Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791536

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects various mammalian species, with farmed minks experiencing the highest number of outbreaks. In Spain, we analyzed 67 whole genome sequences and eight spike sequences from 18 outbreaks, identifying four distinct lineages: B.1, B.1.177, B.1.1.7, and AY.98.1. The potential risk of transmission to humans raises crucial questions about mutation accumulation and its impact on viral fitness. Sequencing revealed numerous not-lineage-defining mutations, suggesting a cumulative mutation process during the outbreaks. We observed that the outbreaks were predominantly associated with different groups of mutations rather than specific lineages. This clustering pattern by the outbreaks could be attributed to the rapid accumulation of mutations, particularly in the ORF1a polyprotein and in the spike protein. Notably, the mutations G37E in NSP9, a potential host marker, and S486L in NSP13 were detected. Spike protein mutations may enhance SARS-CoV-2 adaptability by influencing trimer stability and binding to mink receptors. These findings provide valuable insights into mink coronavirus genetics, highlighting both host markers and viral transmission dynamics within communities.


Asunto(s)
COVID-19 , Genoma Viral , Visón , Mutación , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , COVID-19/virología , COVID-19/epidemiología , COVID-19/transmisión , Animales , SARS-CoV-2/genética , SARS-CoV-2/fisiología , España/epidemiología , Visón/virología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Adaptación al Huésped/genética , Humanos , Brotes de Enfermedades , Pandemias , Filogenia , Secuenciación Completa del Genoma
2.
J Proteome Res ; 18(9): 3512-3520, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31361958

RESUMEN

Peptides generated by proteases in the cytosol must be translocated to endoplasmic reticulum lumen by the transporter associated with antigen processing (TAP) prior to their assembly with major histocompatibility complex (MHC) class I molecules. Nonfunctional TAP complexes produce a drastic decrease of the MHC class I/peptide complexes presented on the cell surface. Previously, the cellular MHC class I ligandome from TAP-deficient cell lines was determined, but similar analysis from normal tissues remains incomplete. Using high-throughput mass spectrometry to analyze the MHC-bound peptide pools isolated from ex vivo spleen cells of TAP-deficient mice, we identified 210 TAP-independent ligands naturally presented by murine MHC class I molecules. This ligandome showed increased peptide lengths, presence of multiple nested set peptides, and low theoretical MHC binding affinity. The gene ontology enrichment analysis of parental proteins of this TAP-independent subligandome showed almost exclusively enrichment in tissue-specific biological processes related to the immune system as would be expected. Also, cellular components of the extracellular space (namely proteins outside the cell but still within the organism excluding the extracellular matrix) were specifically associated with TAP-independent antigen processing from these ex vivo mice cells. In addition, functional protein association network analysis revealed low protein-protein interactions between parental proteins from the TAP-independent ligandome. Finally, predominant endoproteolytic peptidase specificity for Leu/Phe residues in the P1 position of the scissile bond at both ligand termini was found for the ex vivo TAP-independent ligands. These data indicate that the TAP-independent ligandome from ex vivo cells derives from a more diverse collection of both endoprotease activities and parental proteins and where the cell origin and contribution of the extracellular environment are more relevant than in its equivalent cell lines.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/genética , Ligandos , Péptidos/genética , Bazo/metabolismo , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Citosol/metabolismo , Retículo Endoplásmico/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Ratones , Péptidos/aislamiento & purificación , Unión Proteica/genética , Transporte de Proteínas/genética , Proteolisis , Bazo/química
3.
J Proteome Res ; 18(3): 900-911, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30629447

RESUMEN

Protective cellular and humoral immune responses require previous recognition of viral antigenic peptides complexed with human leukocyte antigen (HLA) class II molecules on the surface of the antigen presenting cells. The HLA class II-restricted immune response is important for the control and the clearance of poxvirus infection including vaccinia virus (VACV), the vaccine used in the worldwide eradication of smallpox. In this study, a mass spectrometry analysis was used to identify VACV ligands bound to HLA-DR and -DP class II molecules present on the surface of VACV-infected cells. Twenty-six naturally processed viral ligands among the tens of thousands of cell peptides bound to HLA class II proteins were identified. These viral ligands arose from 19 parental VACV proteins: A4, A5, A18, A35, A38, B5, B13, D1, D5, D7, D12, D13, E3, E8, H5, I2, I3, J2, and K2. The majority of these VACV proteins yielded one HLA ligand and were generated mainly, but not exclusively, by the classical HLA class II antigen processing pathway. Medium-sized and abundant proteins from the virion core and/or involved in the viral gene expression were the major source of VACV ligands bound to HLA-DR and -DP class II molecules. These findings will help to understand the effectiveness of current poxvirus-based vaccines and will be important in the design of new ones.


Asunto(s)
Antígenos de Histocompatibilidad Clase II/metabolismo , Ligandos , Proteómica/métodos , Virus Vaccinia/química , Proteínas Estructurales Virales , Virión/química , Células Cultivadas , Expresión Génica , Humanos , Espectrometría de Masas , Poxviridae/inmunología , Vaccinia/inmunología , Proteínas Virales/inmunología , Proteínas Estructurales Virales/inmunología , Vacunas Virales
4.
PLoS Pathog ; 12(9): e1005859, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27611367

RESUMEN

Human metapneumovirus (hMPV) is a paramyxovirus that is a common cause of bronchiolitis and pneumonia in children less than five years of age. The hMPV fusion (F) glycoprotein is the primary target of neutralizing antibodies and is thus a critical vaccine antigen. To facilitate structure-based vaccine design, we stabilized the ectodomain of the hMPV F protein in the postfusion conformation and determined its structure to a resolution of 3.3 Å by X-ray crystallography. The structure resembles an elongated cone and is very similar to the postfusion F protein from the related human respiratory syncytial virus (hRSV). In contrast, significant differences were apparent with the postfusion F proteins from other paramyxoviruses, such as human parainfluenza type 3 (hPIV3) and Newcastle disease virus (NDV). The high similarity of hMPV and hRSV postfusion F in two antigenic sites targeted by neutralizing antibodies prompted us to test for antibody cross-reactivity. The widely used monoclonal antibody 101F, which binds to antigenic site IV of hRSV F, was found to cross-react with hMPV postfusion F and neutralize both hRSV and hMPV. Despite the cross-reactivity of 101F and the reported cross-reactivity of two other antibodies, 54G10 and MPE8, we found no detectable cross-reactivity in the polyclonal antibody responses raised in mice against the postfusion forms of either hMPV or hRSV F. The postfusion-stabilized hMPV F protein did, however, elicit high titers of hMPV-neutralizing activity, suggesting that it could serve as an effective subunit vaccine. Structural insights from these studies should be useful for designing novel immunogens able to induce wider cross-reactive antibody responses.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Metapneumovirus/inmunología , Proteínas Virales de Fusión/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/química , Antígenos Virales/genética , Reacciones Cruzadas , Cristalografía por Rayos X , Femenino , Ingeniería Genética , Humanos , Metapneumovirus/genética , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Conformación Molecular , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/inmunología , Alineación de Secuencia , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética
5.
J Virol ; 90(11): 5485-5498, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009962

RESUMEN

UNLABELLED: Human respiratory syncytial virus (hRSV) vaccine development has received new impetus from structure-based studies of its main protective antigen, the fusion (F) glycoprotein. Three soluble forms of F have been described: monomeric, trimeric prefusion, and trimeric postfusion. Most human neutralizing antibodies recognize epitopes found exclusively in prefusion F. Although prefusion F induces higher levels of neutralizing antibodies than does postfusion F, postfusion F can also induce protection against virus challenge in animals. However, the immunogenicity and protective efficacy of the three forms of F have not hitherto been directly compared. Hence, BALB/c mice were immunized with a single dose of the three proteins adjuvanted with CpG and challenged 4 weeks later with virus. Serum antibodies, lung virus titers, weight loss, and pulmonary pathology were evaluated after challenge. Whereas small amounts of postfusion F were sufficient to protect mice, larger amounts of monomeric and prefusion F proteins were required for protection. However, postfusion and monomeric F proteins were associated with more pathology after challenge than was prefusion F. Antibodies induced by all doses of prefusion F, in contrast to other F protein forms, reacted predominantly with the prefusion F conformation. At high doses, prefusion F also induced the highest titers of neutralizing antibodies, and all mice were protected, yet at low doses of the immunogen, these antibodies neutralized virus poorly, and mice were not protected. These findings should be considered when developing new hRSV vaccine candidates. IMPORTANCE: Protection against hRSV infection is afforded mainly by neutralizing antibodies, which recognize mostly epitopes found exclusively in the viral fusion (F) glycoprotein trimer, folded in its prefusion conformation, i.e., before activation for membrane fusion. Although prefusion F is able to induce high levels of neutralizing antibodies, highly stable postfusion F (found after membrane fusion) is also able to induce neutralizing antibodies and protect against infection. In addition, a monomeric form of hRSV F that shares epitopes with prefusion F was recently reported. Since each of the indicated forms of hRSV F may have advantages and disadvantages for the development of safe and efficacious subunit vaccines, a direct comparison of the immunogenic properties and protective efficacies of the different forms of hRSV F was made in a mouse model. The results obtained show important differences between the noted immunogens that should be borne in mind when considering the development of hRSV vaccines.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/química , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología , Adyuvantes Inmunológicos , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Relación Dosis-Respuesta Inmunológica , Epítopos/inmunología , Femenino , Humanos , Inmunización , Inmunogenicidad Vacunal , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos/inmunología , Conformación Proteica , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/aislamiento & purificación , Proteínas Virales de Fusión/administración & dosificación
6.
PLoS Pathog ; 11(7): e1005035, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26161532

RESUMEN

Prevention efforts for respiratory syncytial virus (RSV) have been advanced due to the recent isolation and characterization of antibodies that specifically recognize the prefusion conformation of the RSV fusion (F) glycoprotein. These potently neutralizing antibodies are in clinical development for passive prophylaxis and have also aided the design of vaccine antigens that display prefusion-specific epitopes. To date, prefusion-specific antibodies have been shown to target two antigenic sites on RSV F, but both of these sites are also present on monomeric forms of F. Here we present a structural and functional characterization of human antibody AM14, which potently neutralized laboratory strains and clinical isolates of RSV from both A and B subtypes. The crystal structure and location of escape mutations revealed that AM14 recognizes a quaternary epitope that spans two protomers and includes a region that undergoes extensive conformational changes in the pre- to postfusion F transition. Binding assays demonstrated that AM14 is unique in its specific recognition of trimeric furin-cleaved prefusion F, which is the mature form of F on infectious virions. These results demonstrate that the prefusion F trimer contains potent neutralizing epitopes not present on monomers and that AM14 should be particularly useful for characterizing the conformational state of RSV F-based vaccine antigens.


Asunto(s)
Anticuerpos Neutralizantes/ultraestructura , Anticuerpos Antivirales/ultraestructura , Epítopos de Linfocito B/ultraestructura , Virus Sincitiales Respiratorios/inmunología , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Línea Celular , Cromatografía en Gel , Cristalografía por Rayos X , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Epítopos de Linfocito B/química , Epítopos de Linfocito B/inmunología , Citometría de Flujo , Glicoproteínas/química , Glicoproteínas/inmunología , Glicoproteínas/ultraestructura , Humanos , Estructura Cuaternaria de Proteína , Resonancia por Plasmón de Superficie
7.
Antimicrob Agents Chemother ; 60(11): 6498-6509, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27550346

RESUMEN

ALX-0171 is a trivalent Nanobody derived from monovalent Nb017 that binds to antigenic site II of the human respiratory syncytial virus (hRSV) fusion (F) glycoprotein. ALX-0171 is about 6,000 to 10,000 times more potent than Nb017 in neutralization tests with strains of hRSV antigenic groups A and B. To explore the effect of this enhanced neutralization on escape mutant selection, viruses resistant to either ALX-0171 or Nb017 were isolated after serial passage of the hRSV Long strain in the presence of suboptimal concentrations of the respective Nanobodies. Resistant viruses emerged notably faster with Nb017 than with ALX-0171 and in both cases contained amino acid changes in antigenic site II of hRSV F. Detailed binding and neutralization analyses of these escape mutants as well as previously described mutants resistant to certain monoclonal antibodies (MAbs) offered a comprehensive description of site II mutations which are relevant for neutralization by MAbs and Nanobodies. Notably, ALX-0171 showed a sizeable neutralization potency with most escape mutants, even with some of those selected with the Nanobody, and these findings make ALX-0171 an attractive antiviral for treatment of hRSV infections.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Antivirales/farmacología , Antígenos Virales/inmunología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Anticuerpos de Dominio Único/farmacología , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Antivirales/química , Anticuerpos Antivirales/aislamiento & purificación , Antígenos Virales/química , Antígenos Virales/genética , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Células Epiteliales/virología , Epítopos/química , Epítopos/inmunología , Humanos , Evasión Inmune/genética , Sueros Inmunes/química , Modelos Moleculares , Mutación , Pruebas de Neutralización , Unión Proteica , Estructura Secundaria de Proteína , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/inmunología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/aislamiento & purificación , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
8.
Antimicrob Agents Chemother ; 60(1): 6-13, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26438495

RESUMEN

Respiratory syncytial virus (RSV) is an important causative agent of lower respiratory tract infections in infants and elderly individuals. Its fusion (F) protein is critical for virus infection. It is targeted by several investigational antivirals and by palivizumab, a humanized monoclonal antibody used prophylactically in infants considered at high risk of severe RSV disease. ALX-0171 is a trimeric Nanobody that binds the antigenic site II of RSV F protein with subnanomolar affinity. ALX-0171 demonstrated in vitro neutralization superior to that of palivizumab against prototypic RSV subtype A and B strains. Moreover, ALX-0171 completely blocked replication to below the limit of detection for 87% of the viruses tested, whereas palivizumab did so for 18% of the viruses tested at a fixed concentration. Importantly, ALX-0171 was highly effective in reducing both nasal and lung RSV titers when delivered prophylactically or therapeutically directly to the lungs of cotton rats. ALX-0171 represents a potent novel antiviral compound with significant potential to treat RSV-mediated disease.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/efectos de los fármacos , Anticuerpos de Dominio Único/farmacología , Proteínas Virales de Fusión/antagonistas & inhibidores , Administración por Inhalación , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Antivirales/inmunología , Antivirales/metabolismo , Antivirales/farmacología , Femenino , Expresión Génica , Humanos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/virología , Masculino , Modelos Moleculares , Cavidad Nasal/efectos de los fármacos , Cavidad Nasal/inmunología , Cavidad Nasal/virología , Pruebas de Neutralización , Palivizumab/biosíntesis , Palivizumab/inmunología , Palivizumab/farmacología , Pichia/genética , Pichia/metabolismo , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/inmunología , Virus Sincitiales Respiratorios/patogenicidad , Sigmodontinae , Anticuerpos de Dominio Único/biosíntesis , Anticuerpos de Dominio Único/inmunología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
9.
J Virol ; 88(20): 11802-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25078705

RESUMEN

Respiratory syncytial virus (RSV) is the leading infectious cause of severe respiratory disease in infants and a major cause of respiratory illness in the elderly. There remains an unmet vaccine need despite decades of research. Insufficient potency, homogeneity, and stability of previous RSV fusion protein (F) subunit vaccine candidates have hampered vaccine development. RSV F and related parainfluenza virus (PIV) F proteins are cleaved by furin during intracellular maturation, producing disulfide-linked F1 and F2 fragments. During cell entry, the cleaved Fs rearrange from prefusion trimers to postfusion trimers. Using RSV F constructs with mutated furin cleavage sites, we isolated an uncleaved RSV F ectodomain that is predominantly monomeric and requires specific cleavage between F1 and F2 for self-association and rearrangement into stable postfusion trimers. The uncleaved RSV F monomer is folded and homogenous and displays at least two key RSV-neutralizing epitopes shared between the prefusion and postfusion conformations. Unlike the cleaved trimer, the uncleaved monomer binds the prefusion-specific monoclonal antibody D25 and human neutralizing immunoglobulins that do not bind to postfusion F. These observations suggest that the uncleaved RSV F monomer has a prefusion-like conformation and is a potential prefusion subunit vaccine candidate. Importance: RSV is the leading infectious cause of severe respiratory disease in infants and a major cause of respiratory illness in the elderly. Development of an RSV vaccine was stymied when a clinical trial using a formalin-inactivated RSV virus made disease, following RSV infection, more severe. Recent studies have defined the structures that the RSV F envelope glycoprotein adopts before and after virus entry (prefusion and postfusion conformations, respectively). Key neutralization epitopes of prefusion and postfusion RSV F have been identified, and a number of current vaccine development efforts are focused on generating easily produced subunit antigens that retain these epitopes. Here we show that a simple modification in the F ectodomain results in a homogeneous protein that retains critical prefusion neutralizing epitopes. These results improve our understanding of RSV F protein folding and structure and can guide further vaccine design efforts.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Epítopos/inmunología , Virus Sincitiales Respiratorios/inmunología , Humanos , Proteolisis
10.
Proc Natl Acad Sci U S A ; 109(8): 3089-94, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22323598

RESUMEN

Human respiratory syncytial virus (hRSV) is the most important viral agent of pediatric respiratory infections worldwide. The only specific treatment available today is a humanized monoclonal antibody (Palivizumab) directed against the F glycoprotein, administered prophylactically to children at very high risk of severe hRSV infections. Palivizumab, as most anti-F antibodies so far described, recognizes an epitope that is shared by the two conformations in which hRSV_F can fold, the metastable prefusion form and the highly stable postfusion conformation. We now describe a unique class of antibodies specific for the prefusion form of this protein that account for most of the neutralizing activity of either a rabbit serum raised against a vaccinia virus recombinant expressing hRSV_F or a human Ig preparation (Respigam), which was used for prophylaxis before Palivizumab. These antibodies therefore offer unique possibilities for immune intervention against hRSV, and their production should be assessed in trials of hRSV vaccines.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Infecciones por Virus Sincitial Respiratorio/terapia , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología , Secuencia de Aminoácidos , Animales , Humanos , Inmunización , Datos de Secuencia Molecular , Estabilidad Proteica , Conejos , Proteínas Recombinantes/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Vaccinia/inmunología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/ultraestructura
11.
Nat Commun ; 13(1): 1299, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35288548

RESUMEN

The human metapneumovirus (hMPV) fusion (F) protein is essential for viral entry and is a key target of neutralizing antibodies and vaccine development. The prefusion conformation is thought to be the optimal vaccine antigen, but previously described prefusion F proteins expressed poorly and were not well stabilized. Here, we use structures of hMPV F to guide the design of 42 variants containing stabilizing substitutions. Through combinatorial addition of disulfide bonds, cavity-filling substitutions, and improved electrostatic interactions, we describe a prefusion-stabilized F protein (DS-CavEs2) that expresses at 15 mg/L and has a melting temperature of 71.9 °C. Crystal structures of two prefusion-stabilized hMPV F variants reveal that antigenic surfaces are largely unperturbed. Importantly, immunization of mice with DS-CavEs2 elicits significantly higher neutralizing antibody titers against hMPV A1 and B1 viruses than postfusion F. The improved properties of DS-CavEs2 will advance the development of hMPV vaccines and the isolation of therapeutic antibodies.


Asunto(s)
Metapneumovirus , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Humanos , Inmunización , Ratones , Proteínas Virales de Fusión
12.
J Virol ; 84(16): 7970-82, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20534864

RESUMEN

Human respiratory syncytial virus (HRSV) fusion (F) protein is an essential component of the virus envelope that mediates fusion of the viral and cell membranes, and, therefore, it is an attractive target for drug and vaccine development. Our aim was to analyze the neutralizing mechanism of anti-F antibodies in comparison with other low-molecular-weight compounds targeted against the F molecule. It was found that neutralization by anti-F antibodies is related to epitope specificity. Thus, neutralizing and nonneutralizing antibodies could bind equally well to virions and remained bound after ultracentrifugation of the virus, but only the former inhibited virus infectivity. Neutralization by antibodies correlated with inhibition of cell-cell fusion in a syncytium formation assay, but not with inhibition of virus binding to cells. In contrast, a peptide (residues 478 to 516 of F protein [F478-516]) derived from the F protein heptad repeat B (HRB) or the organic compound BMS-433771 did not interfere with virus infectivity if incubated with virus before ultracentrifugation or during adsorption of virus to cells at 4 degrees C. These inhibitors must be present during virus entry to effect HRSV neutralization. These results are best interpreted by asserting that neutralizing antibodies bind to the F protein in virions interfering with its activation for fusion. Binding of nonneutralizing antibodies is not enough to block this step. In contrast, the peptide F478-516 or BMS-433771 must bind to F protein intermediates generated during virus-cell membrane fusion, blocking further development of this process.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antivirales/farmacología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/inmunología , Bencimidazoles/farmacología , Línea Celular , Cricetinae , Humanos , Pruebas de Neutralización , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
13.
PLoS Negl Trop Dis ; 13(7): e0007547, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31276466

RESUMEN

BACKGROUND: Efficient adaptive antiviral cellular and humoral immune responses require previous recognition of viral antigenic peptides bound to human leukocyte antigen (HLA) class I and II molecules, which are exposed on the surface of infected and antigen presenting cells, respectively. The HLA-restricted immune response to Chikungunya virus (CHIKV), a mosquito-borne Alphavirus of the Togaviridae family responsible for severe chronic polyarthralgia and polyarthritis, is largely unknown. METHODOLOGY/PRINCIPAL FINDINGS: In this study, a high-throughput mass spectrometry analysis of complex HLA-bound peptide pools isolated from large amounts of human cells infected with a vaccinia virus (VACV) recombinant expressing CHIKV structural proteins was carried out. Twelve viral ligands from the CHIKV polyprotein naturally presented by different HLA-A, -B, and -C class I, and HLA-DR and -DP class II molecules were identified. CONCLUSIONS/SIGNIFICANCE: The immunoprevalence of the HLA class II but not the HLA class I-restricted cellular immune response against the CHIKV structural polyprotein was greater than that against the VACV vector itself. In addition, most of the CHIKV HLA class I and II ligands detected by mass spectrometry are not conserved compared to its closely related O'nyong-nyong virus. These findings have clear implications for analysis of both cytotoxic and helper immune responses against CHIKV as well as for the future studies focused in the exacerbated T helper response linked to chronic musculoskeletal disorders in CHIKV patients.


Asunto(s)
Fiebre Chikungunya/prevención & control , Antígenos de Histocompatibilidad Clase II/inmunología , Proteómica , Virus Vaccinia , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Animales , Animales Modificados Genéticamente , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Fiebre Chikungunya/inmunología , Virus Chikungunya , Humanos , Inmunidad Celular , Inmunogenicidad Vacunal , Espectrometría de Masas , Ratones , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas Virales/genética
14.
Front Microbiol ; 8: 2301, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29213258

RESUMEN

The influence of age and maternal antibodies on the antibody responses to human respiratory syncytial virus (hRSV) glycoproteins in very young children has been a matter of controversy. Both, immaturity of the immune system at very early age and suppression of the host immune response by high level of maternal antibodies have been claimed to limit the host antibody response to virus infection and to jeopardize the use of hRSV vaccines under development in that age group. Hence, the antibody responses to the two major hRSV glycoproteins (F and G) were evaluated in children younger than 2 years, hospitalized with laboratory confirmed hRSV bronchiolitis. A strong negative correlation was found between the titre of circulating ELISA antibodies directed against either prefusion or postfusion F in the acute phase, but not age, and their fold change at convalescence. These changes correlated also with the level of circulating neutralizing antibodies in sera. As reported in adults, most neutralizing antibodies in a subset of tested sera could not be depleted with postfusion F, suggesting that they were mostly directed against prefusion-specific epitopes. In contrast, a weak negative association was found for group-specific anti-G antibodies in the acute phase and their fold change at convalescence only after correcting for the antigenic group of the infecting virus. In addition, large discrepancies were observed in some individuals between the antibody responses specific for F and G glycoproteins. These results illustrate the complexity of the anti-hRSV antibody responses in children experiencing a primary severe infection and the influence of preexisting maternal antibodies on the host response, factors that should influence hRSV serological studies as well as vaccine development.

15.
J Virol Methods ; 224: 1-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26275682

RESUMEN

Paramyxovirus entry into cells requires fusion of the viral and cell membranes mediated by one of the major virus glycoproteins, the fusion (F) glycoprotein which transits from a metastable pre-fusion conformation to a highly stable post-fusion structure during the membrane fusion process. F protein refolding involves large conformational changes of the protein trimer. One of these changes results in assembly of two heptad repeat sequences (HRA and HRB) from each protomer into a six-helix bundle (6HB) motif. To assist in distinguishing pre- and post-fusion conformations of the Pneumovirinae F proteins, and as extension of previous work (Palomo et al., 2014), a general strategy was designed to obtain polyclonal and particularly monoclonal antibodies specific of the 6HB motif of the Pneumovirinae fusion protein. The antibodies reported here should assist in the characterization of the structural changes that the F protein of human metapneumovirus or respiratory syncytial virus experiences during the process of membrane fusion.


Asunto(s)
Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Antivirales/aislamiento & purificación , Pneumovirinae/inmunología , Proteínas Virales de Fusión/inmunología , Animales , Femenino , Ratones Endogámicos BALB C , Conformación Proteica , Conejos , Proteínas Virales de Fusión/química
16.
PLoS One ; 10(6): e0130829, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26107504

RESUMEN

The respiratory syncytial virus (RSV) fusion protein F is considered an attractive vaccine candidate especially in its prefusion conformation. We studied whether recombinant soluble RSV F proteins could be stabilized in a prefusion-like conformation by mutation of heptad repeat B (HRB). The results show that soluble, trimeric, non-cleaved RSV F protein, produced by expression of the furin cleavage site-mutated F ectodomain extended with a GCN4 trimerization sequence, is efficiently recognized by pre- as well as postfusion-specific antibodies. In contrast, a similar F protein completely lacking HRB displayed high reactivity with prefusion-specific antibodies recognizing antigenic site Ø, but did not expose postfusion-specific antigenic site I, in agreement with this protein maintaining a prefusion-like conformation. These features were dependent on the presence of the GCN4 trimerization domain. Absence of cleavage also contributed to binding of prefusion-specific antibodies. Similar antibody reactivity profiles were observed when the prefusion form of F was stabilized by the introduction of cysteine pairs in HRB. To study whether the inability to form the 6HB was responsible for the prefusion-like antibody reactivity profile, alanine mutations were introduced in HRB. Although introduction of alanine residues in HRB inhibited the formation of the 6HB, the exposure of postfusion-specific antigenic site I was not prevented. In conclusion, proteins that are not able to form the 6HB, due to mutation of HRB, may still display postfusion-specific antigenic site I. Replacement of HRB by the GCN4 trimerization domain in a non-cleaved soluble F protein resulted, however, in a protein with prefusion-like characteristics, suggesting that this HRB-lacking protein may represent a potential prefusion F protein subunit vaccine candidate.


Asunto(s)
Anticuerpos Antivirales/farmacología , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Virus Sincitial Respiratorio Humano/genética , Proteínas Virales de Fusión/genética , Anticuerpos Neutralizantes/farmacología , Sitios de Unión , Línea Celular Tumoral , Células Epiteliales/patología , Células Epiteliales/virología , Expresión Génica , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología , Virus Sincitial Respiratorio Humano/metabolismo , Proteínas Virales de Fusión/antagonistas & inhibidores , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo
17.
Virology ; 460-461: 119-27, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25010277

RESUMEN

Human respiratory syncytial virus (hRSV) has two major surface glycoproteins (G and F) anchored in the lipid envelope. Membrane fusion promoted by hRSV_F occurs via refolding from a pre-fusion form to a highly stable post-fusion state involving large conformational changes of the F trimer. One of these changes results in assembly of two heptad repeat sequences (HRA and HRB) into a six-helix bundle (6HB) motif. To assist in distinguishing pre- and post-fusion conformations of hRSV_F, we have prepared polyclonal (α-6HB) and monoclonal (R145) rabbit antibodies specific for the 6HB. Among other applications, these antibodies were used to explore the requirements of 6HB formation by isolated protein segments or peptides and by truncated mutants of the F protein. Site-directed mutagenesis and electron microscopy located the R145 epitope in the post-fusion hRSV_F at a site distantly located from previously mapped epitopes, extending the repertoire of antibodies that can decorate the F molecule.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/química , Proteínas Virales de Fusión/química , Animales , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/inmunología , Humanos , Estructura Secundaria de Proteína , Conejos , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
18.
J Gen Virol ; 88(Pt 10): 2719-2723, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17872524

RESUMEN

Chimeric 101F (ch101F) is a mouse-human chimeric anti-human respiratory syncytial virus (HRSV) neutralizing antibody that recognizes residues within antigenic site IV, V, VI of the fusion (F) glycoprotein. The binding of ch101F to a series of peptides overlapping aa 422-438 spanning antigenic site IV, V, VI was analysed. Residues 423-436 comprise the minimal peptide sequence for ch101F binding. Substitution analysis revealed that R429 and K433 are critical for ch101F binding, whilst K427 makes a minor contribution. Binding of ch101F to a series of single mutations at positions 427, 429 and 433 in the F protein expressed recombinantly on the cell surface confirmed the peptide results. Sequence analysis of viruses selected for resistance to neutralization by ch101F indicated that a single change (K433T) in the F protein allowed ch101F escape. The results confirm that ch101F and palivizumab have different epitope specificity and define key residues for ch101F recognition.


Asunto(s)
Virus Sincitial Respiratorio Humano/genética , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales , Animales , Anticuerpos Monoclonales , Biotinilación , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Fragmentos de Péptidos/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
19.
J Med Virol ; 76(2): 248-55, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15834867

RESUMEN

Antibodies directed against human respiratory syncytial virus (HRSV) glycoproteins were depleted from a commercial immunoglobulin preparation (RespiGam) by two different methods. The first method consisted of repeated adsorption of RespiGam to Sepharose beads with covalently bound soluble forms of the two major viral glycoproteins (F or G). The second method consisted of adsorption of immunoglobulins to live cells expressing F or G glycoproteins on their surfaces after infection with vaccinia virus recombinants. While the first method removed efficiently antibodies that reacted with F and/or G glycoproteins by ELISA, it was inefficient in the elimination of anti-HRSV neutralizing antibodies. In contrast, the second method removed efficiently anti-HRSV antibodies that both reacted by ELISA and neutralized virus infectivity. These results confirm that human neutralizing antibodies are directed exclusively against HRSV F and G glycoproteins, and, they raise the possibility that F and G glycoproteins inserted into cell membranes differ antigenically from their soluble forms linked covalently to Sepharose beads.


Asunto(s)
Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/inmunología , Eliminación de Componentes Sanguíneos/métodos , Cromatografía de Afinidad , Inmunoglobulinas Intravenosas/inmunología , Técnicas de Inmunoadsorción , Virus Sincitiales Respiratorios/inmunología , Adsorción , Antígenos Virales/inmunología , Ensayo de Inmunoadsorción Enzimática , Glicoproteínas/inmunología , Humanos , Pruebas de Neutralización , Virus Vaccinia
20.
J Clin Microbiol ; 43(5): 2266-73, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15872254

RESUMEN

The genetic and antigenic variability of human respiratory syncytial virus (HRSV) strains isolated in Buenos Aires from 1995 to 2001 was evaluated by partial nucleotide sequencing of the G gene and enzyme-linked immunosorbent assay analysis with anti-G monoclonal antibodies. Phylogenetic analyses showed that 37 group A strains clustered into five genotypes, whereas 20 group B strains clustered into three genotypes. Group A showed more genetic variability than group B. A close correlation between genotypes and antigenic patterns was observed. Changes detected in the G protein of viruses from both groups included (i) amino acid substitutions and(ii) differences in protein length due to either changes in stop codon usage or sequence duplications. Three B strains from 1999 exhibited a duplication of 20 amino acids, while one B strain from 2001 had 2 amino acids duplicated. The comparison among Argentinean HRSV strains and viruses isolated in other geographical areas during different epidemics is discussed.


Asunto(s)
Antígenos Virales/sangre , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Virus Sincitial Respiratorio Humano/genética , Secuencia de Aminoácidos , Argentina , Secuencia de Bases , Línea Celular Tumoral , Preescolar , Cartilla de ADN , Humanos , Datos de Secuencia Molecular , Filogenia , ARN Viral/genética , ARN Viral/aislamiento & purificación , Virus Sincitial Respiratorio Humano/clasificación , Virus Sincitial Respiratorio Humano/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA