Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Virol ; 96(12): e0032022, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35638848

RESUMEN

Vaccination against influenza virus infection can protect the vaccinee and also reduce transmission to contacts. Not all types of vaccines induce sterilizing immunity via neutralizing antibodies; some instead permit low-level, transient infection. There has been concern that infection-permissive influenza vaccines may allow continued spread in the community despite minimizing symptoms in the vaccinee. We have explored that issue for a universal influenza vaccine candidate that protects recipients by inducing T cell responses and nonneutralizing antibodies. Using a mouse model, we have shown previously that an adenoviral vectored vaccine expressing nucleoprotein (NP) and matrix 2 (M2) provides broad protection against diverse strains and subtypes of influenza A viruses and reduces transmission to contacts in an antigen-specific manner. Here, we use this mouse model to further explore the mechanism and features of that reduction in transmission. Passive immunization did not reduce transmission from infected donors to naive contact animals to whom passive serum had been transferred. Vaccination of antibody-deficient mIgTg-JHD-/- mice, which have intact T cell responses and antigen presentation, reduced transmission in an antigen-specific manner, despite the presence of some virus in the lungs and nasal wash, pointing to a role for cellular immunity. Vaccination at ages ranging from 8 to 60 weeks was able to achieve reduction in transmission. Finally, the immune-mediated reduction in transmission persisted for at least a year after a single-dose intranasal vaccination. Thus, this infection-permissive vaccine reduces virus transmission in a long-lasting manner that does not require antibodies. IMPORTANCE Universal influenza virus vaccines targeting antigens conserved among influenza A virus strains can protect from severe disease but do not necessarily prevent infection. Despite allowing low-level infection, intranasal immunization with adenovirus vectors expressing the conserved antigens influenza nucleoprotein (A/NP) and M2 reduces influenza virus transmission from vaccinated to unvaccinated contact mice. Here, we show that antibodies are not required for this transmission reduction, suggesting a role for T cells. We also show that transmission blocking could be achieved in recipients of different ages and remained effective for at least a year following a single-dose vaccination. Such vaccines could have major public health impacts by limiting viral transmission in the community.


Asunto(s)
Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Adenoviridae , Animales , Anticuerpos Antivirales , Humanos , Inmunidad Celular , Virus de la Influenza A/genética , Vacunas contra la Influenza/inmunología , Gripe Humana , Proteínas de la Nucleocápside/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/transmisión , Linfocitos T/inmunología , Vacunación , Proteínas de la Matriz Viral/inmunología , Proteínas Viroporinas/inmunología
2.
Proc Natl Acad Sci U S A ; 119(13): e2025607119, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35320040

RESUMEN

SignificanceAlthough the need for a universal influenza vaccine has long been recognized, only a handful of candidates have been identified so far, with even fewer advancing in the clinical pipeline. The 24-amino acid ectodomain of M2 protein (M2e) has been developed over the past two decades. However, M2e-based vaccine candidates have shortcomings, including the need for several administrations and the lack of sustained antibody titers over time. We report here a vaccine targeting strategy that has the potential to confer sustained and strong protection upon a single shot of a small amount of M2e antigen. The current COVID-19 pandemic has highlighted the importance of developing versatile, powerful platforms for the rapid deployment of vaccines against any incoming threat.


Asunto(s)
COVID-19 , Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Proteínas de la Matriz Viral , Proteínas Viroporinas , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , COVID-19/prevención & control , Células Dendríticas/inmunología , Humanos , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Pandemias/prevención & control , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/inmunología , Proteínas Viroporinas/inmunología
3.
Sci Rep ; 12(1): 1005, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35046461

RESUMEN

The pandemic of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused a public health emergency, and research on the development of various types of vaccines is rapidly progressing at an unprecedented development speed internationally. Some vaccines have already been approved for emergency use and are being supplied to people around the world, but there are still many ongoing efforts to create new vaccines. Virus-like particles (VLPs) enable the construction of promising platforms in the field of vaccine development. Here, we demonstrate that non-infectious SARS-CoV-2 VLPs can be successfully assembled by co-expressing three important viral proteins membrane (M), envelop (E) and nucleocapsid (N) in plants. Plant-derived VLPs were purified by sedimentation through a sucrose cushion. The shape and size of plant-derived VLPs are similar to native SARS-CoV-2 VLPs without spike. Although the assembled VLPs do not have S protein spikes, they could be developed as formulations that can improve the immunogenicity of vaccines including S antigens, and further could be used as platforms that can carry S antigens of concern for various mutations.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Proteínas M de Coronavirus/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , SARS-CoV-2/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Proteínas Viroporinas/inmunología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Proteínas M de Coronavirus/genética , Proteínas M de Coronavirus/metabolismo , Proteínas de la Nucleocápside de Coronavirus/genética , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Humanos , Nicotiana/inmunología , Nicotiana/metabolismo , Nicotiana/virología , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/metabolismo , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo
4.
Mol Immunol ; 142: 11-21, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34959069

RESUMEN

Senecavirus A (SVA) is an oncolytic virus, which can propagate in human tumor cells and has been used as an oncolytic virotherapy candidate in humans. Besides, SVA circulates in pigs and causes vesicles and coalescing erosions on the snouts and coronary bands in infected pigs and results in neonatal morbidity. SVA has evolved the ability to suppress host innate immune response to benefit viral replication. SVA 3Cpro and 2C protein inhibit the production of host type I interferon (IFN) by degradation of several components of innate immune pathway. In this study, for the first time, we determined that SVA 2B antagonized host innate immune response in both human and porcine cells. SVA 2B protein degraded mitochondrial antiviral-signaling protein (MAVS), a key host molecule in the innate immune pathway, and a colocalization and interaction between 2B and MAVS was observed in the context of viral infection. Further study showed that the 1-48 and 100-128 regions of 2B were essential for inhibition of type I IFN expression. In addition, we determined that 2B degraded MAVS depending on caspase-9 and caspase-3. In conclusion, our results revealed a novel strategy for SVA 2B protein to antagonize host innate immune response, which will help for clarification of the pathogenesis of SVA and provide an insight for oncolytic virotherapy of SVA.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata/inmunología , Interferón Tipo I/biosíntesis , Picornaviridae/metabolismo , Proteínas no Estructurales Virales/inmunología , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/inmunología , Viroterapia Oncolítica/métodos , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Porcinos , Proteínas no Estructurales Virales/genética , Proteínas Viroporinas/genética , Proteínas Viroporinas/inmunología , Liberación del Virus/fisiología , Replicación Viral/fisiología
5.
Viruses ; 13(11)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34835009

RESUMEN

ORF3a has been identified as a viroporin of SARS-CoV-2 and is known to be involved in various pathophysiological activities including disturbance of cellular calcium homeostasis, inflammasome activation, apoptosis induction and disruption of autophagy. ORF3a-targeting antibodies may specifically and favorably modulate these viroporin-dependent pathological activities. However, suitable viroporin-targeting antibodies are difficult to generate because of the well-recognized technical challenge associated with isolating antibodies to complex transmembrane proteins. Here we exploited a naïve human single chain antibody phage display library, to isolate binders against carefully chosen ORF3a recombinant epitopes located towards the extracellular N terminal and cytosolic C terminal domains of the protein using peptide antigens. These binders were subjected to further characterization using enzyme-linked immunosorbent assays and surface plasmon resonance analysis to assess their binding affinities to the target epitopes. Binding to full-length ORF3a protein was evaluated by western blot and fluorescent microscopy using ORF3a transfected cells and SARS-CoV-2 infected cells. Co-localization analysis was also performed to evaluate the "pairing potential" of the selected binders as possible alternative diagnostic or prognostic biomarkers for COVID-19 infections. Both ORF3a N and C termini, epitope-specific monoclonal antibodies were identified in our study. Whilst the linear nature of peptides might not always represent their native conformations in the context of full protein, with carefully designed selection protocols, we have been successful in isolating anti-ORF3a binders capable of recognising regions of the transmembrane protein that are exposed either on the "inside" or "outside" of the infected cell. Their therapeutic potential will be discussed.


Asunto(s)
Anticuerpos Monoclonales/inmunología , COVID-19/inmunología , COVID-19/virología , SARS-CoV-2/inmunología , Proteínas Viroporinas/inmunología , Animales , Biomarcadores , Células COS , Técnicas de Visualización de Superficie Celular/métodos , Chlorocebus aethiops , Epítopos/inmunología , Células HEK293 , Humanos , Proteínas de la Membrana/inmunología , Dominios Proteicos , Células Vero
6.
Front Immunol ; 12: 745625, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712234

RESUMEN

Developing influenza vaccines that protect against a broad range of viruses is a global health priority. Several conserved viral proteins or domains have been identified as promising targets for such vaccine development. However, none of the targets is sufficiently immunogenic to elicit complete protection, and vaccine platforms that can enhance immunogenicity and deliver multiple antigens are desperately needed. Here, we report proof-of-concept studies for the development of next-generation influenza vaccines using the bacteriophage T4 virus-like particle (VLP) platform. Using the extracellular domain of influenza matrix protein 2 (M2e) as a readout, we demonstrate that up to ~1,281 M2e molecules can be assembled on a 120 x 86 nanometer phage capsid to generate M2e-T4 VLPs. These M2e-decorated nanoparticles, without any adjuvant, are highly immunogenic, stimulate robust humoral as well as cellular immune responses, and conferred complete protection against lethal influenza virus challenge. Potentially, additional conserved antigens could be incorporated into the M2e-T4 VLPs and mass-produced in E. coli in a short amount of time to deal with an emerging influenza pandemic.


Asunto(s)
Bacteriófago T4/inmunología , Proteínas de la Cápside/inmunología , Vacunas contra la Influenza , Desarrollo de Vacunas/métodos , Proteínas de la Matriz Viral/inmunología , Proteínas Viroporinas/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Líquido del Lavado Bronquioalveolar/inmunología , Proteínas de la Cápside/genética , Femenino , Humanos , Inmunogenicidad Vacunal , Virus de la Influenza A/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C , Sistema de Administración de Fármacos con Nanopartículas , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Biblioteca de Péptidos , Prueba de Estudio Conceptual , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas de la Matriz Viral/genética , Proteínas Viroporinas/genética
7.
Microbiol Spectr ; 9(2): e0141621, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34704808

RESUMEN

The rapid worldwide spread of SARS-CoV-2 has accelerated research and development for controlling the COVID-19 pandemic. A multi-coronavirus protein microarray was created containing full-length proteins, overlapping protein fragments of various lengths, and peptide libraries from SARS-CoV-2 and four other human coronaviruses. Sera from confirmed COVID-19 patients as well as unexposed individuals were applied to multicoronavirus arrays to identify specific antibody reactivity. High-level IgG, IgM, and IgA reactivity to structural proteins S, M, and N of SARS-CoV-2, as well as accessory proteins such as ORF3a and ORF7a, were observed that were specific to COVID-19 patients. Antibody reactivity against overlapping 100-, 50-, and 30-amino acid fragments of SARS-CoV-2 proteins was used to identify antigenic regions. Numerous proteins of SARS-CoV, Middle East respiratory syndrome coronavirus (MERS-CoV), and the endemic human coronaviruses HCoV-NL63 and HCoV-OC43 were also more reactive with IgG, IgM, and IgA in COVID-19 patient sera than in unexposed control sera, providing further evidence of immunologic cross-reactivity between these viruses. Whereas unexposed individuals had minimal reactivity against SARS-CoV-2 proteins that poorly correlated with reactivity against HCoV-NL63 and HCoV-OC43 S2 and N proteins, COVID-19 patient sera had higher correlation between SARS-CoV-2 and HCoV responses, suggesting that de novo antibodies against SARS-CoV-2 cross-react with HCoV epitopes. Array responses were compared with validated spike protein-specific IgG enzyme-linked immunosorbent assays (ELISAs), showing agreement between orthologous methods. SARS-CoV-2 microneutralization titers were low in the COVID-19 patient sera but correlated with array responses against S and N proteins. The multi-coronavirus protein microarray is a useful tool for mapping antibody reactivity in COVID-19 patients. IMPORTANCE With novel mutant SARS-CoV-2 variants of concern on the rise, knowledge of immune specificities against SARS-CoV-2 proteins is increasingly important for understanding the impact of structural changes in antibody-reactive protein epitopes on naturally acquired and vaccine-induced immunity, as well as broader topics of cross-reactivity and viral evolution. A multi-coronavirus protein microarray used to map the binding of COVID-19 patient antibodies to SARS-CoV-2 proteins and protein fragments as well as to the proteins of four other coronaviruses that infect humans has shown specific regions of SARS-CoV-2 proteins that are highly reactive with patient antibodies and revealed cross-reactivity of these antibodies with other human coronaviruses. These data and the multi-coronavirus protein microarray tool will help guide further studies of the antibody response to COVID-19 and to vaccination against this worldwide pandemic.


Asunto(s)
Anticuerpos Antivirales/inmunología , Coronavirus Humano NL63/inmunología , Coronavirus Humano OC43/inmunología , Epítopos/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , SARS-CoV-2/inmunología , Anticuerpos Antivirales/sangre , Sitios de Unión de Anticuerpos/inmunología , COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Reacciones Cruzadas/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Fosfoproteínas/inmunología , Análisis por Matrices de Proteínas , Glicoproteína de la Espiga del Coronavirus/inmunología , Proteínas Virales/inmunología , Proteínas Viroporinas/inmunología
8.
Viruses ; 13(9)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34578289

RESUMEN

Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.


Asunto(s)
Anticuerpos Antivirales/sangre , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Proteínas de la Nucleocápside/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Proteínas de la Matriz Viral/inmunología , Proteínas Viroporinas/inmunología , Animales , Antígenos Virales/inmunología , Protección Cruzada , Femenino , Vectores Genéticos , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/administración & dosificación , Ratones Endogámicos BALB C , Proteínas de la Nucleocápside/administración & dosificación , Infecciones por Orthomyxoviridae/inmunología , Pandemias , Vacunación , Proteínas de la Matriz Viral/administración & dosificación , Proteínas de la Matriz Viral/genética , Proteínas Viroporinas/administración & dosificación
9.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1057-L1063, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33822639

RESUMEN

Viroporins, integral viral membrane ion channel proteins, interact with host-cell proteins deregulating physiological processes and activating inflammasomes. Severity of COVID-19 might be associated with hyperinflammation, thus we aimed at the complete immunoinformatic analysis of the SARS-CoV-2 viroporin E, P0DTC4. We also identified the human proteins interacting with P0DTC4 and the enriched molecular functions of the corresponding genes. The complete sequence of P0DTC4 in FASTA format was processed in 10 databases relative to secondary and tertiary protein structure analyses and prediction of optimal vaccine epitopes. Three more databases were accessed for the retrieval and the molecular functional characterization of the P0DTC4 human interactors. The immunoinformatics analysis resulted in the identification of 4 discontinuous B-cell epitopes along with 1 linear B-cell epitope and 11 T-cell epitopes which were found to be antigenic, immunogenic, nonallergen, nontoxin, and unable to induce autoimmunity thus fulfilling prerequisites for vaccine design. The functional enrichment analysis showed that the predicted host interactors of P0DTC4 target the cellular acetylation network. Two of the identified host-cell proteins - BRD2 and BRD4 - have been shown to be promising targets for antiviral therapy. Thus, our findings have implications for COVID-19 therapy and indicate that viroporin E could serve as a promising vaccine target against SARS-CoV-2. Validation experiments are required to complement these in silico results.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , SARS-CoV-2/inmunología , Proteínas Viroporinas/inmunología , Secuencia de Aminoácidos , COVID-19/prevención & control , Proteínas de Ciclo Celular/inmunología , Simulación por Computador , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Humanos , Factores de Transcripción
10.
Cell Rep ; 34(4): 108666, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33503420

RESUMEN

Although vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are under development, the antigen epitopes on the virus and their immunogenicity are poorly understood. Here, we simulate the 3D structures and predict the B cell epitopes on the spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins of SARS-CoV-2 using structure-based approaches and validate epitope immunogenicity by immunizing mice. Almost all 33 predicted epitopes effectively induce antibody production, six of these are immunodominant epitopes in individuals, and 23 are conserved within SARS-CoV-2, SARS-CoV, and bat coronavirus RaTG13. We find that the immunodominant epitopes of individuals with domestic (China) SARS-CoV-2 are different from those of individuals with imported (Europe) SARS-CoV-2, which may be caused by mutations on the S (G614D) and N proteins. Importantly, we find several epitopes on the S protein that elicit neutralizing antibodies against D614 and G614 SARS-CoV-2, which can contribute to vaccine design against coronaviruses.


Asunto(s)
Proteínas de la Nucleocápside de Coronavirus/inmunología , Epítopos de Linfocito B/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Proteínas de la Matriz Viral/inmunología , Proteínas Viroporinas/inmunología , Adolescente , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , COVID-19/inmunología , COVID-19/terapia , Vacunas contra la COVID-19/inmunología , Niño , Epítopos de Linfocito B/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Adulto Joven
11.
Genome Med ; 13(1): 4, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33413610

RESUMEN

During COVID-19, diagnostic serological tools and vaccines have been developed. To inform control activities in a post-vaccine surveillance setting, we have developed an online "immuno-analytics" resource that combines epitope, sequence, protein and SARS-CoV-2 mutation analysis. SARS-CoV-2 spike and nucleocapsid proteins are both vaccine and serological diagnostic targets. Using the tool, the nucleocapsid protein appears to be a sub-optimal target for use in serological platforms. Spike D614G (and nsp12 L314P) mutations were most frequent (> 86%), whilst spike A222V/L18F have recently increased. Also, Orf3a proteins may be a suitable target for serology. The tool can accessed from: http://genomics.lshtm.ac.uk/immuno (online); https://github.com/dan-ward-bio/COVID-immunoanalytics (source code).


Asunto(s)
SARS-CoV-2/genética , SARS-CoV-2/inmunología , COVID-19/diagnóstico , COVID-19/prevención & control , Prueba de COVID-19 , Vacunas contra la COVID-19 , Simulación por Computador , Proteínas de la Nucleocápside de Coronavirus/genética , Proteínas de la Nucleocápside de Coronavirus/inmunología , Epítopos de Linfocito B/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Mutación , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Proteínas Viroporinas/genética , Proteínas Viroporinas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA