Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Immunol ; 212(12): 1922-1931, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38683124

RÉSUMÉ

Although high titers of neutralizing Abs in human serum are associated with protection from reinfection by SARS-CoV-2, there is considerable heterogeneity in human serum-neutralizing Abs against SARS-CoV-2 during convalescence between individuals. Standard human serum live virus neutralization assays require inactivation of serum/plasma prior to testing. In this study, we report that the SARS-CoV-2 neutralization titers of human convalescent sera were relatively consistent across all disease states except for severe COVID-19, which yielded significantly higher neutralization titers. Furthermore, we show that heat inactivation of human serum significantly lowered neutralization activity in a live virus SARS-CoV-2 neutralization assay. Heat inactivation of human convalescent serum was shown to inactivate complement proteins, and the contribution of complement in SARS-CoV-2 neutralization was often >50% of the neutralizing activity of human sera without heat inactivation and could account for neutralizing activity when standard titers were zero after heat inactivation. This effect was also observed in COVID-19 vaccinees and could be abolished in individuals who were undergoing treatment with therapeutic anti-complement Abs. Complement activity was mainly dependent on the classical pathway with little contributions from mannose-binding lectin and alternative pathways. Our study demonstrates the importance of the complement pathway in significantly increasing viral neutralization activity against SARS-CoV-2 in spike seropositive individuals.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Voie classique d'activation du complément , Tests de neutralisation , SARS-CoV-2 , Humains , SARS-CoV-2/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , COVID-19/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Voie classique d'activation du complément/immunologie , Vaccins contre la COVID-19/immunologie , Mâle , Femelle , Adulte d'âge moyen , Adulte , Convalescence , Sujet âgé , Protéines du système du complément/immunologie
2.
iScience ; 26(10): 107915, 2023 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-37790281

RÉSUMÉ

Older individuals and people with HIV (PWH) were prioritized for COVID-19 vaccination, yet comprehensive studies of the immunogenicity of these vaccines and their effects on HIV reservoirs are not available. Our study on 68 PWH and 23 HIV-negative participants aged 55 and older post-three vaccine doses showed equally strong anti-spike IgG responses in serum and saliva through week 48 from baseline, while PWH salivary IgA responses were low. PWH had diminished live-virus neutralization responses after two vaccine doses, which were 'rescued' post-booster. Spike-specific T cell immunity was enhanced in PWH with normal CD4+ T cell count, suggesting Th1 imprinting. The frequency of detectable HIV viremia increased post-vaccination, but vaccines did not affect the size of the HIV reservoir in most PWH, except those with low-level viremia. Thus, older PWH require three doses of COVID-19 vaccine for maximum protection, while individuals with unsuppressed viremia should be monitored for adverse reactions from HIV reservoirs.

3.
bioRxiv ; 2023 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-37502977

RÉSUMÉ

Older individuals and people with HIV (PWH) were prioritized for COVID-19 vaccination, yet comprehensive studies of the immunogenicity of these vaccines and their effects on HIV reservoirs are not available. We followed 68 PWH aged 55 and older and 23 age-matched HIV-negative individuals for 48 weeks from the first vaccine dose, after the total of three doses. All PWH were on antiretroviral therapy (cART) and had different immune status, including immune responders (IR), immune non-responders (INR), and PWH with low-level viremia (LLV). We measured total and neutralizing Ab responses to SARS-CoV-2 spike and RBD in sera, total anti-spike Abs in saliva, frequency of anti-RBD/NTD B cells, changes in frequency of anti-spike, HIV gag/nef-specific T cells, and HIV reservoirs in peripheral CD4 + T cells. The resulting datasets were used to create a mathematical model for within-host immunization. Various regimens of BNT162b2, mRNA-1273, and ChAdOx1 vaccines elicited equally strong anti-spike IgG responses in PWH and HIV - participants in serum and saliva at all timepoints. These responses had similar kinetics in both cohorts and peaked at 4 weeks post-booster (third dose), while half-lives of plasma IgG also dramatically increased post-booster in both groups. Salivary spike IgA responses were low, especially in INRs. PWH had diminished live virus neutralizing titers after two vaccine doses which were 'rescued' after a booster. Anti-spike T cell immunity was enhanced in IRs even in comparison to HIV - participants, suggesting Th1 imprinting from HIV, while in INRs it was the lowest. Increased frequency of viral 'blips' in PWH were seen post-vaccination, but vaccines did not affect the size of the intact HIV reservoir in CD4 + T cells in most PWH, except in LLVs. Thus, older PWH require three doses of COVID-19 vaccine to maximize neutralizing responses against SARS-CoV-2, although vaccines may increase HIV reservoirs in PWH with persistent viremia.

4.
Sci Adv ; 8(3): eabj9815, 2022 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-35044832

RÉSUMÉ

Safe and effective vaccines are needed to end the COVID-19 pandemic. Here, we report the preclinical development of a lipid nanoparticle­formulated SARS-CoV-2 mRNA vaccine, PTX-COVID19-B. PTX-COVID19-B was chosen among three candidates after the initial mouse vaccination results showed that it elicited the strongest neutralizing antibody response against SARS-CoV-2. Further tests in mice and hamsters indicated that PTX-COVID19-B induced robust humoral and cellular immune responses and completely protected the vaccinated animals from SARS-CoV-2 infection in the lung. Studies in hamsters also showed that PTX-COVID19-B protected the upper respiratory tract from SARS-CoV-2 infection. Mouse immune sera elicited by PTX-COVID19-B vaccination were able to neutralize SARS-CoV-2 variants of concern, including the Alpha, Beta, Gamma, and Delta lineages. No adverse effects were induced by PTX-COVID19-B in either mice or hamsters. Based on these results, PTX-COVID19-B was authorized by Health Canada to enter clinical trials in December 2020 with a phase 2 clinical trial ongoing.


Sujet(s)
Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Vaccins synthétiques/immunologie , Vaccins à ARNm/immunologie , Animaux , Anticorps neutralisants/sang , Anticorps antiviraux/sang , Numération des lymphocytes CD4 , Lymphocytes T CD8+/immunologie , COVID-19/immunologie , Vaccins contre la COVID-19/effets indésirables , Canada , Lignée cellulaire , Cricetinae , Évaluation préclinique de médicament , Femelle , Cellules HEK293 , Humains , Immunité cellulaire/immunologie , Immunité humorale/immunologie , Liposomes/pharmacologie , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Nanoparticules , Glycoprotéine de spicule des coronavirus/génétique , Lymphocytes auxiliaires Th1/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...