Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Article in English | MEDLINE | ID: mdl-34772811

ABSTRACT

Chronic viral infections subvert protective B cell immunity. An early type I interferon (IFN-I)-driven bias to short-lived plasmablast differentiation leads to clonal deletion, so-called "decimation," of antiviral memory B cells. Therefore, prophylactic countermeasures against decimation remain an unmet need. We show that vaccination-induced CD4 T cells prevented the decimation of naïve and memory B cells in chronically lymphocytic choriomeningitis virus (LCMV)-infected mice. Although these B cell responses were largely T independent when IFN-I was blocked, preexisting T help assured their sustainability under conditions of IFN-I-driven inflammation by instructing a germinal center B cell transcriptional program. Prevention of decimation depended on T cell-intrinsic Bcl6 and Tfh progeny formation. Antigen presentation by B cells, interactions with antigen-specific T helper cells, and costimulation by CD40 and ICOS were also required. Importantly, B cell-mediated virus control averted Th1-driven immunopathology in LCMV-challenged animals with preexisting CD4 T cell immunity. Our findings show that vaccination-induced Tfh cells represent a cornerstone of effective B cell immunity to chronic virus challenge, pointing the way toward more effective B cell-based vaccination against persistent viral diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Persistent Infection/immunology , Vaccines/immunology , Virus Diseases/immunology , Animals , Antibodies, Viral/immunology , Antigen Presentation/immunology , Antiviral Agents/immunology , Cells, Cultured , Germinal Center/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Memory B Cells/immunology , Mice , Proto-Oncogene Proteins c-bcl-6/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Vaccination/methods
2.
Rev Med Suisse ; 17(733): 690-696, 2021 Apr 07.
Article in French | MEDLINE | ID: mdl-33830701

ABSTRACT

Many vaccine strategies have been developed to control the COVID-19 pandemic. This article presents the mechanisms of action and the efficacy of different vaccines including mRNA- and adenovirus-based vaccines. We will discuss the different vaccine targets, immune responses and allergic reactions which have been reported during the vaccination campaigns. Finally, the latest recommendations for the prevention and management of severe allergic reactions will be summarized.


De nombreuses stratégies vaccinales ont été développées pour tenter de contrôler la pandémie de Covid-19. Cet article présente le fonctionnement et l'efficacité de différents vaccins, en particulier ceux à ARN messager et adénovecteurs. Nous discutons des cibles vaccinales, des véhicules vaccinaux ainsi que de l'immunité qu'ils induisent. Nous nous penchons également sur la question des allergies aux vaccins qui a rapidement été soulevée après le début des campagnes de vaccination à large échelle. Les allergènes potentiellement en cause et les mécanismes impliqués sont discutés. Enfin, nous proposons des recommandations pour la prévention et la prise en charge des réactions allergiques sévères.


Subject(s)
COVID-19 , Hypersensitivity , Vaccines , COVID-19 Vaccines , Humans , Hypersensitivity/prevention & control , Pandemics , SARS-CoV-2
4.
J Infect ; 87(2): 111-119, 2023 08.
Article in English | MEDLINE | ID: mdl-37321353

ABSTRACT

OBJECTIVES: Intradermal skin test (IDT) with mRNA vaccines may represent a simple, reliable, and affordable tool to measure T cell response in immunocompromised patients who failed to mount serological responses following vaccination with mRNA covid-19 vaccines. METHODS: We compared anti-SARS-CoV-2 antibodies and cellular responses in vaccinated immunocompromised patients (n = 58), healthy seronegative naive controls (NC, n = 8), and healthy seropositive vaccinated controls (VC, n = 32) by Luminex, spike-induced IFN-γ Elispot and an IDT. A skin biopsy 24 h after IDT and single-cell RNAseq was performed in three vaccinated volunteers. RESULTS: Twenty-five percent of seronegative NC had a positive Elispot (2/8) and IDT (1/4), compared to 95% (20/21) and 93% (28/30) in seropositive VC, respectively. Single-cell RNAseq data in the skin of VC showed a predominant mixed population of effector helper and cytotoxic T cells. The TCR repertoire revealed 18/1064 clonotypes with known specificities against SARS-CoV-2, among which six were spike-specific. Seronegative immunocompromised patients with positive Elispot and IDT were in 83% (5/6) treated with B cell-depleting reagents, while those with negative IDT were all transplant recipients. CONCLUSIONS: Our results indicate that delayed local reaction to IDT reflects vaccine-induced T-cell immunity opening new perspectives to monitor seronegative patients and elderly populations with waning immunity.


Subject(s)
COVID-19 , T-Lymphocytes , Aged , Humans , COVID-19 Vaccines , COVID-19/diagnosis , COVID-19/prevention & control , SARS-CoV-2 , Biomarkers , mRNA Vaccines , Antibodies, Viral , Immunocompromised Host , Skin Tests , Vaccination
5.
Cell Rep ; 38(5): 110303, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108544

ABSTRACT

Across the animal kingdom, multivalency discriminates antibodies from all other immunoglobulin superfamily members. The evolutionary forces conserving multivalency above other structural hallmarks of antibodies remain, however, incompletely defined. Here, we engineer monovalent either Fc-competent or -deficient antibody formats to investigate mechanisms of protection of neutralizing antibodies (nAbs) and non-neutralizing antibodies (nnAbs) in virus-infected mice. Antibody bivalency enables the tethering of virions to the infected cell surface, inhibits the release of virions in cell culture, and suppresses viral loads in vivo independently of Fc gamma receptor (FcγR) interactions. In return, monovalent antibody formats either do not inhibit virion release and fail to protect in vivo or their protective efficacy is largely FcγR dependent. Protection in mice correlates with virus-release-inhibiting activity of nAb and nnAb rather than with their neutralizing capacity. These observations provide mechanistic insights into the evolutionary conservation of antibody bivalency and help refining correlates of nnAb protection for vaccine development.


Subject(s)
Antibodies, Viral/pharmacology , Antiviral Agents/pharmacology , HIV Antibodies/pharmacology , Receptors, Fc/drug effects , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Epitopes/drug effects , Epitopes/immunology , HIV Antibodies/immunology , Immunoglobulin G/drug effects , Immunoglobulin G/immunology , Mice, Inbred C57BL , Receptors, IgG/drug effects , Receptors, IgG/immunology
6.
Cell Rep ; 37(9): 110061, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34852228

ABSTRACT

Passive antibody therapy and vectored antibody gene delivery (VAGD) in particular offer an innovative approach to combat persistent viral diseases. Here, we exploit a small animal model to investigate synergies of VAGD with the host's endogenous immune defense for treating chronic viral infection. An adeno-associated virus (AAV) vector delivering the lymphocytic choriomeningitis virus (LCMV)-neutralizing antibody KL25 (AAV-KL25) establishes protective antibody titers for >200 days. When therapeutically administered to chronically infected immunocompetent wild-type mice, AAV-KL25 affords sustained viral load control. In contrast, viral mutational escape thwarts therapeutic AAV-KL25 effects when mice are unable to mount LCMV-specific antibody responses or lack CD8+ T cells. VAGD augments antiviral germinal center B cell and antibody-secreting cell responses and reduces inhibitory receptor expression on antiviral CD8+ T cells. These results indicate that VAGD fortifies host immune defense and synergizes with B cell and CD8 T cell responses to restore immune control of chronic viral infection.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genes, Immunoglobulin , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Germinal Center , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Viral Load
7.
Expert Rev Clin Pharmacol ; 13(11): 1203-1218, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33008265

ABSTRACT

INTRODUCTION: Systemic sclerosis (SSc) is a rare, difficult to treat disease with profound effects on quality of life and high mortality. Complex and incompletely understood pathophysiologic processes and greatly heterogeneous clinical presentations and outcomes have hampered drug development. AREAS COVERED: This review summarizes the currently available immunosuppressive and antifibrotic therapies and discusses novel approaches for the treatment of SSc. We reviewed the literature using the MEDLINE and ClinicalTrial.gov databases between May and September 2020. EXPERT OPINION: Available immunosuppressive and antifibrotic drugs only modestly impact the course of the disease. Most drugs are currently only investigated in the subset of patients with early diffuse cutaneous SSc. In this patient population, hematopoietic stem-cell transplantation is currently the only treatment that has demonstrated reversal of lung involvement, enhanced quality of life and reduced long-term mortality, but carries the risk of short-term treatment-related mortality. A great need to provide better therapeutic options to patients exists also for those patients who have limited cutaneous skin involvement. A better understanding of SSc pathophysiology has enabled the identification of numerous new therapeutic targets. The progress made in the design of clinical trials and outcome parameters will likely result in the improvement of effective management options.


Subject(s)
Drug Development , Immunosuppressive Agents/therapeutic use , Scleroderma, Systemic/drug therapy , Animals , Clinical Trials as Topic/methods , Hematopoietic Stem Cell Transplantation , Humans , Immunosuppressive Agents/pharmacology , Quality of Life , Research Design , Scleroderma, Systemic/mortality , Scleroderma, Systemic/physiopathology
8.
Cell Rep ; 30(4): 1013-1026.e7, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31995746

