Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.270
Filtrer
1.
Science ; 383(6684): eadg0564, 2024 Feb 16.
Article de Anglais | MEDLINE | ID: mdl-38359115

RÉSUMÉ

Influenza viruses escape immunity owing to rapid antigenic evolution, which requires vaccination strategies that allow for broadly protective antibody responses. We found that the lipid globotriaosylceramide (Gb3) expressed on germinal center (GC) B cells is essential for the production of high-affinity antibodies. Mechanistically, Gb3 bound and disengaged CD19 from its chaperone CD81, permitting CD19 to translocate to the B cell receptor complex to trigger signaling. Moreover, Gb3 regulated major histocompatibility complex class II expression to increase diversity of T follicular helper and GC B cells reactive with subdominant epitopes. In influenza infection, elevating Gb3, either endogenously or exogenously, promoted broadly reactive antibody responses and cross-protection. These data demonstrate that Gb3 determines the affinity and breadth of B cell immunity and has potential as a vaccine adjuvant.


Sujet(s)
Anticorps antiviraux , Lymphocytes B , Centre germinatif , Infections à Orthomyxoviridae , Orthomyxoviridae , Trihexosylcéramide , Production d'anticorps , Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes B/immunologie , Centre germinatif/effets des médicaments et des substances chimiques , Centre germinatif/immunologie , Trihexosylcéramide/métabolisme , Trihexosylcéramide/pharmacologie , Animaux , Souris , Souris knockout , Humains , Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/immunologie , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/immunologie
2.
J Virol ; 98(2): e0149423, 2024 Feb 20.
Article de Anglais | MEDLINE | ID: mdl-38294251

RÉSUMÉ

Influenza B viruses (IBV) cocirculate with influenza A viruses (IAV) and cause periodic epidemics of disease, yet antibody and cellular responses following IBV infection are less well understood. Using the ferret model for antisera generation for influenza surveillance purposes, IAV resulted in robust antibody responses following infection, whereas IBV required an additional booster dose, over 85% of the time, to generate equivalent antibody titers. In this study, we utilized primary differentiated ferret nasal epithelial cells (FNECs) which were inoculated with IAV and IBV to study differences in innate immune responses which may result in differences in adaptive immune responses in the host. FNECs were inoculated with IAV (H1N1pdm09 and H3N2 subtypes) or IBV (B/Victoria and B/Yamagata lineages) and assessed for 72 h. Cells were analyzed for gene expression by quantitative real-time PCR, and apical and basolateral supernatants were assessed for virus kinetics and interferon (IFN), respectively. Similar virus kinetics were observed with IAV and IBV in FNECs. A comparison of gene expression and protein secretion profiles demonstrated that IBV-inoculated FNEC expressed delayed type-I/II IFN responses and reduced type-III IFN secretion compared to IAV-inoculated cells. Concurrently, gene expression of Thymic Stromal Lymphopoietin (TSLP), a type-III IFN-induced gene that enhances adaptive immune responses, was significantly downregulated in IBV-inoculated FNECs. Significant differences in other proinflammatory and adaptive genes were suppressed and delayed following IBV inoculation. Following IBV infection, ex vivo cell cultures derived from the ferret upper respiratory tract exhibited reduced and delayed innate responses which may contribute to reduced antibody responses in vivo.IMPORTANCEInfluenza B viruses (IBV) represent nearly one-quarter of all human influenza cases and are responsible for significant clinical and socioeconomic impacts but do not pose the same pandemic risks as influenza A viruses (IAV) and have thus received much less attention. IBV accounts for greater severity and deaths in children, and vaccine efficacy remains low. The ferret can be readily infected with human clinical isolates and demonstrates a similar course of disease and immune responses. IBV, however, generates lower antibodies in ferrets than IAV following the challenge. To determine whether differences in initial innate responses following infection may affect the development of robust adaptive immune responses, ferret respiratory tract cells were isolated, infected with IAV/IBV, and compared. Understanding the differences in the initial innate immune responses to IAV and IBV may be important in the development of more effective vaccines and interventions to generate more robust protective immune responses.


Sujet(s)
Immunité acquise , Cellules épithéliales , Furets , Immunité innée , Virus de la grippe A , Virus influenza B , Interférons , Muqueuse nasale , Animaux , Enfant , Humains , Anticorps antiviraux/analyse , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/immunologie , Modèles animaux de maladie humaine , Cellules épithéliales/cytologie , Cellules épithéliales/immunologie , Cellules épithéliales/virologie , Furets/immunologie , Furets/virologie , Virus de la grippe A/classification , Virus de la grippe A/croissance et développement , Virus de la grippe A/immunologie , Sous-type H1N1 du virus de la grippe A/immunologie , Sous-type H3N2 du virus de la grippe A/immunologie , Virus influenza B/classification , Virus influenza B/croissance et développement , Virus influenza B/immunologie , Vaccins antigrippaux , Grippe humaine/virologie , Interférons/immunologie , Muqueuse nasale/cytologie , Muqueuse nasale/immunologie , Muqueuse nasale/virologie , Lymphopoïétine stromale thymique/génétique , Lymphopoïétine stromale thymique/immunologie , Cellules cultivées
3.
Nature ; 626(7998): 385-391, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38096903

RÉSUMÉ

A limitation of current SARS-CoV-2 vaccines is that they provide minimal protection against infection with current Omicron subvariants1,2, although they still provide protection against severe disease. Enhanced mucosal immunity may be required to block infection and onward transmission. Intranasal administration of current vaccines has proven inconsistent3-7, suggesting that alternative immunization strategies may be required. Here we show that intratracheal boosting with a bivalent Ad26-based SARS-CoV-2 vaccine results in substantial induction of mucosal humoral and cellular immunity and near-complete protection against SARS-CoV-2 BQ.1.1 challenge. A total of 40 previously immunized rhesus macaques were boosted with a bivalent Ad26 vaccine by the intramuscular, intranasal and intratracheal routes, or with a bivalent mRNA vaccine by the intranasal route. Ad26 boosting by the intratracheal route led to a substantial expansion of mucosal neutralizing antibodies, IgG and IgA binding antibodies, and CD8+ and CD4+ T cell responses, which exceeded those induced by Ad26 boosting by the intramuscular and intranasal routes. Intratracheal Ad26 boosting also led to robust upregulation of cytokine, natural killer, and T and B cell pathways in the lungs. After challenge with a high dose of SARS-CoV-2 BQ.1.1, intratracheal Ad26 boosting provided near-complete protection, whereas the other boosting strategies proved less effective. Protective efficacy correlated best with mucosal humoral and cellular immune responses. These data demonstrate that these immunization strategies induce robust mucosal immunity, suggesting the feasibility of developing vaccines that block respiratory viral infections.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Immunité muqueuse , Rappel de vaccin , Macaca mulatta , SARS-CoV-2 , Animaux , Humains , Administration par voie nasale , Anticorps neutralisants/biosynthèse , Anticorps neutralisants/immunologie , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/virologie , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/immunologie , Cytokines/immunologie , Immunité muqueuse/immunologie , Rappel de vaccin/méthodes , Immunoglobuline A/immunologie , Immunoglobuline G/immunologie , Injections musculaires , Cellules tueuses naturelles/immunologie , Poumon/immunologie , Macaca mulatta/immunologie , Macaca mulatta/virologie , Vaccins à ARNm/administration et posologie , Vaccins à ARNm/immunologie , SARS-CoV-2/classification , SARS-CoV-2/immunologie , Trachée/immunologie , Trachée/virologie
4.
J Virol ; 97(7): e0061023, 2023 07 27.
Article de Anglais | MEDLINE | ID: mdl-37367229

RÉSUMÉ

Members of the Sarbecovirus subgenus of Coronaviridae have twice caused deadly threats to humans. There is increasing concern about the rapid mutation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has evolved into multiple generations of epidemic variants in 3 years. Broad neutralizing antibodies are of great importance for pandemic preparedness against SARS-CoV-2 variants and divergent zoonotic sarbecoviruses. Here, we analyzed the structural conservation of the receptor-binding domain (RBD) from representative sarbecoviruses and chose S2H97, a previously reported RBD antibody with ideal breadth and resistance to escape, as a template for computational design to enhance the neutralization activity and spectrum. A total of 35 designs were purified for evaluation. The neutralizing activity of a large proportion of these designs against multiple variants was increased from several to hundreds of times. Molecular dynamics simulation suggested that extra interface contacts and enhanced intermolecular interactions between the RBD and the designed antibodies are established. After light and heavy chain reconstitution, AI-1028, with five complementarity determining regions optimized, showed the best neutralizing activity across all tested sarbecoviruses, including SARS-CoV, multiple SARS-CoV-2 variants, and bat-derived viruses. AI-1028 recognized the same cryptic RBD epitope as the parental prototype antibody. In addition to computational design, chemically synthesized nanobody libraries are also a precious resource for rapid antibody development. By applying distinct RBDs as baits for reciprocal screening, we identified two novel nanobodies with broad activities. These findings provide potential pan-sarbecovirus neutralizing drugs and highlight new pathways to rapidly optimize therapeutic candidates when novel SARS-CoV-2 escape variants or new zoonotic coronaviruses emerge. IMPORTANCE The subgenus Sarbecovirus includes human SARS-CoV, SARS-CoV-2, and hundreds of genetically related bat viruses. The continuous evolution of SARS-CoV-2 has led to the striking evasion of neutralizing antibody (NAb) drugs and convalescent plasma. Antibodies with broad activity across sarbecoviruses would be helpful to combat current SARS-CoV-2 mutations and longer term animal virus spillovers. The study of pan-sarbecovirus NAbs described here is significant for the following reasons. First, we established a structure-based computational pipeline to design and optimize NAbs to obtain more potent and broader neutralizing activity across multiple sarbecoviruses. Second, we screened and identified nanobodies from a highly diversified synthetic library with a broad neutralizing spectrum using an elaborate screening strategy. These methodologies provide guidance for the rapid development of antibody therapeutics against emerging pathogens with highly variable characteristics.


Sujet(s)
Anticorps antiviraux , Anticorps neutralisants à large spectre , Virus du SRAS , Anticorps à domaine unique , Animaux , Humains , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/composition chimique , Anticorps antiviraux/métabolisme , Anticorps neutralisants à large spectre/biosynthèse , Anticorps neutralisants à large spectre/composition chimique , Anticorps neutralisants à large spectre/métabolisme , Chiroptera , COVID-19/virologie , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , Virus du SRAS/immunologie , Virus du SRAS/métabolisme , Structure tertiaire des protéines , Modèles moléculaires , Liaison aux protéines
5.
Nature ; 615(7952): 482-489, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36646114

RÉSUMÉ

The protective efficacy of serum antibodies results from the interplay of antigen-specific B cell clones of different affinities and specificities. These cellular dynamics underlie serum-level phenomena such as original antigenic sin (OAS)-a proposed propensity of the immune system to rely repeatedly on the first cohort of B cells engaged by an antigenic stimulus when encountering related antigens, in detriment to the induction of de novo responses1-5. OAS-type suppression of new, variant-specific antibodies may pose a barrier to vaccination against rapidly evolving viruses such as influenza and SARS-CoV-26,7. Precise measurement of OAS-type suppression is challenging because cellular and temporal origins cannot readily be ascribed to antibodies in circulation; its effect on subsequent antibody responses therefore remains unclear5,8. Here we introduce a molecular fate-mapping approach with which serum antibodies derived from specific cohorts of B cells can be differentially detected. We show that serum responses to sequential homologous boosting derive overwhelmingly from primary cohort B cells, while later induction of new antibody responses from naive B cells is strongly suppressed. Such 'primary addiction' decreases sharply as a function of antigenic distance, allowing reimmunization with divergent viral glycoproteins to produce de novo antibody responses targeting epitopes that are absent from the priming variant. Our findings have implications for the understanding of OAS and for the design and testing of vaccines against evolving pathogens.


Sujet(s)
Production d'anticorps , Lymphocytes B , Rappel de vaccin , Humains , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Antigènes viraux/immunologie , Vaccins antigrippaux/immunologie , SARS-CoV-2/immunologie , Vaccination , Lymphocytes B/immunologie , Vaccins antiviraux/immunologie
6.
Vet Microbiol ; 275: 109593, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-36323175

RÉSUMÉ

Porcine sapelovirus (PSV) is an important emerging swine pathogen that causes diarrhoea, respiratory distress, severe reproductive system and neurological disorders in pigs, posing huge threat to swine industry. However, there are no effective serological diagnostic products and the epitope characterization of PSV VP1 protein is still largely unknown. In current study, we successfully expressed recombinant His-VP1 protein by prokaryotic expression system and the recombinant VP1 protein had good immunogenicity. BALB/C mice were then selected and immunized with purified recombinant VP1 protein, and two monoclonal antibodies (Mabs) 9F10 and 15E4 against VP1 were successfully prepared by hybrioma technology. The isotype of these two Mabs were identified and showed that Mab 9F10 with the heavy chain subtype was IgG1 and the light chain subtype was kappa. Mab 15E4 was identified as IgG2 for the heavy chain subtype and Kappa for the light chain subtype. The antigen epitopes of prepared two VP1 Mabs were clearly identified. The minimal unit of B cell specific epitope recognized by Mab 15E4 was 203YDGDG207 and conserved in different strain genotypes of PSV, indicating this epitope may be a good target for serological detection of PSV. However, the epitope recognized by Mab 9F10 was 8QAIVNRT14 and varied greatly among different PSV strains. Structural modeling analysis showed that the identified two novel B cell epitopes were located on the surface of VP1. Our study provides useful tool for the establishment the serological detection methods of PSV and may support the study of VP1 protein function.


Sujet(s)
Anticorps monoclonaux , Anticorps antiviraux , Déterminants antigéniques des lymphocytes B , Picornaviridae , Protéines virales , Animaux , Souris , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/immunologie , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/immunologie , Déterminants antigéniques des lymphocytes B/immunologie , Immunoglobuline G , Souris de lignée BALB C , Picornaviridae/immunologie , Suidae , Protéines virales/immunologie
7.
Viruses ; 14(7)2022 07 09.
Article de Anglais | MEDLINE | ID: mdl-35891490

RÉSUMÉ

The novel severe acute respiratory syndrome (SARS) coronavirus, SARS-CoV-2, is responsible for the global COVID-19 pandemic. Effective interventions are urgently needed to mitigate the effects of COVID-19 and likely require multiple strategies. Egg-extracted antibody therapies are a low-cost and scalable strategy to protect at-risk individuals from SARS-CoV-2 infection. Commercial laying hens were hyperimmunized against the SARS-CoV-2 S1 protein using three different S1 recombinant proteins and three different doses. Sera and egg yolk were collected at three and six weeks after the second immunization for enzyme-linked immunosorbent assay and plaque-reduction neutralization assay to determine antigen-specific antibody titers and neutralizing antibody titers, respectively. In this study we demonstrate that hens hyperimmunized against the SARS-CoV-2 recombinant S1 and receptor binding domain (RBD) proteins produced neutralizing antibodies against SARS-CoV-2. We further demonstrate that antibody production was dependent on the dose and type of antigen administered. Our data suggests that antibodies purified from the egg yolk of hyperimmunized hens can be used as immunoprophylaxis in humans at risk of exposure to SARS-CoV-2.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Jaune d'œuf , SARS-CoV-2 , Animaux , Anticorps neutralisants/biosynthèse , Anticorps antiviraux/biosynthèse , COVID-19/prévention et contrôle , Poulets , Jaune d'œuf/immunologie , Femelle , Glycoprotéine de spicule des coronavirus
9.
Signal Transduct Target Ther ; 7(1): 61, 2022 02 25.
Article de Anglais | MEDLINE | ID: mdl-35217639

RÉSUMÉ

Variants are globally emerging very quickly following pandemic prototypic SARS-CoV-2. To evaluate the cross-protection of prototypic SARS-CoV-2 vaccine against its variants, we vaccinated rhesus monkeys with three doses of prototypic SARS-CoV-2 inactivated vaccine, followed by challenging with emerging SARS-CoV-2 variants of concern (VOCs). These vaccinated animals produced neutralizing antibodies against Alpha, Beta, Delta, and Omicron variants, although there were certain declinations of geometric mean titer (GMT) as compared with prototypic SARS-CoV-2. Of note, in vivo this prototypic vaccine not only reduced the viral loads in nasal, throat and anal swabs, pulmonary tissues, but also improved the pathological changes in the lung infected by variants of Alpha, Beta, and Delta. In summary, the prototypic SARS-CoV-2 inactivated vaccine in this study protected against VOCs to certain extension, which is of great significance for prevention and control of COVID-19.


Sujet(s)
Anticorps neutralisants/biosynthèse , Anticorps antiviraux/biosynthèse , Vaccins contre la COVID-19/administration et posologie , COVID-19/prévention et contrôle , Protection croisée , SARS-CoV-2/effets des médicaments et des substances chimiques , Vaccination/méthodes , Vaccins inactivés/administration et posologie , Canal anal/virologie , Animaux , Lymphocytes B/immunologie , Lymphocytes B/virologie , COVID-19/immunologie , COVID-19/virologie , Humains , Immunogénicité des vaccins , Poumon/virologie , Macaca mulatta , Mâle , Fosse nasale/virologie , Pharynx/virologie , SARS-CoV-2/croissance et développement , SARS-CoV-2/pathogénicité , Lymphocytes T/immunologie , Lymphocytes T/virologie , Charge virale/effets des médicaments et des substances chimiques
10.
Signal Transduct Target Ther ; 7(1): 44, 2022 02 09.
Article de Anglais | MEDLINE | ID: mdl-35140196

RÉSUMÉ

The wide transmission and host adaptation of SARS-CoV-2 have led to the rapid accumulation of mutations, posing significant challenges to the effectiveness of vaccines and therapeutic antibodies. Although several neutralizing antibodies were authorized for emergency clinical use, convalescent patients derived natural antibodies are vulnerable to SARS-CoV-2 Spike mutation. Here, we describe the screen of a panel of SARS-CoV-2 receptor-binding domain (RBD) targeted nanobodies (Nbs) from a synthetic library and the design of a biparatopic Nb, named Nb1-Nb2, with tight affinity and super-wide neutralization breadth against multiple SARS-CoV-2 variants of concern. Deep-mutational scanning experiments identify the potential binding epitopes of the Nbs on the RBD and demonstrate that biparatopic Nb1-Nb2 has a strong escape-resistant feature against more than 60 tested RBD amino acid substitutions. Using pseudovirion-based and trans-complementation SARS-CoV-2 tools, we determine that the Nb1-Nb2 broadly neutralizes multiple SARS-CoV-2 variants at sub-nanomolar levels, including Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), Lambda (C.37), Kappa (B.1.617.1), and Mu (B.1.621). Furthermore, a heavy-chain antibody is constructed by fusing the human IgG1 Fc to Nb1-Nb2 (designated as Nb1-Nb2-Fc) to improve its neutralization potency, yield, stability, and potential half-life extension. For the new Omicron variant (B.1.1.529) that harbors unprecedented multiple RBD mutations, Nb1-Nb2-Fc keeps a firm affinity (KD < 1.0 × 10-12 M) and strong neutralizing activity (IC50 = 1.46 nM for authentic Omicron virus). Together, we developed a tetravalent biparatopic human heavy-chain antibody with ultrapotent and broad-spectrum SARS-CoV-2 neutralization activity which highlights the potential clinical applications.


Sujet(s)
Anticorps neutralisants/pharmacologie , Anticorps antiviraux/pharmacologie , Fragments Fc des immunoglobulines/pharmacologie , Protéines de fusion recombinantes/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Anticorps à domaine unique/pharmacologie , Anticorps neutralisants/biosynthèse , Anticorps neutralisants/génétique , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/génétique , Affinité des anticorps , Test ELISA , Épitopes/composition chimique , Épitopes/immunologie , Escherichia coli/génétique , Escherichia coli/métabolisme , Expression des gènes , Humains , Fragments Fc des immunoglobulines/biosynthèse , Fragments Fc des immunoglobulines/génétique , Modèles moléculaires , Tests de neutralisation , Liaison aux protéines/effets des médicaments et des substances chimiques , Conformation des protéines , Motifs et domaines d'intéraction protéique , Protéines de fusion recombinantes/biosynthèse , Protéines de fusion recombinantes/génétique , SARS-CoV-2/croissance et développement , SARS-CoV-2/immunologie , Anticorps à domaine unique/biosynthèse , Anticorps à domaine unique/génétique , Glycoprotéine de spicule des coronavirus/antagonistes et inhibiteurs , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie
11.
12.
Microbiol Spectr ; 10(1): e0169521, 2022 02 23.
Article de Anglais | MEDLINE | ID: mdl-35171046

RÉSUMÉ

Global control of COVID-19 will require the deployment of vaccines capable of inducing long-term protective immunity against SARS-CoV-2 variants. In this report, we describe an adjuvanted subunit candidate vaccine that affords elevated, sustained, and cross-variant SARS-CoV-2 neutralizing antibodies (NAbs) in multiple animal models. Alhydroxiquim-II is a Toll-Like Receptor (TLR) 7/8 small-molecule agonist chemisorbed on aluminum hydroxide (Alhydrogel). Vaccination with Alhydroxiquim-II combined with a stabilized, trimeric form of the SARS-CoV-2 spike protein (termed CoVac-II) resulted in high-titer NAbs in mice, with no decay in responses over an 8-month period. NAbs from sera of CoVac-II-immunized mice, horses and rabbits were broadly neutralizing against SARS-CoV-2 variants. Boosting long-term CoVac-II-immunized mice with adjuvanted spike protein from the Beta variant markedly increased levels of NAb titers against multiple SARS-CoV-2 variants; notably, high titers against the Delta variant were observed. These data strongly support the clinical assessment of Alhydroxiquim-II-adjuvanted spike proteins to protect against SARS-CoV-2 variants of concern. IMPORTANCE There is an urgent need for next-generation COVID-19 vaccines that are safe, demonstrate high protective efficacy against SARS-CoV-2 variants and can be manufactured at scale. We describe a vaccine candidate (CoVac-II) that is based on stabilized, trimeric spike antigen produced in an optimized, scalable and chemically defined production process. CoVac-II demonstrates strong and persistent immunity after vaccination of mice, and is highly immunogenic in multiple animal models, including rabbits and horses. We further show that prior immunity can be boosted using a recombinant spike antigen from the Beta variant; importantly, plasma from boosted mice effectively neutralize multiple SARS-CoV-2 variants in vitro, including Delta. The strong humoral and Th1-biased immunogenicity of CoVac-II is driven by use of Alhydroxiquim-II (AHQ-II), the first adjuvant in an authorized vaccine that acts through the dual Toll-like receptor (TLR)7 and TLR8 pathways, as part of the Covaxin vaccine. Our data suggest AHQ-II/spike protein combinations could constitute safe, affordable, and mass-manufacturable COVID-19 vaccines for global distribution.


Sujet(s)
Anticorps neutralisants/biosynthèse , Anticorps antiviraux/biosynthèse , Vaccins contre la COVID-19/immunologie , SARS-CoV-2/immunologie , Animaux , Anticorps neutralisants/sang , Anticorps antiviraux/sang , Lymphocytes T CD4+/immunologie , Equus caballus , Souris , Lapins , Lymphocytes T/immunologie
13.
Oxid Med Cell Longev ; 2022: 5397733, 2022.
Article de Anglais | MEDLINE | ID: mdl-35047106

RÉSUMÉ

The infection of coronavirus disease (COVID-19) seriously threatens human life. It is urgent to generate effective and safe specific antibodies (Abs) against the pathogenic elements of COVID-19. Mice were immunized with SARS-CoV-2 spike protein antigens: S ectodomain-1 (CoV, in short) mixed in Alum adjuvant for 2 times and boosted with CoV weekly for 6 times. A portion of mice were treated with Maotai liquor (MTL, in short) or/and heat stress (HS) together with CoV boosting. We observed that the anti-CoV Ab was successfully induced in mice that received the CoV/Alum immunization for 2 times. However, upon boosting with CoV, the CoV Ab production diminished progressively; spleen CoV Ab-producing plasma cell counts reduced, in which substantial CoV-specific Ab-producing plasma cells (sPC) were apoptotic. Apparent oxidative stress signs were observed in sPCs; the results were reproduced by exposing sPCs to CoV in the culture. The presence of MTL or/and HS prevented the CoV-induced oxidative stress in sPCs and promoted and stabilized the CoV Ab production in mice in re-exposure to CoV. In summary, CoV/Alum immunization can successfully induce CoV Ab production in mice that declines upon reexposure to CoV. Concurrent administration of MTL/HS stabilizes and promotes the CoV Ab production in mice.


Sujet(s)
Anticorps neutralisants/biosynthèse , Anticorps antiviraux/biosynthèse , Apoptose , COVID-19/immunologie , Plasmocytes/immunologie , SARS-CoV-2/physiologie , Superoxide dismutase-1/physiologie , Adjuvants immunologiques , Boissons alcooliques , Alun , Angiotensin-converting enzyme 2/physiologie , Animaux , Anticorps neutralisants/sang , Anticorps antiviraux/sang , COVID-19/enzymologie , Vaccins contre la COVID-19/immunologie , Réaction de choc thermique , Rappel de vaccin , Immunogénicité des vaccins , Kinase Janus-2/physiologie , Mâle , Souris , Souris de lignée C57BL , Stress oxydatif , Plasmocytes/effets des médicaments et des substances chimiques , Plasmocytes/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Facteur de transcription STAT-1/physiologie , Transduction du signal , Organismes exempts d'organismes pathogènes spécifiques , Glycoprotéine de spicule des coronavirus/immunologie , Vaccination
14.
J Immunol ; 208(3): 685-696, 2022 02 01.
Article de Anglais | MEDLINE | ID: mdl-34987111

RÉSUMÉ

Immune response dysregulation plays a key role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis. In this study, we evaluated immune and endothelial blood cell profiles of patients with coronavirus disease 2019 (COVID-19) to determine critical differences between those with mild, moderate, or severe COVID-19 using spectral flow cytometry. We examined a suite of immune phenotypes, including monocytes, T cells, NK cells, B cells, endothelial cells, and neutrophils, alongside surface and intracellular markers of activation. Our results showed progressive lymphopenia and depletion of T cell subsets (CD3+, CD4+, and CD8+) in patients with severe disease and a significant increase in the CD56+CD14+Ki67+IFN-γ+ monocyte population in patients with moderate and severe COVID-19 that has not been previously described. Enhanced circulating endothelial cells (CD45-CD31+CD34+CD146+), circulating endothelial progenitors (CD45-CD31+CD34+/-CD146-), and neutrophils (CD11b+CD66b+) were coevaluated for COVID-19 severity. Spearman correlation analysis demonstrated the synergism among age, obesity, and hypertension with upregulated CD56+ monocytes, endothelial cells, and decreased T cells that lead to severe outcomes of SARS-CoV-2 infection. Circulating monocytes and endothelial cells may represent important cellular markers for monitoring postacute sequelae and impacts of SARS-CoV-2 infection during convalescence and for their role in immune host defense in high-risk adults after vaccination.


Sujet(s)
COVID-19/immunologie , Cellules endothéliales/immunologie , Monocytes/immunologie , SARS-CoV-2 , Adolescent , Adulte , Facteurs âges , Sujet âgé , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/immunologie , Marqueurs biologiques , Antigènes CD56/analyse , COVID-19/sang , COVID-19/épidémiologie , Enfant , Comorbidité , Cellules endothéliales/composition chimique , Femelle , Cytométrie en flux , Humains , Hypertension artérielle/épidémiologie , Hypertension artérielle/immunologie , Immunophénotypage , Activation des lymphocytes , Sous-populations de lymphocytes/immunologie , Lymphopénie/étiologie , Lymphopénie/immunologie , Mâle , Adulte d'âge moyen , Monocytes/composition chimique , Granulocytes neutrophiles/immunologie , Obésité/épidémiologie , Obésité/immunologie , Antigènes CD31/analyse , SARS-CoV-2/immunologie , Indice de gravité de la maladie , Glycoprotéine de spicule des coronavirus/immunologie , Jeune adulte
15.
JCI Insight ; 7(4)2022 02 22.
Article de Anglais | MEDLINE | ID: mdl-35019861

RÉSUMÉ

BACKGROUNDMost individuals with prior COVID-19 disease manifest long-term protective immune responses against reinfection. Accordingly, we tested the hypothesis that humoral immune and reactogenicity responses to a SARS-CoV-2 mRNA vaccine differ in individuals with and without prior COVID-19 disease.METHODSHealth care workers (n = 61) with (n = 30) and without (n = 31) prior COVID-19 disease received two 30 µg doses of Pfizer BNT162b2 vaccine 3 weeks apart. Serum IgG antibody against the spike receptor-binding domain; serum neutralizing activity; and vaccine reactogenicity were assessed longitudinally every 2 weeks for 56 days after the first injection.RESULTSThe COVID-19 group manifested more rapid increases in spike IgG antibody and serum neutralizing activity after the first vaccine dose but showed little or no increase after the second dose compared with the infection-naive group. In fact, spike IgG was at its maximum level after the first dose in 36% of the COVID-19 group versus 0% of the infection-naive group. Peak IgG antibody levels were lower but appeared to fall more slowly in the COVID-19 group versus the infection-naive group. Finally, adverse systemic reactions, e.g., fever, headache, and malaise, were more frequent and lasted longer after both the first and second injection in the COVID-19 group than in the infection-naive group.CONCLUSIONIndividuals with prior COVID-19 disease demonstrate a robust, accelerated humoral immune response to the first dose but an attenuated response to the second dose of BNT162b2 vaccine compared with controls. The COVID-19 group also experienced greater reactogenicity. Humoral responses and reactogenicity to BNT162b2 differ qualitatively and quantitatively in individuals with prior COVID-19 disease compared with infection-naive individuals.FUNDINGThis work was supported by Temple University institutional funds.


Sujet(s)
Anticorps antiviraux/biosynthèse , Vaccin BNT162/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , Adulte , Femelle , Humains , Immunogénicité des vaccins , Mâle , Adulte d'âge moyen
16.
MAbs ; 14(1): 2013594, 2022.
Article de Anglais | MEDLINE | ID: mdl-35000569

RÉSUMÉ

The ongoing SARS-CoV-2 coronavirus pandemic of 2020-2021 underscores the need for manufacturing platforms that can rapidly produce monoclonal antibody (mAb) therapies. As reported here, a platform based on Nicotiana benthamiana produced mAb therapeutics with high batch-to-batch reproducibility and flexibility, enabling production of 19 different mAbs of sufficient purity and safety for clinical application(s). With a single manufacturing run, impurities were effectively removed for a representative mAb product (the ZMapp component c4G7). Our results show for the first time the reproducibility of the platform for production of multiple batches of clinical-grade mAb, manufactured under current Good Manufacturing Practices, from Nicotiana benthamiana. The flexibility of the system was confirmed by the results of release testing of 19 different mAbs generated with the platform. The process from plant infection to product can be completed within 10 days. Therefore, with a constant supply of plants, response to the outbreak of an infectious disease could be initiated within a matter of weeks. Thus, these data demonstrated that this platform represents a reproducible, flexible system for rapid production of mAb therapeutics to support clinical development.


Sujet(s)
Anticorps monoclonaux , Anticorps antiviraux , COVID-19/immunologie , Nicotiana , Végétaux génétiquement modifiés , SARS-CoV-2/immunologie , Anticorps monoclonaux/biosynthèse , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/génétique , Anticorps monoclonaux/immunologie , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/composition chimique , Anticorps antiviraux/génétique , Anticorps antiviraux/immunologie , Humains , Végétaux génétiquement modifiés/composition chimique , Végétaux génétiquement modifiés/génétique , Végétaux génétiquement modifiés/croissance et développement , Végétaux génétiquement modifiés/immunologie , Protéines recombinantes/biosynthèse , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/immunologie , Nicotiana/composition chimique , Nicotiana/génétique , Nicotiana/croissance et développement , Nicotiana/immunologie , Traitements médicamenteux de la COVID-19
17.
JCI Insight ; 7(4)2022 02 22.
Article de Anglais | MEDLINE | ID: mdl-35030101

RÉSUMÉ

BACKGROUNDVaccine-elicited adaptive immunity is a prerequisite for control of SARS-CoV-2 infection. Multiple sclerosis (MS) disease-modifying therapies (DMTs) differentially target humoral and cellular immunity. A comprehensive comparison of the effects of MS DMTs on SARS-CoV-2 vaccine-specific immunity is needed, including quantitative and functional B and T cell responses.METHODSSpike-specific Ab and T cell responses were measured before and following SARS-CoV-2 vaccination in a cohort of 80 study participants, including healthy controls and patients with MS in 6 DMT groups: untreated and treated with glatiramer acetate (GA), dimethyl fumarate (DMF), natalizumab (NTZ), sphingosine-1-phosphate (S1P) receptor modulators, and anti-CD20 mAbs. Anti-spike-Ab responses were assessed by Luminex assay, VirScan, and pseudovirus neutralization. Spike-specific CD4+ and CD8+ T cell responses were characterized by activation-induced marker and cytokine expression and tetramer.RESULTSAnti-spike IgG levels were similar between healthy control participants and patients with untreated MS and those receiving GA, DMF, or NTZ but were reduced in anti-CD20 mAb- and S1P-treated patients. Anti-spike seropositivity in anti-CD20 mAb-treated patients was correlated with CD19+ B cell levels and inversely correlated with cumulative treatment duration. Spike epitope reactivity and pseudovirus neutralization were reduced in anti-CD20 mAb- and S1P-treated patients. Spike-specific CD4+ and CD8+ T cell reactivity remained robust across all groups, except in S1P-treated patients, in whom postvaccine CD4+ T cell responses were attenuated.CONCLUSIONThese findings from a large cohort of patients with MS exposed to a wide spectrum of MS immunotherapies have important implications for treatment-specific COVID-19 clinical guidelines.FUNDINGNIH grants 1K08NS107619, K08NS096117, R01AI159260, R01NS092835, R01AI131624, and R21NS108159; NMSS grants TA-1903-33713 and RG1701-26628; Westridge Foundation; Chan Zuckerberg Biohub; Maisin Foundation.


Sujet(s)
Anticorps antiviraux/biosynthèse , Vaccins contre la COVID-19/immunologie , Sclérose en plaques/thérapie , SARS-CoV-2/immunologie , Lymphocytes T/immunologie , Anticorps antiviraux/immunologie , Humains , Sclérose en plaques/immunologie
18.
J Autoimmun ; 127: 102792, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34995958

RÉSUMÉ

The emergence and rapid global spread of the new Delta and, more recently, Omicron variants of SARS-CoV-2 pose a daunting public health emergency. Being an RNA virus, the Covid-19 virus is continuing to mutate, resulting in the emergence of new variants with high transmissibility, such as the recently discovered Omicron variant. In this paper, we consider the conditions that may facilitate viral mutations and the emergence of variants with the ability to evade immunity. Here, we have discussed the importance of vaccination with the currently available vaccines. These vaccines are highly effective at preventing serious disease, hospitalization, and death from Covid-19. However, the antibody response induced by these vaccines is short-lasting and there are reports of breakthrough infections. A stable and persistent interaction between T follicular helper cells and germinal center B cells is needed for robust B cell memory response. We discussed the potential reasons behind the breakthrough infections and underscored the importance of developing better second-generation vaccines that may not necessitate frequent booster immunizations and are preventive in nature. This may involve the development of multivalent vaccines and creating vaccines against other viral proteins including conserved proteins. Vaccine hesitancy remains a notable hurdle for implementing vaccination. Furthermore, we recommend different approaches to increase vaccine acceptance, which is a critical translational component of a successful vaccine strategy. These perspectives on overcoming the pandemic's current challenges provide strategies to contain SARS-CoV-2 globally.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19/prévention et contrôle , Pandémies/prévention et contrôle , SARS-CoV-2/immunologie , Réticence à l'égard de la vaccination , Anticorps antiviraux/biosynthèse , Anticorps antiviraux/immunologie , Sous-populations de lymphocytes B/immunologie , COVID-19/transmission , Vaccins contre la COVID-19/immunologie , Interactions hôte-pathogène/immunologie , Humains , Immunogénicité des vaccins , Mémoire immunologique , SARS-CoV-2/génétique , Lymphocytes T/immunologie , Vaccination , Développement de vaccin
19.
Cancer Cell ; 40(1): 103-108.e2, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-34990570

RÉSUMÉ

Patients with cancer are more likely to have impaired immune responses to SARS-CoV-2 vaccines. We study the breadth of responses against SARS-CoV-2 variants after primary vaccination in 178 patients with a variety of tumor types and after booster doses in a subset. Neutralization of alpha, beta, gamma, and delta SARS-CoV-2 variants is impaired relative to wildtype, regardless of vaccine type. Regardless of viral variant, mRNA1273 is the most immunogenic, followed by BNT162b2, and then Ad26.COV2.S. Neutralization of more variants (breadth) is associated with a greater magnitude of wildtype neutralization, and increases with time since vaccination; advancing age associates with a lower breadth. The concentrations of anti-spike protein antibody are a good surrogate for breadth (positive predictive value of =90% at >1,000 U/mL). Booster SARS-CoV-2 vaccines confer enhanced breadth. These data suggest that achieving a high antibody titer is desirable to achieve broad neutralization; a single booster dose with the current vaccines increases the breadth of responses against variants.


Sujet(s)
Anticorps neutralisants/biosynthèse , Anticorps antiviraux/biosynthèse , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , Tumeurs/immunologie , SARS-CoV-2/immunologie , Sujet âgé , Vieillissement/immunologie , Antigènes viraux/immunologie , Femelle , Humains , Rappel de vaccin , Sujet immunodéprimé , Immunogénicité des vaccins , Techniques in vitro , Mâle , Adulte d'âge moyen , Tumeurs/thérapie , Glycoprotéine de spicule des coronavirus/immunologie , Charge virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...