Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(94): eadg7549, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38640252

ABSTRACT

Vedolizumab (VDZ) is a first-line treatment in ulcerative colitis (UC) that targets the α4ß7- mucosal vascular addressin cell adhesion molecule 1 (MAdCAM-1) axis. To determine the mechanisms of action of VDZ, we examined five distinct cohorts of patients with UC. A decrease in naïve B and T cells in the intestines and gut-homing (ß7+) plasmablasts in circulation of VDZ-treated patients suggested that VDZ targets gut-associated lymphoid tissue (GALT). Anti-α4ß7 blockade in wild-type and photoconvertible (KikGR) mice confirmed a loss of GALT size and cellularity because of impaired cellular entry. In VDZ-treated patients with UC, treatment responders demonstrated reduced intestinal lymphoid aggregate size and follicle organization and a reduction of ß7+IgG+ plasmablasts in circulation, as well as IgG+ plasma cells and FcγR-dependent signaling in the intestine. GALT targeting represents a previously unappreciated mechanism of action of α4ß7-targeted therapies, with major implications for this therapeutic paradigm in UC.


Subject(s)
Colitis, Ulcerative , Humans , Animals , Mice , Colitis, Ulcerative/drug therapy , Integrins , Intestinal Mucosa , Peyer's Patches , Immunoglobulin G/therapeutic use
2.
Immunity ; 56(10): 2373-2387.e8, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37714151

ABSTRACT

Immunoglobulin A (IgA) maintains commensal communities in the intestine while preventing dysbiosis. IgA generated against intestinal microbes assures the simultaneous binding to multiple, diverse commensal-derived antigens. However, the exact mechanisms by which B cells mount broadly reactive IgA to the gut microbiome remains elusive. Here, we have shown that IgA B cell receptor (BCR) is required for B cell fitness during the germinal center (GC) reaction in Peyer's patches (PPs) and for generation of gut-homing plasma cells (PCs). We demonstrate that IgA BCR drove heightened intracellular signaling in mouse and human B cells, and as a consequence, IgA+ B cells received stronger positive selection cues. Mechanistically, IgA BCR signaling offset Fas-mediated death, possibly rescuing low-affinity B cells to promote a broad humoral response to commensals. Our findings reveal an additional mechanism linking BCR signaling, B cell fate, and antibody production location, which have implications for how intestinal antigen recognition shapes humoral immunity.


Subject(s)
B-Lymphocytes , Peyer's Patches , Mice , Humans , Animals , Antigens/metabolism , Receptors, Antigen, B-Cell/metabolism , Immunoglobulin A , Intestinal Mucosa
3.
bioRxiv ; 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36711839

ABSTRACT

Targeting the α4ß7-MAdCAM-1 axis with vedolizumab (VDZ) is a front-line therapeutic paradigm in ulcerative colitis (UC). However, mechanism(s) of action (MOA) of VDZ remain relatively undefined. Here, we examined three distinct cohorts of patients with UC (n=83, n=60, and n=21), to determine the effect of VDZ on the mucosal and peripheral immune system. Transcriptomic studies with protein level validation were used to study drug MOA using conventional and transgenic murine models. We found a significant decrease in colonic and ileal naïve B and T cells and circulating gut-homing plasmablasts (ß7+) in VDZ-treated patients, pointing to gut-associated lymphoid tissue (GALT) targeting by VDZ. Murine Peyer's patches (PP) demonstrated a significant loss cellularity associated with reduction in follicular B cells, including a unique population of epithelium-associated B cells, following anti-α4ß7 antibody (mAb) administration. Photoconvertible (KikGR) mice unequivocally demonstrated impaired cellular entry into PPs in anti-α4ß7 mAb treated mice. In VDZ-treated, but not anti-tumor necrosis factor-treated UC patients, lymphoid aggregate size was significantly reduced in treatment responders compared to non-responders, with an independent validation cohort further confirming these data. GALT targeting represents a novel MOA of α4ß7-targeted therapies, with major implications for this therapeutic paradigm in UC, and for the development of new therapeutic strategies.

4.
Immunity ; 54(10): 2273-2287.e6, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644558

ABSTRACT

Diets high in cholesterol alter intestinal immunity. Here, we examined how the cholesterol metabolite 25-hydroxycholesterol (25-HC) impacts the intestinal B cell response. Mice lacking cholesterol 25-hydroxylase (CH25H), the enzyme generating 25-HC, had higher frequencies of immunoglobulin A (IgA)-secreting antigen-specific B cells upon immunization or infection. 25-HC did not affect class-switch recombination but rather restrained plasma cell (PC) differentiation. 25-HC was produced by follicular dendritic cells and increased in response to dietary cholesterol. Mechanistically, 25-HC restricted activation of the sterol-sensing transcription factor SREBP2, thereby regulating B cell cholesterol biosynthesis. Ectopic expression of SREBP2 in germinal center B cells induced rapid PC differentiation, whereas SREBP2 deficiency reduced PC output in vitro and in vivo. High-cholesterol diet impaired, whereas Ch25h deficiency enhanced, the IgA response against Salmonella and the resulting protection from systemic bacterial dissemination. Thus, a 25-HC-SREBP2 axis shapes the humoral response at the intestinal barrier, providing insight into the effect of high dietary cholesterol in intestinal immunity.


Subject(s)
Cell Differentiation/immunology , Hydroxycholesterols/metabolism , Immunoglobulin A/immunology , Plasma Cells/immunology , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Cholesterol, Dietary/immunology , Cholesterol, Dietary/metabolism , Hydroxycholesterols/immunology , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Peyer's Patches/immunology , Peyer's Patches/metabolism , Plasma Cells/metabolism
5.
J Immunol ; 205(7): 1810-1818, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32859730

ABSTRACT

Systemic lupus erythematosus (SLE) is defined by loss of B cell tolerance, resulting in production of autoantibodies against nucleic acids and other cellular Ags. Aberrant activation of TLRs by self-derived RNA and DNA is strongly associated with SLE in patients and in mouse models, but the mechanism by which TLR signaling to self-ligands is regulated remains poorly understood. In this study, we show that αv integrin plays a critical role in regulating B cell TLR signaling to self-antigens in mice. We show that deletion of αv from B cells accelerates autoantibody production and autoimmune kidney disease in the Tlr7.1 transgenic mouse model of SLE. Increased autoimmunity was associated with specific expansion of transitional B cells, extrafollicular IgG2c-producing plasma cells, and activation of CD4 and CD8 T cells. Our data show that αv-mediated regulation of TLR signaling in B cells is critical for preventing autoimmunity and indicate that loss of αv promotes escape from tolerance. Thus, we identify a new regulatory pathway in autoimmunity and elucidate upstream signals that adjust B cell activation to prevent development of autoimmunity in a mouse model.


Subject(s)
B-Lymphocytes/physiology , Integrin alphaV/metabolism , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/metabolism , Toll-Like Receptor 7/metabolism , Animals , Autoantibodies/metabolism , Autoimmunity , Cells, Cultured , Disease Models, Animal , Humans , Immunoglobulin G/metabolism , Immunomodulation , Integrin alphaV/genetics , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Toll-Like Receptor 7/genetics
6.
J Exp Med ; 216(11): 2531-2545, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31506281

ABSTRACT

B cells in germinal centers (GCs) cycle between light zone (LZ) and dark zone (DZ). The cues in the GC microenvironment that regulate the transition from LZ to DZ have not been well characterized. In Peyer's patches (PPs), transforming growth factor-ß (TGFß) promotes IgA induction in activated B cells that can then differentiate into GC B cells. We show here that TGFß signaling occurs in B cells in GCs and is distinct from signaling that occurs in activated B cells in PPs. Whereas in activated B cells TGFß signaling is required for IgA induction, in the GC it was instead required for the transition from LZ to DZ. In the absence of TGFß signaling, there was an accumulation of LZ GC B cells and reduced antibody affinity maturation likely due to reduced activation of Foxo1. This work identifies TGFß as a microenvironmental cue that is critical for GC homeostasis and function.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Peyer's Patches/immunology , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Animals , B-Lymphocytes/metabolism , Forkhead Box Protein O1/immunology , Forkhead Box Protein O1/metabolism , Germinal Center/cytology , Germinal Center/metabolism , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peyer's Patches/cytology , Peyer's Patches/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/immunology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Receptors, CCR6/metabolism , Transforming Growth Factor beta/metabolism
7.
J Clin Invest ; 128(9): 4163-4178, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29999501

ABSTRACT

Germinal centers (GCs) are major sites of clonal B cell expansion and generation of long-lived, high-affinity antibody responses to pathogens. Signaling through TLRs on B cells promotes many aspects of GC B cell responses, including affinity maturation, class switching, and differentiation into long-lived memory and plasma cells. A major challenge for effective vaccination is identifying strategies to specifically promote GC B cell responses. Here, we have identified a mechanism of regulation of GC B cell TLR signaling, mediated by αv integrins and noncanonical autophagy. Using B cell-specific αv-KO mice, we show that loss of αv-mediated TLR regulation increased GC B cell expansion, somatic hypermutation, class switching, and generation of long-lived plasma cells after immunization with virus-like particles (VLPs) or antigens associated with TLR ligand adjuvants. Furthermore, targeting αv-mediated regulation increased the magnitude and breadth of antibody responses to influenza virus vaccination. These data therefore identify a mechanism of regulation of GC B cells that can be targeted to enhance antibody responses to vaccination.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Integrin alphaV/immunology , Animals , Autophagy/immunology , Female , Germinal Center/cytology , Immunization , Immunoglobulin Class Switching , Immunoglobulin G/blood , Immunologic Memory , Influenza A virus/immunology , Integrin alphaV/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Signal Transduction/immunology , Somatic Hypermutation, Immunoglobulin , Toll-Like Receptors/immunology , Vaccines, Virus-Like Particle/immunology
8.
PLoS Pathog ; 14(5): e1007060, 2018 02.
Article in English | MEDLINE | ID: mdl-29782535

ABSTRACT

Containment of Mycobacterium tuberculosis (Mtb) infection requires T cell recognition of infected macrophages. Mtb has evolved to tolerate, evade, and subvert host immunity. Despite a vigorous and sustained CD8+ T cell response during Mtb infection, CD8+ T cells make limited contribution to protection. Here, we ask whether the ability of Mtb-specific T cells to restrict Mtb growth is related to their capacity to recognize Mtb-infected macrophages. We derived CD8+ T cell lines that recognized the Mtb immunodominant epitope TB10.44-11 and compared them to CD4+ T cell lines that recognized Ag85b240-254 or ESAT63-17. While the CD4+ T cells recognized Mtb-infected macrophages and inhibited Mtb growth in vitro, the TB10.4-specific CD8+ T cells neither recognized Mtb-infected macrophages nor restricted Mtb growth. TB10.4-specific CD8+ T cells recognized macrophages infected with Listeria monocytogenes expressing TB10.4. However, over-expression of TB10.4 in Mtb did not confer recognition by TB10.4-specific CD8+ T cells. CD8+ T cells recognized macrophages pulsed with irradiated Mtb, indicating that macrophages can efficiently cross-present the TB10.4 protein and raising the possibility that viable bacilli might suppress cross-presentation. Importantly, polyclonal CD8+ T cells specific for Mtb antigens other than TB10.4 recognized Mtb-infected macrophages in a MHC-restricted manner. As TB10.4 elicits a dominant CD8+ T cell response that poorly recognizes Mtb-infected macrophages, we propose that TB10.4 acts as a decoy antigen. Moreover, it appears that this response overshadows subdominant CD8+ T cell response that can recognize Mtb-infected macrophages. The ability of Mtb to subvert the CD8+ T cell response may explain why CD8+ T cells make a disproportionately small contribution to host defense compared to CD4+ T cells. The selection of Mtb antigens for vaccines has focused on antigens that generate immunodominant responses. We propose that establishing whether vaccine-elicited, Mtb-specific T cells recognize Mtb-infected macrophages could be a useful criterion for preclinical vaccine development.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Macrophages, Peritoneal/microbiology , Mycobacterium tuberculosis/growth & development , Tuberculosis, Pulmonary/immunology , Animals , Antigens, Bacterial/immunology , Blotting, Western , Cell Line , Flow Cytometry , Listeria/physiology , Lung/cytology , Lung/microbiology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/radiation effects , Thioglycolates/pharmacology , Tuberculosis, Pulmonary/microbiology
9.
Nat Commun ; 7: 10917, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26965188

ABSTRACT

Integrin signalling triggers cytoskeletal rearrangements, including endocytosis and exocytosis of integrins and other membrane proteins. In addition to recycling integrins, this trafficking can also regulate intracellular signalling pathways. Here we describe a role for αv integrins in regulating Toll-like receptor (TLR) signalling by modulating intracellular trafficking. We show that deletion of αv or ß3 causes increased B-cell responses to TLR stimulation in vitro, and αv-conditional knockout mice have elevated antibody responses to TLR-ligand-associated antigens. αv regulates TLR signalling by promoting recruitment of the autophagy component LC3 (microtubule-associated proteins 1 light chain 3) to TLR-containing endosomes, which is essential for progression from NF-κB to IRF signalling, and ultimately for traffic to lysosomes where signalling is terminated. Disruption of LC3 recruitment leads to prolonged NF-κB signalling and increased B-cell proliferation and antibody production. This work identifies a previously unrecognized role for αv and the autophagy components LC3 and atg5 in regulating TLR signalling and B-cell immunity.


Subject(s)
B-Lymphocytes/immunology , Integrin alphaV/immunology , Microtubule-Associated Proteins/immunology , Protein Transport/immunology , Toll-Like Receptors/immunology , Animals , Autophagy , Autophagy-Related Protein 5 , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , In Vitro Techniques , Integrin alphaV/genetics , Integrin beta3/genetics , Mice , Mice, Knockout , Microscopy, Confocal , Signal Transduction/immunology
10.
Org Lett ; 16(8): 2122-5, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24684381

ABSTRACT

OvoA in ovothiol biosynthesis is a mononuclear non-heme iron enzyme catalyzing the oxidative coupling between histidine and cysteine. It can also catalyze the oxidative coupling between hercynine and cysteine, yet with a different regio-selectivity. Due to the potential application of this reaction for industrial ergothioneine production, in this study, we systematically characterized OvoA by a combination of three different assays. Our studies revealed that OvoA can also catalyze the oxidation of cysteine to either cysteine sulfinic acid or cystine. Remarkably, these OvoA-catalyzed reactions can be systematically modulated by a slight modification of one of its substrates, histidine.


Subject(s)
Cysteine/analogs & derivatives , Methylhistidines/metabolism , Mycobacterium smegmatis/metabolism , Betaine/analogs & derivatives , Betaine/metabolism , Catalysis , Cysteine/chemistry , Cysteine/metabolism , Cysteine Dioxygenase/metabolism , Heme/metabolism , Histidine/analogs & derivatives , Histidine/metabolism , Humans , Models, Biological , Molecular Structure , Mycobacterium smegmatis/enzymology , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...