ABSTRACT

Persistent viral infections subvert key elements of adaptive immunity. To compare germinal center (GC) B cell responses in chronic and acute lymphocytic choriomeningitis virus infection, we exploit activation-induced deaminase (AID) fate-reporter mice and perform adoptive B cell transfer experiments. Chronic infection yields GC B cell responses of higher cellularity than acute infections do, higher memory B cell and antibody secreting cell output for longer periods of time, a better representation of the late B cell repertoire in serum immunoglobulin, and higher titers of protective neutralizing antibodies. GC B cells of chronically infected mice are similarly hypermutated as those emerging from acute infection. They efficiently adapt to viral escape variants and even in hypermutation-impaired AID mutant mice, chronic infection selects for GC B cells with hypermutated B cell receptors (BCRs) and neutralizing antibody formation. These findings demonstrate that, unlike for CD8+ T cells, chronic viral infection drives a functional, productive, and protective GC B cell response.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Single-Domain Antibodies/genetics , Acute Disease , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line , Chronic Disease , Cricetinae , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Germinal Center/cytology , High-Throughput Nucleotide Sequencing , Immunoglobulin Joining Region/genetics , Immunohistochemistry , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Somatic Hypermutation, Immunoglobulin
9.
Cell Host Microbe ; 22(3): 354-365.e5, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28826838

ABSTRACT

Arenaviruses such as Lassa virus (LASV) cause hemorrhagic fever. Terminal shock is associated with a systemic cytokine storm, but the mechanisms are ill defined. Here we used HLA-A2-expressing mice infected with a monkey-pathogenic strain of lymphocytic choriomeningitis virus (LCMV-WE), a close relative of LASV, to investigate the pathophysiology of arenavirus hemorrhagic fever (AHF). AHF manifested as pleural effusions, edematous skin swelling, and serum albumin loss, culminating in hypovolemic shock. A characteristic cytokine storm included numerous pro-inflammatory cytokines and nitric oxide (NO) metabolites. Edema formation and terminal shock were abrogated in mice lacking inducible nitric oxide synthase (iNOS), although the cytokine storm persisted. iNOS was upregulated in the liver in a T cell- and interferon-γ (IFN-γ)-dependent fashion. Accordingly, blockade of IFN-γ or depletion of T cells repressed hepatic iNOS and prevented disease despite unchecked high-level viremia. We identify the IFN-γ-iNOS axis as an essential and potentially druggable molecular pathway to AHF-induced shock.


Subject(s)
Hemorrhagic Fevers, Viral/immunology , Interferon-gamma/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Nitric Oxide Synthase Type II/immunology , Animals , Disease Models, Animal , Female , Hemorrhagic Fevers, Viral/genetics , Hemorrhagic Fevers, Viral/virology , Humans , Interferon-gamma/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/genetics
10.
Sci Immunol ; 1(4)2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27872905

ABSTRACT

Inadequate antibody responses and perturbed B cell compartments represent hallmarks of persistent microbial infections, but the mechanisms whereby persisting pathogens suppress humoral immunity remain poorly defined. Using adoptive transfer experiments in the context of a chronic lymphocytic choriomeningitis virus (LCMV) infection of mice, we have documented rapid depletion of virus-specific B cells that coincided with the early type I interferon response to infection. We found that the loss of activated B cells was driven by type I interferon (IFN-I) signaling to several cell types including dendritic cells, T cells and myeloid cells. Intriguingly, this process was independent of B cell-intrinsic IFN-I sensing and resulted from biased differentiation of naïve B cells into short-lived antibody-secreting cells. The ability to generate robust B cell responses was restored upon IFN-I receptor blockade or, partially, when experimentally depleting myeloid cells or the IFN-I-induced cytokines interleukin 10 and tumor necrosis factor alpha. We have termed this IFN-I-driven depletion of B cells "B cell decimation". Strategies to counter "B cell decimation" should thus help us better leverage humoral immunity in the combat against persistent microbial diseases.

11.
Sci Immunol ; 1(4)2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27868108

ABSTRACT

Antibodies are critical for protection against viral infections. However, several viruses, such as lymphocytic choriomeningitis virus (LCMV), avoid the induction of early protective antibody responses by poorly understood mechanisms. Here we analyzed the spatiotemporal dynamics of B cell activation to show that, upon subcutaneous infection, LCMV-specific B cells readily relocate to the interfollicular and T cell areas of the draining lymph node where they extensively interact with CD11b+Ly6Chi inflammatory monocytes. These myeloid cells were recruited to lymph nodes draining LCMV infection sites in a type I interferon-, CCR2-dependent fashion and they suppressed antiviral B cell responses by virtue of their ability to produce nitric oxide. Depletion of inflammatory monocytes, inhibition of their lymph node recruitment or impairment of their nitric oxide-producing ability enhanced LCMV-specific B cell survival and led to robust neutralizing antibody production. In conclusion, our results identify inflammatory monocytes as critical gatekeepers that prevent antiviral B cell responses and suggest that certain viruses take advantage of these cells to prolong their persistence within the host.

SELECTION OF CITATIONS
SEARCH DETAIL