Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 184(19): 4848-4856, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34480864

RESUMEN

Since the first reports of a novel severe acute respiratory syndrome (SARS)-like coronavirus in December 2019 in Wuhan, China, there has been intense interest in understanding how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in the human population. Recent debate has coalesced around two competing ideas: a "laboratory escape" scenario and zoonotic emergence. Here, we critically review the current scientific evidence that may help clarify the origin of SARS-CoV-2.


Asunto(s)
SARS-CoV-2/fisiología , Animales , Evolución Biológica , COVID-19/virología , Humanos , Laboratorios , SARS-CoV-2/genética , Zoonosis/virología
2.
Biotechnol Bioeng ; 118(5): 2067-2075, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33615450

RESUMEN

Heat treatment denatures viral proteins that comprise the virion, making the virus incapable of infecting a host. Coronavirus (CoV) virions contain single-stranded RNA genomes with a lipid envelope and four proteins, three of which are associated with the lipid envelope and thus are thought to be easily denatured by heat or surfactant-type chemicals. Prior studies have shown that a temperature as low as 75°C with a treatment duration of 15 min can effectively inactivate CoV. The degree of CoV heat inactivation greatly depends on the length of heat treatment time and the temperature applied. With the goal of finding whether sub-second heat exposure of CoV can sufficiently inactivate CoV, we designed and developed a simple fluidic system that can measure sub-second heat inactivation of CoV. The system is composed of a stainless-steel capillary immersed in a temperature-controlled oil bath followed by an ice bath, through which virus solution can flow at various speeds. Flowing virus solution at different speeds, along with temperature control and monitoring system, allows the virus to be exposed to the desired temperature and treatment durations with high accuracy. Using mouse hepatitis virus, a betacoronavirus, as a model CoV system, we identified that 71.8°C for 0.51 s exposure is sufficient to obtain >5 Log10 reduction in viral titer (starting titer: 5 × 107 PFU/ml), and that when exposed to 83.4°C for 1.03 s, the virus was completely inactivated (>6 Log10 reduction).


Asunto(s)
Betacoronavirus/fisiología , Calor , Inactivación de Virus , Virus de la Hepatitis Murina/fisiología , Ensayo de Placa Viral
3.
J Gen Virol ; 96(Pt 3): 494-506, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25252685

RESUMEN

Coronaviruses (CoVs) have been studied for over 60 years, but have only recently gained notoriety as deadly human pathogens with the emergence of severe respiratory syndrome CoV and Middle East respiratory syndrome virus. The rapid emergence of these viruses has demonstrated the need for good models to study severe CoV respiratory infection and pathogenesis. There are, currently, different methods and models for the study of CoV disease. The available genetic methods for the study and evaluation of CoV genetics are reviewed here. There are several animal models, both mouse and alternative animals, for the study of severe CoV respiratory disease that have been examined, each with different pros and cons relative to the actual pathogenesis of the disease in humans. A current limitation of these models is that no animal model perfectly recapitulates the disease seen in humans. Through the review and analysis of the available disease models, investigators can employ the most appropriate available model to study various aspects of CoV pathogenesis and evaluate possible antiviral treatments that may potentially be successful in future treatment and prevention of severe CoV respiratory infections.


Asunto(s)
Infecciones por Coronavirus/virología , Coronavirus/genética , Coronavirus/patogenicidad , Neumonía/virología , Animales , Infecciones por Coronavirus/genética , Humanos
4.
Pharmaceutics ; 16(4)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38675125

RESUMEN

New antiviral agents are essential to improving treatment and control of SARS-CoV-2 infections that can lead to the disease COVID-19. Antimicrobial peptoids are sequence-specific oligo-N-substituted glycine peptidomimetics that emulate the structure and function of natural antimicrobial peptides but are resistant to proteases. We demonstrate antiviral activity of a new peptoid (TM9) against the coronavirus, murine hepatitis virus (MHV), as a closely related model for the structure and antiviral susceptibility profile of SARS-CoV-2. This peptoid mimics the human cathelicidin LL-37, which has also been shown to have antimicrobial and antiviral activity. In this study, TM9 was effective against three murine coronavirus strains, demonstrating that the therapeutic window is large enough to allow the use of TM9 for treatment. All three isolates of MHV generated infection in mice after 15 min of exposure by aerosol using the Madison aerosol chamber, and all three viral strains could be isolated from the lungs throughout the 5-day observation period post-infection, with the peak titers on day 2. MHV-A59 and MHV-A59-GFP were also isolated from the liver, heart, spleen, olfactory bulbs, and brain. These data demonstrate that MHV serves as a valuable natural murine model of coronavirus pathogenesis in multiple organs, including the brain.

5.
J Biol Chem ; 287(10): 7063-73, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22241479

RESUMEN

Coronavirus (CoV) nucleocapsid (N) protein contains two structurally independent RNA binding domains. These are denoted N-terminal domain (NTD) and C-terminal domain and are joined by a charged linker region rich in serine and arginine residues (SR linker). In mouse hepatitis virus (MHV), the NTD binds the transcriptional regulatory sequence (TRS) RNA, a conserved hexanucleotide sequence required for subgenomic RNA synthesis. The NTD is also capable of disrupting a short RNA duplex. We show here that three residues on the ß3 (Arg-125 and Tyr-127) and ß5 (Tyr-190) strands play key roles in TRS RNA binding and helix destabilization with Ala substitutions of these residues lethal to the virus. NMR studies of the MHV NTD·TRS complex revealed that this region defines a major RNA binding interface in MHV with site-directed spin labeling studies consistent with a model in which the adenosine-rich 3'-region of TRS is anchored by Arg-125, Tyr-127, and Tyr-190 in a way that is critical for efficient subgenomic RNA synthesis in MHV. Characterization of CoV N NTDs from infectious bronchitis virus and from severe acute respiratory syndrome CoV revealed that, although detailed NTD-TRS determinants are distinct from those of MHV NTD, rapid helix destabilization activity of CoV N NTDs is most strongly correlated with CoV function and virus viability.


Asunto(s)
Modelos Moleculares , Virus de la Hepatitis Murina/química , Proteínas de la Nucleocápside/química , ARN Viral/química , Proteínas de Unión al ARN/química , Sustitución de Aminoácidos , Animales , Línea Celular , Ratones , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/metabolismo , Mutación Missense , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN Viral/genética , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
J Virol ; 86(8): 4294-304, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301153

RESUMEN

Coronaviruses encode an endoribonuclease, Nsp15, which has a poorly defined role in infection. Sequence analysis revealed a retinoblastoma protein-binding motif (LXCXE/D) in the majority of the Nsp15 of the severe acute respiratory syndrome coronavirus (SARS-CoV) and its orthologs in the alpha and beta coronaviruses. The endoribonuclease activity of the SARS-CoV Nsp15 (sNsp15) was stimulated by retinoblastoma protein (pRb) in vitro, and the two proteins can be coimmunoprecipitated from cellular extracts. Mutations in the pRb-binding motif rendered sNsp15 to be differentially modified by ubiquitin in cells, and cytotoxicity was observed upon its expression. Expression of the sNsp15 in cells resulted in an increased abundance of pRb in the cytoplasm, decreased overall levels of pRb, an increased proportion of cells in the S phase of the cell cycle, and an enhanced expression from a promoter normally repressed by pRb. The endoribonuclease activity of the mouse hepatitis virus (MHV) A59 Nsp15 was also increased by pRb in vitro, and an MHV with mutations in the LXCXE/D-motif, named vLC, exhibited a smaller plaque diameter and reduced the virus titer by ∼1 log. Overexpression of pRb delayed the viral protein production by wild-type MHV but not by vLC. This study reveals that pRb and its interaction with Nsp15 can affect coronavirus infection and adds coronaviruses to a small but growing family of RNA viruses that encode a protein to interact with pRb.


