Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 289(41): 28489-504, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25135641

RESUMEN

The initial recognition between influenza virus and the host cell is mediated by interactions between the viral surface protein hemagglutinin and sialic acid-terminated glycoconjugates on the host cell surface. The sialic acid residues can be linked to the adjacent monosaccharide by α2-3- or α2-6-type glycosidic bonds. It is this linkage difference that primarily defines the species barrier of the influenza virus infection with α2-3 binding being associated with avian influenza viruses and α2-6 binding being associated with human strains. The ferret has been extensively used as an animal model to study the transmission of influenza. To better understand the validity of this model system, we undertook glycomic characterization of respiratory tissues of ferret, which allows a comparison of potential viral receptors to be made between humans and ferrets. To complement the structural analysis, lectin staining experiments were performed to characterize the regional distributions of glycans along the respiratory tract of ferrets. Finally, the binding between the glycans identified and the hemagglutinins of different strains of influenza viruses was assessed by glycan array experiments. Our data indicated that the respiratory tissues of ferret heterogeneously express both α2-3- and α2-6-linked sialic acids. However, the respiratory tissues of ferret also expressed the Sda epitope (NeuAcα2-3(GalNAcß1-4)Galß1-4GlcNAc) and sialylated N,N'-diacetyllactosamine (NeuAcα2-6GalNAcß1-4GlcNAc), which have not been observed in the human respiratory tract surface epithelium. The presence of the Sda epitope reduces potential binding sites for avian viruses and thus may have implications for the usefulness of the ferret in the study of influenza virus infection.


Asunto(s)
Glicómica , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Virus de la Influenza A/química , Polisacáridos/química , Sistema Respiratorio/química , Ácidos Siálicos/química , Animales , Sitios de Unión , Secuencia de Carbohidratos , Modelos Animales de Enfermedad , Femenino , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Virus de la Influenza A/metabolismo , Lectinas/química , Masculino , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/virología , Unión Proteica , Sistema Respiratorio/virología , Ácidos Siálicos/metabolismo , Especificidad de la Especie , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
2.
PLoS Pathog ; 9(3): e1003223, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23516363

RESUMEN

The first step in influenza infection of the human respiratory tract is binding of the virus to sialic (Sia) acid terminated receptors. The binding of different strains of virus for the receptor is determined by the α linkage of the sialic acid to galactose and the adjacent glycan structure. In this study the N- and O-glycan composition of the human lung, bronchus and nasopharynx was characterized by mass spectrometry. Analysis showed that there was a wide spectrum of both Sia α2-3 and α2-6 glycans in the lung and bronchus. This glycan structural data was then utilized in combination with binding data from 4 of the published glycan arrays to assess whether these current glycan arrays were able to predict replication of human, avian and swine viruses in human ex vivo respiratory tract tissues. The most comprehensive array from the Consortium for Functional Glycomics contained the greatest diversity of sialylated glycans, but was not predictive of productive replication in the bronchus and lung. Our findings indicate that more comprehensive but focused arrays need to be developed to investigate influenza virus binding in an assessment of newly emerging influenza viruses.


Asunto(s)
Glicómica , Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Polisacáridos/aislamiento & purificación , Sistema Respiratorio/química , Adulto , Animales , Aves , Bronquios/química , Bronquios/virología , Línea Celular , Perros , Galactosa/metabolismo , Humanos , Gripe Humana/virología , Pulmón/química , Pulmón/virología , Análisis por Micromatrices , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/química , Receptores de Superficie Celular/metabolismo , Sistema Respiratorio/virología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Porcinos , Tropismo Viral , Acoplamiento Viral , Replicación Viral
3.
J Virol ; 87(16): 8962-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740997

RESUMEN

The human parainfluenza virus (hPIV) hemagglutinin-neuraminidase (HN) protein binds (H) oligosaccharide receptors that contain N-acetylneuraminic acid (Neu5Ac) and cleaves (N) Neu5Ac from these oligosaccharides. In order to determine if one of HN's two functions is predominant, we measured the affinity of H for its ligands by a solid-phase binding assay with two glycoprotein substrates and by surface plasmon resonance with three monovalent glycans. We compared the dissociation constant (Kd) values from these experiments with previously determined Michaelis-Menten constants (Kms) for the enzyme activity. We found that glycoprotein substrates and monovalent glycans containing Neu5Acα2-3Galß1-4GlcNAc bind HN with Kd values in the 10 to 100 µM range. Km values for HN were previously determined to be on the order of 1 mM (M. M. Tappert, D. F. Smith, and G. M. Air, J. Virol. 85:12146-12159, 2011). A Km value greater than the Kd value indicates that cleavage occurs faster than the dissociation of binding and will dominate under N-permissive conditions. We propose, therefore, that HN is a neuraminidase that can hold its substrate long enough to act as a binding protein. The N activity can therefore regulate binding by reducing virus-receptor interactions when the concentration of receptor is high.


Asunto(s)
Proteína HN/metabolismo , Virus de la Parainfluenza 1 Humana/enzimología , Receptores Virales/metabolismo , Humanos , Hidrólisis , Cinética , Unión Proteica , Resonancia por Plasmón de Superficie
4.
Nature ; 453(7195): 667-71, 2008 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-18449194

RESUMEN

Pre-existing neutralizing antibody provides the first line of defence against pathogens in general. For influenza virus, annual vaccinations are given to maintain protective levels of antibody against the currently circulating strains. Here we report that after booster vaccination there was a rapid and robust influenza-specific IgG+ antibody-secreting plasma cell (ASC) response that peaked at approximately day 7 and accounted for up to 6% of peripheral blood B cells. These ASCs could be distinguished from influenza-specific IgG+ memory B cells that peaked 14-21 days after vaccination and averaged 1% of all B cells. Importantly, as much as 80% of ASCs purified at the peak of the response were influenza specific. This ASC response was characterized by a highly restricted B-cell receptor (BCR) repertoire that in some donors was dominated by only a few B-cell clones. This pauci-clonal response, however, showed extensive intraclonal diversification from accumulated somatic mutations. We used the immunoglobulin variable regions isolated from sorted single ASCs to produce over 50 human monoclonal antibodies (mAbs) that bound to the three influenza vaccine strains with high affinity. This strategy demonstrates that we can generate multiple high-affinity mAbs from humans within a month after vaccination. The panel of influenza-virus-specific human mAbs allowed us to address the issue of original antigenic sin (OAS): the phenomenon where the induced antibody shows higher affinity to a previously encountered influenza virus strain compared with the virus strain present in the vaccine. However, we found that most of the influenza-virus-specific mAbs showed the highest affinity for the current vaccine strain. Thus, OAS does not seem to be a common occurrence in normal, healthy adults receiving influenza vaccination.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos/inmunología , Memoria Inmunológica/inmunología , Orthomyxoviridae/inmunología , Anticuerpos Monoclonales/genética , Especificidad de Anticuerpos/inmunología , Células Clonales/inmunología , Células Clonales/metabolismo , Clonación Molecular , Humanos , Inmunización Secundaria , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/inmunología , Modelos Inmunológicos , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Hipermutación Somática de Inmunoglobulina/genética , Factores de Tiempo , Vacunación
5.
J Biol Chem ; 287(53): 44784-99, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23115247

RESUMEN

Human milk contains a large diversity of free glycans beyond lactose, but their functions are not well understood. To explore their functional recognition, here we describe a shotgun glycan microarray prepared from isolated human milk glycans (HMGs), and our studies on their recognition by viruses, antibodies, and glycan-binding proteins (GBPs), including lectins. The total neutral and sialylated HMGs were derivatized with a bifunctional fluorescent tag, separated by multidimensional HPLC, and archived in a tagged glycan library, which was then used to print a shotgun glycan microarray (SGM). This SGM was first interrogated with well defined GBPs and antibodies. These data demonstrated both the utility of the array and provided preliminary structural information (metadata) about this complex glycome. Anti-TRA-1 antibodies that recognize human pluripotent stem cells specifically recognized several HMGs that were then further structurally defined as novel epitopes for these antibodies. Human influenza viruses and Parvovirus Minute Viruses of Mice also specifically recognized several HMGs. For glycan sequencing, we used a novel approach termed metadata-assisted glycan sequencing (MAGS), in which we combine information from analyses of glycans by mass spectrometry with glycan interactions with defined GBPs and antibodies before and after exoglycosidase treatments on the microarray. Together, these results provide novel insights into diverse recognition functions of HMGs and show the utility of the SGM approach and MAGS as resources for defining novel glycan recognition by GBPs, antibodies, and pathogens.


Asunto(s)
Biomarcadores/química , Glicómica , Leche Humana/química , Polisacáridos/química , Receptores Virales/análisis , Animales , Secuencia de Carbohidratos , Línea Celular , Células Madre Embrionarias/metabolismo , Humanos , Ratones , Leche Humana/metabolismo , Datos de Secuencia Molecular , Polisacáridos/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo
6.
J Virol ; 86(10): 5857-66, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22398283

RESUMEN

Genetic drift of influenza virus genomic sequences occurs through the combined effects of sequence alterations introduced by a low-fidelity polymerase and the varying selective pressures experienced as the virus migrates through different host environments. While traditional phylogenetic analysis is useful in tracking the evolutionary heritage of these viruses, the specific genetic determinants that dictate important phenotypic characteristics are often difficult to discern within the complex genetic background arising through evolution. Here we describe a novel influenza virus sequence feature variant type (Flu-SFVT) approach, made available through the public Influenza Research Database resource (www.fludb.org), in which variant types (VTs) identified in defined influenza virus protein sequence features (SFs) are used for genotype-phenotype association studies. Since SFs have been defined for all influenza virus proteins based on known structural, functional, and immune epitope recognition properties, the Flu-SFVT approach allows the rapid identification of the molecular genetic determinants of important influenza virus characteristics and their connection to underlying biological functions. We demonstrate the use of the SFVT approach to obtain statistical evidence for effects of NS1 protein sequence variations in dictating influenza virus host range restriction.


Asunto(s)
Especificidad del Huésped , Virus de la Influenza A/genética , Gripe Humana/virología , Proteínas no Estructurales Virales/genética , Secuencia de Aminoácidos , Variación Genética , Humanos , Virus de la Influenza A/química , Virus de la Influenza A/clasificación , Virus de la Influenza A/fisiología , Datos de Secuencia Molecular , Filogenia , Estructura Secundaria de Proteína , Alineación de Secuencia , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo
7.
J Biol Chem ; 286(36): 31610-22, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21757734

RESUMEN

Many glycan-binding proteins in animals and pathogens recognize sialic acid or its modified forms, but their molecular recognition is poorly understood. Here we describe studies on sialic acid recognition using a novel sialylated glycan microarray containing modified sialic acids presented on different glycan backbones. Glycans terminating in ß-linked galactose at the non-reducing end and with an alkylamine-containing fluorophore at the reducing end were sialylated by a one-pot three-enzyme system to generate α2-3- and α2-6-linked sialyl glycans with 16 modified sialic acids. The resulting 77 sialyl glycans were purified and quantified, characterized by mass spectrometry, covalently printed on activated slides, and interrogated with a number of key sialic acid-binding proteins and viruses. Sialic acid recognition by the sialic acid-binding lectins Sambucus nigra agglutinin and Maackia amurensis lectin-I, which are routinely used for detecting α2-6- and α2-3-linked sialic acids, are affected by sialic acid modifications, and both lectins bind glycans terminating with 2-keto-3-deoxy-D-glycero-D-galactonononic acid (Kdn) and Kdn derivatives stronger than the derivatives of more common N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc). Three human parainfluenza viruses bind to glycans terminating with Neu5Ac or Neu5Gc and some of their derivatives but not to Kdn and its derivatives. Influenza A virus also does not bind glycans terminating in Kdn or Kdn derivatives. An especially novel aspect of human influenza A virus binding is its ability to equivalently recognize glycans terminated with either α2-6-linked Neu5Ac9Lt or α2-6-linked Neu5Ac. Our results demonstrate the utility of this sialylated glycan microarray to investigate the biological importance of modified sialic acids in protein-glycan interactions.


Asunto(s)
Polisacáridos/metabolismo , Análisis por Matrices de Proteínas , Proteínas/metabolismo , Ácidos Siálicos/metabolismo , Virus/metabolismo , Sitios de Unión , Humanos , Virus de la Influenza A , Lectinas/metabolismo , Respirovirus , Ácidos Siálicos/química
8.
BMC Struct Biol ; 12: 7, 2012 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-22559154

RESUMEN

BACKGROUND: Influenza neuraminidase (NA) is an important target for antiviral inhibitors since its active site is highly conserved such that inhibitors can be cross-reactive against multiple types and subtypes of influenza. Here, we discuss the crystal structure of neuraminidase subtype N9 complexed with a new benzoic acid based inhibitor (2) that was designed to add contacts by overpacking one side of the active site pocket. Inhibitor 2 uses benzoic acid to mimic the pyranose ring, a bis-(hydroxymethyl)-substituted 2-pyrrolidinone ring in place of the N-acetyl group of the sialic acid, and a branched aliphatic structure to fill the sialic acid C6 subsite. RESULTS: Inhibitor 2 {4-[2,2-bis(hydroxymethyl)-5-oxo-pyrrolidin-1-yl]-3-[(dipropylamino)methyl)]benzoic acid} was soaked into crystals of neuraminidase of A/tern/Australia/G70c/75 (N9), and the structure refined with 1.55 Å X-ray data. The benzene ring of the inhibitor tilted 8.9° compared to the previous compound (1), and the number of contacts, including hydrogen bonds, increased. However, the IC50 for compound 2 remained in the low micromolar range, likely because one propyl group was disordered. In this high-resolution structure of NA isolated from virus grown in chicken eggs, we found electron density for additional sugar units on the N-linked glycans compared to previous neuraminidase structures. In particular, seven mannoses and two N-acetylglucosamines are visible in the glycan attached to Asn200. This long, branched high-mannose glycan makes significant contacts with the neighboring subunit. CONCLUSIONS: We designed inhibitor 2 with an extended substituent at C4-corresponding to C6 of sialic acid-to increase the contact surface in the C6-subsite and to force the benzene ring to tilt to maximize these interactions while retaining the interactions of the carboxylate and the pyrolidinone substituents. The crystal structure at 1.55 Å showed that we partially succeeded in that the ring in 2 is tilted relative to 1 and the number of contacts increased, but one hydrophobic branch makes no contacts, perhaps explaining why the IC50 did not decrease. Future design efforts will include branches of unequal length so that both branches may be accommodated in the C6-subsite without conformational disorder. The high-mannose glycan attached to Asn200 makes several inter-subunit contacts and appears to stabilize the tetramer.


Asunto(s)
Ácido Benzoico/química , Ácido Benzoico/farmacología , Dominio Catalítico , Virus de la Influenza A/enzimología , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/química , Animales , Antivirales/química , Antivirales/farmacología , Cristalografía por Rayos X , Glucosa/química , Concentración 50 Inhibidora , Modelos Moleculares , Neuraminidasa/metabolismo , Polisacáridos/química , Unión Proteica/efectos de los fármacos , Difracción de Rayos X
9.
J Virol ; 85(23): 12146-59, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21917945

RESUMEN

The hemagglutinin-neuraminidase (HN) protein of human parainfluenza viruses (hPIVs) both binds (H) and cleaves (N) oligosaccharides that contain N-acetylneuraminic acid (Neu5Ac). H is thought to correspond to receptor binding and N to receptor-destroying activity. At present, N's role in infection remains unclear: does it destroy only receptors, or are there other targets? We previously demonstrated that hPIV1 and 3 HNs bind to oligosaccharides containing the motif Neu5Acα2-3Galß1-4GlcNAc (M. Amonsen, D. F. Smith, R. D. Cummings, and G. M. Air, J. Virol. 81:8341-8345, 2007). In the present study, we tested the binding specificity of hPIV2 on the Consortium for Functional Glycomics' glycan array and found that hPIV2 binds to oligosaccharides containing the same motif. We determined the specificities of N on red blood cells, soluble small-molecule and glycoprotein substrates, and the glycan array and compared them to the specificities of H. hPIV2 and -3, but not hPIV1, cleaved their ligands on red blood cells. hPIV1, -2, and -3 cleaved their NeuAcα2-3 ligands on the glycan array; hPIV2 and -3 also cleaved NeuAcα2-6 ligands bound by influenza A virus. While all three HNs exhibited similar affinities for all cleavable soluble substrates, their activities were 5- to 10-fold higher on small molecules than on glycoproteins. In addition, some soluble glycoproteins were not cleaved, despite containing oligosaccharides that were cleaved on the glycan array. We conclude that the susceptibility of an oligosaccharide substrate to N increases when the substrate is fixed to a surface. These findings suggest that HN may undergo a conformational change that activates N upon receptor binding at a cell surface.


Asunto(s)
Hemaglutininas/metabolismo , Neuraminidasa/metabolismo , Oligosacáridos/metabolismo , Virus de la Parainfluenza 1 Humana/metabolismo , Virus de la Parainfluenza 2 Humana/metabolismo , Virus de la Parainfluenza 3 Humana/metabolismo , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Pollos , Eritrocitos/metabolismo , Glicosilación , Pruebas de Hemaglutinación , Humanos , Análisis por Micromatrices , Datos de Secuencia Molecular , Virus de la Parainfluenza 1 Humana/aislamiento & purificación , Virus de la Parainfluenza 2 Humana/aislamiento & purificación , Virus de la Parainfluenza 3 Humana/aislamiento & purificación , Filogenia , Polisacáridos/metabolismo , Receptores Virales/metabolismo , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/metabolismo , Infecciones por Respirovirus/virología , Homología de Secuencia de Aminoácido , Turquía
10.
Bioorg Med Chem ; 20(14): 4582-9, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22677529

RESUMEN

Neuraminidase (NA) plays a critical role in the life cycle of influenza virus and is a target for new therapeutic agents. A series of influenza neuraminidase inhibitors with the pyrrolidinobenzoic acid scaffold containing lipophilic side chains at the C3 position have been synthesized and evaluated for influenza neuraminidase inhibitory activity. The size and geometry of the C3 side chains have been modified in order to investigate structure-activity relationships. The results indicated that size and geometry of the C3-side chain are important for selectivity of inhibition against N1 versus N2 NA, important type A influenza variants that infect man, including the highly lethal avian influenza.


Asunto(s)
Antivirales/química , Ácido Benzoico/química , Virus de la Influenza A/enzimología , Neuraminidasa/antagonistas & inhibidores , Pirrolidinonas/química , Antivirales/síntesis química , Antivirales/farmacología , Ácido Benzoico/síntesis química , Ácido Benzoico/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Interacciones Hidrofóbicas e Hidrofílicas , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/metabolismo , Relación Estructura-Actividad
11.
Arthritis Rheum ; 63(8): 2396-406, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21598235

RESUMEN

OBJECTIVE: Vaccination against common pathogens, such as influenza, is recommended for patients with systemic lupus erythematosus (SLE) to decrease infections and improve health. However, most reports describing the vaccination response are limited to evaluations of SLE patients with quiescent disease. This study focuses on understanding the clinical, serologic, therapeutic, and demographic factors that influence the response to influenza vaccination in SLE patients with a broad range of disease activity. METHODS: Blood specimens and information on disease activity were collected from 72 patients with SLE, at baseline and at 2, 6, and 12 weeks after influenza vaccination. Influenza-specific antibody responses were assessed by determining the total serum antibody concentration (B(max)), relative affinity (K(a)), and level of hemagglutination inhibition in the plasma. Using a cumulative score, the patients were evenly divided into groups of high or low vaccine responders. Autoantibody levels were evaluated at each time point using immunofluorescence tests and standard enzyme-linked immunosorbent assays. RESULTS: Compared to high responders, low responders to the vaccine were more likely to have hematologic criteria (P = 0.009), to have more American College of Rheumatology classification criteria for SLE (P = 0.05), and to be receiving concurrent prednisone treatment (P = 0.04). Interestingly, European American patients were more likely to be low responders than were African American patients (P = 0.03). Following vaccination, low responders were more likely to experience disease flares (P = 0.01) and to have increased titers of antinuclear antibodies (P = 0.04). Serum interferon-α activity at baseline was significantly higher in patients in whom a flare occurred after vaccination compared to a matched group of patients who did not experience a disease flare (P = 0.04). CONCLUSION: Ancestral background, prednisone treatment, hematologic criteria, and evidence of increased likelihood of disease flares were associated with low antibody responses to influenza vaccination in SLE patients.


Asunto(s)
Vacunas contra la Influenza/inmunología , Lupus Eritematoso Sistémico/inmunología , Adulto , Autoanticuerpos/sangre , Femenino , Humanos , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/prevención & control , Masculino , Persona de Mediana Edad , Vacunación
12.
Proc Natl Acad Sci U S A ; 106(2): 540-5, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19122146

RESUMEN

Cytotoxic T lymphocytes (CTL) limit influenza virus replication and prevent morbidity and mortality upon recognition of HLA class I presented epitopes on the surface of virus infected cells, yet the number and origin of the viral epitopes that decorate the infected cell are unknown. To understand the presentation of influenza virus ligands by human MHC class I molecules, HLA-B*0702-presented viral peptides were directly identified following influenza infection. After transfection with soluble class I molecules, peptide ligands unique to infected cells were eluted from isolated MHC molecules and identified by comparative mass spectrometry (MS). Then CTL were gathered following infection with influenza and viral peptides were tested for immune recognition. We found that the class I molecule B*0702 presents 3-6 viral ligands following infection with different strains of influenza. Peptide ligands derived from the internal viral nucleoprotein (NP(418-426) and NP(473-481)) and from the internal viral polymerase subunit PB1 (PB1(329-337)) were presented by B*0702 following infection with each of 3 different influenza strains; ligands NP(418-426), NP(473-481), and PB1(329-337) derived from internal viral proteins were consistently revealed by class I HLA. In contrast, ligands derived from hemagglutinin (HA) and matrix protein (M1) were presented intermittently on a strain-by-strain basis. When tested for immune recognition, HLA-B*0702 transgenic mice responded to NP(418-426) and PB1(329-337) consistently and NP(473-481) intermittently while ligands from HA and M1 were not recognized. These data demonstrate an emerging pattern whereby class I HLA reveal a handful of internal viral ligands and whereby CTL recognize consistently presented influenza ligands.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos Virales/inmunología , Antígenos HLA-B/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Orthomyxoviridae/inmunología , Animales , Antígenos Virales/análisis , Antígenos HLA/inmunología , Antígeno HLA-B7 , Células HeLa , Humanos , Espectrometría de Masas , Ratones , Ratones Transgénicos , Nucleoproteínas/inmunología , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Proteínas del Núcleo Viral/inmunología , Proteínas Virales/inmunología
13.
Retrovirology ; 8: 101, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22151792

RESUMEN

BACKGROUND: Bevirimat, the prototype Human Immunodeficiency Virus type 1 (HIV-1) maturation inhibitor, is highly potent in cell culture and efficacious in HIV-1 infected patients. In contrast to inhibitors that target the active site of the viral protease, bevirimat specifically inhibits a single cleavage event, the final processing step for the Gag precursor where p25 (CA-SP1) is cleaved to p24 (CA) and SP1. RESULTS: In this study, photoaffinity analogs of bevirimat and mass spectrometry were employed to map the binding site of bevirimat to Gag within immature virus-like particles. Bevirimat analogs were found to crosslink to sequences overlapping, or proximal to, the CA-SP1 cleavage site, consistent with previous biochemical data on the effect of bevirimat on Gag processing and with genetic data from resistance mutations, in a region predicted by NMR and mutational studies to have α-helical character. Unexpectedly, a second region of interaction was found within the Major Homology Region (MHR). Extensive prior genetic evidence suggests that the MHR is critical for virus assembly. CONCLUSIONS: This is the first demonstration of a direct interaction between the maturation inhibitor, bevirimat, and its target, Gag. Information gained from this study sheds light on the mechanisms by which the virus develops resistance to this class of drug and may aid in the design of next-generation maturation inhibitors.


Asunto(s)
Fármacos Anti-VIH/farmacología , VIH-1/efectos de los fármacos , Succinatos/farmacología , Triterpenos/farmacología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana , Sitios de Unión , Línea Celular , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , VIH-1/fisiología , Humanos , Espectrometría de Masas , Datos de Secuencia Molecular , Unión Proteica , Ensamble de Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo
14.
J Clin Immunol ; 31(5): 900-12, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21732013

RESUMEN

INTRODUCTION: The 2009 swine-origin H1N1 influenza virus (swH1N1) provided an opportunity to study immune responses to a new influenza strain in the context of seasonal influenza vaccination. Our goals were: to assess whether analyzing multiple parameters of immune responsiveness to influenza has an advantage over evaluating hemagglutination inhibition (HAI) titer alone, to determine whether vaccination with the seasonal vaccine induced cross-reactive immunity to swH1N1 in some individuals, and to determine whether the immune response against swH1N1 is higher after infection than vaccination. METHODS: Antibody and T cell responses were studied in ten subjects who were first immunized with the 2009-2010 seasonal influenza subunit vaccine, then 6 weeks later with the swH1N1 monovalent subunit vaccine. The amount of antibody against native virus glycoproteins, overall avidity of these antibodies, and HAI titer were measured. T cells were evaluated for proliferation and IFNγ secretion in response to the vaccine in vitro. Individuals with influenza-like illness were also evaluated, adding a microplate neuraminidase inhibition (NAI) test. RESULTS: The immune response to influenza was highly variable and immune parameters did not increase in parallel. The seasonal vaccine induced antibodies recognizing the pandemic virus in 50% of subjects. Antibody affinity and NAI activity to swH1N1 were higher after natural infection than vaccination. CONCLUSIONS: The evaluation of several immune parameters gives a more complete measure of immune responsiveness to influenza infection or vaccination than the HAI test alone.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza , Gripe Humana/epidemiología , Gripe Humana/inmunología , Pandemias , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Células Cultivadas , Reacciones Cruzadas , Pruebas de Inhibición de Hemaglutinación/métodos , Humanos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/diagnóstico , Gripe Humana/fisiopatología , Monitorización Inmunológica/métodos , Porcinos , Linfocitos T/inmunología , Linfocitos T/virología , Estados Unidos , Vacunación , Zoonosis
15.
Virus Res ; 304: 198545, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34391827

RESUMEN

The influenza A virus genome contains 8 gene segments encoding 10 commonly recognized proteins. Additional protein products have been identified, including PB1-F2 and PA-X. We report the in-silico identification of novel isoforms of PB1-F2 and PA-X in influenza virus genomes sequenced from avian samples. The isoform observed in PA-X includes a mutated stop codon that should extend the protein product by 8 amino acids. The isoform observed in PB1-F2 includes two nonsense mutations that should truncate the N-terminal region of the protein product and remove the entire mitochondrial targeting domain. Both isoforms were uncovered during automatic annotation of CEIRS sequence data. Nominally termed PA-X8 and PB1-F2-Cterm, both predicted isoforms were subsequently found in other annotated influenza genomes previously deposited in GenBank. Both isoforms were noticed due to discrepant annotations output by two annotation engines, indicating a benefit of incorporating multiple algorithms during gene annotation.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Secuencia de Bases , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Virales/metabolismo
16.
Trends Microbiol ; 16(4): 149-57, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18375125

RESUMEN

Sialic acids (Sias) are regarded as receptors for influenza viruses and are usually bound to galactose (Gal) in an alpha2-3 or alpha2-6 configuration. The detection of these Sia configurations in tissues has commonly been through the use of plant lectins that are able to identify which cells contain Siaalpha2-3- and Siaalpha2-6-linked glycans, although other techniques for receptor distribution have been used. Initial experiments indicated that avian versus human influenza virus binding was determined by either Siaalpha2-6 or Siaalpha2-3 expression. In this review, we suggest that the distribution and detection of these terminal Siaalpha2-3- and Siaalpha2-6-linked receptors within the respiratory tract might not be as clear cut as has been reported. We will also review how other viral and receptor components might act as determinants for successful viral replication and transmission. Understanding these additional components is important in comprehending the infection and the transmission of both existing human influenza viruses and newly emerging avian influenza viruses.


Asunto(s)
Virus de la Influenza A/metabolismo , Receptores Virales/metabolismo , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Humanos , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Virus de la Influenza A/genética , Modelos Moleculares , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Virales/química , Receptores Virales/genética , Porcinos
17.
Virol J ; 6: 22, 2009 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-19216793

RESUMEN

BACKGROUND: Influenza viruses attach to cells via sialic acid receptors. The viral neuraminidase (NA) is needed to remove sialic acids so that newly budded virions can disperse. Known mechanisms of resistance to NA inhibitors include mutations in the inhibitor binding site, or mutations in the hemagglutinin that reduce avidity for sialic acid and therefore reduce the requirement for NA activity. RESULTS: Influenza H3N2 isolates A/Oklahoma/323/03 (Fujian-like), A/Oklahoma/1992/05 (California-like), and A/Oklahoma/309/06 (Wisconsin-like) lost NA activity on passage in MDCK cells due to internal deletions in the NA-coding RNA segment. The viruses grow efficiently in MDCK cells despite diminished NA activity. The full length NA enzyme activity is sensitive to oseltamivir but replication of A/Oklahoma/323/03 and A/Oklahoma/309/06 in MDCK cells was resistant to this inhibitor, indicating that NA is not essential for replication. There was no change in HA activity or sequence after the NA activity was lost but the three viruses show distinct, quite restricted patterns of receptor specificity by Glycan Array analysis. Extensive predicted secondary structure in RNA segment 6 that codes for NA suggests the deletions are generated by polymerase skipping over base-paired stem regions. In general the NA deletions were not carried into subsequent passages, and we were unable to plaque-purify virus with a deleted NA RNA segment. CONCLUSION: H3N2 viruses from 2003 to the present have reduced requirement for NA when passaged in MDCK cells and are resistant to NA inhibitors, possibly by a novel mechanism of narrow receptor specificity such that virus particles do not self-aggregate. These viruses delete internal regions of the NA RNA during passage and are resistant to oseltamivir. However, deletions are independently generated at each passage, suggesting that virus with a full length NA RNA segment initiates the first round of infection.


Asunto(s)
Farmacorresistencia Viral , Inhibidores Enzimáticos/farmacología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/enzimología , Neuraminidasa/metabolismo , Infecciones por Orthomyxoviridae/virología , Oseltamivir/farmacología , Animales , Línea Celular , Eliminación de Gen , Subtipo H3N2 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/enzimología , Pase Seriado , Especificidad por Sustrato
18.
J Mol Biol ; 356(3): 651-63, 2006 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-16384583

RESUMEN

The crystal structure of the complex between neuraminidase (NA) of influenza virus A/Memphis/31/98 (H3N2) and Fab of monoclonal antibody Mem5 has been determined at 2.1A resolution and shows a novel pattern of interactions compared to other NA-Fab structures. The interface buries a large area of 2400 A2 and the surfaces have high complementarity. However, the interface is also highly hydrated. There are 33 water molecules in the interface>or=95% buried from bulk solvent, but only 13 of these are isolated from other water molecules. The rest are involved in an intricate network of water-mediated hydrogen bonds throughout the interface, stabilizing the complex. Glu199 on NA, the most critical side-chain to the interaction as previously determined by escape mutant analysis and site-directed mutation, is located in a non-aqueous island. Glu199 and three other residues that contribute the major part of the antigen buried surface of the complex have mutated in human influenza viruses isolated after 1998, confirming that Mem5 identifies an epidemiologically important antigenic site. We conclude that antibody selection of NA variants is a significant component of recent antigenic drift in human H3N2 influenza viruses, supporting the idea that influenza vaccines should contain NA in addition to hemagglutinin.


Asunto(s)
Anticuerpos Antivirales/química , Complejo Antígeno-Anticuerpo/química , Epítopos/química , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/epidemiología , Neuraminidasa/química , Orthomyxoviridae/enzimología , Orthomyxoviridae/inmunología , Agua/metabolismo , Animales , Anticuerpos Monoclonales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Cristalografía por Rayos X , Epítopos/inmunología , Flujo Genético , Humanos , Enlace de Hidrógeno , Fragmentos Fab de Inmunoglobulinas/química , Gripe Humana/inmunología , Ratones , Neuraminidasa/inmunología , Unión Proteica , Estructura Terciaria de Proteína , Propiedades de Superficie
19.
Virol J ; 4: 42, 2007 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-17490484

RESUMEN

BACKGROUND: Human influenza viruses are known to bind to sialic acid linked alpha2-6 to galactose, but the binding specificity beyond that linkage has not been systematically examined. H3N2 human influenza isolates lost binding to chicken red cells in the 1990s but viruses isolated since 2003 have re-acquired the ability to agglutinate chicken erythrocytes. We have investigated specificity of binding, changes in hemagglutinin sequence of the recent viruses and the role of sialic acid in productive infection. RESULTS: Viruses that agglutinate, or do not agglutinate, chicken red cells show identical binding to a Glycan Array of 264 oligosaccharides, binding exclusively to a subset of alpha2-6-sialylsaccharides. We identified an amino acid change in hemagglutinin that seemed to correlate with chicken red cell binding but when tested by mutagenesis there was no effect. Recombinant hemagglutinins expressed on Sf-9 cells bound chicken red cells but the released recombinant baculoviruses agglutinated only human red cells. Similarly, an isolate that does not agglutinate chicken red cells show hemadsorption of chicken red cells to infected MDCK cells. We suggest that binding of chicken red cells to cell surface hemagglutinin but not to virions is due to a more favorable hemagglutinin density on the cell surface. We investigated whether a virus specific for alpha2-6 sialyloligosaccharides shows differential entry into cells that have varying proportions of alpha2-6 and alpha2-3 sialic acids, including human A549 and HeLa cells with high levels of alpha2-6 sialic acid, and CHO cells that have only alpha2-3 sialic acid. We found that the virus enters all cell types tested and synthesizes viral nucleoprotein, localized in the nucleus, and hemagglutinin, transported to the cell surface, but infectious progeny viruses were released only from MDCK cells. CONCLUSION: Agglutination of chicken red cells does not correlate with altered binding to any oligosaccharide on the Glycan Array, and may result from increased avidity due to density of hemagglutinin and not increased affinity. Absence of alpha2-6 sialic acid does not protect a cell from influenza infection and the presence of high levels of alpha2-6-sialic acids on a cell surface does not guarantee productive replication of a virus with alpha2-6 receptor specificity.


Asunto(s)
Subtipo H3N2 del Virus de la Influenza A/fisiología , Receptores Virales/fisiología , Acoplamiento Viral , Animales , Línea Celular , Pollos , Cricetinae , Perros , Eritrocitos/virología , Hemaglutinación por Virus , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Macaca mulatta , Ácido N-Acetilneuramínico/metabolismo , Polisacáridos/metabolismo , Ensamble de Virus , Internalización del Virus , Replicación Viral
20.
Sci Rep ; 6: 36216, 2016 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-27796371

RESUMEN

Since the emergence of human H3N2 influenza A viruses in the pandemic of 1968, these viruses have become established as strains of moderate severity. A decline in virulence has been accompanied by glycan accumulation on the hemagglutinin globular head, and hemagglutinin receptor binding has changed from recognition of a broad spectrum of glycan receptors to a narrower spectrum. The relationship between increased glycosylation, binding changes, and reduction in H3N2 virulence is not clear. We evaluated the effect of hemagglutinin glycosylation on receptor binding and virulence of engineered H3N2 viruses. We demonstrate that low-binding virus is as virulent as higher binding counterparts, suggesting that H3N2 infection does not require either recognition of a wide variety of, or high avidity binding to, receptors. Among the few glycans recognized with low-binding virus, there were two structures that were bound by the vast majority of H3N2 viruses isolated between 1968 and 2012. We suggest that these two structures support physiologically relevant binding of H3N2 hemagglutinin and that this physiologically relevant binding has not changed since the 1968 pandemic. Therefore binding changes did not contribute to reduced severity of seasonal H3N2 viruses. This work will help direct the search for factors enhancing influenza virulence.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza , Subtipo H3N2 del Virus de la Influenza A , Acoplamiento Viral , Células A549 , Animales , Chlorocebus aethiops , Perros , Glicosilación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Células de Riñón Canino Madin Darby , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA