Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
J Virol ; 92(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29514915

RESUMEN

The spike glycoprotein (S) of murine coronavirus mouse hepatitis virus (MHV) strain A59 uses murine carcinoembryonic antigen-related cell adhesion molecule 1a as its receptor for cell entry, but S protein can also be triggered in the absence of receptor by pH 8.0 alone at 37°C. The mechanism by which conformational changes of this S glycoprotein can be triggered by pH 8.0 has not yet been determined. Here, we show that MHV-A59 S protein is triggered by pH 8.0 at 37°C to induce receptor-independent syncytium (RIS) formation on 293T cells, and that the conformational changes in S proteins triggered by pH 8.0 are very similar to those triggered by receptor binding. We systemically mutated each of 15 histidine residues in S protein and found that H209 is essential for pH 8.0-triggered RIS formation, while H179, H441, H643, and H759 also play important roles in this process. Replacement of H209 with Ala had no effect on receptor binding, but in murine 17Cl.1 cells mutant H209A MHV-A59 showed delayed growth kinetics and was readily outcompeted by wild-type virus when mixed together, indicating that the H209A mutation caused a defect in virus fitness. Finally, the H209A mutation significantly increased the thermostability of S protein in its prefusion conformation, which may raise the energy barrier for conformational change of S protein required for membrane fusion and lead to a decrease in virus fitness in cell culture. Thus, MHV-A59 may have evolved to lower the stability of its S protein in order to increase virus fitness.IMPORTANCE Enveloped viruses enter cells through fusion of viral and cellular membranes, and the process is mediated by interactions between viral envelope proteins and their host receptors. In the prefusion conformation, viral envelope proteins are metastable, and activation to the fusion conformation is tightly regulated, since premature activation would lead to loss of viral infectivity. The stability of viral envelope proteins greatly influences their activation and virus fitness. Here, we report that, similar to the A82V mutation in Ebola glycoprotein, in the S glycoprotein of murine coronavirus MHV-A59, the histidine residue at position of 209 significantly affects the thermal stability of the S protein, determines whether S protein can be activated at 37°C by either pH 8.0 alone or by receptor binding, and affects viral fitness in cell culture. Thus, the spike glycoprotein of MHV-A59 has evolved to retain histidine at position 209 to optimize virus fitness.


Asunto(s)
Sustitución de Aminoácidos/genética , Células Gigantes/virología , Virus de la Hepatitis Murina/crecimiento & desarrollo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Gatos , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Fusión de Membrana/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones , Virus de la Hepatitis Murina/genética , Mutación/genética , Unión Proteica/genética
2.
J Biol Chem ; 292(6): 2174-2181, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28035001

RESUMEN

Hosts and pathogens are locked in an evolutionary arms race. To infect mice, mouse hepatitis coronavirus (MHV) has evolved to recognize mouse CEACAM1a (mCEACAM1a) as its receptor. To elude MHV infections, mice may have evolved a variant allele from the Ceacam1a gene, called Ceacam1b, producing mCEACAM1b, which is a much poorer MHV receptor than mCEACAM1a. Previous studies showed that sequence differences between mCEACAM1a and mCEACAM1b in a critical MHV-binding CC' loop partially account for the low receptor activity of mCEACAM1b, but detailed structural and molecular mechanisms for the differential MHV receptor activities of mCEACAM1a and mCEACAM1b remained elusive. Here we have determined the crystal structure of mCEACAM1b and identified the structural differences and additional residue differences between mCEACAM1a and mCEACAM1b that affect MHV binding and entry. These differences include conformational alterations of the CC' loop as well as residue variations in other MHV-binding regions, including ß-strands C' and C'' and loop C'C''. Using pseudovirus entry and protein-protein binding assays, we show that substituting the structural and residue features from mCEACAM1b into mCEACAM1a reduced the viral receptor activity of mCEACAM1a, whereas substituting the reverse changes from mCEACAM1a into mCEACAM1b increased the viral receptor activity of mCEACAM1b. These results elucidate the detailed molecular mechanism for how mice may have kept pace in the evolutionary arms race with MHV by undergoing structural and residue changes in the MHV receptor, providing insight into this possible example of pathogen-driven evolution of a host receptor protein.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Virus de la Hepatitis Murina/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Antígeno Carcinoembrionario/química , Cristalografía por Rayos X , Fusión de Membrana , Ratones , Virus de la Hepatitis Murina/fisiología , Mutación , Conformación Proteica , Receptores Virales/química , Homología de Secuencia de Aminoácido
3.
J Virol ; 90(12): 5586-5600, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27030273

RESUMEN

UNLABELLED: The fusion peptides (FP) play an essential role in fusion of viral envelope with cellular membranes. The location and properties of the FPs in the spike (S) glycoproteins of different coronaviruses (CoV) have not yet been determined. Through amino acid sequence analysis of S proteins of representative CoVs, we identified a common region as a possible FP (pFP) that shares the characteristics of FPs of class I viral fusion proteins, including high Ala/Gly content, intermediate hydrophobicity, and few charged residues. To test the hypothesis that this region contains the CoV FP, we systemically mutated every residue in the pFP of Middle East respiratory syndrome betacoronavirus (MERS-CoV) and found that 11 of the 22 residues in the pFP (from G953 to L964, except for A956) were essential for S protein-mediated cell-cell fusion and virus entry. The synthetic MERS-CoV pFP core peptide (955IAGVGWTAGL964) induced extensive fusion of liposome membranes, while mutant peptide failed to induce any lipid mixing. We also selectively mutated residues in pFPs of two other ß-CoVs, severe acute respiratory syndrome coronavirus (SARS-CoV) and mouse hepatitis virus (MHV). Although the amino acid sequences of these two pFPs differed significantly from that of MERS-CoV and each other, most of the pFP mutants of SARS-CoV and MHV also failed to mediate membrane fusion, suggesting that these pFPs are also the functional FPs. Thus, the FPs of 3 different lineages of ß-CoVs are conserved in location within the S glycoproteins and in their functions, although their amino acid sequences have diverged significantly during CoV evolution. IMPORTANCE: Within the class I viral fusion proteins of many enveloped viruses, the FP is the critical mediator of fusion of the viral envelope with host cell membranes leading to virus infection. FPs from within a virus family, like influenza viruses or human immunodeficiency viruses (HIV), tend to share high amino acid sequence identity. In this study, we determined the location and amino acid sequences of the FPs of S glycoproteins of 3 ß-CoVs, MERS-CoV, SARS-CoV, and MHV, and demonstrated that they were essential for mediating cell-cell fusion and virus entry. Interestingly, in marked contrast to the FPs of influenza and HIV, the primary amino acid sequences of the FPs of ß-CoVs in 3 different lineages differed significantly. Thus, during evolution the FPs of ß-CoVs have diverged significantly in their primary sequences while maintaining the same essential biological functions. Our findings identify a potential new target for development of drugs against CoVs.


Asunto(s)
Coronavirus del Síndrome Respiratorio de Oriente Medio/química , Virus de la Hepatitis Murina/química , Péptidos/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Secuencia de Aminoácidos , Animales , Evolución Molecular , Células HEK293 , Humanos , Fusión de Membrana , Ratones , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Virus de la Hepatitis Murina/genética , Mutación , Péptidos/síntesis química , Péptidos/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Alineación de Secuencia , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus
4.
Biopolymers ; 106(2): 144-159, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26799790

RESUMEN

We have utilized a de novo designed two-stranded α-helical coiled-coil template to display conserved α-helical epitopes from the stem region of hemagglutinin (HA) glycoproteins of influenza A. The immunogens have all the surface-exposed residues of the native α-helix in the native HA protein of interest displayed on the surface of the two-stranded α-helical coiled-coil template. This template when used as an immunogen elicits polyclonal antibodies which bind to the α-helix in the native protein. We investigated the highly conserved sequence region 421-476 of HA by inserting 21 or 28 residue sequences from this region into our template. The cross-reactivity of the resulting rabbit polyclonal antibodies prepared to these immunogens was determined using a series of HA proteins from H1N1, H2N2, H3N2, H5N1, H7N7, and H7N9 virus strains which are representative of Group 1 and Group 2 virus subtypes of influenza A. Antibodies from region 449-476 were Group 1 specific. Antibodies to region 421-448 showed the greatest degree of cross-reactivity to Group 1 and Group 2 and suggested that this region has a great potential as a "universal" synthetic peptide vaccine for influenza A. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 144-159, 2016.

5.
J Virol ; 89(17): 8816-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26085157

RESUMEN

UNLABELLED: Coronavirus spike (S) glycoproteins mediate receptor binding, membrane fusion, and virus entry and determine host range. Murine betacoronavirus (ß-CoV) in group A uses the N-terminal domain (NTD) of S protein to bind to its receptor, whereas the ß-CoVs severe acute respiratory syndrome CoV in group B and Middle East respiratory syndrome CoV in group C and several α-CoVs use the downstream C domain in their S proteins to recognize their receptor proteins. To identify the receptor-binding domain in the spike of human ß-CoV HKU1 in group A, we generated and mapped a panel of monoclonal antibodies (MAbs) to the ectodomain of HKU1 spike protein. They did not cross-react with S proteins of any other CoV tested. Most of the HKU1 spike MAbs recognized epitopes in the C domain between amino acids 535 and 673, indicating that this region is immunodominant. Two of the MAbs blocked HKU1 virus infection of primary human tracheal-bronchial epithelial (HTBE) cells. Preincubation of HTBE cells with a truncated HKU1 S protein that includes the C domain blocked infection with HKU1 virus, but preincubation of cells with truncated S protein containing only the NTD did not block infection. These data suggest that the receptor-binding domain (RBD) of HKU1 spike protein is located in the C domain, where the spike proteins of α-CoVs and ß-CoVs in groups B and C bind to their specific receptor proteins. Thus, two ß-CoVs in group A, HKU1 and murine CoV, have evolved to use different regions of their spike glycoproteins to recognize their respective receptor proteins. IMPORTANCE: Mouse hepatitis virus, a ß-CoV in group A, uses the galectin-like NTD in its spike protein to bind its receptor protein, while HCoV-OC43, another ß-CoV in group A, uses the NTD to bind to its sialic-acid containing receptor. In marked contrast, the NTD of the spike glycoprotein of human respiratory ß-CoV HKU1, which is also in group A, does not bind sugar. In this study, we showed that for the spike protein of HKU1, the purified C domain, downstream of the NTD, could block HKU1 virus infection of human respiratory epithelial cells, and that several monoclonal antibodies that mapped to the C domain neutralized virus infectivity. Thus, the receptor-binding domain of HKU1 spike glycoprotein is located in the C domain. Surprisingly, two ß-CoVs in group A, mouse hepatitis virus and HKU1, have evolved to use different regions of their spike glycoproteins to recognize their respective receptors.


Asunto(s)
Infecciones por Coronavirus/virología , Coronavirus/metabolismo , Receptores Virales/genética , Glicoproteína de la Espiga del Coronavirus/genética , Tropismo Viral/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular Transformada , Chlorocebus aethiops , Coronavirus/genética , Coronavirus/inmunología , Perros , Células Epiteliales/virología , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/metabolismo , Estructura Terciaria de Proteína , Mucosa Respiratoria/citología , Mucosa Respiratoria/virología , Alineación de Secuencia , Glicoproteína de la Espiga del Coronavirus/inmunología , Células Vero , Internalización del Virus
6.
Viruses ; 6(5): 1897-910, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24784569

RESUMEN

The recent discovery of genetically distinct hantaviruses in multiple species of shrews and moles prompted a further exploration of their host diversification by analyzing frozen, ethanol-fixed and RNAlater®-preserved archival tissues and fecal samples from 533 bats (representing seven families, 28 genera and 53 species in the order Chiroptera), captured in Asia, Africa and the Americas in 1981-2012, using RT-PCR. Hantavirus RNA was detected in Pomona roundleaf bats (Hipposideros pomona) (family Hipposideridae), captured in Vietnam in 1997 and 1999, and in banana pipistrelles (Neoromicia nanus) (family Vespertilionidae), captured in Côte d'Ivoire in 2011. Phylogenetic analysis, based on the full-length S- and partial M- and L-segment sequences using maximum likelihood and Bayesian methods, demonstrated that the newfound hantaviruses formed highly divergent lineages, comprising other recently recognized bat-borne hantaviruses in Sierra Leone and China. The detection of bat-associated hantaviruses opens a new era in hantavirology and provides insights into their evolutionary origins.


Asunto(s)
Quirópteros/virología , Variación Genética , Orthohantavirus/clasificación , Orthohantavirus/genética , Filogenia , Animales , Análisis por Conglomerados , Côte d'Ivoire , Orthohantavirus/aislamiento & purificación , ARN Viral/genética , ARN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Vietnam
7.
J Gen Virol ; 95(Pt 4): 836-848, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24394697

RESUMEN

From 1 January 2009 to 31 May 2013, 15 287 respiratory specimens submitted to the Clinical Virology Laboratory at the Children's Hospital Colorado were tested for human coronavirus RNA by reverse transcription-PCR. Human coronaviruses HKU1, OC43, 229E and NL63 co-circulated during each of the respiratory seasons but with significant year-to-year variability, and cumulatively accounted for 7.4-15.6 % of all samples tested during the months of peak activity. A total of 79 (0.5 % prevalence) specimens were positive for human betacoronavirus HKU1 RNA. Genotypes HKU1 A and B were both isolated from clinical specimens and propagated on primary human tracheal-bronchial epithelial cells cultured at the air-liquid interface and were neutralized in vitro by human intravenous immunoglobulin and by polyclonal rabbit antibodies to the spike glycoprotein of HKU1. Phylogenetic analysis of the deduced amino acid sequences of seven full-length genomes of Colorado HKU1 viruses and the spike glycoproteins from four additional HKU1 viruses from Colorado and three from Brazil demonstrated remarkable conservation of these sequences with genotypes circulating in Hong Kong and France. Within genotype A, all but one of the Colorado HKU1 sequences formed a unique subclade defined by three amino acid substitutions (W197F, F613Y and S752F) in the spike glycoprotein and exhibited a unique signature in the acidic tandem repeat in the N-terminal region of the nsp3 subdomain. Elucidating the function of and mechanisms responsible for the formation of these varying tandem repeats will increase our understanding of the replication process and pathogenicity of HKU1 and potentially of other coronaviruses.


Asunto(s)
Infecciones por Coronaviridae/epidemiología , Infecciones por Coronaviridae/virología , Coronaviridae/clasificación , Coronaviridae/aislamiento & purificación , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Células Cultivadas , Análisis por Conglomerados , Colorado , Coronaviridae/genética , Genotipo , Humanos , Datos de Secuencia Molecular , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Cultivo de Virus
8.
PLoS One ; 8(10): e76469, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098509

RESUMEN

Little is known about the biology of the emerging human group c betacoronavirus, Middle East Respiratory Syndrome coronavirus (MERS-CoV). Because coronavirus spike glycoproteins (S) mediate virus entry, affect viral host range, and elicit neutralizing antibodies, analyzing the functions of MERS-CoV S protein is a high research priority. MERS-CoV S on lentivirus pseudovirions mediated entry into a variety of cell types including embryo cells from New World Eptesicus fuscus bats. Surprisingly, a polyclonal antibody to the S protein of MHV, a group a murine betacoronavirus, cross-reacted in immunoblots with the S2 domain of group c MERS-CoV spike protein. MERS pseudovirions released from 293T cells contained only uncleaved S, and pseudovirus entry was blocked by lysosomotropic reagents NH4Cl and bafilomycin and inhibitors of cathepsin L. However, when MERS pseudovirions with uncleaved S protein were adsorbed at 4°C to Vero E6 cells, brief trypsin treatment at neutral pH triggered virus entry at the plasma membrane and syncytia formation. When 293T cells producing MERS pseudotypes co-expressed serine proteases TMPRSS-2 or -4, large syncytia formed at neutral pH, and the pseudovirions produced were non-infectious and deficient in S protein. These experiments show that if S protein on MERS pseudovirions is uncleaved, then viruses enter by endocytosis in a cathepsin L-dependent manner, but if MERS-CoV S is cleaved, either during virus maturation by serine proteases or on pseudovirions by trypsin in extracellular fluids, then viruses enter at the plasma membrane at neutral pH and cause massive syncytia formation even in cells that express little or no MERS-CoV receptor. Thus, whether MERS-CoV enters cells within endosomes or at the plasma membrane depends upon the host cell type and tissue, and is determined by the location of host proteases that cleave the viral spike glycoprotein and activate membrane fusion.


Asunto(s)
Coronavirus/fisiología , Células Gigantes/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/virología , Chlorocebus aethiops , Endosomas/metabolismo , Eliminación de Gen , Expresión Génica , Orden Génico , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Receptores Virales/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Tripsina/metabolismo , Células Vero , Tropismo Viral
9.
Am J Respir Cell Mol Biol ; 48(6): 742-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23418343

RESUMEN

Severe acute respiratory syndrome (SARS)-coronavirus (CoV) produces a devastating primary viral pneumonia with diffuse alveolar damage and a marked increase in circulating cytokines. One of the major cell types to be infected is the alveolar type II cell. However, the innate immune response of primary human alveolar epithelial cells infected with SARS-CoV has not been defined. Our objectives included developing a culture system permissive for SARS-CoV infection in primary human type II cells and defining their innate immune response. Culturing primary human alveolar type II cells at an air-liquid interface (A/L) improved their differentiation and greatly increased their susceptibility to infection, allowing us to define their primary interferon and chemokine responses. Viral antigens were detected in the cytoplasm of infected type II cells, electron micrographs demonstrated secretory vesicles filled with virions, virus RNA concentrations increased with time, and infectious virions were released by exocytosis from the apical surface of polarized type II cells. A marked increase was evident in the mRNA concentrations of interferon-ß and interferon-λ (IL-29) and in a large number of proinflammatory cytokines and chemokines. A surprising finding involved the variability of expression of angiotensin-converting enzyme-2, the SARS-CoV receptor, in type II cells from different donors. In conclusion, the cultivation of alveolar type II cells at an air-liquid interface provides primary cultures in which to study the pulmonary innate immune responses to infection with SARS-CoV, and to explore possible therapeutic approaches to modulating these innate immune responses.


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Innata , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Enzima Convertidora de Angiotensina 2 , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Diferenciación Celular , Citoplasma/inmunología , Citoplasma/ultraestructura , Citoplasma/virología , Células Epiteliales/virología , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Interferones , Interleucinas/inmunología , Interleucinas/metabolismo , Peptidil-Dipeptidasa A/inmunología , Peptidil-Dipeptidasa A/metabolismo , Cultivo Primario de Células , Alveolos Pulmonares/citología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/virología , ARN Mensajero/metabolismo , Receptores Virales/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Factores de Tiempo , Liberación del Virus
10.
mBio ; 4(1): e00548-12, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23300251

RESUMEN

A new human betacoronavirus in lineage c, tentatively called HCoV-EMC, was isolated from a patient from the Kingdom of Saudi Arabia who died from acute severe pneumonia and renal failure. The viral RNA has been detected in eight additional cases. Sequencing and bioinformatic analysis of the viral genomic RNA showed that it is a novel virus not previously detected in any other species and that its closest relatives are two Asian bat coronaviruses. HCoV-EMC may represent a sporadic spillover to humans from an unknown animal reservoir. In a recent article, van Boheemen et al. demonstrated how state-of-the-art sequencing technology and bioinformatic analysis can quickly provide critical insight into the viral genome sequence, phylogeny, replication strategy, and potential drug and vaccine targets and generate tools to evaluate the possible epidemic risk associated with this novel human virus.


Asunto(s)
Coronavirus/clasificación , Coronavirus/aislamiento & purificación , Genoma Viral , Neumonía Viral/virología , ARN Viral/genética , Síndrome de Dificultad Respiratoria/virología , Humanos
11.
Immunity ; 37(5): 930-46, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23123061

RESUMEN

Carcinoembryonic antigen cell adhesion molecule like I (CEACAM1) is expressed on activated T cells and signals through either a long (L) cytoplasmic tail containing immune receptor tyrosine based inhibitory motifs, which provide inhibitory function, or a short (S) cytoplasmic tail with an unknown role. Previous studies on peripheral T cells show that CEACAM1-L isoforms predominate with little to no detectable CEACAM1-S isoforms in mouse and human. We show here that this was not the case in tissue resident T cells of intestines and gut associated lymphoid tissues, which demonstrated predominant expression of CEACAM1-S isoforms relative to CEACAM1-L isoforms in human and mouse. This tissue resident predominance of CEACAM1-S expression was determined by the intestinal environment where it served a stimulatory function leading to the regulation of T cell subsets associated with the generation of secretory IgA immunity, the regulation of mucosal commensalism, and defense of the barrier against enteropathogens.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Inmunidad Mucosa/inmunología , Intestinos/inmunología , Linfocitos T/inmunología , Secuencias de Aminoácidos/genética , Secuencias de Aminoácidos/inmunología , Animales , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Citoplasma/genética , Citoplasma/inmunología , Citoplasma/metabolismo , Homeostasis , Inmunidad Mucosa/genética , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Mucosa Intestinal/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Activación de Linfocitos , Metagenoma/inmunología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Isoformas de Proteínas , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T/metabolismo , Tirosina/genética , Tirosina/inmunología , Tirosina/metabolismo
12.
J Biol Chem ; 287(50): 41931-8, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23091051

RESUMEN

The spike protein N-terminal domains (NTDs) of bovine coronavirus (BCoV) and mouse hepatitis coronavirus (MHV) recognize sugar and protein receptors, respectively, despite their significant sequence homology. We recently determined the crystal structure of MHV NTD complexed with its protein receptor murine carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), which surprisingly revealed a human galectin (galactose-binding lectin) fold in MHV NTD. Here, we have determined at 1.55 Å resolution the crystal structure of BCoV NTD, which also has the human galectin fold. Using mutagenesis, we have located the sugar-binding site in BCoV NTD, which overlaps with the galactose-binding site in human galectins. Using a glycan array screen, we have identified 5-N-acetyl-9-O-acetylneuraminic acid as the preferred sugar substrate for BCoV NTD. Subtle structural differences between BCoV and MHV NTDs, primarily involving different conformations of receptor-binding loops, explain why BCoV NTD does not bind CEACAM1 and why MHV NTD does not bind sugar. These results suggest a successful viral evolution strategy in which coronaviruses stole a galectin from hosts, incorporated it into their spike protein, and evolved it into viral receptor-binding domains with altered sugar specificity in contemporary BCoV or novel protein specificity in contemporary MHV.


Asunto(s)
Coronavirus Bovino/química , Evolución Molecular , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Animales , Sitios de Unión , Bovinos , Coronavirus Bovino/genética , Coronavirus Bovino/metabolismo , Cristalografía por Rayos X , Humanos , Lectinas/química , Lectinas/genética , Lectinas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ácidos Neuramínicos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Glicoproteína de la Espiga del Coronavirus , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
13.
J Gen Virol ; 93(Pt 11): 2387-2398, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22837419

RESUMEN

This study compared the complete genome sequences of 16 NL63 strain human coronaviruses (hCoVs) from respiratory specimens of paediatric patients with respiratory disease in Colorado, USA, and characterized the epidemiology and clinical characteristics associated with circulating NL63 viruses over a 3-year period. From 1 January 2009 to 31 December 2011, 92 of 9380 respiratory specimens were found to be positive for NL63 RNA by PCR, an overall prevalence of 1 %. NL63 viruses were circulating during all 3 years, but there was considerable yearly variation in prevalence and the month of peak incidence. Phylogenetic analysis comparing the genome sequences of the 16 Colorado NL63 viruses with those of the prototypical hCoV-NL63 and three other NL63 viruses from the Netherlands demonstrated that there were three genotypes (A, B and C) circulating in Colorado from 2005 to 2010, and evidence of recombination between virus strains was found. Genotypes B and C co-circulated in Colorado in 2005, 2009 and 2010, but genotype A circulated only in 2005 when it was the predominant NL63 strain. Genotype C represents a new lineage that has not been described previously. The greatest variability in the NL63 virus genomes was found in the N-terminal domain (NTD) of the spike gene (nt 1-600, aa 1-200). Ten different amino acid sequences were found in the NTD of the spike protein among these NL63 strains and the 75 partial published sequences of NTDs from strains found at different times throughout the world.


Asunto(s)
Coronavirus Humano NL63/genética , Variación Genética , Genotipo , Glicoproteínas de Membrana/genética , Recombinación Genética , Proteínas del Envoltorio Viral/genética , Adolescente , Niño , Preescolar , Colorado/epidemiología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Femenino , Genoma Viral , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Filogenia , Estructura Terciaria de Proteína , Glicoproteína de la Espiga del Coronavirus , Factores de Tiempo
14.
J Gen Virol ; 93(Pt 3): 494-503, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22090214

RESUMEN

Human coronavirus strain 229E (HCoV-229E) commonly causes upper respiratory tract infections. However, lower respiratory tract infections can occur in some individuals, indicating that cells in the distal lung are susceptible to HCoV-229E. This study determined the virus susceptibility of primary cultures of human alveolar epithelial cells and alveolar macrophages (AMs). Fluorescent antibody staining indicated that HCoV-229E could readily infect AMs, but no evidence was found for infection in differentiated alveolar epithelial type II cells and only a very low level of infection in type II cells transitioning to the type I-like cell phenotype. However, a human bronchial epithelial cell line (16HBE) was readily infected. The innate immune response of AMs to HCoV-229E infection was evaluated for cytokine production and interferon (IFN) gene expression. AMs secreted significant amounts of tumour necrosis factor alpha (TNF-α), regulated on activation normal T-cell expressed and secreted (RANTES/CCL5) and macrophage inflammatory protein 1ß (MIP-1ß/CCL4) in response to HCoV-229E infection, but these cells exhibited no detectable increase in IFN-ß or interleukin-29 in mRNA levels. AMs from smokers had reduced secretion of TNF-α compared with non-smokers in response to HCoV-229E infection. Surfactant protein A (SP-A) and SP-D are part of the innate immune system in the distal lung. Both surfactant proteins bound to HCoV-229E, and pre-treatment of HCoV-229E with SP-A or SP-D inhibited infection of 16HBE cells. In contrast, there was a modest reduction in infection in AMs by SP-A, but not by SP-D. In summary, AMs are an important target for HCoV-229E, and they can mount a pro-inflammatory innate immune response to infection.


Asunto(s)
Coronavirus Humano 229E/patogenicidad , Macrófagos Alveolares/virología , Células Cultivadas , Citocinas/biosíntesis , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/virología , Técnica del Anticuerpo Fluorescente Directa , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Macrófagos Alveolares/inmunología , Ensayo de Placa Viral
15.
PLoS One ; 6(7): e21611, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21760897

RESUMEN

BACKGROUND: Allogeneic bone marrow transplantation (allo-BMT) is a potentially curative therapy for a variety of hematologic diseases, but benefits, including graft-versus-tumor (GVT) activity are limited by graft-versus-host-disease (GVHD). Carcinoembryonic antigen related cell adhesion molecule 1 (Ceacam1) is a transmembrane glycoprotein found on epithelium, T cells, and many tumors. It regulates a variety of physiologic and pathological processes such as tumor biology, leukocyte activation, and energy homeostasis. Previous studies suggest that Ceacam1 negatively regulates inflammation in inflammatory bowel disease models. METHODS: We studied Ceacam1 as a regulator of GVHD and GVT after allogeneic bone marrow transplantation (allo-BMT) in mouse models. In vivo, Ceacam1(-/-) T cells caused increased GVHD mortality and GVHD of the colon, and greater numbers of donor T cells were positive for activation markers (CD25(hi), CD62L(lo)). Additionally, Ceacam1(-/-) CD8 T cells had greater expression of the gut-trafficking integrin α(4)ß(7), though both CD4 and CD8 T cells were found increased numbers in the gut post-transplant. Ceacam1(-/-) recipients also experienced increased GVHD mortality and GVHD of the colon, and alloreactive T cells displayed increased activation. Additionally, Ceacam1(-/-) mice had increased mortality and decreased numbers of regenerating small intestinal crypts upon radiation exposure. Conversely, Ceacam1-overexpressing T cells caused attenuated target-organ and systemic GVHD, which correlated with decreased donor T cell numbers in target tissues, and mortality. Finally, graft-versus-tumor survival in a Ceacam1(+) lymphoma model was improved in animals receiving Ceacam1(-/-) vs. control T cells. CONCLUSIONS: We conclude that Ceacam1 regulates T cell activation, GVHD target organ damage, and numbers of donor T cells in lymphoid organs and GVHD target tissues. In recipients of allo-BMT, Ceacam1 may also regulate tissue radiosensitivity. Because of its expression on both the donor graft and host tissues, this suggests that targeting Ceacam1 may represent a potent strategy for the regulation of GVHD and GVT after allogeneic transplantation.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Antígeno Carcinoembrionario/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Efecto Injerto vs Tumor/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Polaridad Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Citotoxicidad Inmunológica/efectos de la radiación , Células Dendríticas/inmunología , Células Dendríticas/efectos de la radiación , Enfermedad Injerto contra Huésped/complicaciones , Enfermedad Injerto contra Huésped/mortalidad , Efecto Injerto vs Tumor/efectos de la radiación , Humanos , Integrinas/metabolismo , Intestino Delgado/patología , Intestino Delgado/efectos de la radiación , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Recuento de Linfocitos , Tejido Linfoide/citología , Tejido Linfoide/efectos de la radiación , Ratones , Especificidad de Órganos/inmunología , Especificidad de Órganos/efectos de la radiación , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Radiación Ionizante , Trasplante Homólogo
16.
Proc Natl Acad Sci U S A ; 108(26): 10696-701, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21670291

RESUMEN

Coronaviruses have evolved diverse mechanisms to recognize different receptors for their cross-species transmission and host-range expansion. Mouse hepatitis coronavirus (MHV) uses the N-terminal domain (NTD) of its spike protein as its receptor-binding domain. Here we present the crystal structure of MHV NTD complexed with its receptor murine carcinoembryonic antigen-related cell adhesion molecule 1a (mCEACAM1a). Unexpectedly, MHV NTD contains a core structure that has the same ß-sandwich fold as human galectins (S-lectins) and additional structural motifs that bind to the N-terminal Ig-like domain of mCEACAM1a. Despite its galectin fold, MHV NTD does not bind sugars, but instead binds mCEACAM1a through exclusive protein-protein interactions. Critical contacts at the interface have been confirmed by mutagenesis, providing a structural basis for viral and host specificities of coronavirus/CEACAM1 interactions. Sugar-binding assays reveal that galectin-like NTDs of some coronaviruses such as human coronavirus OC43 and bovine coronavirus bind sugars. Structural analysis and mutagenesis localize the sugar-binding site in coronavirus NTDs to be above the ß-sandwich core. We propose that coronavirus NTDs originated from a host galectin and retained sugar-binding functions in some contemporary coronaviruses, but evolved new structural features in MHV for mCEACAM1a binding.


Asunto(s)
Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Antígeno Carcinoembrionario/química , Antígeno Carcinoembrionario/metabolismo , Cristalografía por Rayos X , Evolución Molecular , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Receptores de Coronavirus , Receptores Virales/química , Homología de Secuencia de Aminoácido
17.
PLoS One ; 6(5): e19156, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21589915

RESUMEN

Bats are reservoirs for many different coronaviruses (CoVs) as well as many other important zoonotic viruses. We sampled feces and/or anal swabs of 1,044 insectivorous bats of 2 families and 17 species from 21 different locations within Colorado from 2007 to 2009. We detected alphacoronavirus RNA in bats of 4 species: big brown bats (Eptesicus fuscus), 10% prevalence; long-legged bats (Myotis volans), 8% prevalence; little brown bats (Myotis lucifugus), 3% prevalence; and western long-eared bats (Myotis evotis), 2% prevalence. Overall, juvenile bats were twice as likely to be positive for CoV RNA as adult bats. At two of the rural sampling sites, CoV RNAs were detected in big brown and long-legged bats during the three sequential summers of this study. CoV RNA was detected in big brown bats in all five of the urban maternity roosts sampled throughout each of the periods tested. Individually tagged big brown bats that were positive for CoV RNA and later sampled again all became CoV RNA negative. Nucleotide sequences in the RdRp gene fell into 3 main clusters, all distinct from those of Old World bats. Similar nucleotide sequences were found in amplicons from gene 1b and the spike gene in both a big-brown and a long-legged bat, indicating that a CoV may be capable of infecting bats of different genera. These data suggest that ongoing evolution of CoVs in bats creates the possibility of a continued threat for emergence into hosts of other species. Alphacoronavirus RNA was detected at a high prevalence in big brown bats in roosts in close proximity to human habitations (10%) and known to have direct contact with people (19%), suggesting that significant potential opportunities exist for cross-species transmission of these viruses. Further CoV surveillance studies in bats throughout the Americas are warranted.


Asunto(s)
Quirópteros/virología , Coronaviridae/aislamiento & purificación , Animales , Coronaviridae/clasificación , Coronaviridae/genética , Humanos , Filogenia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Virol ; 84(13): 6654-66, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410265

RESUMEN

Although most inbred mouse strains are highly susceptible to mouse hepatitis virus (MHV) infection, the inbred SJL line of mice is highly resistant to its infection. The principal receptor for MHV is murine CEACAM1 (mCEACAM1). Susceptible strains of mice are homozygous for the 1a allele of mCeacam1, while SJL mice are homozygous for the 1b allele. mCEACAM1a (1a) has a 10- to 100-fold-higher receptor activity than does mCEACAM1b (1b). To explore the hypothesis that MHV susceptibility is due to the different MHV receptor activities of 1a and 1b, we established a chimeric C57BL/6 mouse (cB61ba) in which a part of the N-terminal immunoglobulin (Ig)-like domain of the mCeacam1a (1a) gene, which is responsible for MHV receptor function, is replaced by the corresponding region of mCeacam1b (1b). We compared the MHV susceptibility of these chimeric mice to that of SJL and B6 mice. B6 mice that are homozygous for 1a are highly susceptible to MHV-A59 infection, with a 50% lethal dose (LD(50)) of 10(2.5) PFU, while chimeric cB61ba mice and SJL mice homozygous for 1ba and 1b, respectively, survived following inoculation with 10(5) PFU. Unexpectedly, cB61ba mice were more resistant to MHV-A59 infection than SJL mice as measured by virus replication in target organs, including liver and brain. No infectious virus or viral RNA was detected in the organs of cB61ba mice, while viral RNA and infectious virus were detected in target organs of SJL mice. Furthermore, SJL mice produced antiviral antibodies after MHV-A59 inoculation with 10(5) PFU, but cB61ba mice did not. Thus, cB61ba mice are apparently completely resistant to MHV-A59 infection, while SJL mice permit low levels of MHV-A59 virus replication during self-limited, asymptomatic infection. When expressed on cultured BHK cells, the mCEACAM1b and mCEACAM1ba proteins had similar levels of MHV-A59 receptor activity. These results strongly support the hypothesis that although alleles of mCEACAM1 are the principal determinants of mouse susceptibility to MHV-A59, other as-yet-unidentified murine genes may also play a role in susceptibility to MHV.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Glicoproteínas/metabolismo , Inmunidad Innata , Virus de la Hepatitis Murina/patogenicidad , Internalización del Virus , Alelos , Animales , Antígeno Carcinoembrionario/genética , Moléculas de Adhesión Celular , Glicoproteínas/genética , Homocigoto , Dosificación Letal Mediana , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análisis de Supervivencia
19.
J Gen Virol ; 90(Pt 12): 2956-2964, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19741068

RESUMEN

The rat coronavirus sialodacryoadenitis virus (SDAV) causes respiratory infection and provides a system for investigating respiratory coronaviruses in a natural host. A viral suspension in the form of a microspray aerosol was delivered by intratracheal instillation into the distal lung of 6-8-week-old Fischer 344 rats. SDAV inoculation produced a 7 % body weight loss over a 5 day period that was followed by recovery over the next 7 days. SDAV caused focal lesions in the lung, which were most severe on day 4 post-inoculation (p.i.). Immunofluorescent staining showed that four cell types supported SDAV virus replication in the lower respiratory tract, namely Clara cells, ciliated cells in the bronchial airway and alveolar type I and type II cells in the lung parenchyma. In bronchial alveolar lavage fluid (BALF) a neutrophil influx increased the population of neutrophils to 45 % compared with 6 % of the cells in control samples on day 2 after mock inoculation. Virus infection induced an increase in surfactant protein SP-D levels in BALF of infected rats on days 4 and 8 p.i. that subsided by day 12. The concentrations of chemokines MCP-1, LIX and CINC-1 in BALF increased on day 4 p.i., but returned to control levels by day 8. Intratracheal instillation of rats with SDAV coronavirus caused an acute, self-limited infection that is a useful model for studying the early events of the innate immune response to respiratory coronavirus infections in lungs of the natural virus host.


Asunto(s)
Infecciones por Coronavirus , Coronavirus de la Rata/patogenicidad , Células Epiteliales/virología , Pulmón/virología , Alveolos Pulmonares/virología , Replicación Viral , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/fisiopatología , Infecciones por Coronavirus/virología , Coronavirus de la Rata/fisiología , Citocinas/metabolismo , Inmunidad Innata , Pulmón/citología , Masculino , Alveolos Pulmonares/citología , Surfactantes Pulmonares/metabolismo , Ratas , Ratas Endogámicas F344 , Pérdida de Peso
20.
J Leukoc Biol ; 86(5): 1145-51, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19638499

RESUMEN

Viruses that infect the lung are a significant cause of morbidity and mortality in animals and humans worldwide. Coronaviruses are being associated increasingly with severe diseases in the lower respiratory tract. Alveolar epithelial cells are an important target for coronavirus infection in the lung, and infected cells can initiate innate immune responses to viral infection. In this overview, we describe in vitro models of highly differentiated alveolar epithelial cells that are currently being used to study the innate immune response to coronavirus infection. We have shown that rat coronavirus infection of rat alveolar type I epithelial cells in vitro induces expression of CXC chemokines, which may recruit and activate neutrophils. Although neutrophils are recruited early in infection in several coronavirus models including rat coronavirus. However, their role in viral clearance and/or immune-mediated tissue damage is not understood. Primary cultures of differentiated alveolar epithelial cells will be useful for identifying the interactions between coronaviruses and alveolar epithelial cells that influence the innate immune responses to infection in the lung. Understanding the molecular details of these interactions will be critical for the design of effective strategies to prevent and treat coronavirus infections in the lung.


Asunto(s)
Infecciones por Coronavirus/fisiopatología , Coronavirus/fisiología , Células Epiteliales/virología , Interacciones Huésped-Patógeno/fisiología , Alveolos Pulmonares/virología , Animales , Diferenciación Celular , Quimiocinas CXC/fisiología , Coronavirus/aislamiento & purificación , Infecciones por Coronavirus/inmunología , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/fisiología , Humanos , Pulmón/virología , Enfermedades Pulmonares/virología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/fisiopatología , Ratas , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/fisiopatología , Virosis/epidemiología , Virosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA