Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Eur J Pharm Biopharm ; 133: 85-95, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30312742

RESUMEN

Bird to human transmission of high pathogenicity avian influenza virus (HPAIV) poses a significant risk of triggering a flu pandemic in the human population. Therefore, vaccination of susceptible poultry during an HPAIV outbreak might be the best remedy to prevent such transmissions. To this end, suitable formulations and an effective mass vaccination method that can be translated to field settings needs to be developed. Our previous study in chickens has shown that inhalation of a non-adjuvanted dry powder influenza vaccine formulation during normal breathing results in partial protection against lethal influenza challenge. The aim of the present study was to improve the effectiveness of pulmonary vaccination by increasing the vaccine dose deposited in the lungs and by the use of suitable adjuvants. Two adjuvants, namely, Bacterium-like Particles (BLP) and Advax, were spray freeze dried with influenza vaccine into dry powder formulations. Delivery of dry formulations directly at the syrinx revealed that BLP and Advax had the potential to boost either systemic or mucosal immune responses or both. Upon passive inhalation of dry influenza vaccine formulations in an optimized set-up, BLP and Advax/BLP adjuvanted formulations induced significantly higher systemic immune responses than the non-adjuvanted formulation. Remarkably, all vaccinated animals not only survived a lethal influenza challenge, but also did not show any shedding of challenge virus except for two out of six animals in the Advax group. Overall, our results indicate that passive inhalation is feasible, effective and suitable for mass vaccination of chickens if it can be adapted to field settings.


Asunto(s)
Pollos/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Polvos/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Administración por Inhalación , Animales , Anticuerpos Antivirales/inmunología , Química Farmacéutica/métodos , Pollos/virología , Inmunidad Mucosa/inmunología , Inmunización/métodos , Vacunación/métodos , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
2.
J Gen Virol ; 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30067172

RESUMEN

Avian coronavirus infectious bronchitis virus (IBV) infects domestic fowl, resulting in respiratory disease and causing serious losses in unprotected birds. Its control is mainly achieved by using live attenuated vaccines. Here we explored the possibilities for rationally attenuating IBV to improve our knowledge regarding the function of IBV accessory proteins and for the development of next-generation vaccines with the recently established reverse genetic system for IBV H52 based on targeted RNA recombination and selection of recombinant viruses in embryonated eggs. To this aim, we selectively removed accessory genes 3a, 3b, 5a and 5b individually, and rescued the resulting recombinant (r) rIBV-Δ3a, rIBV-Δ3b, rIBV-Δ5a and rIBV-Δ5b. In vitro inoculation of chicken embryo kidney cells with recombinant and wild-type viruses demonstrated that the accessory protein 5b is involved in the delayed activation of the interferon response of the host after IBV infection. Embryo mortality after the inoculation of 8-day-old embryonated chicken eggs with recombinant and wild-type viruses showed that rIBV-Δ3b, rIBV-Δ5a and rIBV-Δ5b had an attenuated phenotype in ovo, with reduced titres at 6 h p.i. and 12 h p.i. for all viruses, while growing to the same titre as wild-type rIBV at 48 h p.i. When administered to 1-day-old chickens, rIBV-Δ3a, rIBV-Δ3b, rIBV-Δ5a and rIBV-Δ5b showed reduced ciliostasis in comparison to the wild-type viruses. In conclusion, individual deletion of accessory genes in IBV H52 resulted in mutant viruses with an attenuated phenotype.

3.
Virus Res ; 255: 14-23, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29936068

RESUMEN

The ability to infect and replicate in monocytes/macrophages is a critically distinguishing feature between the two feline coronavirus (FCoV) pathotypes: feline enteric coronavirus (FECV; low-virulent) and feline infectious peritonitis virus (FIPV; lethal). Previously, by comparing serotype II strains FIPV 79-1146 and FECV 79-1683 and recombinant chimeric forms thereof in cultured feline bone marrow macrophages, we mapped this difference to the C-terminal part of the viral spike (S) protein (S2). In view of the later identified diagnostic difference in this very part of the S protein of serotype I FCoV pathotypes, the present study aimed to further define the contribution of the earlier observed ten amino acids difference to the serotype II virus phenotype in macrophages. Using targeted RNA recombination as a reverse genetics system we introduced the mutations singly and in combinations into the S gene and evaluated their effects on the infection characteristics of the mutant viruses in macrophages. While some of the single mutations had a significant effect, none of them fully reverted the infection phenotype. Only by combining five specific mutations the infections mediated by the FIPV and FECV spike proteins could be fully blocked or potentiated, respectively. Hence, the differential macrophage infection phenotype is caused by the cooperative effect of five mutations, which occur in five functionally different domains of the spike fusion subunit S2. The significance of these observations will be discussed, taking into account also some questions related to the identity of the virus strains used.


Asunto(s)
Coronavirus Felino/patogenicidad , Peritonitis Infecciosa Felina/virología , Macrófagos/virología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Sustitución de Aminoácidos , Animales , Gatos , Células Cultivadas , Coronavirus Felino/genética , Coronavirus Felino/fisiología , Ratones , Dominios Proteicos , ARN Viral/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Recombinación Genética , Serogrupo , Tropismo Viral , Internalización del Virus , Replicación Viral
4.
Vaccine ; 36(8): 1085-1092, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29366709

RESUMEN

Avian coronavirus infectious bronchitis virus (IBV) is a respiratory pathogen of chickens, causing severe economic losses in poultry industry worldwide. Live attenuated viruses are widely used in both the broiler and layer industry because of their efficacy and ability to be mass applied. Recently, we established a novel reverse genetics system based on targeted RNA recombination to manipulate the genome of IBV strain H52. Here we explore the possibilities to attenuate IBV in a rational way in order to generate safe and effective vaccines against virulent IBV (van Beurden et al., 2017). To this end, we deleted the nonessential group-specific accessory genes 3 and/or 5 in the IBV genome by targeted RNA recombination and selected the recombinant viruses in embryonated eggs. The resulting recombinant (r) rIBV-Δ3ab, rIBV-Δ5ab, and rIBV-Δ3ab5ab could be rescued and grew to the same virus titer as recombinant and wild type IBV strain H52. Thus, genes 3ab and 5ab are not essential for replication in ovo. When administered to one-day-old chickens, rIBV-Δ3ab, rIBV-Δ5ab, and rIBV-Δ3ab5ab showed reduced ciliostasis as compared to rIBV H52 and wild type H52, indicating that the accessory genes contribute to the pathogenicity of IBV. After homologous challenge with the virulent IBV strain M41, all vaccinated chickens were protected against disease based on reduced loss of ciliary movement in the trachea compared to the non-vaccinated but challenged controls. Taken together, deletion of accessory genes 3ab and/or 5ab in IBV resulted in mutant viruses with an attenuated phenotype and the ability to induce protection in chickens. Hence, targeted RNA recombination based on virulent IBV provides opportunities for the development of a next generation of rationally designed live attenuated IBV vaccines.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Virus de la Bronquitis Infecciosa/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas Virales/inmunología , Animales , Embrión de Pollo , Pollos , Eliminación de Gen , Virus de la Bronquitis Infecciosa/genética , Enfermedades de las Aves de Corral/virología , ARN Viral/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/inmunología , Carga Viral , Vacunas Virales/administración & dosificación
5.
Vet Microbiol ; 212: 31-38, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29173585

RESUMEN

Canine infectious respiratory disease (CIRD) is a major cause of morbidity in dogs worldwide, and is associated with a number of new and emerging pathogens. In a large multi-centre European study the prevalences of four key emerging CIRD pathogens; canine respiratory coronavirus (CRCoV), canine pneumovirus (CnPnV), influenza A, and Mycoplasma cynos (M. cynos); were estimated, and risk factors for exposure, infection and clinical disease were investigated. CIRD affected 66% (381/572) of the dogs studied, including both pet and kennelled dogs. Disease occurrence and severity were significantly reduced in dogs vaccinated against classic CIRD agents, canine distemper virus (CDV), canine adenovirus 2 (CAV-2) and canine parainfluenza virus (CPIV), but substantial proportions (65.7%; 201/306) of vaccinated dogs remained affected. CRCoV and CnPnV were highly prevalent across the different dog populations, with overall seropositivity and detection rates of 47% and 7.7% for CRCoV, and 41.7% and 23.4% for CnPnV, respectively, and their presence was associated with increased occurrence and severity of clinical disease. Antibodies to CRCoV had a protective effect against CRCoV infection and more severe clinical signs of CIRD but antibodies to CnPnV did not. Involvement of M. cynos and influenza A in CIRD was less apparent. Despite 45% of dogs being seropositive for M. cynos, only 0.9% were PCR positive for M. cynos. Only 2.7% of dogs were seropositive for Influenza A, and none were positive by PCR.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Enfermedades de los Perros/epidemiología , Infecciones por Mycoplasma/veterinaria , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Pneumovirus/veterinaria , Infecciones del Sistema Respiratorio/veterinaria , Animales , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/microbiología , Enfermedades Transmisibles Emergentes/veterinaria , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Coronavirus Canino/aislamiento & purificación , Enfermedades de los Perros/microbiología , Perros , Monitoreo Epidemiológico , Europa (Continente)/epidemiología , Virus de la Influenza A/aislamiento & purificación , Mycoplasma/aislamiento & purificación , Infecciones por Mycoplasma/epidemiología , Infecciones por Mycoplasma/microbiología , Infecciones por Orthomyxoviridae/epidemiología , Infecciones por Orthomyxoviridae/virología , Pneumovirus/aislamiento & purificación , Infecciones por Pneumovirus/epidemiología , Infecciones por Pneumovirus/virología , Prevalencia , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología
6.
Virol J ; 14(1): 109, 2017 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-28606144

RESUMEN

BACKGROUND: Avian coronavirus infectious bronchitis virus (IBV) is a respiratory pathogen of chickens that causes severe economic losses in the poultry industry worldwide. Major advances in the study of the molecular biology of IBV have resulted from the development of reverse genetics systems for the highly attenuated, cell culture-adapted, IBV strain Beaudette. However, most IBV strains, amongst them virulent field isolates, can only be propagated in embryonated chicken eggs, and not in continuous cell lines. METHODS: We established a reverse genetics system for the IBV strain H52, based on targeted RNA recombination in a two-step process. First, a genomic and a chimeric synthetic, modified IBV RNA were co-transfected into non-susceptible cells to generate a recombinant chimeric murinized (m) IBV intermediate (mIBV). Herein, the genomic part coding for the spike glycoprotein ectodomain was replaced by that of the coronavirus mouse hepatitis virus (MHV), allowing for the selection and propagation of recombinant mIBV in murine cells. In the second step, mIBV was used as the recipient. To this end a recombination with synthetic RNA comprising the 3'-end of the IBV genome was performed by introducing the complete IBV spike gene, allowing for the rescue and selection of candidate recombinants in embryonated chicken eggs. RESULTS: Targeted RNA recombination allowed for the modification of the 3'-end of the IBV genome, encoding all structural and accessory genes. A wild-type recombinant IBV was constructed, containing several synonymous marker mutations. The in ovo growth kinetics and in vivo characteristics of the recombinant virus were similar to those of the parental IBV strain H52. CONCLUSIONS: Targeted RNA recombination allows for the generation of recombinant IBV strains that are not able to infect and propagate in continuous cell lines. The ability to introduce specific mutations holds promise for the development of rationally designed live-attenuated IBV vaccines and for studies into the biology of IBV in general.


Asunto(s)
Virus de la Bronquitis Infecciosa/crecimiento & desarrollo , Virus de la Bronquitis Infecciosa/genética , ARN Viral/genética , Recombinación Genética , Genética Inversa/métodos , Animales , Línea Celular , Pollos , Marcación de Gen/métodos , Ratones
7.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28381581

RESUMEN

Porcine epidemic diarrhea virus (PEDV) causes enteric disease in pigs, resulting in significant economic losses to the swine industry worldwide. Current vaccination approaches against this emerging coronavirus are only partially effective, though natural infection protects pigs against reinfection and provides lactogenic immunity to suckling piglets. The viral spike (S) glycoprotein, responsible for receptor binding and cell entry, is the major target for neutralizing antibodies. However, knowledge of antibody epitopes, their nature and location in the spike structure, and the mechanisms by which the antibodies interfere with infection is scarce. Here we describe the generation and characterization of 10 neutralizing and nonneutralizing mouse monoclonal antibodies raised against the S1 receptor binding subunit of the S protein. By expression of different S1 protein fragments, six antibody epitope classes distributed over the five structural domains of the S1 subunit were identified. Characterization of antibodies for cross-reactivity and cross-neutralization revealed antigenic differences among PEDV strains. The epitopes of potent neutralizing antibodies segregated into two epitope classes and mapped within the N-terminal sialic acid binding domain and in the more C-terminal receptor binding domain. Antibody neutralization escape mutants displayed single amino acid substitutions that impaired antibody binding and neutralization and defined the locations of the epitopes. Our observations picture the antibody epitope landscape of the PEDV S1 subunit and reveal that its cell attachment domains are key targets of neutralizing antibodies.IMPORTANCE Porcine epidemic diarrhea virus (PEDV), an emerging porcine coronavirus, causes an economically important enteric disease in pigs. Effective PEDV vaccines for disease control are currently lacking. The spike (S) glycoprotein on the virion surface is the key player in virus cell entry and, therefore, the main target of neutralizing antibodies. To understand the antigenic landscape of the PEDV spike protein, we developed monoclonal antibodies against the spike protein's S1 receptor binding region and characterized their epitopes, neutralizing activity, and cross-reactivity toward multiple PEDV strains. Epitopes of antibodies segregated into six epitope classes dispersed over the multidomain S1 structure. Monoclonal antibodies revealed antigenic variability in B-cell epitopes between PEDV strains. The epitopes of neutralizing antibodies mapped to two distinct domains in S1 that are involved in binding to carbohydrate and proteinaceous cell surface molecules, respectively, indicating the importance of these cell attachment sites on the PEDV spike protein in eliciting a protective humoral immune response.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Virus de la Diarrea Epidémica Porcina/química , Virus de la Diarrea Epidémica Porcina/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Sustitución de Aminoácidos , Animales , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/metabolismo , Afinidad de Anticuerpos , Epítopos de Linfocito B/química , Epítopos de Linfocito B/clasificación , Epítopos de Linfocito B/inmunología , Ratones , Mutación , Pruebas de Neutralización , Virus de la Diarrea Epidémica Porcina/genética , Virus de la Diarrea Epidémica Porcina/fisiología , Glicoproteína de la Espiga del Coronavirus/genética , Porcinos
8.
Virus Res ; 235: 6-13, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28363778

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is an emerging pathogenic coronavirus that causes a significant economic burden to the swine industry. The virus infects the intestinal epithelium and causes villous atrophy, resulting in diarrhea and dehydration. Interaction of the viral spike (S) surface glycoprotein - through its S1 subunit - with the host cell receptor is the first step in infection and the main determinant for virus tropism. As for several other alphacoronaviruses including the porcine transmissible gastroenteritis virus (TGEV) and the human coronavirus 229E (HCoV-229E), the aminopeptidase N (APN) protein was reported to be a functional receptor for PEDV. In this study we examined the role of APN as a receptor. We show that overexpression of porcine APN renders MDCK cells susceptible to TGEV, but not to PEDV. Consistently, unlike TGEV-S1, PEDV-S1 exhibited no binding to cell-surface expressed APN or to a soluble version of APN. Moreover, preincubation of these viruses with soluble APN or pretreatment of APN expressing ST cells with soluble TGEV-S1 blocked TGEV infection, but had no effect on infection by PEDV. The combined observations indicated that APN is not required for PEDV infection. To definitively prove this conclusion, we applied CRISPR/Cas9 genome engineering to knock out APN expression in PEDV-susceptible porcine (ST) and human cell lines (Huh7 and HeLa). As a consequence these cells no longer bound TGEV-S1 and HCoV-229E-S1 at their surface and were resistant to infection by the corresponding viruses. However, genetic ablation of APN expression had no effect on their infectability by PEDV, demonstrating that APN is not essential for PEDV cell entry.


Asunto(s)
Antígenos CD13/metabolismo , Virus de la Diarrea Epidémica Porcina/fisiología , Receptores Virales/metabolismo , Internalización del Virus , Animales , Antígenos CD13/genética , Línea Celular , Perros , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Receptores Virales/genética , Porcinos
9.
Virology ; 503: 83-93, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135661

RESUMEN

Highly pathogenic H5N1 avian influenza A viruses display a remarkable genetic and antigenic diversity. We examined to what extent genetic distances between several H5N1 viruses from different clades correlate with antigenic differences and vaccine performance. H5-specific antisera were generated, and cross-reactivity and antigenic distances between 12 different viruses were determined. In general, antigenic distances increased proportional to genetic distances although notable exceptions were observed. Antigenic distances correlated better with genetic variation in 27 selected, antigenically-relevant H5 residues, than in the complete HA1 domain. Variation in these selected residues could accurately predict the antigenic distances for a novel H5N8 virus. Protection provided by vaccines against heterologous H5N1 challenge viruses indicated that cross-protection also correlates better with genetic variation in the selected antigenically-relevant residues than in complete HA1. When time is limited, variation at these selected residues may be used to accurately predict antigenic distance and vaccine performance.


Asunto(s)
Anticuerpos Antivirales/inmunología , Variación Antigénica/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Animales , Variación Antigénica/genética , Antígenos Virales/genética , Antígenos Virales/inmunología , Línea Celular , Pollos/virología , Protección Cruzada/inmunología , Reacciones Cruzadas/genética , Reacciones Cruzadas/inmunología , Perros , Variación Genética/genética , Variación Genética/inmunología , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/inmunología , Gripe Aviar/virología , Células de Riñón Canino Madin Darby , Enfermedades de las Aves de Corral/virología , Pruebas Serológicas , Células Sf9 , Spodoptera , Vacunación
10.
Protein Sci ; 26(1): 113-121, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27667334

RESUMEN

The tremendous pandemic potential of coronaviruses was demonstrated twice in the last 15 years by two global outbreaks of deadly pneumonia. Entry of coronaviruses into cells is mediated by the transmembrane spike glycoprotein S, which forms a trimer carrying receptor-binding and membrane fusion functions. Despite their biomedical importance, coronavirus S glycoproteins have proven difficult targets for structural characterization, precluding high-resolution studies of the biologically relevant trimer. Recent technological developments in single particle cryo-electron microscopy allowed us to determine the first structure of a coronavirus S glycoprotein trimer which provided a framework to understand the mechanisms of viral entry and suggested potential inhibition strategies for this family of viruses. Here, we describe the key factors that enabled this breakthrough.


Asunto(s)
Microscopía por Crioelectrón/métodos , Coronavirus del Síndrome Respiratorio de Oriente Medio/ultraestructura , Modelos Moleculares , Multimerización de Proteína , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Coronavirus del Síndrome Respiratorio de Oriente Medio/química , Estructura Cuaternaria de Proteína , Glicoproteína de la Espiga del Coronavirus/química
11.
Virus Res ; 226: 117-127, 2016 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-27317167

RESUMEN

Porcine epidemic diarrhea virus (PEDV), a coronavirus discovered more than 40 years ago, regained notoriety recently by its devastating outbreaks in East Asia and the Americas, causing substantial economic losses to the swine husbandry. The virus replicates extensively and almost exclusively in the epithelial cells of the small intestine resulting in villus atrophy, malabsorption and severe diarrhea. Cellular entry of this enveloped virus is mediated by the large spike (S) glycoprotein, trimers of which mediate virus attachment to the target cell and subsequent membrane fusion. The S protein has a multidomain architecture and has been reported to bind to carbohydrate (sialic acid) and proteinaceous (aminopeptidase N) cell surface molecules. PEDV propagation in vitro requires the presence of trypsin(-like) proteases in the culture medium, which capacitates the fusion function of the S protein. Here we review the current data on PEDV entry into its host cell, including therein our new observations regarding the functional role of the sialic acid binding activity of the S protein in virus infection. Moreover, we summarize the recent progress on the proteolytic activation of PEDV S proteins, and discuss factors that may determine tissue tropism of PEDV in vivo.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Virus de la Diarrea Epidémica Porcina/fisiología , Enfermedades de los Porcinos/virología , Acoplamiento Viral , Animales , Ácido N-Acetilneuramínico/metabolismo , Virus de la Diarrea Epidémica Porcina/ultraestructura , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Receptores Virales , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Relación Estructura-Actividad , Porcinos , Enfermedades de los Porcinos/epidemiología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/metabolismo
12.
J Virol ; 90(13): 5965-5977, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27099320

RESUMEN

UNLABELLED: Antibodies against the fusion (F) protein of respiratory syncytial virus (RSV) play an important role in the protective immune response to this important respiratory virus. Little is known, however, about antibody levels against multiple F-specific epitopes induced by infection or after vaccination against RSV, while this is important to guide the evaluation of (novel) vaccines. In this study, we analyzed antibody levels against RSV proteins and F-specific epitopes in human sera and in sera of vaccinated and experimentally infected cotton rats and the correlation thereof with virus neutralization. Analysis of human sera revealed substantial diversity in antibody levels against F-, G (attachment)-, and F-specific epitopes between individuals. The highest correlation with virus neutralization was observed for antibodies recognizing prefusion-specific antigenic site Ø. Nevertheless, our results indicate that high levels of antibodies targeting other parts of the F protein can also mediate a potent antiviral antibody response. In agreement, sera of experimentally infected cotton rats contained high neutralizing activity despite lacking antigenic site Ø-specific antibodies. Strikingly, vaccination with formalin-inactivated RSV (FI-RSV) exclusively resulted in the induction of poorly neutralizing antibodies against postfusion-specific antigenic site I, although antigenic sites I, II, and IV were efficiently displayed in FI-RSV. The apparent immunodominance of antigenic site I in FI-RSV likely explains the low levels of neutralizing antibodies upon vaccination and challenge and may play a role in the vaccination-induced enhancement of disease observed with such preparations. IMPORTANCE: RSV is an importance cause of hospitalization of infants. The development of a vaccine against RSV has been hampered by the disastrous results obtained with FI-RSV vaccine preparations in the 1960s that resulted in vaccination-induced enhancement of disease. To get a better understanding of the antibody repertoire induced after infection or after vaccination against RSV, we investigated antibody levels against fusion (F) protein, attachment (G) protein, and F-specific epitopes in human and animal sera. The results indicate the importance of prefusion-specific antigenic site Ø antibodies as well as of antibodies targeting other epitopes in virus neutralization. However, vaccination of cotton rats with FI-RSV specifically resulted in the induction of weakly neutralizing, antigenic site I-specific antibodies, which may play a role in the enhancement of disease observed after vaccination with such preparations.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Epítopos/inmunología , Inmunidad Innata , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Anticuerpos Antivirales/inmunología , Formaldehído , Humanos , Epítopos Inmunodominantes/sangre , Epítopos Inmunodominantes/inmunología , Ratas , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/química , Virus Sincitial Respiratorio Humano/química , Sigmodontinae , Vacunación/efectos adversos , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/efectos adversos , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Proteínas Virales/inmunología
13.
Nature ; 531(7592): 114-117, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26855426

RESUMEN

The tremendous pandemic potential of coronaviruses was demonstrated twice in the past few decades by two global outbreaks of deadly pneumonia. Entry of coronaviruses into cells is mediated by the transmembrane spike glycoprotein S, which forms a trimer carrying receptor-binding and membrane fusion functions. S also contains the principal antigenic determinants and is the target of neutralizing antibodies. Here we present the structure of a mouse coronavirus S trimer ectodomain determined at 4.0 Å resolution by single particle cryo-electron microscopy. It reveals the metastable pre-fusion architecture of S and highlights key interactions stabilizing it. The structure shares a common core with paramyxovirus F proteins, implicating mechanistic similarities and an evolutionary connection between these viral fusion proteins. The accessibility of the highly conserved fusion peptide at the periphery of the trimer indicates potential vaccinology strategies to elicit broadly neutralizing antibodies against coronaviruses. Finally, comparison with crystal structures of human coronavirus S domains allows rationalization of the molecular basis for species specificity based on the use of spatially contiguous but distinct domains.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Hepatitis Murina/química , Virus de la Hepatitis Murina/ultraestructura , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Línea Celular , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Drosophila melanogaster , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/inmunología , Multimerización de Proteína , Estructura Terciaria de Proteína , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Virales/química , Vacunas Virales/inmunología , Internalización del Virus
15.
J Invertebr Pathol ; 134: 15-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26743500

RESUMEN

Baculoviruses are a group of enveloped, double-stranded DNA insect viruses with budded (BV) and occlusion-derived (ODV) virions produced during their infection cycle. BVs are commonly described as rod shaped particles with a high apical density of protein extensions (spikes) on the lipid envelope surface. However, due to the fragility of BVs the conventional purification and electron microscopy (EM) staining methods considerably distort the native viral structure. Here, we use cryo-EM analysis to reveal the near-native morphology of two intensively studied baculoviruses, Autographa californica multicapsid nucleopolyhedrovirus (AcMNPV) and Spodoptera exigua MNPV (SeMNPV), as models for BVs carrying GP64 and F as envelope fusion protein on the surface. The now well-preserved AcMNPV and SeMNPV BV particles have a remarkable elongated, ovoid shape leaving a large, lateral space between nucleocapsid (NC) and envelope. Consistent with previous findings the NC has a distinctive cap and base structure interacting tightly with the envelope. This tight interaction may explain the partial retaining of the envelope on both ends of the NC and the disappearance of the remainder of the BV envelope in the negative-staining EM images. Cryo-EM also reveals that the viral envelope contains two layers with a total thickness of ≈ 6-7 nm, which is significantly thicker than a usual biological membrane (<4 nm) as measured by X-ray scanning. Most spikes are densely clustered at the two apical ends of the virion although some envelope proteins are also found more sparsely on the lateral regions. The spikes on the surface of AcMNPV BVs appear distinctly different from those of SeMNPV. Based on our observations we propose a new near-native structural model of baculovirus BVs.


Asunto(s)
Nucleopoliedrovirus/ultraestructura , Virión/ultraestructura , Microscopía por Crioelectrón , Tamaño del Genoma , Genoma Viral , Nucleocápside/ultraestructura , Nucleopoliedrovirus/genética , Nucleopoliedrovirus/fisiología
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.
J Virol ; 89(15): 8077-81, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25972540

RESUMEN

The emerging porcine epidemic diarrhea virus (PEDV) requires trypsin supplementation to activate its S protein for membrane fusion and virus propagation in cell culture. By substitution of a single amino acid in the S protein, we created a recombinant PEDV with an artificial furin protease cleavage site N terminal of the putative fusion peptide (PEDV-SFCS). PEDV-SFCS exhibited trypsin-independent cell-cell fusion and was able to replicate in culture cells independently of trypsin, though to low titer.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Furina/metabolismo , Mutación Puntual , Virus de la Diarrea Epidémica Porcina/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Enfermedades de los Porcinos/enzimología , Tripsina/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Infecciones por Coronavirus/enzimología , Infecciones por Coronavirus/virología , Virus de la Diarrea Epidémica Porcina/química , Virus de la Diarrea Epidémica Porcina/fisiología , Procesamiento Proteico-Postraduccional , Glicoproteína de la Espiga del Coronavirus/química , Porcinos , Enfermedades de los Porcinos/virología
18.
Emerg Infect Dis ; 21(5): 842-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25897518

RESUMEN

In 2014, novel highly pathogenic avian influenza A H5N2, H5N5, H5N6, and H5N8 viruses caused outbreaks in Asia, Europe, and North America. The H5 genes of these viruses form a monophyletic group that evolved from a clade 2.3.4 H5N1 variant. This rapid emergence of new H5Nx combinations is unprecedented in the H5N1 evolutionary history.


Asunto(s)
Variación Genética , Hemaglutininas/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Gripe Aviar/virología , Gripe Humana/virología , Animales , Brotes de Enfermedades , Genotipo , Humanos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/patogenicidad , Filogenia , Aves de Corral , Virus Reordenados/clasificación , Virus Reordenados/genética
19.
J Virol ; 89(8): 4434-48, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25653449

RESUMEN

UNLABELLED: In addition to transporting ions, the multisubunit Na(+),K(+)-ATPase also functions by relaying cardiotonic steroid (CTS)-binding-induced signals into cells. In this study, we analyzed the role of Na(+),K(+)-ATPase and, in particular, of its ATP1A1 α subunit during coronavirus (CoV) infection. As controls, the vesicular stomatitis virus (VSV) and influenza A virus (IAV) were included. Using gene silencing, the ATP1A1 protein was shown to be critical for infection of cells with murine hepatitis virus (MHV), feline infectious peritonitis virus (FIPV), and VSV but not with IAV. Lack of ATP1A1 did not affect virus binding to host cells but resulted in inhibited entry of MHV and VSV. Consistently, nanomolar concentrations of the cardiotonic steroids ouabain and bufalin, which are known not to affect the transport function of Na(+),K(+)-ATPase, inhibited infection of cells with MHV, FIPV, Middle East respiratory syndrome (MERS)-CoV, and VSV, but not IAV, when the compounds were present during virus inoculation. Cardiotonic steroids were shown to inhibit entry of MHV at an early stage, resulting in accumulation of virions close to the cell surface and, as a consequence, in reduced fusion. In agreement with an early block in infection, the inhibition of VSV by CTSs could be bypassed by low-pH shock. Viral RNA replication was not affected when these compounds were added after virus entry. The antiviral effect of ouabain could be relieved by the addition of different Src kinase inhibitors, indicating that Src signaling mediated via ATP1A1 plays a crucial role in the inhibition of CoV and VSV infections. IMPORTANCE: Coronaviruses (CoVs) are important pathogens of animals and humans, as demonstrated by the recent emergence of new human CoVs of zoonotic origin. Antiviral drugs targeting CoV infections are lacking. In the present study, we show that the ATP1A1 subunit of Na(+),K(+)-ATPase, an ion transporter and signaling transducer, supports CoV infection. Targeting ATP1A1 either by gene silencing or by low concentrations of the ATP1A1-binding cardiotonic steroids ouabain and bufalin resulted in inhibition of infection with murine, feline, and MERS-CoVs at an early entry stage. Infection with the control virus VSV was also inhibited. Src signaling mediated by ATP1A1 was shown to play a crucial role in the inhibition of virus entry by ouabain and bufalin. These results suggest that targeting the Na(+),K(+)-ATPase using cardiotonic steroids, several of which are FDA-approved compounds, may be an attractive therapeutic approach against CoV and VSV infections.


Asunto(s)
Glicósidos Cardíacos/farmacología , Infecciones por Coronaviridae/fisiopatología , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Animales , Bufanólidos/farmacología , Línea Celular , Chlorocebus aethiops , Coronavirus Felino/fisiología , Silenciador del Gen , Humanos , Concentración de Iones de Hidrógeno , Ratones , Virus de la Hepatitis Murina/fisiología , Ouabaína/farmacología , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Vero
20.
Methods Mol Biol ; 1282: 213-29, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25720483

RESUMEN

Over the last 2 decades, yeast two-hybrid became an invaluable technique to decipher protein-protein interaction networks. In the field of virology, it has proven instrumental to identify virus-host interactions that are involved in viral embezzlement of cellular functions and inhibition of immune mechanisms. Here, we present a yeast two-hybrid protocol that has been used in our laboratory since 2006 to search for cellular partners of more than 300 viral proteins. Our aim was to develop a robust and straightforward pipeline, which minimizes false-positive interactions with a decent coverage of target cDNA libraries, and only requires a minimum of equipment. We also discuss reasons that motivated our technical choices and compromises that had to be made. This protocol has been used to screen most non-structural proteins of murine hepatitis virus (MHV), a member of betacoronavirus genus, against a mouse brain cDNA library. Typical results were obtained and are presented in this report.


Asunto(s)
Virus de la Hepatitis Murina/fisiología , Proteínas del Tejido Nervioso/metabolismo , Técnicas del Sistema de Dos Híbridos , Proteínas Virales/metabolismo , Animales , Interacciones Huésped-Patógeno , Ratones , Acoplamiento Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...