Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Immunol ; 9(93): eadi8150, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38517953

ABSTRACT

In autoreactive germinal centers (GC) initiated by a single rogue B cell clone, wild-type B cells expand and give rise to clones that target other autoantigens, known as epitope spreading. The chronic, progressive nature of epitope spreading calls for early interventions to limit autoimmune pathologies, but the kinetics and molecular requirements for wild-type B cell invasion and participation in GC remain largely unknown. With parabiosis and adoptive transfer approaches in a murine model of systemic lupus erythematosus, we demonstrate that wild-type B cells join existing GCs rapidly, clonally expand, persist, and contribute to autoantibody production and diversification. The invasion of autoreactive GCs by wild-type B cells required TLR7, B cell receptor specificity, antigen presentation, and type I interferon signaling. The adoptive transfer model provides a tool for identifying early events in the breaking of B cell tolerance in autoimmunity.


Subject(s)
B-Lymphocytes , Lupus Erythematosus, Systemic , Mice , Animals , Germinal Center , Autoimmunity , Epitopes
2.
Clin Exp Immunol ; 210(3): 263-272, 2022 12 31.
Article in English | MEDLINE | ID: mdl-35960996

ABSTRACT

Obesity increases the risk of type 2 diabetes mellitus, cardiovascular disease, fatty liver disease, and cancer. It is also linked with more severe complications from infections, including COVID-19, and poor vaccine responses. Chronic, low-grade inflammation and associated immune perturbations play an important role in determining morbidity in people living with obesity. The contribution of B cells to immune dysregulation and meta-inflammation associated with obesity has been documented by studies over the past decade. With a focus on human studies, here we consolidate the observations demonstrating that there is altered B cell subset composition, differentiation, and function both systemically and in the adipose tissue of individuals living with obesity. Finally, we discuss the potential factors that drive B cell dysfunction in obesity and propose a model by which altered B cell subset composition in obesity underlies dysfunctional B cell responses to novel pathogens.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/metabolism , Obesity/complications , Obesity/metabolism , Inflammation , Adipose Tissue , Immunity
3.
Front Immunol ; 12: 611795, 2021.
Article in English | MEDLINE | ID: mdl-33995344

ABSTRACT

Regulatory B cells (Bregs) is a term that encompasses all B cells that act to suppress immune responses. Bregs contribute to the maintenance of tolerance, limiting ongoing immune responses and reestablishing immune homeostasis. The important role of Bregs in restraining the pathology associated with exacerbated inflammatory responses in autoimmunity and graft rejection has been consistently demonstrated, while more recent studies have suggested a role for this population in other immune-related conditions, such as infections, allergy, cancer, and chronic metabolic diseases. Initial studies identified IL-10 as the hallmark of Breg function; nevertheless, the past decade has seen the discovery of other molecules utilized by human and murine B cells to regulate immune responses. This new arsenal includes other anti-inflammatory cytokines such IL-35 and TGF-ß, as well as cell surface proteins like CD1d and PD-L1. In this review, we examine the main suppressive mechanisms employed by these novel Breg populations. We also discuss recent evidence that helps to unravel previously unknown aspects of the phenotype, development, activation, and function of IL-10-producing Bregs, incorporating an overview on those questions that remain obscure.


Subject(s)
B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Immunomodulation , Animals , B-Lymphocyte Subsets/metabolism , B-Lymphocytes, Regulatory/cytology , Biomarkers , Cell Differentiation , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
4.
Cell Rep ; 29(7): 1878-1892.e7, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31722204

ABSTRACT

Regulatory B cells (Bregs) play a critical role in the control of autoimmunity and inflammation. IL-10 production is the hallmark for the identification of Bregs. However, the molecular determinants that regulate the transcription of IL-10 and control the Breg developmental program remain unknown. Here, we demonstrate that aryl hydrocarbon receptor (AhR) regulates the differentiation and function of IL-10-producing CD19+CD21hiCD24hiBregs and limits their differentiation into B cells that contribute to inflammation. Chromatin profiling and transcriptome analyses show that loss of AhR in B cells reduces expression of IL-10 by skewing the differentiation of CD19+CD21hiCD24hiB cells into a pro-inflammatory program, under Breg-inducing conditions. B cell AhR-deficient mice develop exacerbated arthritis, show significant reductions in IL-10-producing Bregs and regulatory T cells, and show an increase in T helper (Th) 1 and Th17 cells compared with B cell AhR-sufficient mice. Thus, we identify AhR as a relevant contributor to the transcriptional regulation of Breg differentiation.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation/immunology , Interleukin-10/immunology , Receptors, Aryl Hydrocarbon/immunology , Transcription, Genetic/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B-Lymphocytes, Regulatory/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Interleukin-10/genetics , Mice , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology
5.
Nat Rev Nephrol ; 15(1): 11-26, 2019 01.
Article in English | MEDLINE | ID: mdl-30443016

ABSTRACT

B cells have a central role in many autoimmune diseases, including in those with renal involvement, as well as in the immunological response to kidney transplantation. The majority of B cell studies have focused on their pathological role as antibody producers. However, these cells have broad functions in immune responses beyond immunoglobulin secretion, including antigen presentation to T cells and cytokine production. Importantly, not all B cell subsets enhance immune responses. Regulatory B (Breg) cells attenuate inflammation and contribute to the maintenance of immune tolerance. Breg cells are numerically deficient and/or dysfunctional in several autoimmune diseases that can affect the kidneys, including systemic lupus erythematosus and anti-neutrophil cytoplasmic antibody-associated vasculitis, as well as in some groups of renal transplant recipients with alloimmune graft damage. B cell-targeting biologics have been trialled with promising results in diverse immune-mediated renal conditions. These therapies can affect both pro-inflammatory B cells and Breg cells, potentially limiting their long-term efficacy. Future strategies might involve the modulation of pro-inflammatory B cells in combination with the stimulation of regulatory subsets. Additionally, the monitoring of individual B cell subsets in patients may lead to the discovery of novel biomarkers that could help to predict disease relapse or progression.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes, Regulatory/immunology , Kidney Diseases/immunology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/metabolism , Autoimmune Diseases/therapy , Biological Factors/therapeutic use , Biomarkers/metabolism , Humans , Kidney Diseases/diagnosis , Kidney Diseases/metabolism , Kidney Diseases/therapy , Lymphocyte Depletion/methods
6.
Cell Rep ; 15(12): 2756-70, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27292648

ABSTRACT

The transcription factor T-bet directs Th1 cell differentiation, but the molecular mechanisms that underlie this lineage-specific gene regulation are not completely understood. Here, we show that T-bet acts through enhancers to allow the recruitment of Mediator and P-TEFb in the form of the super elongation complex (SEC). Th1 genes are occupied by H3K4me3 and RNA polymerase II in Th2 cells, while T-bet-mediated recruitment of P-TEFb in Th1 cells activates transcriptional elongation. P-TEFb is recruited to both genes and enhancers, where it activates enhancer RNA transcription. P-TEFb inhibition and Mediator and SEC knockdown selectively block activation of T-bet target genes, and P-TEFb inhibition abrogates Th1-associated experimental autoimmune uveitis. T-bet activity is independent of changes in NF-κB RelA and Brd4 binding, with T-bet- and NF-κB-mediated pathways instead converging to allow P-TEFb recruitment. These data provide insight into the mechanism through which lineage-specifying factors promote differentiation of alternative T cell fates.


Subject(s)
Gene Expression Regulation , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Transcription Elongation, Genetic , Animals , Cell Lineage/genetics , Enhancer Elements, Genetic/genetics , Humans , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/genetics , RNA/genetics , RNA/metabolism , Th2 Cells/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Uveitis/genetics
7.
Nat Med ; 20(11): 1334-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25326801

ABSTRACT

Regulatory B cells (Breg cells) differentiate in response to inflammation and subsequently restrain excessive immune responses via the release of interleukin-10 (IL-10). However, the precise inflammatory signals governing their differentiation remain to be elucidated. Here we show that the gut microbiota promotes the differentiation of Breg cells in the spleen as well as in the mesenteric lymph nodes. Perturbation of the gut microbiome imposed either by antibiotic treatment or by changes in the sterility of housing conditions reduces the number and function of Breg cells. Following the induction of arthritis, IL-1ß and IL-6 are produced only in conventionally housed mice and both cytokines directly promote Breg cell differentiation and IL-10 production. Mice lacking IL-6 receptor (IL-6R) or IL-1 receptor 1 (IL-1R1) specifically on B cells have a reduced number of IL-10-producing B cells and develop exacerbated arthritis compared to control animals. Thus, in response to inflammatory signals induced by both the gut flora and arthritis, Breg cells increase in number and restrain excessive inflammation.


Subject(s)
B-Lymphocytes, Regulatory/metabolism , Gastrointestinal Tract/microbiology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Microbiota , Animals , Anti-Bacterial Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes, Regulatory/drug effects , Cell Differentiation/drug effects , Flow Cytometry , Gastrointestinal Tract/drug effects , Inflammation/pathology , Interleukin-10/biosynthesis , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Mice, Inbred C57BL , Microbiota/drug effects , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL
...