis in swine of all ages, and especially in suckling piglets. Small intestinal inflammation is considered a central event in the pathogenesis of TGEV infections, and nuclear factor-kappa B (NF-κB) is a key transcription factor in the inflammatory response. However, it is unclear whether NF-κB is crucial for inducing inflammation during a TGEV infection. Our results show that NF-κB was activated in swine testicular (ST) cells and intestinal epithelial cell lines J2 (IPEC-J2) cells infected with TGEV, which is consistent with the up-regulation of pro-inflammatory cytokines. Treatment of TGEV-infected ST cells and IPEC-J2 cells with the NF-κB-specific inhibitor caused the down-regulation of pro-inflammatory cytokine expression, but did not significantly affect TGEV replication. Individual TGEV protein screening results demonstrated that Nsp2 exhibited a high potential for activating NF-κB and enhancing the expression of pro-inflammatory cytokines. Functional domain analyzes indicated that the first 120 amino acid residues of Nsp2 were essential for NF-κB activation. Taken together, these data suggested that NF-κB activation was a major contributor to TGEV infection-induced inflammation, and that Nsp2 was the key viral protein involved in the regulation of inflammation, with amino acids 1-120 playing a critical role in activating NF-κB. Abbreviations: TCID50: 50% tissue culture infectious dose; DMEM: Dulbecco's Modified Eagle Medium; eNOS: Endothelial nitric oxide synthase; FBS: fetal bovine serum; IFA: Indirect immunofluorescence; IκB: inhibitor of nuclear factor kappa-B; IL: interleukin; IPEC-J2: intestinal epithelial cell lines J2; IKK: IκB kinase; Luc: luciferase reporter gene; mAbs: monoclonal antibodies; MOI: multiple of infection; Nsp: nonstructural protein; NF-κB: nuclear factor-kappa ; ORFs: open reading frames; PBS: phosphate-buffered saline; p65 p-p65: phosphorylated; RT-PCR: reverse transcription PC; SeV: Sendai virus; ST: swine testicular; TGEV: Transmissible gastroenteritis virus; TNF-α: tumor necrosis factor α; UV-TGEV: Ultraviolet light-inactivated TGEV; ZnF: zinc finger.


Asunto(s)
Gastroenteritis Porcina Transmisible/inmunología , Inflamación , FN-kappa B/inmunología , Virus de la Gastroenteritis Transmisible/química , Proteínas no Estructurales Virales/inmunología , Animales , Línea Celular , Citocinas/inmunología , Células Epiteliales/virología , Regulación Viral de la Expresión Génica , Intestinos/citología , Intestinos/virología , FN-kappa B/antagonistas & inhibidores , Fosforilación , Porcinos , Virus de la Gastroenteritis Transmisible/inmunología , Replicación Viral
3.
J Virol ; 92(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29237834

RESUMEN

Porcine epidemic diarrhea virus (PEDV), an enteropathogenic Alphacoronavirus, has caused enormous economic losses in the pork industry. Nonstructural protein 1 (nsp1) is a characteristic feature of alpha- and betacoronaviruses, which exhibits both functional conservation and mechanistic diversity in inhibiting host gene expression and antiviral responses. However, the detailed structure and molecular mechanisms underlying the Alphacoronavirus nsp1 inhibition of host gene expression remain unclear. Here, we report the first full-length crystal structure of Alphacoronavirus nsp1 from PEDV. The structure displays a six-stranded ß-barrel fold in the middle of two α-helices. The core structure of PEDV nsp1 shows high similarity to those of severe acute respiratory syndrome coronavirus (SARS-CoV) nsp1 and transmissible gastroenteritis virus (TGEV) nsp1, despite its low degree of sequence homology. Using ribopuromycylation and Renilla luciferase reporter assays, we showed that PEDV nsp1 can dramatically inhibit general host gene expression. Furthermore, three motifs (amino acids [aa] 67 to 71, 78 to 85, and 103 to 110) of PEDV nsp1 create a stable functional region for inhibiting protein synthesis, differing considerably from Betacoronavirus nsp1. These results elucidate the detailed structural basis through which PEDV nsp1 inhibits host gene expression, providing insight into the development of a new attenuated vaccine with nsp1 modifications.IMPORTANCE Porcine epidemic diarrhea virus (PEDV) has led to tremendous economic losses in the global swine industry. PEDV nsp1 plays a crucial role in inhibiting host gene expression, but its functional mechanism remains unclear. Here, we report the full-length structure of PEDV nsp1, the first among coronaviruses to be reported. The 1.25-Å resolution crystal structure of PEDV nsp1 shows high similarity to severe acute respiratory syndrome coronavirus (SARS-CoV) nsp113-128 and transmissible gastroenteritis virus (TGEV) nsp11-104, despite a lack of sequence homology. Structural and biochemical characterization demonstrated that PEDV nsp1 possesses a stable functional region for inhibition of host protein synthesis, which is formed by loops at residues 67 to 71, 78 to 85, and 103 to 110. The different functional regions in PEDV nsp1 and SARS-CoV nsp1 may explain their distinct mechanisms. Importantly, our structural data are conducive to understanding the mechanism of PEDV nsp1 inhibition of the expression of host genes and may aid in the development of a new attenuated vaccine.


Asunto(s)
Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Virus de la Diarrea Epidémica Porcina/química , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Animales , Línea Celular , Coronavirus/genética , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/virología , Cristalografía por Rayos X , Células HEK293 , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Modelos Moleculares , Virus de la Diarrea Epidémica Porcina/genética , Pliegue de Proteína , Estructura Terciaria de Proteína , ARN Polimerasa Dependiente del ARN , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Alineación de Secuencia , Homología de Secuencia , Porcinos , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/virología , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/genética , Vacunas Atenuadas , Proteínas no Estructurales Virales/clasificación , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
4.
Biomed Res Int ; 2017: 7089091, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29201911

RESUMEN

Coronaviruses (CoVs), such as human coronavirus NL63 (HCoV-NL63), severe acute respiratory syndrome CoV (SARS-CoV), murine hepatitis virus (MHV), porcine epidemic diarrhea virus (PEDV), and Middle East Respiratory Syndrome Coronavirus (MERS-CoV), encode papain-like (PL) proteases that inhibit Sendai virus- (SeV-) induced interferon (IFN-ß) production. Recently, the crystal structure of transmissible gastroenteritis virus (TGEV) PL1 has been solved, which was similar to that of SARS-CoV PL2pro, which may antagonize host innate immunity. However, very little is known about whether TGEV PL1 can antagonize host innate immune response. Here, we presented evidence that TGEV PL1 encoded by the replicase gene could suppress the IFN-ß expression and inhibit the nuclear translocation of interferon regulatory factor 3 (IRF3). The ability to antagonize IFN-ß production was dependent on the intact catalytic activity of PL1. Furthermore, TGEV PL1 exerted deubiquitinase (DUB) activity which strongly inhibited the retinoic acid-induced gene I- (RIG-1-) and stimulator of interferon gene- (STING-) dependent IFN expression. Our data collectively suggest that TGEV PL1 can inhibit the IFN-ß expression and interfere with RIG-1- and STING-mediated signaling through a viral DUB activity. Our study has yielded strong evidence for the TGEV PL1 mechanisms that counteract the host innate immunity.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Interferón beta/genética , Papaína/genética , Virus de la Gastroenteritis Transmisible/genética , Animales , Proteasas Similares a la Papaína de Coronavirus , Proteína 58 DEAD Box/genética , Enzimas Desubicuitinizantes/genética , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Interferón beta/biosíntesis , Proteínas de la Membrana/genética , Papaína/química , Papaína/inmunología , ARN Polimerasa Dependiente del ARN/genética , Receptores Inmunológicos , Porcinos , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/patogenicidad , Ubiquitina/genética
5.
Antiviral Res ; 131: 74-84, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26988122

RESUMEN

Transmissible gastroenteritis (TGE) causes severe diarrhea in suckling piglets, results in enormous economic loss in swine-producing areas of the world. To develop an effective, safe, and convenient vaccine for the prevention of TGE, we have constructed a recombinant Bacillus subtilis strain (B. subtilis CotGSG) displaying the transmissible gastroenteritis virus (TGEV) spike (S) protein and discussed its immune function to intestinal submucosal dendritic cells (DCs). Our results showed that the recombinant B. subtilis had the ability to recruit more DCs to sample B. subtilis CotGSG, migrate to MLNs, and induce immune responses. Immunized piglets with B. subtilis CotGSG could significantly elevate the specific SIgA titers in feces, IgG titers and neutralizing antibodies in serum. Collectively, our results suggested that recombinant B. subtilis CotGSG expressing the TGEV S protein could effectively induce immune responses via DCs, and provided a perspective on potential novel strategy and approach that may be applicable to the development of the next generation of TGEV vaccines.


Asunto(s)
Anticuerpos Antivirales/sangre , Bacillus subtilis/inmunología , Gastroenteritis Porcina Transmisible/inmunología , Virus de la Gastroenteritis Transmisible/química , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Antígenos Virales/inmunología , Bacillus subtilis/genética , Movimiento Celular , Células Dendríticas/inmunología , Gastroenteritis Porcina Transmisible/prevención & control , Inmunoglobulina A Secretora/análisis , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/inmunología , Vacunas Sintéticas/inmunología , Proteínas Virales/administración & dosificación , Proteínas Virales/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
6.
J Virol ; 89(6): 3332-42, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25589635

RESUMEN

UNLABELLED: Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) are economically important swine enteropathogenic coronaviruses. These two viruses belong to two distinct species of the Alphacoronavirus genus within Coronaviridae and induce similar clinical signs and pathological lesions in newborn piglets, but they are presumed to be antigenically distinct. In the present study, two-way antigenic cross-reactivity examinations between the prototype PEDV CV777 strain, three distinct U.S. PEDV strains (the original highly virulent PC22A, S indel Iowa106, and S 197del PC177), and two representative U.S. TGEV strains (Miller and Purdue) were conducted by cell culture immunofluorescent (CCIF) and viral neutralization (VN) assays. None of the pig TGEV antisera neutralized PEDV and vice versa. One-way cross-reactions were observed by CCIF between TGEV Miller hyperimmune pig antisera and all PEDV strains. Enzyme-linked immunosorbent assays, immunoblotting using monoclonal antibodies and Escherichia coli-expressed recombinant PEDV and TGEV nucleocapsid (N) proteins, and sequence analysis suggested at least one epitope on the N-terminal region of PEDV/TGEV N protein that contributed to this cross-reactivity. Biologically, PEDV strain CV777 induced greater cell fusion in Vero cells than did U.S. PEDV strains. Consistent with the reported genetic differences, the results of CCIF and VN assays also revealed higher antigenic variation between PEDV CV777 and U.S. strains. IMPORTANCE: Evidence of antigenic cross-reactivity between porcine enteric coronaviruses, PEDV and TGEV, in CCIF assays supports the idea that these two species are evolutionarily related, but they are distinct species defined by VN assays. Identification of PEDV- or TGEV-specific antigenic regions allows the development of more specific immunoassays for each virus. Antigenic and biologic variations between the prototype and current PEDV strains could explain, at least partially, the recurrence of PEDV epidemics. Information on the conserved antigenicity among PEDV strains is important for the development of PEDV vaccines to protect swine from current highly virulent PEDV infections.


Asunto(s)
Anticuerpos Antivirales/inmunología , Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/inmunología , Enfermedades de los Porcinos/inmunología , Virus de la Gastroenteritis Transmisible/inmunología , Secuencia de Aminoácidos , Animales , Variación Antigénica , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Reacciones Cruzadas , Gastroenteritis Porcina Transmisible/inmunología , Gastroenteritis Porcina Transmisible/virología , Datos de Secuencia Molecular , Virus de la Diarrea Epidémica Porcina/química , Virus de la Diarrea Epidémica Porcina/clasificación , Virus de la Diarrea Epidémica Porcina/genética , Alineación de Secuencia , Porcinos , Enfermedades de los Porcinos/virología , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/clasificación , Virus de la Gastroenteritis Transmisible/genética
7.
Virology ; 464-465: 397-405, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25113909

RESUMEN

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.


Asunto(s)
Proteína S/química , Proteína S/metabolismo , Enfermedades de los Porcinos/virología , Virus de la Gastroenteritis Transmisible/metabolismo , Proteínas de la Matriz Viral/metabolismo , Virión/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Lipoilación , Datos de Secuencia Molecular , Unión Proteica , Proteína S/genética , Estructura Terciaria de Proteína , Alineación de Secuencia , Porcinos , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/genética , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética , Virión/química , Virión/genética
8.
J Virol ; 87(5): 2949-55, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269811

RESUMEN

Coronavirus nsp1 has been shown to induce suppression of host gene expression and to interfere with the host immune response. However, the mechanism is currently unknown. The only available structural information on coronavirus nsp1 is the nuclear magnetic resonance (NMR) structure of the N-terminal domain of nsp1 from severe acute respiratory syndrome coronavirus (SARS-CoV) from the betacoronavirus genus. Here we present the first nsp1 structure from an alphacoronavirus, transmissible gastroenteritis virus (TGEV) nsp1. It displays a six-stranded ß-barrel fold with a long alpha helix on the rim of the barrel, a fold shared with SARS-CoV nsp1(13-128). Contrary to previous speculation, the TGEV nsp1 structure suggests that coronavirus nsp1s have a common origin, despite the lack of sequence homology. However, comparisons of surface electrostatics, shape, and amino acid conservation between the alpha- and betacoronaviruses lead us to speculate that the mechanism for nsp1-induced suppression of host gene expression might be different in these two genera.


Asunto(s)
ARN Polimerasa Dependiente del ARN/química , Virus de la Gastroenteritis Transmisible/química , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Expresión Génica , Modelos Moleculares , Datos de Secuencia Molecular , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Alineación de Secuencia , Virus de la Gastroenteritis Transmisible/clasificación , Virus de la Gastroenteritis Transmisible/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
9.
New Microbiol ; 35(4): 487-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23109017

RESUMEN

The coronavirus heptide repeat (HR) region in the spike protein induces neutralizing antibodies that block the postfusion core formation and inhibit virus entry into target cells. The HR2 regions for coronaviruses of the same serogroup share high homology. We found that polyclonal antibodies derived from transmissible gastroenteritis coronavirus HR2 and upstream region were cross-reactive with the S proteins of the same serogroup in western blotting. The polyclonal antibodies also potently cross-neutralized viruses from the same serogroup. This study provides new insight for designing vaccine and therapeutic reagents against coronavirus infections.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Coronavirus/inmunología , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/inmunología , Secuencias Repetitivas de Aminoácido , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Coronavirus/clasificación , Coronavirus/genética , Reacciones Cruzadas , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Glicoproteína de la Espiga del Coronavirus , Secuencias Repetidas en Tándem , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/inmunología , Proteínas del Envoltorio Viral/genética
10.
Virus Genes ; 43(3): 335-41, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21701858

RESUMEN

The spike (S) protein is a key structural protein of coronaviruses including, the porcine transmissible gastroenteritis virus (TGEV). The S protein is a type I membrane glycoprotein located in the viral envelope and is responsible for mediating the binding of viral particles to specific cell receptors and therefore specific cell types. It is also an important immune target for the host in neutralizing the virus. Four antigenic sites A, B, C, and D that reside near the N-terminal domain have been defined in the S protein. Of these, the region encoding antigenic sites A and to a lesser extent D, herein defined as S-AD, are most critical in eliciting host neutralizing antibodies. Herein, we enzymatically amplified, cloned, and expressed the S-AD fragment from TGEV in the prokaryotic expression vector, pET-30a. Maximum protein expression was achieved at 30°C over a 5-h period post-induction. Rabbit polyclonal antiserum was generated using recombinant S-AD (rS-AD) protein. In contrast to prior studies showing no activity with bacterially produced S protein, results indicated that polyclonal serum recognized TGEV-infected cells and reduced infection by 100%. Furthermore, the truncated rS-AD peptide was able to bind to the surface of cells from swine testes in a competitive manner and completely inhibit viral infection.


Asunto(s)
Antígenos Virales/genética , Antígenos Virales/metabolismo , Gastroenteritis Porcina Transmisible/prevención & control , Expresión Génica , Virus de la Gastroenteritis Transmisible/fisiología , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Animales , Antígenos Virales/química , Línea Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Gastroenteritis Porcina Transmisible/virología , Unión Proteica , Conejos , Porcinos , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/inmunología , Proteínas Virales/química , Proteínas Virales/genética
11.
J Virol Methods ; 175(1): 7-13, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21513742

RESUMEN

Coronavirus RNA synthesis is a sophisticated process performed by a viral multienzymatic replicase complex, together with cellular factors. A key enzyme of this replication complex is the RNA dependent RNA polymerase (RdRp). To study the replication of coronavirus genome, six monoclonal antibodies (mAbs) specific for transmissible gastroenteritis virus (TGEV) RdRp were generated and characterized. His-tagged RdRp was expressed in baculovirus, purified and used as immunogen to produce mAbs. The TGEV RdRp was recognized by these mAbs in the context of virus infection by immunofluorescence analysis and Western blot. Epitope mapping by Pepscan indicated that RdRp mAbs recognized four non-overlapping linear epitopes located in a 62-amino acid region of the N-terminal domain, suggesting that this region may constitute an immunodominant domain. The availability of TGEV RdRp mAbs will be instrumental to study coronavirus replication and to analyze the function of RdRp in pathogenesis.


Asunto(s)
Mapeo Epitopo/métodos , ARN Polimerasa Dependiente del ARN/análisis , ARN Polimerasa Dependiente del ARN/inmunología , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos Virales/inmunología , Baculoviridae/genética , Western Blotting , Línea Celular , Coronavirus/genética , Coronavirus/inmunología , Epítopos/genética , Epítopos/inmunología , Técnica del Anticuerpo Fluorescente , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Porcinos , Virus de la Gastroenteritis Transmisible/genética
12.
Antiviral Res ; 88(3): 311-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20951168

RESUMEN

Cholesterol is a major constituent of detergent-resistant membrane microdomains (DRMs). We localized transmissible gastroenteritis virus (TGEV) spike (S) protein in DRMs in the viral envelope. Though S protein was not solubilized by cold non-ionic detergents, this behavior was unchanged when cholesterol was depleted from viral membrane by methyl-ß-cyclodextrin (MßCD) and the protein did not comigrate with cellular DRM marker proteins in flotation analyses. Therefore, the S protein is not anchored in the viral membrane DRMs as they are known to occur in the plasma membrane. Cholesterol depletion from viral membrane may not affect the adsorption process as neither the sialic acid binding activity nor the binding to aminopeptidase N was reduced post-MßCD treatment. Reduced infectivity of cholesterol-depleted TGEV was observed only when the adsorption process occurred at 37°C but not when the virus was applied at 4°C. Cholesterol is important for a post-adsorption step, allowing membrane rearrangements that facilitate virus entry.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/metabolismo , Gastroenteritis Porcina Transmisible/metabolismo , Microdominios de Membrana/metabolismo , Virus de la Gastroenteritis Transmisible/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Acoplamiento Viral , Internalización del Virus , Animales , Línea Celular , Membrana Celular/virología , Colesterol/química , Cricetinae , Detergentes , Gastroenteritis Porcina Transmisible/virología , Porcinos , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/patogenicidad
13.
J Gen Virol ; 90(Pt 7): 1724-1729, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19264610

RESUMEN

The surface proteins S of severe acute respiratory syndrome coronavirus (SARS-CoV) and transmissible gastroenteritis virus (TGEV) were compared for their ability to mediate infection of viral pseudotypes based on vesicular stomatitis virus (VSV). The cell tropism of the respective pseudotypes corresponded to the tropism of the viruses from which the S protein was derived. Higher infectivity values were obtained with the SARS-CoV S protein than with the TGEV S protein. Differences were observed with respect to the importance of the cytoplasmic tail and the membrane anchor of the S proteins. In the case of the SARS-CoV S protein, truncation of the cytoplasmic tail resulted in increased infectivity. For the TGEV S protein, the inactivation of an intracellular retention signal in the cytoplasmic tail was required. Exchange of the membrane anchor of the S proteins led to a low infection efficiency. Our results indicate that related glycoproteins may show substantial differences in their ability to mediate pseudotype infection.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Virus de la Gastroenteritis Transmisible/química , Vesiculovirus/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Cricetinae , Humanos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Eliminación de Secuencia , Glicoproteína de la Espiga del Coronavirus , Porcinos , Virus de la Gastroenteritis Transmisible/genética , Vesiculovirus/genética , Virulencia
14.
J Virol ; 79(4): 2506-16, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15681451

RESUMEN

The generation of subgenomic mRNAs in coronavirus involves a discontinuous mechanism of transcription by which the common leader sequence, derived from the genome 5' terminus, is fused to the 5' end of the mRNA coding sequence (body). Transcription-regulating sequences (TRSs) precede each gene and include a conserved core sequence (CS) surrounded by relatively variable sequences (5' TRS and 3' TRS). Regulation of transcription in coronaviruses has been studied by reverse-genetics analysis of the sequences immediately flanking a unique CS in the Transmissible gastroenteritis virus genome (CS-S2), located inside the S gene, that does not lead to detectable amounts of the corresponding mRNA, in spite of its canonical sequence. The transcriptional inactivity of CS-S2 was genome position independent. The presence of a canonical CS was not sufficient to drive transcription, but subgenomic synthesis requires a minimum base pairing between the leader TRS (TRS-L) and the complement of the body TRS (cTRS-B) provided by the CS and its adjacent nucleotides. A good correlation was observed between the free energy of TRS-L and cTRS-B duplex formation and the levels of subgenomic mRNA S2, demonstrating that base pairing between the leader and body beyond the CS is a determinant regulation factor in coronavirus transcription. In TRS mutants with increasing complementarity between TRS-L and cTRS-B, a tendency to reach a plateau in DeltaG values was observed, suggesting that a more precise definition of the TRS limits might be proposed, specifically that it consists of the central CS and around 4 nucleotides flanking 5' and 3' the CS. Sequences downstream of the CS exert a stronger influence on the template-switching decision according to a model of polymerase strand transfer and template switching during minus-strand synthesis.


Asunto(s)
Región de Flanqueo 3'/genética , Regulación Viral de la Expresión Génica , Genoma Viral , ARN Viral/biosíntesis , Transcripción Genética/genética , Virus de la Gastroenteritis Transmisible/genética , Animales , Secuencia de Bases , Línea Celular , Cricetinae , ARN Mensajero/biosíntesis , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/metabolismo
15.
J Virol ; 78(2): 980-94, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14694129

RESUMEN

Coronavirus transcription leads to the synthesis of a nested set of mRNAs with a leader sequence derived from the 5' end of the genome. The mRNAs are produced by a discontinuous transcription in which the leader is linked to the mRNA coding sequences. This process is regulated by transcription-regulating sequences (TRSs) preceding each mRNA, including a highly conserved core sequence (CS) with high identity to sequences present in the virus genome and at the 3' end of the leader (TRS-L). The role of TRSs was analyzed by reverse genetics using a full-length infectious coronavirus cDNA and site-directed mutagenesis of the CS. The canonical CS-B was nonessential for the generation of subgenomic mRNAs (sgmRNAs), but its presence led to transcription levels at least 10(3)-fold higher than those in its absence. The data obtained are compatible with a transcription mechanism including three steps: (i) formation of 5'-3' complexes in the genomic RNA, (ii) base-pairing scanning of the nascent negative RNA strand by the TRS-L, and (iii) template switching during synthesis of the negative strand to complete the negative sgRNA. This template switch takes place after copying the CS sequence and was predicted in silico based on high base-pairing score between the nascent negative RNA strand and the TRS-L and minimum DeltaG.


Asunto(s)
Secuencia de Bases , Regulación Viral de la Expresión Génica , Genoma Viral , ARN Mensajero/biosíntesis , Transcripción Genética , Virus de la Gastroenteritis Transmisible/metabolismo , Regiones no Traducidas 3'/química , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/química , Regiones no Traducidas 5'/genética , Animales , Emparejamiento Base , Cricetinae , ADN Complementario/genética , Humanos , Modelos Genéticos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , ARN Mensajero/química , ARN Mensajero/genética , ARN Viral/biosíntesis , ARN Viral/química , ARN Viral/genética , Moldes Genéticos , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/genética
16.
Acta Pharmacol Sin ; 24(6): 497-504, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12791174

RESUMEN

AIM: To constructed a three-dimensional (3D) model for the 3C like (3CL) proteinase of SARS coronavirus (SARS-CoV), and to design inhibitors of the 3CL proteinase based on the 3D model. METHODS: Bioinformatics analyses were performed to search the homologous proteins of the SARS-CoV 3CL proteinase from the GenBank and PDB database. A 3D model of the proteinase was constructed by using homology modeling technique. Targeting to the 3D model and its X-ray crystal structure of the main proteinase (Mpro) of transmissible gastroenteritis virus (TGEV), virtual screening was performed employing molecular docking method to identify possible 3CL proteinase inhibitors from small molecular databases. RESULTS: Sequence alignment indicated that the SARS-CoV 3CL proteinase was extremely homologous to TGEV Mpro, especially the substrate-binding pocket (active site). Accordingly, a 3D model for the SARS-CoV 3CL proteinase was constructed based on the crystal structure of TGEV Mpro. The 3D model adopts a similar fold of the TGEV Mpro, its structure and binding pocket feature are almost as same as that of TGEV Mpro. The tested virtual screening indicated that 73 available proteinase inhibitors in the MDDR database might dock into both the binding pockets of the TGEV Mpro and the SARS-CoV 3CL proteinase. CONCLUSIONS: Either the 3D model of the SARS-CoV 3CL proteinase or the X-ray crystal structure of the TGEV Mpro may be used as a starting point for design anti-SARS drugs. Screening the known proteinase inhibitors may be an appreciated shortcut to discover anti-SARS drugs.


Asunto(s)
Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/aislamiento & purificación , Síndrome Respiratorio Agudo Grave/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Biología Computacional , Proteasas 3C de Coronavirus , Cristalografía por Rayos X , Cisteína Endopeptidasas/genética , Diseño de Fármacos , Humanos , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/aislamiento & purificación , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Homología Estructural de Proteína , Virus de la Gastroenteritis Transmisible/química
17.
Vet Microbiol ; 94(3): 195-206, 2003 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-12814887

RESUMEN

The spike (S) glycoprotein of transmissible gastroenteritis virus (TGEV) is the predominant inducer of neutralizing antibodies and has been implicated in virulence and host cell tropism. In this study, the nucleotide and deduced amino acid sequences of the amino terminal half of the S glycoprotein gene of one Korean field TGEV strain (133) isolated in 1997 and three Korean field TGEV strains (KT2, KT3 and KT4) isolated in 2000 and HKT2 strain, KT2 passaged 104 times in ST cells, were determined. The amino terminal half of the S glycoprotein gene including antigenic sites A, B, C and D, were amplified by reverse transcriptase-polymerase chain reaction (RT-PCR). Amplified PCR products were cloned, sequenced, and compared with published sequences for non-Korean TGEV strains. Korea TGEV field strains had 98.5-99.5% nucleotide sequence and 97.2-99.0% amino acid sequence similarity with each other. They had 96.5-99.0% nucleotide sequence similarity and 94.9-97.6% amino acid sequence similarity compared to non-Korean TGEV strains. Korean TGEV strains had several specific nucleotide and amino acid sequences which were not found in foreign TGEV or PRCV strains. HKT2 strain differed by 0.89% in nucleotide and 2.03% amino acid sequences compared to original KT2 strain although the regions forming four antigenic sites were not changed. By phylogenetic tree analysis, Korean field TGEV strains were branched into different groups from non-Korean TGEV or PRCV strains. Korean TGEV field strains KT2 and 133 were branched in separate groups that were differentiated from the other Korean TGEV strains. The Korean TGEV strains seemed to be evolved from a separate lineage of TGEV strain.


Asunto(s)
Gastroenteritis Porcina Transmisible/virología , Genoma Viral , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/genética , Proteínas Virales/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Clonación Molecular , Corea (Geográfico) , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Pase Seriado , Porcinos , Virus de la Gastroenteritis Transmisible/clasificación , Proteínas Virales/química
18.
J Virol ; 75(24): 12228-40, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11711614

RESUMEN

The difference in membrane (M) protein compositions between the transmissible gastroenteritis coronavirus (TGEV) virion and the core has been studied. The TGEV M protein adopts two topologies in the virus envelope, a Nexo-Cendo topology (with the amino terminus exposed to the virus surface and the carboxy terminus inside the virus particle) and a Nexo-Cexo topology (with both the amino and carboxy termini exposed to the virion surface). The existence of a population of M molecules adopting a Nexo-Cexo topology in the virion envelope was demonstrated by (i) immunopurification of (35)S-labeled TGEV virions using monoclonal antibodies (MAbs) specific for the M protein carboxy terminus (this immunopurification was inhibited only by deletion mutant M proteins that maintained an intact carboxy terminus), (ii) direct binding of M-specific MAbs to the virus surface, and (iii) mass spectrometry analysis of peptides released from trypsin-treated virions. Two-thirds of the total number of M protein molecules found in the virion were associated with the cores, and one-third was lost during core purification. MAbs specific for the M protein carboxy terminus were bound to native virions through the M protein in a Nexo-Cexo conformation, and these molecules were removed when the virus envelope was disrupted with NP-40 during virus core purification. All of the M protein was susceptible to N-glycosidase F treatment of the native virions, which indicates that all the M protein molecules are exposed to the virus surface. Cores purified from glycosidase-treated virions included M protein molecules that completely or partially lost the carbohydrate moiety, which strongly suggests that the M protein found in the cores was also exposed in the virus envelope and was not present exclusively in the virus interior. A TGEV virion structure integrating all the data is proposed. According to this working model, the TGEV virion consists of an internal core, made of the nucleocapsid and the carboxy terminus of the M protein, and the envelope, containing the spike (S) protein, the envelope (E) protein, and the M protein in two conformations. The two-thirds of the molecules that are in a Nexo-Cendo conformation (with their carboxy termini embedded within the virus core) interact with the internal core, and the remaining third of the molecules, whose carboxy termini are in a Nexo-Cexo conformation, are lost during virus core purification.


Asunto(s)
Virus de la Gastroenteritis Transmisible/química , Proteínas de la Matriz Viral/química , Virión/química , Secuencia de Aminoácidos , Animales , Glicósido Hidrolasas/farmacología , Masculino , Datos de Secuencia Molecular , Porcinos
19.
J Virol ; 75(2): 844-9, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11134297

RESUMEN

The sedimentation behavior of transmissible gastroenteritis coronavirus (TGEV) was analyzed. Upon sucrose gradient centrifugation, the major virus band was found at a density of 1.20 to 1.22 g/cm(3). This high density was observed only when TGEV with a functional sialic acid binding activity was analyzed. Mutants of TGEV that lacked sialic acid binding activity due to a point mutation in the sialic acid binding site of the S protein were mainly recovered at a lower-density position on the sucrose gradient (1.18 to 1.19 g/cm(3)). Neuraminidase treatment of purified virions resulted in a shift of the sedimentation value from the higher to the lower density. These results suggest that binding of sialoglycoproteins to the virion surface is responsible for the sedimentation behavior of TGEV. When purified virions were treated with octylglucoside to solubilize viral glycoproteins, ultracentrifugation resulted in sedimentation of the S protein of TGEV. However, when neuraminidase-treated virions or mutants with a defective sialic acid binding activity were analyzed, the S protein remained in the supernatant rather than in the pellet fraction. These results indicate that the interaction of the surface protein S with sialoglycoconjugates is maintained after solubilization of this viral glycoprotein by detergent treatment.


Asunto(s)
Ácido N-Acetilneuramínico/metabolismo , Virus de la Gastroenteritis Transmisible/química , Virus de la Gastroenteritis Transmisible/metabolismo , Proteínas Virales/química , Animales , Centrifugación por Gradiente de Densidad/métodos , Glucósidos/farmacología , Microscopía Electrónica , Neuraminidasa/metabolismo , Solubilidad , Porcinos , Virus de la Gastroenteritis Transmisible/efectos de los fármacos , Proteínas Virales/metabolismo , Virión/química , Virión/efectos de los fármacos , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...