Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Antiviral Res ; 223: 105823, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38331200

RESUMEN

Mucosal immunity has regained its spotlight amidst the ongoing Coronavirus disease 19 (COVID-19) pandemic, with numerous studies highlighting the crucial role of mucosal secretory IgA (SIgA) in protection against Severe acute respiratory syndrome coronavirus-2 or SARS-CoV-2 infections. The observed limitations in the efficacy of currently authorized COVID-19 vaccines in inducing effective mucosal immune responses remind us of the limitations of systemic vaccination in promoting protective mucosal immunity. This resurgence of interest has motivated the development of vaccine platforms capable of enhancing mucosal responses, specifically the SIgA response, and the development of IgA-based therapeutics. Recognizing viral respiratory infections as a global threat, we would like to comprehensively review the existing knowledge on mucosal immunity, with a particular emphasis on SIgA, in the context of SARS-CoV-2, influenza, and Respiratory Syncytial Virus (RSV) infections. This review aims to describe the structural and functional specificities of SIgA, along with its nuanced role in combating influenza, RSV, and SARS-CoV-2 infections. Subsequent sections further elaborate promising vaccine strategies, including mucosal vaccines against Influenza, RSV, and SARS-CoV-2 respiratory viruses, currently undergoing preclinical and clinical development. Additionally, we address the challenges associated with mucosal vaccine development, concluding with a discussion on IgA-based therapeutics as a promising platform for the treatment of viral respiratory infections. This comprehensive review not only synthesizes current insights into mucosal immunity but also identifies critical knowledge gaps, strengthening the way for further advancements in our current understanding and approaches to combat respiratory viral threats.


Asunto(s)
COVID-19 , Vacunas contra la Influenza , Gripe Humana , Infecciones por Virus Sincitial Respiratorio , Humanos , Inmunoglobulina A Secretora , Vacunas contra la COVID-19 , COVID-19/prevención & control , SARS-CoV-2
2.
Vaccines (Basel) ; 11(4)2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37112685

RESUMEN

Antibody-dependent enhancement (ADE) can increase the rates and severity of infection with various viruses, including coronaviruses, such as MERS. Some in vitro studies on COVID-19 have suggested that prior immunization enhances SARS-CoV-2 infection, but preclinical and clinical studies have demonstrated the contrary. We studied a cohort of COVID-19 patients and a cohort of vaccinated individuals with a heterologous (Moderna/Pfizer) or homologous (Pfizer/Pfizer) vaccination scheme. The dependence on IgG or IgA of ADE of infection was evaluated on the serum samples from these subjects (twenty-six vaccinated individuals and twenty-one PCR-positive SARS-CoV-2-infected patients) using an in vitro model with CD16- or CD89-expressing cells and the Delta (B.1.617.2 lineage) and Omicron (B.1.1.529 lineage) variants of SARS-CoV-2. Sera from COVID-19 patients did not show ADE of infection with any of the tested viral variants. Some serum samples from vaccinated individuals displayed a mild IgA-ADE effect with Omicron after the second dose of the vaccine, but this effect was abolished after the completion of the full vaccination scheme. In this study, FcγRIIIa- and FcαRI-dependent ADE of SARS-CoV-2 infection after prior immunization, which might increase the risk of severe disease in a second natural infection, was not observed.

3.
NPJ Vaccines ; 8(1): 57, 2023 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-37080996

RESUMEN

This study reports the 6-month humoral immune response in vaccinated patients concomitantly infected with Delta and Omicron BA.1 variants of SARS-CoV-2. Interestingly, the simultaneous exposure to the Delta and BA.1 S proteins does not confer an additional immune advantage compared to exposure to the BA.1 S protein alone.

5.
Mucosal Immunol ; 15(6): 1170-1180, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36195658

RESUMEN

Systemic and mucosal humoral immune responses are crucial to fight respiratory viral infections in the current pandemic of COVID-19 caused by the SARS-CoV-2 virus. During SARS-CoV-2 infection, the dynamics of systemic and mucosal antibody infections are affected by patient characteristics, such as age, sex, disease severity, or prior immunity to other human coronaviruses. Patients suffering from severe disease develop higher levels of anti-SARS-CoV-2 antibodies in serum and mucosal tissues than those with mild disease, and these antibodies are detectable for up to a year after symptom onset. In hospitalized patients, the aberrant glycosylation of anti-SARS-CoV-2 antibodies enhances inflammation-associated antibody Fc-dependent effector functions, thereby contributing to COVID-19 pathophysiology. Current vaccines elicit robust humoral immune responses, principally in the blood. However, they are less effective against new viral variants, such as Delta and Omicron. This review provides an overview of current knowledge about the humoral immune response to SARS-CoV-2, with a particular focus on the protective and pathological role of humoral immunity in COVID-19 severity. We also discuss the humoral immune response elicited by COVID-19 vaccination and protection against emerging viral variants.


Asunto(s)
COVID-19 , Inmunidad Humoral , Humanos , SARS-CoV-2 , Vacunas contra la COVID-19 , Anticuerpos Antivirales , Anticuerpos Neutralizantes
6.
Vaccines (Basel) ; 10(6)2022 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-35746511

RESUMEN

Broadly neutralizing antibodies (bNAbs) offer promising opportunities for preventing HIV-1 infection. The protection mechanisms of bNAbs involve the Fc domain, as well as their Fab counterpart. Here, different bNAb isotypes including IgG1, IgA1, IgA2, and IgA122 (IgA2 with the hinge of IgA1) were generated and then produced in CHO cells. Their ability to neutralize pseudovirus and primary HIV-1 isolates were measured, as well as their potential ADCC-like activity using a newly developed assay. In our work, gp41-specific IgA seems to be more efficient than IgG1 in inducing ADCC-like activity, but not in its virus neutralization effect. We show that either gp120-specific IgA or IgG1 isotypes are both efficient in neutralizing different viral strains. In contrast, gp120-specific IgG1 was a better ADCC-like inducer than IgA isotypes. These results provide new insights into the neutralization and ADCC-like activity of different bNAbs that might be taken into consideration when searching for new treatments or antibody-based vaccines.

7.
Nature ; 600(7890): 701-706, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34673755

RESUMEN

Following severe adverse reactions to the AstraZeneca ChAdOx1-S-nCoV-19 vaccine1,2, European health authorities recommended that patients under the age of 55 years who received one dose of ChAdOx1-S-nCoV-19 receive a second dose of the Pfizer BNT162b2 vaccine as a booster. However, the effectiveness and the immunogenicity of this vaccination regimen have not been formally tested. Here we show that the heterologous ChAdOx1-S-nCoV-19 and BNT162b2 combination confers better protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection than the homologous BNT162b2 and BNT162b2 combination in a real-world observational study of healthcare workers (n = 13,121). To understand the underlying mechanism, we conducted a longitudinal survey of the anti-spike immunity conferred by each vaccine combination. Both combinations induced strong anti-spike antibody responses, but sera from heterologous vaccinated individuals displayed a stronger neutralizing activity regardless of the SARS-CoV-2 variant. This enhanced neutralizing potential correlated with increased frequencies of switched and activated memory B cells that recognize the SARS-CoV-2 receptor binding domain. The ChAdOx1-S-nCoV-19 vaccine induced a weaker IgG response but a stronger T cell response than the BNT162b2 vaccine after the priming dose, which could explain the complementarity of both vaccines when used in combination. The heterologous vaccination regimen could therefore be particularly suitable for immunocompromised individuals.


Asunto(s)
Vacuna BNT162/administración & dosificación , Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/prevención & control , ChAdOx1 nCoV-19/administración & dosificación , ChAdOx1 nCoV-19/inmunología , SARS-CoV-2/inmunología , Vacunación/estadística & datos numéricos , Adulto , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Femenino , Francia/epidemiología , Hospitales Universitarios , Humanos , Memoria Inmunológica/inmunología , Incidencia , Masculino , Células B de Memoria/inmunología , Células T de Memoria/inmunología , Persona de Mediana Edad , Glicoproteína de la Espiga del Coronavirus/inmunología
8.
Viral Immunol ; 30(5): 350-358, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28418786

RESUMEN

Our group has developed a subunit vaccine candidate against Dengue virus (DENV) based on two different viral regions, the domain III of the envelope protein and the capsid protein. The chimeric proteins for each serotype (DIIIC1-4), aggregated with the oligodeoxynucleotide 39 M, form the tetravalent formulation named Tetra DIIIC. Tetra DIIIC induces a protective immune response in mice when it is inoculated by intraperitoneal route. However, if children are the main targets for a DENV vaccine, then a needle-free route of administration should be attractive and advantageous. In this study, we evaluated for the first time, in vivo, a vaccine candidate against DENV based on recombinant proteins using the intranasal route. After three doses of Tetra DIIIC in mice, we measured the humoral immune response against the four DENV serotypes and the corresponding recombinant proteins. Moreover, the functionality of these antibodies was evaluated through a plaque reduction neutralization test. Finally, to assess the cellular immune response induced, we measured the IFN-γ-levels secreted by spleen cells after in vitro stimulation with DENV. The results presented in this study indicate that the intranasal immunization with Tetra DIIIC favors the generation of DENV-specific cell-mediated immunity. On the other hand, the immunization using intraperitoneal and intranasal routes, simultaneously, generate functional antibodies (anti-DIIIC and anti-DENV) and an in vitro response of IFN-γ secretion.


Asunto(s)
Virus del Dengue/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Administración Intranasal , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Ratones Endogámicos BALB C , Pruebas de Neutralización , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/inmunología , Ensayo de Placa Viral , Vacunas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...