Asunto(s)
Endorribonucleasas/metabolismo , Virus de la Hepatitis Murina/enzimología , Proteína de Retinoblastoma/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Sitios de Unión , Línea Celular , Infecciones por Coronavirus/metabolismo , Cricetinae , Endorribonucleasas/genética , Regulación de la Expresión Génica , Humanos , Ratones , Modelos Moleculares , Virus de la Hepatitis Murina/genética , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteína de Retinoblastoma/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
7.
Am J Vet Res ; 84(9)2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37442546

RESUMEN

OBJECTIVE: To examine the susceptibility of cultured primary equine bronchial epithelial cells (EBECs) to a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudovirus relative to human bronchial epithelial cells (HBECs). SAMPLE: Primary EBEC cultures established from healthy adult horses and commercially sourced human bronchial epithelial cells (HBECs) were used as a positive control. METHODS: Angiotensin-converting enzyme 2 (ACE2) expression by EBECs was demonstrated using immunofluorescence, western immunoblot, and flow cytometry. EBECs were transduced with a lentivirus pseudotyped with the SARS-CoV-2 spike protein that binds to ACE2 and expresses the enhanced green fluorescent protein (eGFP) as a reporter. Cells were transduced with the pseudovirus at a multiplicity of infection of 0.1 for 6 hours, washed, and maintained in media for 96 hours. After 96 hours, eGFP expression in EBECs was assessed by fluorescence microscopy of cell cultures and quantitative PCR. RESULTS: ACE2 expression in EBECs detected by immunofluorescence, western immunoblotting, and flow cytometry was lower in EBECs than in HBECs. After 96 hours, eGFP expression in EBECs was demonstrated by fluorescence microscopy, and mean ΔCt values from quantitative PCR were significantly (P < .0001) higher in EBECs (8.78) than HBECs (3.24) indicating lower infectivity in EBECs. CLINICAL RELEVANCE: Equine respiratory tract cells were susceptible to cell entry with a SARS-CoV-2 pseudovirus. Lower replication efficiency in EBECs suggests that horses are unlikely to be an important zoonotic host of SARS-CoV-2, but viral mutations could render some strains more infective to horses. Serological and virological monitoring of horses in contact with persons shedding SARS-CoV-2 is warranted.


Asunto(s)
COVID-19 , Enfermedades de los Caballos , Caballos , Animales , Humanos , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Internalización del Virus , COVID-19/veterinaria , Células Epiteliales
8.
J Virol ; 84(18): 9278-91, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20631137

RESUMEN

We report here investigation into the genetic basis of mouse hepatitis virus strain 1 (MHV-1) pneumovirulence. Sequencing of the 3' one-third of the MHV-1 genome demonstrated that the genetic organization of MHV-1 was similar to that of other strains of MHV. The hemagglutinin esterase (HE) protein was truncated, and reverse transcription-PCR (RT-PCR) studies confirmed previous work that suggested that the MHV-1 HE is a pseudogene. Targeted recombination was used to select chimeric viruses containing either the MHV-1 S gene or genes encoding all of the MHV-1 structural proteins, on an MHV-A59 background. Challenge studies in mice demonstrated that expression of the MHV-1 S gene within the MHV-A59 background (rA59/S(MHV-1)) increased the pneumovirulence of MHV-A59, and mice infected with this recombinant virus developed pulmonary lesions that were similar to those observed with MHV-1, although rA59/S(MHV-1) was significantly less virulent. Chimeras containing all of the MHV-1 structural genes on an MHV-A59 background were able to reproduce the severe acute respiratory syndrome (SARS)-like pathology observed with MHV-1 and reproducibly increased pneumovirulence relative to rA59/S(MHV-1), but were still much less virulent than MHV-1. These data suggest that important determinants of pneumopathogenicity are contained within the 3' one-third of the MHV-1 genome, but additional important virulence factors must be encoded in the genome upstream of the S gene. The severity of the pulmonary lesions observed correlates better with elevated levels of inflammatory cytokines than with viral replication in the lungs, suggesting that pulmonary disease has an important immunological component.


Asunto(s)
Pulmón/patología , Pulmón/virología , Glicoproteínas de Membrana/fisiología , Virus de la Hepatitis Murina/patogenicidad , Proteínas del Envoltorio Viral/fisiología , Factores de Virulencia/fisiología , Animales , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Femenino , Orden Génico , Genes Virales , Glicoproteínas de Membrana/genética , Ratones , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/genética , Neumonía Viral/patología , Neumonía Viral/virología , ARN Viral/química , ARN Viral/genética , Recombinación Genética , Análisis de Secuencia de ADN , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral/genética , Factores de Virulencia/genética
9.
J Virol ; 83(23): 12084-93, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19759148

RESUMEN

Stem-loop 2 (SL2) of the 5'-untranslated region of the mouse hepatitis virus (MHV) contains a highly conserved pentaloop (C47-U48-U49-G50-U51) stacked on a 5-bp stem. Solution nuclear magnetic resonance experiments are consistent with a 5'-uYNMG(U)a or uCUYG(U)a tetraloop conformation characterized by an anti-C47-syn-G50 base-pairing interaction, with U51 flipped out into solution and G50 stacked on A52. Previous studies showed that U48C and U48A substitutions in MHV SL2 were lethal, while a U48G substitution was viable. Here, we characterize viruses harboring all remaining single-nucleotide substitutions in the pentaloop of MHV SL2 and also investigate the degree to which the sequence context of key pentaloop point mutations influences the MHV replication phenotype. U49 or U51 substitution mutants all are viable; C47 substitution mutants also are viable but produce slightly smaller plaques than wild-type virus. In contrast, G50A and G50C viruses are severely crippled and form much smaller plaques. Virus could not be recovered from G50U-containing mutants; rather, only true wild-type revertants or a virus, G50U/C47A, containing a second site mutation were recovered. These functional data suggest that the Watson-Crick edges of C47 and G50 (or A47 and U50 in the G50U/C47A mutant) are in close enough proximity to a hydrogen bond with U51 flipped out of the hairpin. Remarkably, increasing the helical stem stability rescues the previously lethal mutants U48C and G50U. These studies suggest that SL2 functions as an important, but rather plastic, structural element in stimulating subgenomic RNA synthesis in coronaviruses.


Asunto(s)
Virus de la Hepatitis Murina/fisiología , Conformación de Ácido Nucleico , Mutación Puntual , ARN Viral/química , ARN Viral/genética , Regiones no Traducidas 5' , Animales , Emparejamiento Base , Línea Celular , Cricetinae , Espectroscopía de Resonancia Magnética , Ensayo de Placa Viral , Replicación Viral
10.
Lab Chip ; 20(9): 1628-1638, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32196032

RESUMEN

Identifying antibodies (Abs) that neutralize infectious agents is the first step for developing therapeutics, vaccines, and diagnostic tools for these infectious agents. However, current approaches for identifying neutralizing Abs (nAbs) typically rely on dilution-based assays that are costly, inefficient, and only survey a small subset of the entire repertoire. There are also intrinsic biases in many steps of conventional nAb identification processes. More importantly, conventional assays rely on simple Ab-antigen binding assays, which may not result in identifying the most potent nAbs, as the strongest binder may not be the most potent nAb. Droplet microfluidic systems have the capability to overcome such limitations by conducting complex multi-step assays with high reliability, resolution, and throughput in a pico-liter volume water-in-oil emulsion droplet format. Here, we describe the development of PRESCIENT (Platform for the Rapid Evaluation of antibody SucCess using Integrated microfluidics ENabled Technology), a droplet microfluidic system that can enable high-throughput single-cell resolution identification of nAb repertoires elicited in response to viral infection. We demonstrate PRESCIENT's ability to identify Abs that neutralize a model viral agent, Murine coronavirus (murine hepatitis virus), which causes high mortality rates in experimentally infected mice. In-droplet infection of host cells by the virus was first demonstrated, followed by demonstration of in-droplet neutralization by nAbs produced from a single Ab-producing hybridoma cell. Finally, fluorescence intensity analyses of two populations of hybridoma cell lines (nAb-producing and non-nAb-producing hybridoma cell lines) successfully discriminated between the two populations. The presented strategy and platform have the potential to identify and investigate neutralizing activities against a broad range of potential infectious agents for which nAbs have yet to be discovered, significantly advancing the nAb identification process as well as reinvigorating the field of Ab discovery, characterization, and development.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Coronavirus/inmunología , Microfluídica/métodos , Animales , Línea Celular , Coronavirus/aislamiento & purificación , Dispositivos Laboratorio en un Chip , Ratones , Virosis/diagnóstico , Virosis/veterinaria , Virosis/virología
11.
J Virol ; 81(24): 13587-97, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17898055

RESUMEN

The goal of this project was to better define the relationship between the endoribonuclease activity of murine hepatitis virus (MHV) Nsp15 (mNsp15) and its role in virus infection. Molecular modeling demonstrated that the catalytic residues of mNsp15 are superimposable with its severe acute respiratory syndrome coronavirus ortholog. Alanine substitutions at three key residues in the mNsp15 catalytic pocket (H262, H277, and G275) and a double-mutant version (H262P and H277A) generated proteins with greatly reduced but detectable endoribonuclease activities. Furthermore, these mutant proteins demonstrated lower cleavage specificities for uridylate than wild-type (WT) mNsp15. These mutations were successfully incorporated into viruses named vH262A, vH277A, vG275A, and vH262P+H277A. All four mutant viruses formed plaques with diameters similar to that of MHV-A59 1000 (WT virus) on several different cell lines. Interestingly, viruses with a mutation at a noncatalytic residue, D324A, could not be recovered despite repeated attempts, and expression of mNsp15 containing the D324A mutation in Escherichia coli resulted in an insoluble protein. Plaques derived from vH262A produced approximately 6- to 13-fold fewer PFU than those from WT virus. Cells infected with mNsp15 mutant viruses accumulated lesser amounts of plus- and minus-sense subgenomic RNAs and spike protein than WT virus. The expression of mNsp15 in trans by transient transfection partially restored RNA synthesis by vH262A. These results demonstrate that mNsp15 is required for optimal infection by MHV.


Asunto(s)
Endorribonucleasas/metabolismo , Virus de la Hepatitis Murina/patogenicidad , Proteínas Virales/metabolismo , Animales , Línea Celular , Cricetinae , Endorribonucleasas/química , Endorribonucleasas/genética , Ratones , Modelos Moleculares , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/metabolismo , Mutación , Plásmidos/genética , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ensayo de Placa Viral , Proteínas Virales/química , Proteínas Virales/genética
12.
Virus Res ; 206: 120-33, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-25736566

RESUMEN

Coronaviruses (CoVs) are an important cause of illness in humans and animals. Most human coronaviruses commonly cause relatively mild respiratory illnesses; however two zoonotic coronaviruses, SARS-CoV and MERS-CoV, can cause severe illness and death. Investigations over the past 35 years have illuminated many aspects of coronavirus replication. The focus of this review is the functional analysis of conserved RNA secondary structures in the 5' and 3' of the betacoronavirus genomes. The 5' 350 nucleotides folds into a set of RNA secondary structures which are well conserved, and reverse genetic studies indicate that these structures play an important role in the discontinuous synthesis of subgenomic RNAs in the betacoronaviruses. These cis-acting elements extend 3' of the 5'UTR into ORF1a. The 3'UTR is similarly conserved and contains all of the cis-acting sequences necessary for viral replication. Two competing conformations near the 5' end of the 3'UTR have been shown to make up a potential molecular switch. There is some evidence that an association between the 3' and 5'UTRs is necessary for subgenomic RNA synthesis, but the basis for this association is not yet clear. A number of host RNA proteins have been shown to bind to the 5' and 3' cis-acting regions, but the significance of these in viral replication is not clear. Two viral proteins have been identified as binding to the 5' cis-acting region, nsp1 and N protein. A genetic interaction between nsp8 and nsp9 and the region of the 3'UTR that contains the putative molecular switch suggests that these two proteins bind to this region.


Asunto(s)
Regiones no Traducidas 3' , Regiones no Traducidas 5' , Coronavirus/fisiología , Genoma Viral , Interacciones Huésped-Patógeno , ARN Viral/metabolismo , Replicación Viral , Animales , Secuencia Conservada , Coronavirus/genética , Humanos , Modelos Biológicos , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , ARN Viral/química , ARN Viral/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética
13.
Virology ; 475: 15-27, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25462342

RESUMEN

SHAPE technology was used to analyze RNA secondary structure of the 5' most 474 nts of the MHV-A59 genome encompassing the minimal 5' cis-acting region required for defective interfering RNA replication. The structures generated were in agreement with previous characterizations of SL1 through SL4 and two recently predicted secondary structure elements, S5 and SL5A. SHAPE provided biochemical support for four additional stem-loops not previously functionally investigated in MHV. Secondary structure predictions for 5' regions of MHV-A59, BCoV and SARS-CoV were similar despite high sequence divergence. The pattern of SHAPE reactivity of in virio genomic RNA, ex virio genomic RNA, and in vitro synthesized RNA was similar, suggesting that binding of N protein or other proteins to virion RNA fails to protect the RNA from reaction with lipid permeable SHAPE reagent. Reverse genetic experiments suggested that SL5C and SL6 within the nsp1 coding sequence are not required for viral replication.


Asunto(s)
Regiones no Traducidas 5'/genética , Regulación Viral de la Expresión Génica/fisiología , Virus de la Hepatitis Murina/metabolismo , ARN Viral/química , Proteínas no Estructurales Virales/metabolismo , Animales , Ratones , Virus de la Hepatitis Murina/genética , Conformación de Ácido Nucleico , ARN Viral/metabolismo , Proteínas no Estructurales Virales/genética
14.
Virus Res ; 189: 177-88, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-24910120

RESUMEN

Mouse Hepatitis Virus (MHV) is a single-stranded positive sense RNA virus with the ability to promote acute and chronic diseases in mice. The MHV spike protein (S) is a major virulence determinant which in addition to binding to cellular receptors to mediate cell entry and facilitate virus spread to adjacent cells by cell-cell fusion, also is a molecular mimic of the FcγRII receptor. This molecular mimicry of FcγRII by the MHV S protein is also exhibited by other lineage 2a betacoronaviruses, with the exception of the human coronavirus HCoV-OC43. In this work we undertook a mutational analysis to attempt to identify specific amino acid sequences within the spike glycoprotein crucial for molecular mimicry of FcγRII. Although we were unsuccessful in isolating mutant viruses which were specifically defective in that property, we identified several mutations with interesting phenotypes. Mutation of the cysteine in position 547 to alanine and alanine replacements at residues 581-586 was lethal. Replacing proline 939 with the corresponding HCoV-OC43 residue, leucine, decreased the ability MHV to induce cell-cell fusion, providing experimental support for an earlier proposal that residues 929-944 make up the fusion peptide of the MHV S protein.


Asunto(s)
Imitación Molecular , Virus de la Hepatitis Murina/fisiología , Receptores de IgG/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , Línea Celular , Análisis Mutacional de ADN , Humanos , Ratones , Viabilidad Microbiana , Virus de la Hepatitis Murina/genética , Mutación Missense , Receptores de IgG/genética , Glicoproteína de la Espiga del Coronavirus/genética , Internalización del Virus
15.
Virology ; 443(1): 40-7, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23683838

RESUMEN

We designed a series of mutations to separately destabilize two helical stems (designated S3 and S4) predicted by a covariation-based model of the coronavirus 3'UTR (Zust et al., 2008). Mouse hepatitis virus genomes containing three or four nucleotide mutations that destabilize either S3 or S4 were viable, whereas genomes carrying these mutations in both S3 and S4 were not viable. A genome carrying these mutations in S3 and S4 plus compensatory mutations restoring base-pairing yielded a virus with wild type phenotype. Larger mutations which completely disrupt S3 or S4 generated various phenotypes. Mutations opening up S3 were lethal. Disruptions of S4 generated both viable and lethal mutants. Genomes carrying the original mutations in S3 or S4 plus compensatory mutations restoring base pairing were viable and had robust growth phenotypes. These results support the Zust model for the coronavirus 3'UTR and suggest that the S3 stem is required for virus viability.


Asunto(s)
Regiones no Traducidas 3' , Viabilidad Microbiana , Virus de la Hepatitis Murina/fisiología , ARN Viral/genética , Animales , Emparejamiento Base , Línea Celular , Análisis Mutacional de ADN , Ratones , Modelos Moleculares , Virus de la Hepatitis Murina/genética , Conformación de Ácido Nucleico , ARN Bicatenario/genética
18.
Adv Virus Res ; 81: 85-164, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22094080

RESUMEN

Coronaviruses infect many species of animals including humans, causing acute and chronic diseases. This review focuses primarily on the pathogenesis of murine coronavirus mouse hepatitis virus (MHV) and severe acute respiratory coronavirus (SARS-CoV). MHV is a collection of strains, which provide models systems for the study of viral tropism and pathogenesis in several organs systems, including the central nervous system, the liver, and the lung, and has been cited as providing one of the few animal models for the study of chronic demyelinating diseases such as multiple sclerosis. SARS-CoV emerged in the human population in China in 2002, causing a worldwide epidemic with severe morbidity and high mortality rates, particularly in older individuals. We review the pathogenesis of both viruses and the several reverse genetics systems that made much of these studies possible. We also review the functions of coronavirus proteins, structural, enzymatic, and accessory, with an emphasis on roles in pathogenesis. Structural proteins in addition to their roles in virion structure and morphogenesis also contribute significantly to viral spread in vivo and in antagonizing host cell responses. Nonstructural proteins include the small accessory proteins that are not at all conserved between MHV and SARS-CoV and the 16 conserved proteins encoded in the replicase locus, many of which have enzymatic activities in RNA metabolism or protein processing in addition to functions in antagonizing host response.


Asunto(s)
Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Virus de la Hepatitis Murina/patogenicidad , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Animales , Humanos , Ratones , Virus de la Hepatitis Murina/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo , Tropismo Viral , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
19.
J Mol Biol ; 394(3): 544-57, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19782089

RESUMEN

All coronaviruses (CoVs), including the causative agent of severe acute respiratory syndrome (SARS), encode a nucleocapsid (N) protein that harbors two independent RNA binding domains of known structure, but poorly characterized RNA binding properties. We show here that the N-terminal domain (NTD) of N protein from mouse hepatitis virus (MHV), a virus most closely related to SARS-CoV, employs aromatic amino acid-nucleobase stacking interactions with a triple adenosine motif to mediate high-affinity binding to single-stranded RNAs containing the transcriptional regulatory sequence (TRS) or its complement (cTRS). Stoichiometric NTD fully unwinds a TRS-cTRS duplex that mimics a transiently formed transcription intermediate in viral subgenomic RNA synthesis. Mutation of the solvent-exposed Y127, positioned on the beta-platform surface of our 1.75 A structure, binds the TRS far less tightly and is severely crippled in its RNA unwinding activity. In contrast, the C-terminal domain (CTD) exhibits no RNA unwinding activity. Viruses harboring Y127A N mutation are strongly selected against and Y127A N does not support an accessory function in MHV replication. We propose that the helix melting activity of the coronavirus N protein NTD plays a critical accessory role in subgenomic RNA synthesis and other processes requiring RNA remodeling.


Asunto(s)
Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/metabolismo , ARN Viral/química , ARN Viral/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de la Nucleocápside de Coronavirus , Cristalografía por Rayos X , Cinética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/metabolismo , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Proteínas de la Nucleocápside/genética , Unión Proteica , Estructura Terciaria de Proteína , ARN Viral/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos Reguladores de la Transcripción , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Homología de Secuencia de Aminoácido , Electricidad Estática , Termodinámica
20.
J Mol Biol ; 377(3): 790-803, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18289557

RESUMEN

The leader RNA of the 5' untranslated region (UTR) of coronaviral genomes contains two stem-loop structures denoted SL1 and SL2. Herein, we show that SL1 is functionally and structurally bipartite. While the upper region of SL1 is required to be paired, we observe strong genetic selection against viruses that contain a deletion of A35, an extrahelical nucleotide that destabilizes SL1, in favor of genomes that contain a diverse panel of destabilizing second-site mutations, due to introduction of a noncanonical base pair near A35. Viruses containing destabilizing SL1-DeltaA35 mutations also contain one of two specific mutations in the 3' UTR. Thermal denaturation and imino proton solvent exchange experiments reveal that the lower half of SL1 is unstable and that second-site SL1-DeltaA35 substitutions are characterized by one or more features of the wild-type SL1. We propose a "dynamic SL1" model, in which the base of SL1 has an optimized lability required to mediate a physical interaction between the 5' UTR and the 3' UTR that stimulates subgenomic RNA synthesis. Although not conserved at the nucleotide sequence level, these general structural characteristics of SL1 appear to be conserved in other coronaviral genomes.


Asunto(s)
Regiones no Traducidas 3'/metabolismo , Regiones no Traducidas 5'/metabolismo , Virus de la Hepatitis Murina/fisiología , ARN Viral/metabolismo , Replicación Viral , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Animales , Emparejamiento Base , Línea Celular , Cricetinae , Ratones , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/genética , Mutación , Conformación de Ácido Nucleico , ARN Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA