Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Vet J ; 305: 106122, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641200

RESUMEN

The generation of genetically engineered recombinant viruses from modified DNA/RNA is commonly referred to as reverse genetics, which allows the introduction of desired mutations into the viral genome. Reverse genetics systems (RGSs) are powerful tools for studying fundamental viral processes, mechanisms of infection, pathogenesis and vaccine development. However, establishing RGS for coronaviruses (CoVs) and toroviruses (ToVs), which have the largest genomes among vertebrate RNA viruses, is laborious and hampered by technical constraints. Hence, little research has focused on animal CoVs and ToVs using RGSs, especially in large domestic animals such as pigs and cattle. In the last decade, however, studies of porcine CoVs and bovine ToVs using RGSs have been reported. In addition, the coronavirus disease-2019 pandemic has prompted the development of new and simple CoV RGSs, which will accelerate RGS-based research on animal CoVs and ToVs. In this review, we summarise the general characteristics of CoVs and ToVs, the RGSs available for CoVs and ToVs and the progress made in the last decade in RGS-based research on porcine CoVs and bovine ToVs.


Asunto(s)
Coronavirus , Genética Inversa , Torovirus , Animales , Genética Inversa/métodos , Porcinos , Bovinos , Torovirus/genética , Coronavirus/genética , Infecciones por Torovirus/veterinaria , Infecciones por Torovirus/virología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/epidemiología , Enfermedades de los Porcinos/virología , Enfermedades de los Bovinos/virología , Animales Domésticos/virología
2.
Microbiol Spectr ; 12(1): e0192023, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38051050

RESUMEN

IMPORTANCE: Since the pandemic of coronavirus diseases 2019, the use of real-time PCR assay has become widespread among people who were not familiar with it in virus detection. As a result, whether a high real-time PCR value in one time test indicates virus transmissibly became a complicated social problem, regardless of the difference in assays and/or amplification conditions, the time and number of diagnostic test during the time course of infection. In addition, the multiple positives in the test of respiratory viruses further add to the confusion in the interpretation of the infection. To address this issue, we performed virus isolation using pediatric SARI (severe acute respiratory infections) specimens on air-liquid interface culture of human bronchial/tracheal epithelial cell culture. The result of this study can be a strong evidence that the specimens showing positivity for multiple agents in real-time PCR tests possibly contain infectious viruses.


Asunto(s)
Neumonía , Infecciones del Sistema Respiratorio , Virosis , Virus , Humanos , Niño , Infecciones del Sistema Respiratorio/diagnóstico , Virus/genética , Virosis/diagnóstico , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Microbiol Spectr ; : e0459022, 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36744940

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic virus that causes MERS, which is endemic in the Middle East. The absence of human cases in Africa despite the presence of MERS-CoV suggests virological differences between MERS-CoVs in Africa and the Middle East. In fact, in the laboratory, recombinant MERS-CoV carrying the spike (S) protein of Ethiopian isolates exhibits attenuated properties, being more easily neutralized and replicating slower than viruses carrying the S protein of Middle Eastern isolate, EMC. In this study, to identify the amino acids that define the different virological features between Ethiopian and Middle Eastern MERS-CoVs, neutralization titers and viral replication were evaluated using recombinant MERS-CoVs carrying amino acid substitution(s) in the S protein. A single amino acid difference introduced into the receptor binding domain was sufficient to reverse the difference in the neutralizing properties of the S protein between Ethiopian and Middle Eastern MERS-CoVs. Furthermore, amino acid mutations in the S1 and S2 regions of S protein were collectively involved in slow viral replication. Since even a single amino acid difference in S protein can reverse the viral properties of MERS-CoV, it should be noted that multiple mutations may induce a significant change. Careful monitoring of genetic alterations in MERS-CoVs in Africa is therefore required to detect the emergence of virulent strains generated by a few genetic differences. IMPORTANCE There have been no reported cases of human Middle East respiratory syndrome (MERS) in Africa, despite the presence of MERS coronavirus (MERS-CoV). Previous studies have shown that recombinant MERS-CoV carrying the S protein of an Ethiopian isolate replicated slower and was more easily neutralized relative to MERS-CoV carrying the S protein of a Middle Eastern isolate. In this study, we investigated the amino acid(s) in S protein associated with the different viral characteristics between Ethiopian and Middle Eastern MERS-CoVs. The results revealed that a single amino acid difference in the receptor binding domain was sufficient to reverse the neutralization profile. This implies that slight genetic changes can alter the predominant population of MERS-CoV, similar to the transition of variants of severe acute respiratory syndrome coronavirus-2. Careful genetic monitoring of isolates is important to detect the spread of possible virulent MERS-CoVs generated by mutation(s).

4.
Jpn J Infect Dis ; 76(3): 204-206, 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-36575025

RESUMEN

The Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late 2021 and gradually overtook the Delta variant, which was the predominant variant at that time. The Omicron variant has been consecutively replaced by related sublineages. The real-time RT-PCR assays developed by the National Institute of Infectious Diseases (NIID), Japan (i.e., the NIID-N2 and NIID-S2 assays) are the reference assays that have been used in Japan since the outbreak of SARS-CoV-2. To evaluate the applicability of the NIID assays for the Omicron variants, trends in the prevalence of nucleotide mismatches in the primer/probe sequences were traced using sequences registered in the Global Initiative on Sharing Avian Influenza Data database. Approximately 99% of the deposited Omicron variant sequences did not have any mismatches in the NIID assay primer/probes from January to August 2022. This indicates that the NIID assays have been able to detect the changing SARS-CoV-2 Omicron variants.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Animales , SARS-CoV-2/genética , Japón/epidemiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , COVID-19/diagnóstico , COVID-19/epidemiología , Prueba de COVID-19
5.
Nature ; 609(7927): 582-589, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36071157

RESUMEN

Increased levels of proteases, such as trypsin, in the distal intestine have been implicated in intestinal pathological conditions1-3. However, the players and mechanisms that underlie protease regulation in the intestinal lumen have remained unclear. Here we show that Paraprevotella strains isolated from the faecal microbiome of healthy human donors are potent trypsin-degrading commensals. Mechanistically, Paraprevotella recruit trypsin to the bacterial surface through type IX secretion system-dependent polysaccharide-anchoring proteins to promote trypsin autolysis. Paraprevotella colonization protects IgA from trypsin degradation and enhances the effectiveness of oral vaccines against Citrobacter rodentium. Moreover, Paraprevotella colonization inhibits lethal infection with murine hepatitis virus-2, a mouse coronavirus that is dependent on trypsin and trypsin-like proteases for entry into host cells4,5. Consistently, carriage of putative genes involved in trypsin degradation in the gut microbiome was associated with reduced severity of diarrhoea in patients with SARS-CoV-2 infection. Thus, trypsin-degrading commensal colonization may contribute to the maintenance of intestinal homeostasis and protection from pathogen infection.


Asunto(s)
Microbioma Gastrointestinal , Intestino Grueso , Simbiosis , Tripsina , Administración Oral , Animales , Sistemas de Secreción Bacterianos , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Bacteroidetes/aislamiento & purificación , Bacteroidetes/metabolismo , COVID-19/complicaciones , Citrobacter rodentium/inmunología , Diarrea/complicaciones , Heces/microbiología , Microbioma Gastrointestinal/genética , Humanos , Inmunoglobulina A/metabolismo , Intestino Grueso/metabolismo , Intestino Grueso/microbiología , Ratones , Virus de la Hepatitis Murina/metabolismo , Virus de la Hepatitis Murina/patogenicidad , Proteolisis , SARS-CoV-2/patogenicidad , Tripsina/metabolismo , Internalización del Virus
6.
Viruses ; 14(2)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35215991

RESUMEN

Deletions in the spike gene of mouse hepatitis virus (MHV) produce several variants with diverse biological characteristics, highlighting the significance of the spike gene in viral pathogenesis. In this study, we characterized the JHM-X strain, which has a deletion in the hypervariable region (HVR) of the spike gene, compared with the cl-2 strain, which has a full spike gene. Cytopathic effects (CPEs) induced by the two strains revealed that the size of the CPE produced by cl-2 is much greater than that produced by JHM-X in delayed brain tumor (DBT) cells. Thus, this finding explains the greater fusion activity of cl-2 than JHM-X in cultured cells, and we speculate that the deletion region of the spike protein is involved in the fusion activity differences. In contrast with the fusion activity, a comparison of the virus growth kinetics revealed that the titer of JHM-X was approximately 100 times higher than that of cl-2. We found that the deletion region of the spike protein was involved in fusion activity differences, whereas cl-2 produced significantly higher luciferase activity than JHM-X upon similar expression levels of the spike protein. However, the reason behind the growth difference is still unknown. Overall, we discovered that deletion in the HVR of the spike gene could be involved in the fusion activity differences between the two strains.


Asunto(s)
Fusión Celular , Virus de la Hepatitis Murina/patogenicidad , Glicoproteína de la Espiga del Coronavirus/fisiología , Animales , Línea Celular , Ratones , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/fisiología , Eliminación de Secuencia , Glicoproteína de la Espiga del Coronavirus/genética
7.
J Virol ; 96(3): e0156121, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34817201

RESUMEN

Historically part of the coronavirus (CoV) family, torovirus (ToV) was recently classified in the new family Tobaniviridae. While reverse genetics systems have been established for various CoVs, none exist for ToVs. Here, we developed a reverse genetics system using an infectious full-length cDNA clone of bovine ToV (BToV) in a bacterial artificial chromosome (BAC). Recombinant BToV harboring genetic markers had the same phenotype as wild-type (wt) BToV. To generate two types of recombinant virus, the hemagglutinin-esterase (HE) gene was edited, as cell-adapted wtBToV generally loses full-length HE (HEf), resulting in soluble HE (HEs). First, recombinant viruses with HEf and hemagglutinin (HA)-tagged HEf or HEs genes were rescued. These exhibited no significant differences in their effect on virus growth in HRT18 cells, suggesting that HE is not essential for viral replication in these cells. Thereafter, we generated a recombinant virus (rEGFP) wherein HE was replaced by the enhanced green fluorescent protein (EGFP) gene. rEGFP expressed EGFP in infected cells but showed significantly lower levels of viral growth than wtBToV. Moreover, rEGFP readily deleted the EGFP gene after one passage. Interestingly, rEGFP variants with two mutations (C1442F and I3562T) in nonstructural proteins (NSPs) that emerged during passage exhibited improved EGFP expression, EGFP gene retention, and viral replication. An rEGFP into which both mutations were introduced displayed a phenotype similar to that of these variants, suggesting that the mutations contributed to EGFP gene acceptance. The current findings provide new insights into BToV, and reverse genetics will help advance the current understanding of this neglected pathogen. IMPORTANCE ToVs are diarrhea-causing pathogens detected in various species, including humans. Through the development of a BAC-based BToV, we introduced the first reverse genetics system for Tobaniviridae. Utilizing this system, recombinant BToVs with a full-length HE gene were generated. Remarkably, although clinical BToVs generally lose the HE gene after a few passages, some recombinant viruses generated in the current study retained the HE gene for up to 20 passages while accumulating mutations in NSPs, which suggested that these mutations may be involved in HE gene retention. The EGFP gene of recombinant viruses was unstable, but rEGFP into which two NSP mutations were introduced exhibited improved EGFP expression, gene retention, and viral replication. These data suggested the existence of an NSP-based acceptance or retention mechanism for exogenous RNA or HE genes. Recombinant BToVs and reverse genetics are powerful tools for understanding fundamental viral processes, pathogenesis, and BToV vaccine development.


Asunto(s)
ADN Complementario , Genoma Viral , Genética Inversa , Torovirus/genética , Animales , Bovinos , Enfermedades de los Bovinos/virología , Línea Celular , Células Cultivadas , Cromosomas Artificiales Bacterianos , Clonación Molecular , Genes Reporteros , Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Mutación , Plásmidos/genética , Torovirus/aislamiento & purificación , Infecciones por Torovirus , Transfección
8.
Viruses ; 13(3)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33800523

RESUMEN

Torovirus (ToV) has recently been classified into the new family Tobaniviridae, although it belonged to the Coronavirus (CoV) family historically. ToVs are associated with enteric diseases in animals and humans. In contrast to CoVs, which are recognised as pathogens of veterinary and medical importance, little attention has been paid to ToVs because their infections are usually asymptomatic or not severe; for a long time, only one equine ToV could be propagated in cultured cells. However, bovine ToVs, which predominantly cause diarrhoea in calves, have been detected worldwide, leading to economic losses. Porcine ToVs have also spread globally; although they have not caused serious economic losses, coinfections with other pathogens can exacerbate their symptoms. In addition, frequent inter- or intra-recombination among ToVs can increase pathogenesis or unpredicted host adaptation. These findings have highlighted the importance of ToVs as pathogens and the need for basic ToV research. Here, we review recent progress in the study of ToV molecular biology including reverse genetics, focusing on the similarities and differences between ToVs and CoVs.


Asunto(s)
Infecciones por Torovirus/virología , Torovirus/fisiología , Animales , Coronavirus/genética , Coronavirus/fisiología , Infecciones por Coronavirus/virología , Humanos , Torovirus/genética
9.
J Virol ; 95(3)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33177195

RESUMEN

Torovirus (ToV) has recently been classified into the new family Tobaniviridae, although historically, it belonged to the Coronavirus (CoV) family. The nucleocapsid (N) proteins of CoVs are predominantly localized in the cytoplasm, where the viruses replicate, but in some cases the proteins are partially located in the nucleolus. Many studies have investigated the subcellular localization and nucleocytoplasmic trafficking signals of the CoV N proteins, but little is known about ToV N proteins. Here, we studied the subcellular localization of the bovine ToV (BToV) N protein (BToN) and characterized its nucleocytoplasmic trafficking signals. Unlike other CoVs, BToN in infected cells was transported mainly to the nucleolus during early infection but was distributed predominantly in the nucleoplasm rather than in the nucleolus during late infection. Interestingly, a small quantity of BToN was detected in the cytoplasm during infection. Examination of a comprehensive set of substitution or deletion mutants of BToN fused with enhanced green fluorescent protein (EGFP) revealed that clusters of arginine (R) residues comprise nuclear/nucleolar localization signals (NLS/NoLS), and the C-terminal region served as a chromosomal maintenance 1 (CRM1)-independent nuclear export signal (NES). Moreover, recombinant viruses with mutations in the NLS/NoLS, but retaining nuclear accumulation, were successfully rescued and showed slightly reduced growth ability, while the virus that lost the NLS/NoLS-mediated nuclear accumulation of BToN was not rescued. These results indicate that BToN uniquely accumulates mainly in nuclear compartments during infection, regulated by an R-rich NLS/NoLS and a CRM1-independent NES, and that the BToN accumulation in the nuclear compartment driven by NLS/NoLS is important for virus growth.IMPORTANCE ToVs are diarrhea-causing pathogens detected in many species, including humans. BToV has spread worldwide, leading to economic loss, and there is currently no treatment or vaccine available. Positive-stranded RNA viruses, including ToVs, replicate in the cytoplasm, and their structural proteins generally accumulate in the cytoplasm. Interestingly, BToN accumulated predominantly in the nucleus/nucleolus during all infectious processes, with only a small fraction accumulating in the cytoplasm despite being a major structural protein. Furthermore, we identified unique nucleocytoplasmic trafficking signals and demonstrated the importance of NLS/NoLS for virus growth. This study is the first to undertake an in-depth investigation of the subcellular localization and intracellular trafficking signals of BToN. Our findings additionally suggest that the NLS/NoLS-mediated nuclear accumulation of BToN is important for virus replication. An understanding of the unique features of BToV may provide novel insights into the assembly mechanisms of not only ToVs but also other positive-stranded RNA viruses.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/metabolismo , Torovirus/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Nucléolo Celular/metabolismo , Citoplasma/metabolismo , Humanos , Mutación , Señales de Exportación Nuclear , Señales de Localización Nuclear , Proteínas de la Nucleocápside/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Torovirus/crecimiento & desarrollo , Torovirus/metabolismo , Replicación Viral/genética
10.
J Virol ; 95(1)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33055254

RESUMEN

Here, we screened steroid compounds to obtain a drug expected to block host inflammatory responses and Middle East respiratory syndrome coronavirus (MERS-CoV) replication. Ciclesonide, an inhaled corticosteroid, suppressed the replication of MERS-CoV and other coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of coronavirus disease 2019 (COVID-19), in cultured cells. The 90% effective concentration (EC90) of ciclesonide for SARS-CoV-2 in differentiated human bronchial tracheal epithelial cells was 0.55 µM. Eight consecutive passages of 43 SARS-CoV-2 isolates in the presence of ciclesonide generated 15 resistant mutants harboring single amino acid substitutions in nonstructural protein 3 (nsp3) or nsp4. Of note, ciclesonide suppressed the replication of all these mutants by 90% or more, suggesting that these mutants cannot completely overcome ciclesonide blockade. Under a microscope, the viral RNA replication-transcription complex in cells, which is thought to be detectable using antibodies specific for nsp3 and double-stranded RNA, was observed to fall in the presence of ciclesonide in a concentration-dependent manner. These observations indicate that the suppressive effect of ciclesonide on viral replication is specific to coronaviruses, highlighting it as a candidate drug for the treatment of COVID-19 patients.IMPORTANCE The outbreak of SARS-CoV-2, the cause of COVID-19, is ongoing. New and effective antiviral agents that combat the disease are needed urgently. Here, we found that an inhaled corticosteroid, ciclesonide, suppresses the replication of coronaviruses, including betacoronaviruses (murine hepatitis virus type 2 [MHV-2], MERS-CoV, SARS-CoV, and SARS-CoV-2) and an alphacoronavirus (human coronavirus 229E [HCoV-229E]), in cultured cells. Ciclesonide is safe; indeed, it can be administered to infants at high concentrations. Thus, ciclesonide is expected to be a broad-spectrum antiviral drug that is effective against many members of the coronavirus family. It could be prescribed for the treatment of MERS and COVID-19.


Asunto(s)
COVID-19/metabolismo , Pregnenodionas/farmacología , ARN Bicatenario/biosíntesis , ARN Viral/biosíntesis , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos , Animales , Chlorocebus aethiops , Perros , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Células Vero , Tratamiento Farmacológico de COVID-19
11.
Infect Genet Evol ; 75: 103975, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31344488

RESUMEN

Enterovirus G (EV-G) belongs to the family of Picornaviridae. Two types of recombinant porcine EV-Gs carrying papain-like cysteine protease (PLCP) gene of porcine torovirus, a virus in Coronaviridae, are reported. Type 1 recombinant EV-Gs are detected in pig feces in Japan, USA, and Belgium and carry the PLPC gene at the junction site of 2C/3A genes, while PLPC gene replaces the viral structural genes in type 2 recombinant EV-G detected in pig feces in a Chinese farm. We identified a novel type 2 recombinant EV-G carrying the PLCP gene with flanking sequences in place of the viral structural genes in pig feces in Japan. The ~0.3 kb-long upstream flanking sequence had no sequence homology with any proteins deposited in GenBank, while the downstream ~0.9 kb-long flanking sequence included a domain having high amino acid sequence homology with a baculoviral inhibitor of apoptosis repeat superfamily. The pig feces, where the novel type 2 recombinant EV-G was detected, also carried type 1 recombinant EV-G. The amount of type 1 and type 2 recombinant EV-G genomes was almost same in the pig feces. Although the phylogenetic analysis suggested that these two recombinant EV-Gs have independently evolved, type 1 recombinant EV-G might have served as a helper virus by providing viral structural proteins for dissemination of the type 2 recombinant EV-G.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteasas de Cisteína/genética , Infecciones por Enterovirus/veterinaria , Enterovirus Porcinos/genética , Proteínas Estructurales Virales/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteasas de Cisteína/metabolismo , Infecciones por Enterovirus/epidemiología , Infecciones por Enterovirus/virología , Heces/virología , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Filogenia , Porcinos , Enfermedades de los Porcinos/epidemiología , Enfermedades de los Porcinos/virología
12.
J Vet Med Sci ; 79(7): 1155-1162, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28529244

RESUMEN

Cancer stem cells or tumor-initiating cells (TICs) are a small subpopulation of cells that have the capacity to self-renew, differentiate and initiate tumors. These cells may function in tumor initiation, aggression and recurrence. Whether spheres derived from canine rhabdomyosarcoma cells have stem cell-like properties is unclear. We induced sphere formation in the canine rhabdomyosarcoma cell lines, CMS-C and CMS-J, and characterized the spheres in vitro and in vivo. Sphere-forming cells were more resistant to vincristine, mitoxantrone and doxorubicin than adherent cells. Xenograft transplantation demonstrated that 1 × 103 sphere-forming cells derived from CMS-C were sufficient for tumor formation. The sphere assay showed that the sphere-forming cells were present in these tumors. These results suggest that the spheres derived from canine rhabdomyosarcoma cells may possess characteristics of TICs. This study provides the foundation for elucidating the contribution of TICs to rhabdomyosarcoma tumorigenesis.


Asunto(s)
Enfermedades de los Perros/patología , Neoplasias de los Músculos/veterinaria , Células Madre Neoplásicas/citología , Rabdomiosarcoma/veterinaria , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Perros , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Femenino , Ratones , Ratones Endogámicos BALB C , Mitoxantrona/farmacología , Neoplasias de los Músculos/tratamiento farmacológico , Neoplasias de los Músculos/patología , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Vincristina/farmacología
13.
Virus Res ; 222: 113-119, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27292080

RESUMEN

To evaluate the mechanism by which a large outbreak of porcine epidemic diarrhea (PED) occurred in Japan, where the majority of sows are vaccinated, we isolated two new strains of PED virus (PEDV) from the intestines of piglets and found that they showed greater similarity to US isolates (group II PEDV) than to the Japanese vaccine strain (group I PEDV). We compared the antigenicity of the vaccine type strain and newly isolated strains by means of a neutralization test using sera from a number of pigs from various farms; the results revealed that they are antigenically similar. This is the first report of the similarity of group I and II viruses using sera from individual pigs vaccinated with group I virus. These data suggest that the large outbreak of PED in Japan cannot be attributed to inefficient vaccination but may be due to the extremely high virulence of the newly appearing viruses.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/inmunología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Japón , Pruebas de Neutralización , Filogenia , Virus de la Diarrea Epidémica Porcina/clasificación , Virus de la Diarrea Epidémica Porcina/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Porcinos , Enfermedades de los Porcinos/epidemiología
14.
J Gen Virol ; 97(8): 1853-1864, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27145752

RESUMEN

The cytoplasmic tails of some coronavirus (CoV) spike (S) proteins contain an endoplasmic reticulum retrieval signal (ERRS) that can retrieve S proteins from the Golgi to the endoplasmic reticulum (ER); this process is thought to accumulate S proteins at the CoV budding site, the ER-Golgi intermediate compartment (ERGIC), and to facilitate S protein incorporation into virions. However, we showed previously that porcine epidemic diarrhoea CoV S proteins lacking the ERRS were efficiently incorporated into virions, similar to the original virus. Thus, the precise role of the ERRS in virus assembly remains unclear. Here, the roles of the S protein ERRS in severe acute respiratory syndrome CoV (SARS-CoV) intracellular trafficking and S incorporation into virus-like particles (VLPs) are described. Intracellular trafficking and indirect immunofluorescence analysis suggested that when M protein was present, wild-type S protein (wtS) could be retained in the pre- and post-medial Golgi compartments intracellularly and co-localized with M protein in the Golgi. In contrast, mutant S protein lacking the ERRS was distributed throughout the ER and only partially co-localized with M protein. Moreover, the intracellular accumulation of mutant S protein, particularly at the post-medial Golgi compartment, was significantly reduced compared with wtS. A VLP assay suggested that wtS that reached the post-medial compartment could be returned to the ERGIC for subsequent incorporation into VLPs, while mutant S protein could not. These results suggest that the ERRS of SARS-CoV contributes to intracellular S protein accumulation specifically in the post-medial Golgi compartment and to S protein incorporation into VLPs.


Asunto(s)
Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Virosomas/metabolismo , Ensamble de Virus , Animales , Línea Celular , Proteínas M de Coronavirus , Aparato de Golgi/química , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Señales de Clasificación de Proteína , Transporte de Proteínas , Glicoproteína de la Espiga del Coronavirus/genética , Proteínas de la Matriz Viral/metabolismo
15.
Virus Res ; 210: 213-26, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26277777

RESUMEN

Human respiratory syncytial virus (RSV) is a major causative agent of respiratory tract infections in children worldwide. Preterm children or those with underlying cardiopulmonary disorders are at particularly high risk of developing severe and lethal RSV respiratory tract infections; however, there are currently no effective vaccines or anti-viral drugs. To identify targets for the development of drugs to treat RSV infections, we investigated host cell factors involved in the replication of RSV. To this end, MDCK cells with low susceptibility to RSV were transfected with cDNA libraries derived from RSV-susceptible human lung or HeLa cells. A microarray analysis was subsequently performed on parental MDCK cells and MDCK cells that were converted to an RSV-susceptible form. Among the genes identified, chemokine (C-C motif) ligand 2 (CCL2), retinoic acid receptor responder protein 2 (RARRES2) and ephrin-B2 (EFNB2) had a positive effect on RSV replication. Expression of these genes in MDCK cells resulted in a 10- to 100-fold increase in RSV replication. CCL2 expression also disrupted the distribution of claudin-1, a tight junction protein, suggesting that CCL2 plays a role in claudin-based tight junction formation during RSV replication. The knockdown of EFNB2 and RARRES2 by siRNA in RSV-susceptible cell lines (HEp-2 and A549) resulted in reduced RSV replication, suggesting that EFNB2 and RARRES2 participate in RSV replication. Together, our findings suggest that CCL2, RARRES2 and EFNB2 are host cell factors involved in RSV replication.


Asunto(s)
Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Efrina-B2/metabolismo , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Virus Sincitial Respiratorio Humano/fisiología , Replicación Viral , Animales , Línea Celular , Perfilación de la Expresión Génica , Humanos , Análisis por Micromatrices
16.
PLoS One ; 10(6): e0130208, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26079133

RESUMEN

A/Narita/1/2009 (A/N) was the first H1N1 virus from the 2009 pandemic (H1pdm) to be isolated in Japan. To better understand and predict the possible development of this virus strain, the effect of passaging A/N was investigated in Madin-Darby canine kidney cells, chicken eggs and mice. A/N that had been continuously passaged in cells, eggs, or mice obtained the ability to grow efficiently in each host. Moreover, A/N grown in mice had both a high level of pathogenicity in mice and an increased growth rate in cells and eggs. Changes in growth and pathogenicity were accompanied by amino acid substitutions in viral hemagglutinin (HA) and PB2. In addition, the adapted viruses exhibited a reduced ability to react with ferret antisera against A/N. In conclusion, prolonged passaging allowed influenza A/N to adapt to different hosts, as indicated by a high increase in proliferative capacity that was accompanied by an antigenic alteration leading to amino acid substitutions.


Asunto(s)
Adaptación Fisiológica , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/fisiología , Sustitución de Aminoácidos , Animales , Pollos , Perros , Femenino , Hurones/virología , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Japón , Cinética , Células de Riñón Canino Madin Darby , Ratones , Óvulo/virología
17.
Viruses ; 7(4): 1700-25, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25855243

RESUMEN

The envelopes of coronaviruses (CoVs) contain primarily three proteins; the two major glycoproteins spike (S) and membrane (M), and envelope (E), a non-glycosylated protein. Unlike other enveloped viruses, CoVs bud and assemble at the endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC). For efficient virion assembly, these proteins must be targeted to the budding site and to interact with each other or the ribonucleoprotein. Thus, the efficient incorporation of viral envelope proteins into CoV virions depends on protein trafficking and protein-protein interactions near the ERGIC. The goal of this review is to summarize recent findings on the mechanism of incorporation of the M and S glycoproteins into the CoV virion, focusing on protein trafficking and protein-protein interactions.


Asunto(s)
Coronavirus/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo , Ensamble de Virus , Unión Proteica , Multimerización de Proteína , Transporte de Proteínas
18.
Arch Virol ; 158(7): 1561-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23420207

RESUMEN

Bovine torovirus (BToV)-Aichi, recently isolated in cultured cells, showed hemagglutination (HA) activity, although the virus has a truncated hemagglutinin-esterase (HE) protein, judging from its gene structure, indicating the existence of another viral protein with HA activity. We examined whether the spike (S) protein possesses HA activity. A BToV antiserum used in this study, reactive to S but not to HE, inhibited HA activity. Furthermore, cells infected with BToV and those expressing S showed hemadsorption (HAD) activity, which was inhibited by the anti-BToV serum; however, HAD activity by expressed HE was not blocked. These data indicate that the S protein of BToV-Aichi is responsible for its HA activity.


Asunto(s)
Hemaglutinación , Glicoproteínas de Membrana/metabolismo , Torovirus/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Factores de Virulencia/metabolismo , Animales , Eritrocitos/virología , Glicoproteína de la Espiga del Coronavirus , Acoplamiento Viral
19.
J Gen Virol ; 93(Pt 4): 823-828, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238235

RESUMEN

The endodomain of several coronavirus (CoV) spike (S) proteins contains palmitylated cysteine residues and enables co-localization and interaction with the CoV membrane (M) protein. Depalmitylation of mouse hepatitis virus S proteins abolished this interaction, resulting in the failure of S incorporation into virions. In contrast, an immunofluorescence assay (IFA) showed that depalmitylated severe acute respiratory syndrome coronavirus (SCoV) S proteins still co-localized with the M protein in the budding site. Here, we determined the ability of depalmitylated SCoV S mutants to incorporate S into virus-like particles (VLPs). IFA confirmed that all SCoV S mutants co-localized with the M protein intracellularly. However, the mutants lacking two cysteine residues (C(1234/1235)) failed to incorporate S into VLPs. This indicated that these palmitylated cysteines are essential for S incorporation, but are not involved in S co-localization mediated by the M protein. Our findings suggest that M-S co-localization and S incorporation occur independently of one another in SCoV virion assembly.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Síndrome Respiratorio Agudo Grave/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas de la Matriz Viral/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas M de Coronavirus , Cisteína/metabolismo , Ratones , Ácido Palmítico/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Glicoproteína de la Espiga del Coronavirus , Virión/metabolismo , Virión/fisiología , Ensamble de Virus/fisiología
20.
J Med Virol ; 84(2): 365-70, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22170560

RESUMEN

Previously, it was reported that productive viral infection, viral protein synthesis, and viral RNA replication of respiratory syncytial virus (RSV) operated efficiently in two human epithelial cell lines (HEp-2 and A549), but not in a human mast-cell line, HMC-1. Based on these observations, it was hypothesized that HMC-1 cells lack the machinery required for RSV replication. To identify the host factors required for RSV replication, cDNA subtraction using A549, HEp-2, and HMC-1 cells was performed, and cytokeratin 18 (C18) was identified as a candidate host factor. Because C18 is generally expressed in simple epithelia with cytokeratin 8 (C8), HMC-1 cells that constitutively express C18 and C8 (HMC-1-C8/18) were established to evaluate the role of C8/18 in RSV replication. In HMC-1-C8/18 cells, RSV RNA replication was increased, and the amount of infective virus produced was also increased in the cellular fraction after RSV spinoculation, whereas RSV production was decreased in A549 cells in which C18 expression was knocked down. These data suggest that the replication of RSV increases in the presence of C8/18.


Asunto(s)
Queratina-18/metabolismo , Queratina-8/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Replicación Viral , Línea Celular , Humanos , Queratina-18/genética , Queratina-8/genética , ARN Viral/biosíntesis , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA