Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 91
Filtrer
1.
Cell ; 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39197450

RÉSUMÉ

Immunization with mosaic-8b (nanoparticles presenting 8 SARS-like betacoronavirus [sarbecovirus] receptor-binding domains [RBDs]) elicits more broadly cross-reactive antibodies than homotypic SARS-CoV-2 RBD-only nanoparticles and protects against sarbecoviruses. To investigate original antigenic sin (OAS) effects on mosaic-8b efficacy, we evaluated the effects of prior COVID-19 vaccinations in non-human primates and mice on anti-sarbecovirus responses elicited by mosaic-8b, admix-8b (8 homotypics), or homotypic SARS-CoV-2 immunizations, finding the greatest cross-reactivity for mosaic-8b. As demonstrated by molecular fate mapping, in which antibodies from specific cohorts of B cells are differentially detected, B cells primed by WA1 spike mRNA-LNP dominated antibody responses after RBD-nanoparticle boosting. While mosaic-8b- and homotypic-nanoparticles boosted cross-reactive antibodies, de novo antibodies were predominantly induced by mosaic-8b, and these were specific for variant RBDs with increased identity to RBDs on mosaic-8b. These results inform OAS mechanisms and support using mosaic-8b to protect COVID-19-vaccinated/infected humans against as-yet-unknown SARS-CoV-2 variants and animal sarbecoviruses with human spillover potential.

2.
Nature ; 633(8029): 451-458, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39112706

RÉSUMÉ

Cancer cells frequently alter their lipids to grow and adapt to their environment1-3. Despite the critical functions of lipid metabolism in membrane physiology, signalling and energy production, how specific lipids contribute to tumorigenesis remains incompletely understood. Here, using functional genomics and lipidomic approaches, we identified de novo sphingolipid synthesis as an essential pathway for cancer immune evasion. Synthesis of sphingolipids is surprisingly dispensable for cancer cell proliferation in culture or in immunodeficient mice but required for tumour growth in multiple syngeneic models. Blocking sphingolipid production in cancer cells enhances the anti-proliferative effects of natural killer and CD8+ T cells partly via interferon-γ (IFNγ) signalling. Mechanistically, depletion of glycosphingolipids increases surface levels of IFNγ receptor subunit 1 (IFNGR1), which mediates IFNγ-induced growth arrest and pro-inflammatory signalling. Finally, pharmacological inhibition of glycosphingolipid synthesis synergizes with checkpoint blockade therapy to enhance anti-tumour immune response. Altogether, our work identifies glycosphingolipids as necessary and limiting metabolites for cancer immune evasion.


Sujet(s)
Lymphocytes T CD8+ , Glycosphingolipides , , Interféron gamma , Protéines proto-oncogènes p21(ras) , Transduction du signal , Échappement de la tumeur à la surveillance immunitaire , Animaux , Glycosphingolipides/métabolisme , Glycosphingolipides/biosynthèse , Souris , Humains , Interféron gamma/métabolisme , Interféron gamma/immunologie , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Femelle , Lignée cellulaire tumorale , Tumeurs/immunologie , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Prolifération cellulaire , Récepteur interféron/métabolisme , Récepteur interféron/génétique , Récepteur interféron/déficit , Mâle , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Sphingolipides/métabolisme , Sphingolipides/biosynthèse , Échappement immunitaire , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris de lignée C57BL
3.
Immunity ; 57(7): 1618-1628.e4, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38838672

RÉSUMÉ

Re-exposure to an antigen generates abundant antibody responses and drives the formation of secondary germinal centers (GCs). Recall GCs in mice consist almost entirely of naïve B cells, whereas recall antibodies derive overwhelmingly from memory B cells. Here, we examine this division between cellular and serum compartments. After repeated immunization with the same antigen, tetramer analyses of recall GCs revealed a marked decrease in the ability of B cells in these structures to bind the antigen. Boosting with viral variant proteins restored antigen binding in recall GCs, as did genetic ablation of primary-derived antibody-secreting cells through conditional deletion of Prdm1, demonstrating suppression of GC recall responses by pre-existing antibodies. In hapten-carrier experiments in which B and T cell specificities were uncoupled, memory T cell help allowed B cells with undetectable antigen binding to access GCs. Thus, antibody-mediated feedback steers recall GC B cells away from previously targeted epitopes and enables specific targeting of variant epitopes, with implications for vaccination protocols.


Sujet(s)
Lymphocytes B , Centre germinatif , Mémoire immunologique , Centre germinatif/immunologie , Animaux , Souris , Mémoire immunologique/immunologie , Lymphocytes B/immunologie , Cellules T mémoire/immunologie , Souris de lignée C57BL , Facteur-1 liant le domaine de régulation positive I/immunologie , Facteur-1 liant le domaine de régulation positive I/génétique , Production d'anticorps/immunologie , Cellules B mémoire/immunologie , Souris knockout
4.
bioRxiv ; 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38798523

RÉSUMÉ

Nucleoside-modified mRNA vaccines elicit protective antibodies through their ability to promote T follicular helper (Tfh) cells. The lipid nanoparticle (LNP) component of mRNA vaccines possesses inherent adjuvant activity. However, to what extent the nucleoside-modified mRNA can be sensed and contribute to Tfh cell responses remains largely undefined. Herein, we deconvoluted the signals induced by LNP and mRNA that instruct dendritic cells (DCs) to promote Tfh cell differentiation. We demonstrated that the nucleoside-modified mRNA drives the production of type I interferons that act on DCs to induce their maturation and the induction of Th1-biased Tfh responses. Conversely, LNP favors the acquisition of a Tfh cell-inducing program in DCs, a stronger Th2 polarization in Tfh cells, and allows for rapid mRNA translation by DCs within the draining lymph node. Our work unravels distinct adjuvant features of mRNA and LNP necessary for the induction of Tfh cells, with implications for vaccine design.

5.
Nature ; 627(8003): 399-406, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38448581

RÉSUMÉ

Immune cells rely on transient physical interactions with other immune and non-immune populations to regulate their function1. To study these 'kiss-and-run' interactions directly in vivo, we previously developed LIPSTIC (labelling immune partnerships by SorTagging intercellular contacts)2, an approach that uses enzymatic transfer of a labelled substrate between the molecular partners CD40L and CD40 to label interacting cells. Reliance on this pathway limited the use of LIPSTIC to measuring interactions between CD4+ T helper cells and antigen-presenting cells, however. Here we report the development of a universal version of LIPSTIC (uLIPSTIC), which can record physical interactions both among immune cells and between immune and non-immune populations irrespective of the receptors and ligands involved. We show that uLIPSTIC can be used, among other things, to monitor the priming of CD8+ T cells by dendritic cells, reveal the steady-state cellular partners of regulatory T cells and identify germinal centre-resident T follicular helper cells on the basis of their ability to interact cognately with germinal centre B cells. By coupling uLIPSTIC with single-cell transcriptomics, we build a catalogue of the immune populations that physically interact with intestinal epithelial cells at the steady state and profile the evolution of the interactome of lymphocytic choriomeningitis virus-specific CD8+ T cells in multiple organs following systemic infection. Thus, uLIPSTIC provides a broadly useful technology for measuring and understanding cell-cell interactions across multiple biological systems.


Sujet(s)
Lymphocytes B , Lymphocytes T CD8+ , Communication cellulaire , Cellules dendritiques , Cellules épithéliales , Lymphocytes T auxiliaires folliculaires , Lymphocytes T régulateurs , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Communication cellulaire/immunologie , Cellules dendritiques/cytologie , Cellules dendritiques/immunologie , Ligands , Lymphocytes T régulateurs/cytologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T auxiliaires folliculaires/cytologie , Lymphocytes T auxiliaires folliculaires/immunologie , Lymphocytes B/cytologie , Lymphocytes B/immunologie , Centre germinatif/cytologie , Analyse de l'expression du gène de la cellule unique , Cellules épithéliales/cytologie , Cellules épithéliales/immunologie , Muqueuse intestinale/cytologie , Muqueuse intestinale/immunologie , Virus de la chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/virologie , Spécificité d'organe
6.
bioRxiv ; 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38370696

RÉSUMÉ

Immunization with mosaic-8b [60-mer nanoparticles presenting 8 SARS-like betacoronavirus (sarbecovirus) receptor-binding domains (RBDs)] elicits more broadly cross-reactive antibodies than homotypic SARS-CoV-2 RBD-only nanoparticles and protects against sarbecoviruses. To investigate original antigenic sin (OAS) effects on mosaic-8b efficacy, we evaluated effects of prior COVID-19 vaccinations in non-human primates and mice on anti-sarbecovirus responses elicited by mosaic-8b, admix-8b (8 homotypics), or homotypic SARS-CoV-2 immunizations, finding greatest cross-reactivity for mosaic-8b. As demonstrated by molecular fate-mapping in which antibodies from specific cohorts of B cells are differentially detected, B cells primed by WA1 spike mRNA-LNP dominated antibody responses after RBD-nanoparticle boosting. While mosaic-8b- and homotypic-nanoparticles boosted cross-reactive antibodies, de novo antibodies were predominantly induced by mosaic-8b, and these were specific for variant RBDs with increased identity to RBDs on mosaic-8b. These results inform OAS mechanisms and support using mosaic-8b to protect COVID-19 vaccinated/infected humans against as-yet-unknown SARS-CoV-2 variants and animal sarbecoviruses with human spillover potential.

7.
bioRxiv ; 2023 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-37873082

RÉSUMÉ

Immunoglobulin A (IgA) is the most abundant antibody isotype produced across mammals and plays a specialized role in mucosal homeostasis 1 . Constantly secreted into the lumen of the intestine, IgA binds commensal microbiota to regulate their colonization and function 2,3 , with unclear implications for health. IgA deficiency is common in humans but is difficult to study due to its complex etiology and comorbidities 4-8 . Using genetically and environmentally controlled mice, here we show that IgA-deficient animals have a baseline alteration in the colon epithelium that increases susceptibility to multiple models of colorectal cancer. Transcriptome, imaging, and flow cytometry-based analyses revealed that, in the absence of IgA, colonic epithelial cells induce antibacterial factors and accelerate cell cycling in response to the microbiota. Oral treatment with IgA was sufficient to suppress aberrant epithelial proliferation independently of bacterial binding, suggesting that IgA provides a feedback signal to epithelial cells in parallel with its known roles in microbiome shaping. In a primary colonic organoid culture system, IgA directly suppresses epithelial growth. Conversely, the susceptibility of IgA-deficient mice to colorectal cancer was reversed by Notch inhibition to suppress the absorptive colonocyte developmental program, or by inhibition of the cytokine MIF, the receptor for which was upregulated in stem cells of IgA-deficient animals. These studies demonstrate a homeostatic function for IgA in tempering physiological epithelial responses to microbiota to maintain mucosal health.

8.
Nat Microbiol ; 8(6): 1051-1063, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37188812

RÉSUMÉ

Human monoclonal antibodies (mAbs) that target the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein have been isolated from convalescent individuals and developed into therapeutics for SARS-CoV-2 infection. However, therapeutic mAbs for SARS-CoV-2 have been rendered obsolete by the emergence of mAb-resistant virus variants. Here we report the generation of a set of six human mAbs that bind the human angiotensin-converting enzyme-2 (hACE2) receptor, rather than the SARS-CoV-2 spike protein. We show that these antibodies block infection by all hACE2 binding sarbecoviruses tested, including SARS-CoV-2 ancestral, Delta and Omicron variants at concentrations of ~7-100 ng ml-1. These antibodies target an hACE2 epitope that binds to the SARS-CoV-2 spike, but they do not inhibit hACE2 enzymatic activity nor do they induce cell-surface depletion of hACE2. They have favourable pharmacology, protect hACE2 knock-in mice against SARS-CoV-2 infection and should present a high genetic barrier to the acquisition of resistance. These antibodies should be useful prophylactic and treatment agents against any current or future SARS-CoV-2 variants and might be useful to treat infection with any hACE2-binding sarbecoviruses that emerge in the future.


Sujet(s)
COVID-19 , Virus du SRAS , Humains , Animaux , Souris , SARS-CoV-2 , COVID-19/prévention et contrôle , Anticorps monoclonaux/pharmacologie
9.
Methods Mol Biol ; 2618: 71-80, 2023.
Article de Anglais | MEDLINE | ID: mdl-36905509

RÉSUMÉ

Interactions between different cell types are key for immune function. Traditionally, interactions have been investigated in vivo by intravital two-photon microscopy, but the molecular characterization of the cells participating in a specific interaction is limited by the inability to retrieve the cells for downstream analysis. We recently developed an approach to label cells undergoing specific interactions in vivo, which we called LIPSTIC (Labeling Immune Partnership by Sortagging Intercellular Contacts). Here, we provide detailed instructions on how to track CD40-CD40L interactions between dendritic cells (DCs) and CD4+ T cells using genetically engineered LIPSTIC mice. This protocol requires expertise in animal experimentation and multicolor flow cytometry. Once mouse crossing has been achieved, it takes 3 days or more to complete, depending on the kinetics of the interactions that the researcher wishes to investigate.


Sujet(s)
Ligand de CD40 , Lymphocytes T , Souris , Animaux , Lymphocytes T/métabolisme , Ligand de CD40/métabolisme , Antigènes CD40 , Cellules dendritiques , Lymphocytes T CD4+
10.
bioRxiv ; 2023 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-36993443

RÉSUMÉ

Cellular interactions are essential for tissue organization and functionality. In particular, immune cells rely on direct and usually transient interactions with other immune and non-immune populations to specify and regulate their function. To study these "kiss-and-run" interactions directly in vivo, we previously developed LIPSTIC (Labeling Immune Partnerships by SorTagging Intercellular Contacts), an approach that uses enzymatic transfer of a labeled substrate between the molecular partners CD40L and CD40 to label interacting cells. Reliance on this pathway limited the use of LIPSTIC to measuring interactions between CD4+ helper T cells and antigen presenting cells, however. Here, we report the development of a universal version of LIPSTIC (uLIPSTIC), which can record physical interactions both among immune cells and between immune and non-immune populations irrespective of the receptors and ligands involved. We show that uLIPSTIC can be used, among other things, to monitor the priming of CD8+ T cells by dendritic cells, reveal the cellular partners of regulatory T cells in steady state, and identify germinal center (GC)-resident T follicular helper (Tfh) cells based on their ability to interact cognately with GC B cells. By coupling uLIPSTIC with single-cell transcriptomics, we build a catalog of the immune populations that physically interact with intestinal epithelial cells (IECs) and find evidence of stepwise acquisition of the ability to interact with IECs as CD4+ T cells adapt to residence in the intestinal tissue. Thus, uLIPSTIC provides a broadly useful technology for measuring and understanding cell-cell interactions across multiple biological systems.

11.
Science ; 379(6629): eabj7412, 2023 01 20.
Article de Anglais | MEDLINE | ID: mdl-36656933

RÉSUMÉ

Multicellular life requires altruistic cooperation between cells. The adaptive immune system is a notable exception, wherein germinal center B cells compete vigorously for limiting positive selection signals. Studying primary human lymphomas and developing new mouse models, we found that mutations affecting BTG1 disrupt a critical immune gatekeeper mechanism that strictly limits B cell fitness during antibody affinity maturation. This mechanism converted germinal center B cells into supercompetitors that rapidly outstrip their normal counterparts. This effect was conferred by a small shift in MYC protein induction kinetics but resulted in aggressive invasive lymphomas, which in humans are linked to dire clinical outcomes. Our findings reveal a delicate evolutionary trade-off between natural selection of B cells to provide immunity and potentially dangerous features that recall the more competitive nature of unicellular organisms.


Sujet(s)
Lymphocytes B , Transformation cellulaire néoplasique , Lymphome B diffus à grandes cellules , Protéines tumorales , Animaux , Humains , Souris , Affinité des anticorps/génétique , Lymphocytes B/anatomopathologie , Centre germinatif , Mutation , Protéines tumorales/génétique , Lymphome B diffus à grandes cellules/génétique , Transformation cellulaire néoplasique/génétique , Sélection génétique
12.
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
13.
Cell ; 186(1): 131-146.e13, 2023 01 05.
Article de Anglais | MEDLINE | ID: mdl-36565697

RÉSUMÉ

Germinal centers (GCs) form in secondary lymphoid organs in response to infection and immunization and are the source of affinity-matured B cells. The duration of GC reactions spans a wide range, and long-lasting GCs (LLGCs) are potentially a source of highly mutated B cells. We show that rather than consisting of continuously evolving B cell clones, LLGCs elicited by influenza virus or SARS-CoV-2 infection in mice are sustained by progressive replacement of founder clones by naive-derived invader B cells that do not detectably bind viral antigens. Rare founder clones that resist replacement for long periods are enriched in clones with heavily mutated immunoglobulins, including some with very high affinity for antigen, that can be recalled by boosting. Our findings reveal underappreciated aspects of the biology of LLGCs generated by respiratory virus infection and identify clonal replacement as a potential constraint on the development of highly mutated antibodies within these structures.


Sujet(s)
Lymphocytes B , Centre germinatif , Infections à virus à ARN , Animaux , Souris , Lymphocytes B/cytologie , Lymphocytes B/immunologie , Clones cellulaires , COVID-19 , Centre germinatif/cytologie , Centre germinatif/immunologie , SARS-CoV-2 , Grippe humaine , Infections à virus à ARN/immunologie , Infections à virus à ARN/anatomopathologie , Infections à virus à ARN/virologie
14.
bioRxiv ; 2023 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-38168231

RÉSUMÉ

Re-exposure to an antigen generates serum antibody responses that greatly exceed in magnitude those elicited by primary antigen encounter, while simultaneously driving the formation of recall germinal centers (GCs). Although recall GCs in mice are composed almost entirely of naïve B cells, recall antibody titers derive overwhelmingly from memory B cells, suggesting a division between cellular and serum compartments. Here, we show that this schism is at least partly explained by a marked decrease in the ability of recall GC B cells to detectably bind antigen. Variant priming and plasmablast ablation experiments show that this decrease is largely due to suppression by pre-existing antibody, whereas hapten-carrier experiments reveal a role for memory T cell help in allowing B cells with undetectable antigen binding to access GCs. We propose a model in which antibody-mediated feedback steers recall GC B cells away from previously targeted epitopes, thus enabling specific targeting of variant epitopes.

15.
Front Immunol ; 13: 1007080, 2022.
Article de Anglais | MEDLINE | ID: mdl-36451809

RÉSUMÉ

Efficient mouse models to study SARS-CoV-2 infection are critical for the development and assessment of vaccines and therapeutic approaches to mitigate the current pandemic and prevent reemergence of COVID-19. While the first generation of mouse models allowed SARS-CoV-2 infection and pathogenesis, they relied on ectopic expression and non-physiological levels of human angiotensin-converting enzyme 2 (hACE2). Here we generated a mouse model carrying the minimal set of modifications necessary for productive infection with multiple strains of SARS-CoV-2. Substitution of only three amino acids in the otherwise native mouse Ace2 locus (Ace2 TripleMutant or Ace2™), was sufficient to render mice susceptible to both SARS-CoV-2 strains USA-WA1/2020 and B.1.1.529 (Omicron). Infected Ace2™ mice exhibited weight loss and lung damage and inflammation, similar to COVID-19 patients. Previous exposure to USA-WA1/2020 or mRNA vaccination generated memory B cells that participated in plasmablast responses during breakthrough B.1.1.529 infection. Thus, the Ace2™ mouse replicates human disease after SARS-CoV-2 infection and provides a tool to study immune responses to sequential infections in mice.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , Souris , Animaux , Angiotensin-converting enzyme 2/génétique , Modèles animaux de maladie humaine , Pandémies
16.
Adv Immunol ; 155: 95-131, 2022.
Article de Anglais | MEDLINE | ID: mdl-36357013

RÉSUMÉ

Most antibody produced by humans originates from mucosal B cell responses. The rules, mechanisms, and outcomes of this process are distinct from B cell responses to infection. Within the context of the intestine, we discuss the induction of follicular B cell responses by microbiota, the development and maintenance of mucosal antibody-secreting cells, and the unusual impacts of mucosal antibody on commensal bacteria. Much remains to be learned about the interplay between B cells and the microbiota, but past and present work hints at a complex, nuanced relationship that may be critical to the way the mammalian gut fosters a beneficial microbial ecosystem.


Sujet(s)
Microbiome gastro-intestinal , Humains , Animaux , Muqueuse intestinale , Immunoglobuline A , Écosystème , Lymphocytes B , Immunité muqueuse , Mammifères
18.
bioRxiv ; 2022 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-36093344

RÉSUMÉ

The ability of serum antibody to protect against pathogens arises from the interplay of antigen-specific B cell clones of different affinities and fine specificities. These cellular dynamics are ultimately responsible for serum-level phenomena such as antibody imprinting or "Original Antigenic Sin" (OAS), a proposed propensity of the immune system to rely repeatedly on the first cohort of B cells that responded to a stimulus upon exposure to related antigens. Imprinting/OAS is thought to pose a barrier to vaccination against rapidly evolving viruses such as influenza and SARS-CoV-2. Precise measurement of the extent to which imprinting/OAS inhibits the recruitment of new B cell clones by boosting is challenging because cellular and temporal origins cannot readily be assigned to antibodies in circulation. Thus, the extent to which imprinting/OAS impacts the induction of new responses in various settings remains unclear. To address this, we developed a "molecular fate-mapping" approach in which serum antibodies derived from specific cohorts of B cells can be differentially detected. We show that, upon sequential homologous boosting, the serum antibody response strongly favors reuse of the first cohort of B cell clones over the recruitment of new, naÏve-derived B cells. This "primary addiction" decreases as a function of antigenic distance, allowing secondary immunization with divergent influenza virus or SARS-CoV-2 glycoproteins to overcome imprinting/OAS by targeting novel epitopes absent from the priming variant. Our findings have implications for the understanding of imprinting/OAS, and for the design and testing of vaccines aimed at eliciting antibodies to evolving antigens.

19.
Science ; 377(6606): 660-666, 2022 08 05.
Article de Anglais | MEDLINE | ID: mdl-35926021

RÉSUMÉ

The microbiome contributes to the development and maturation of the immune system. In response to commensal bacteria, intestinal CD4+ T lymphocytes differentiate into functional subtypes with regulatory or effector functions. The development of small intestine intraepithelial lymphocytes that coexpress CD4 and CD8αα homodimers (CD4IELs) depends on the microbiota. However, the identity of the microbial antigens recognized by CD4+ T cells that can differentiate into CD4IELs remains unknown. We identified ß-hexosaminidase, a conserved enzyme across commensals of the Bacteroidetes phylum, as a driver of CD4IEL differentiation. In a mouse model of colitis, ß-hexosaminidase-specific lymphocytes protected against intestinal inflammation. Thus, T cells of a single specificity can recognize a variety of abundant commensals and elicit a regulatory immune response at the intestinal mucosa.


Sujet(s)
Bacteroidetes , Lymphocytes T CD4+ , Colite , Muqueuse intestinale , beta-N-Acetylhexosaminidases , Animaux , Bacteroidetes/enzymologie , Bacteroidetes/immunologie , Lymphocytes T CD4+/immunologie , Antigènes CD8/immunologie , Colite/immunologie , Colite/microbiologie , Modèles animaux de maladie humaine , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Souris , Souris de lignée C57BL , beta-N-Acetylhexosaminidases/immunologie
20.
Annu Rev Immunol ; 40: 413-442, 2022 04 26.
Article de Anglais | MEDLINE | ID: mdl-35113731

RÉSUMÉ

Germinal centers (GCs) are microanatomical sites of B cell clonal expansion and antibody affinity maturation. Therein, B cells undergo the Darwinian process of somatic diversification and affinity-driven selection of immunoglobulins that produces the high-affinity antibodies essential for effective humoral immunity. Here, we review recent developments in the field of GC biology, primarily as it pertains to GCs induced by infection or immunization. First, we summarize the phenotype and function of the different cell types that compose the GC, focusing on GC B cells. Then, we review the cellular and molecular bases of affinity-dependent selection within the GC and the export of memory and plasma cells. Finally, we present an overview of the emerging field of GC clonal dynamics, focusing on how GC and post-GC selection shapes the diversity of antibodies secreted into serum.


Sujet(s)
Lymphocytes B , Centre germinatif , Animaux , Anticorps , Affinité des anticorps , Humains , Immunité humorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE