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










Publication year range
1.
J Immunol ; 201(2): 371-382, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29866702

ABSTRACT

Lysosomes maintain immune homeostasis through the degradation of phagocytosed apoptotic debris; however, the signaling events regulating lysosomal maturation remain undefined. In this study, we show that lysosome acidification, key to the maturation process, relies on mTOR complex 2 (mTORC2), activation of caspase-1, and cleavage of Rab39a. Mechanistically, the localization of cofilin to the phagosome recruits caspase-11, which results in the localized activation of caspase-1. Caspase-1 subsequently cleaves Rab39a on the phagosomal membrane, promoting lysosome acidification. Although caspase-1 is critical for lysosome acidification, its activation is independent of inflammasomes and cell death mediated by apoptosis-associated speck-like protein containing a caspase recruitment domain, revealing a role beyond pyroptosis. In lupus-prone murine macrophages, chronic mTORC2 activity decouples the signaling pathway, leaving Rab39a intact. As a result, the lysosome does not acidify, and degradation is impaired, thereby heightening the burden of immune complexes that activate FcγRI and sustain mTORC2 activity. This feedforward loop promotes chronic immune activation, leading to multiple lupus-associated pathologies. In summary, these findings identify the key molecules in a previously unappreciated signaling pathway that promote lysosome acidification. It also shows that this pathway is disrupted in systemic lupus erythematosus.


Subject(s)
Caspase 1/metabolism , Lupus Erythematosus, Systemic/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Antigen-Antibody Complex/metabolism , Apoptosis/physiology , Homeostasis/physiology , Inflammasomes/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phagocytosis/physiology , Phagosomes/metabolism , Pyroptosis/physiology , Signal Transduction/physiology
2.
J Immunol ; 198(7): 2602-2611, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28235864

ABSTRACT

Tissue-specific immune responses play an important role in the pathology of autoimmune diseases. In systemic lupus erythematosus, deposits of IgG-immune complexes and the activation of complement in the kidney have long been thought to promote inflammation and lupus nephritis. However, the events that localize cells in non-lymphoid tertiary organs and sustain tissue-specific immune responses remain undefined. In this manuscript, we show that BAFF promotes events leading to lupus nephritis. Using an inducible model of systemic lupus erythematosus, we found that passive transfer of antinucleosome IgG into AID-/-MRL/lpr mice elevated autoantibody levels and promoted lupus nephritis by inducing BAFF production in the kidneys, and the formation of renal tertiary lymphoid structures (TLSs). Reducing BAFF in vivo prevented the formation of TLSs and lupus nephritis; however, it did not reduce immune cell infiltrates, or the deposits of IgG and complement in the kidney. Mechanistically, lowering BAFF levels also diminished the number of T cells positioned inside the glomeruli and reduced inflammation. Thus, BAFF plays a previously unappreciated role in lupus nephritis by inducing renal TLSs and regulating the position of T cells within the glomeruli.


Subject(s)
B-Cell Activating Factor/immunology , Kidney Glomerulus/immunology , Lupus Nephritis/immunology , Tertiary Lymphoid Structures/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred MRL lpr
3.
J Immunol ; 198(3): 1263-1273, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28031339

ABSTRACT

Infection with Staphylococcus aureus does not induce long-lived protective immunity for reasons that are not completely understood. Human and murine vaccine studies support a role for Abs in protecting against recurring infections, but S. aureus modulates the B cell response through expression of staphylococcus protein A (SpA), a surface protein that drives polyclonal B cell expansion and induces cell death in the absence of costimulation. In this murine study, we show that SpA altered the fate of plasmablasts and plasma cells (PCs) by enhancing the short-lived extrafollicular response and reducing the pool of bone marrow (BM)-resident long-lived PCs. The absence of long-lived PCs was associated with a rapid decline in Ag-specific class-switched Ab. In contrast, when previously inoculated mice were challenged with an isogenic SpA-deficient S. aureus mutant, cells proliferated in the BM survival niches and sustained long-term Ab titers. The effects of SpA on PC fate were limited to the secondary response, because Ab levels and the formation of B cell memory occurred normally during the primary response in mice inoculated with wild-type or SpA-deficient S. aureus mutant. Thus, failure to establish long-term protective Ab titers against S. aureus was not a consequence of diminished formation of B cell memory; instead, SpA reduced the proliferative capacity of PCs that entered the BM, diminishing the number of cells in the long-lived pool.


Subject(s)
Plasma Cells/drug effects , Staphylococcal Protein A/pharmacology , Animals , Antibody-Producing Cells/immunology , Immunoglobulin G/biosynthesis , Immunologic Memory , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Spleen/immunology , Staphylococcus aureus/immunology
4.
J Immunol ; 196(10): 4030-9, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27059595

ABSTRACT

Apoptotic debris, autoantibody, and IgG-immune complexes (ICs) have long been implicated in the inflammation associated with systemic lupus erythematosus (SLE); however, it remains unclear whether they initiate immune-mediated events that promote disease. In this study, we show that PBMCs from SLE patients experiencing active disease, and hematopoietic cells from lupus-prone MRL/lpr and NZM2410 mice accumulate markedly elevated levels of surface-bound nuclear self-antigens. On dendritic cells (DCs) and macrophages (MFs), the self-antigens are part of IgG-ICs that promote FcγRI-mediated signal transduction. Accumulation of IgG-ICs is evident on ex vivo myeloid cells from MRL/lpr mice by 10 wk of age and steadily increases prior to lupus nephritis. IgG and FcγRI play a critical role in disease pathology. Passive transfer of pathogenic IgG into IgG-deficient MRL/lpr mice promotes the accumulation of IgG-ICs prior to significant B cell expansion, BAFF secretion, and lupus nephritis. In contrast, diminishing the burden IgG-ICs in MRL/lpr mice through deficiency in FcγRI markedly improves these lupus pathologies. Taken together, our findings reveal a previously unappreciated role for the cell surface accumulation of IgG-ICs in human and murine lupus.


Subject(s)
Apoptosis , Blood Cells/immunology , Dendritic Cells/immunology , Lupus Erythematosus, Systemic/immunology , Macrophages/immunology , Adult , Animals , Autoantigens/immunology , Autoantigens/metabolism , B-Cell Activating Factor/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Middle Aged , Receptors, IgG/genetics , Young Adult
5.
Proc Natl Acad Sci U S A ; 113(15): E2142-51, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27035940

ABSTRACT

Defects in clearing apoptotic debris disrupt tissue and immunological homeostasis, leading to autoimmune and inflammatory diseases. Herein, we report that macrophages from lupus-prone MRL/lpr mice have impaired lysosomal maturation, resulting in heightened ROS production and attenuated lysosomal acidification. Impaired lysosomal maturation diminishes the ability of lysosomes to degrade apoptotic debris contained within IgG-immune complexes (IgG-ICs) and promotes recycling and the accumulation of nuclear self-antigens at the membrane 72 h after internalization. Diminished degradation of IgG-ICs prolongs the intracellular residency of nucleic acids, leading to the activation of Toll-like receptors. It also promotes phagosomal membrane permeabilization, allowing dsDNA and IgG to leak into the cytosol and activate AIM2 and TRIM21. Collectively, these events promote the accumulation of nuclear antigens and activate innate sensors that drive IFNα production and heightened cell death. These data identify a previously unidentified defect in lysosomal maturation that provides a mechanism for the chronic activation of intracellular innate sensors in systemic lupus erythematosus.


Subject(s)
Lupus Erythematosus, Systemic/immunology , Lysosomes/immunology , Macrophages/immunology , Animals , Antigen-Antibody Complex/immunology , Cell Membrane Permeability , DNA/metabolism , DNA-Binding Proteins/immunology , Escherichia coli/immunology , Haptens , Hemocyanins/immunology , Immunity, Innate , Immunoglobulin G/immunology , Interferon-alpha/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Transgenic , Reactive Oxygen Species/metabolism , Ribonucleoproteins/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
6.
Clin Immunol ; 168: 16-24, 2016 07.
Article in English | MEDLINE | ID: mdl-27102764

ABSTRACT

The immunopathological events in the kidneys of lupus nephritis (LN) patients are poorly understood due in part to the difficulty in acquiring serial biopsies and the inherent limitations in their analysis. To identify a means to circumvent these limitations, we investigated whether immune cells of kidney origin are present in patient urine and whether they correlate with kidney pathology. Flow cytometry analysis was performed on peripheral blood and urine cells of 69 SLE patients, of whom 41 were LN patients. In addition, type I IFN (IFNα/ß) levels were determined in plasma and urine by bioassay. Approximately 60% of non-LN patients had urine lymphocytes. In these patients, T cells were always present and predominantly CD8(+), while B cells were either absent or a mixture of naïve and memory B cells. In contrast, >90% of LN patients had urine lymphocytes. In half, the B and T cells resembled those in non-LN patient urine; however, in the remaining patients, the B cells were exclusively Ig-secreting plasmablasts or plasma cells (PB/PCs) and the T cells were predominantly CD4(+). In addition, pDCs and IFNα/ß frequently accompanied PB/PCs. The majority of patients with urine PB/PCs presented with proliferative nephritis and a significant loss of kidney function, which in some cases had progressed to end stage renal disease (ESRD). In conclusion, urine can provide access to cells of kidney resident populations for phenotypic and functional characterization. Analysis of these cells provides insight into the kidney immunopathology and may serve as biomarkers to identify patients at risk for developing LN and progressing to ESRD.


Subject(s)
Immune System/cytology , Interferon Type I/immunology , Kidney/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Urine/cytology , Adult , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Female , Flow Cytometry , Humans , Immune System/immunology , Immunologic Memory/immunology , Interferon Type I/blood , Interferon Type I/urine , Interferon-alpha/blood , Interferon-alpha/immunology , Interferon-alpha/urine , Interferon-beta/blood , Interferon-beta/immunology , Interferon-beta/urine , Kidney/pathology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/urine , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/urine , Lupus Nephritis/blood , Lupus Nephritis/urine , Male , Plasma Cells/immunology
7.
J Immunol ; 196(1): 196-206, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26621863

ABSTRACT

Memory B cell responses are vital for protection against infections but must also be regulated to prevent autoimmunity. Cognate T cell help, somatic hypermutation, and affinity maturation within germinal centers (GCs) are required for high-affinity memory B cell formation; however, the signals that commit GC B cells to the memory pool remain unclear. In this study, we identify a role for IgG-immune complexes (ICs), FcγRs, and BAFF during the formation of memory B cells in mice. We found that early secretion of IgG in response to immunization with a T-dependent Ag leads to IC-FcγR interactions that induce dendritic cells to secrete BAFF, which acts at or upstream of Bcl-6 in activated B cells. Loss of CD16, hematopoietic cell-derived BAFF, or blocking IC:FcγR regions in vivo diminished the expression of Bcl-6, the frequency of GC and memory B cells, and secondary Ab responses. BAFF also contributed to the maintenance and/or expansion of the follicular helper T cell population, although it was dispensable for their formation. Thus, early Ab responses contribute to the optimal formation of B cell memory through IgG-ICs and BAFF. Our work defines a new role for FcγRs in GC and memory B cell responses.


Subject(s)
Antigen-Antibody Complex/immunology , B-Cell Activating Factor/biosynthesis , Immunoglobulin G/immunology , Immunologic Memory/immunology , Receptors, IgG/immunology , Adoptive Transfer , Animals , B-Cell Activating Factor/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/immunology , Germinal Center/cytology , Germinal Center/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/biosynthesis , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Receptors, IgG/genetics , T-Lymphocytes, Helper-Inducer/immunology
8.
J Immunol ; 189(8): 3859-68, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22984080

ABSTRACT

To maintain tolerance, autoreactive B cells must regulate signal transduction from the BCR and TLRs. We recently identified that dendritic cells and macrophages regulate autoreactive cells during TLR4 activation by releasing IL-6 and soluble CD40 ligand (sCD40L). These cytokines selectively repress Ab secretion from autoreactive, but not antigenically naive, B cells. How IL-6 and sCD40L repress autoantibody production is unknown. In this work, we show that IL-6 and sCD40L are required for low-affinity/avidity autoreactive B cells to maintain tolerance through a mechanism involving receptor cross-talk between the BCR, TLR4, and the IL-6R or CD40. We show that acute signaling through IL-6R or CD40 integrates with chronic BCR-mediated ERK activation to restrict p-ERK from the nucleus and represses TLR4-induced Blimp-1 and XBP-1 expression. Tolerance is disrupted in 2-12H/MRL/lpr mice where IL-6 and sCD40L fail to spatially restrict p-ERK and fail to repress TLR4-induced Ig secretion. In the case of CD40, acute signaling in B cells from 2-12H/MRL/lpr mice is intact, but the chronic activation of p-ERK emanating from the BCR is attenuated. Re-establishing chronically active ERK through retroviral expression of constitutively active MEK1 restores tolerance upon sCD40L, but not IL-6, stimulation, indicating that regulation by IL-6 requires another signaling effector. These data define the molecular basis for the regulation of low-affinity autoreactive B cells during TLR4 stimulation; they explain how autoreactive but not naive B cells are repressed by IL-6 and sCD40L; and they identify B cell defects in lupus-prone mice that lead to TLR4-induced autoantibody production.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lupus Nephritis/metabolism , Receptor Cross-Talk/immunology , Toll-Like Receptor 4/physiology , Animals , B-Lymphocyte Subsets/pathology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cells, Cultured , Coculture Techniques , Female , Immune Tolerance/genetics , Lupus Nephritis/enzymology , Lupus Nephritis/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Transgenic , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Transport/genetics , Protein Transport/immunology , Receptors, Antigen, B-Cell/physiology , Receptors, Interleukin-6/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
9.
J Immunol ; 189(2): 711-20, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22675201

ABSTRACT

The ability to induce Ab responses to pathogens while maintaining the quiescence of autoreactive cells is an important aspect of immune tolerance. During activation of TLR4, dendritic cells (DCs) and macrophages (MFs) repress autoantibody production through their secretion of IL-6 and soluble CD40L (sCD40L). These soluble mediators selectively repress B cells chronically exposed to Ag, but not naive cells, suggesting a means to maintain tolerance during TLR4 stimulation, yet allow immunity. In this study, we identify TNF-α as a third repressive factor, which together with IL-6 and CD40L account for nearly all the repression conferred by DCs and MFs. Similar to IL-6 and sCD40L, TNF-α did not alter B cell proliferation or survival. Instead, it reduced the number of Ab-secreting cells. To address whether the soluble mediators secreted by DCs and MFs functioned in vivo, we generated mice lacking IL-6, CD40L, and TNF-α. Compared to wild-type mice, these mice showed prolonged anti-nuclear Ab responses following TLR4 stimulation. Furthermore, adoptive transfer of autoreactive B cells into chimeric IL-6(-/-) × CD40L(-/-) × TNF-α(-/-) mice showed that preplasma cells secreted autoantibodies independent of germinal center formation or extrafollicular foci. These data indicate that in the absence of genetic predisposition to autoimmunity, loss of endogenous IL-6, CD40L, and TNF-α promotes autoantibody secretion during TLR4 stimulation.


Subject(s)
Autoantibodies/biosynthesis , Dendritic Cells/immunology , Immune Tolerance , Macrophages/immunology , Plasma Cells/immunology , Stem Cells/immunology , Adoptive Transfer , Animals , Antigens, Nuclear/genetics , Antigens, Nuclear/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD40 Ligand/deficiency , Cells, Cultured , Dendritic Cells/metabolism , Immune Tolerance/genetics , Interleukin-6/deficiency , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Mice, Transgenic , Plasma Cells/metabolism , Plasma Cells/transplantation , Radiation Chimera/immunology , Stem Cells/metabolism , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/deficiency
10.
J Immunol ; 184(12): 6637-48, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483720

ABSTRACT

The peritoneal cavity is recognized as an important site for autoreactive B cells prior to their transit to other immune tissues; however, little is known of the genes that may regulate this process. Mice lacking the receptor tyrosine kinase, Mertk, display a lupus-like autoimmune phenotype with splenomegaly and high autoantibodies titers. In this study, we investigate whether Mertk regulates the composition of peritoneal cells that favor an autoimmune phenotype. We found an increase in the number of macrophages, dendritic cells (DCs), plasmacytoid DCs, T cells, and B cells in the peritoneal cavity of mertk-/- mice when compared with wild-type mice. This disparity in cell numbers was not due to changes in cell proliferation or cell death. In adoptive transfer experiments, we showed an increase in migration of labeled donor cells into the mertk-/- peritoneal cavity. In addition, bone marrow chimeric mice showed hematopoietic-derived factors were also critical for T cell migration. Consistent with this migration and the increase in the number of cells, we identified elevated expression of CXCL9, its receptor CXCR3, and IL-7R on peritoneal cells from mertk-/- mice. To corroborate the migratory function of CXCR3 on cells, the depletion of CXCR3 donor cells significantly reduced the number of adoptively transferred cells that entered into the peritoneum of mertk-/- mice. This control of peritoneal cells numbers correlated with autoantibody production and was exclusively attributed to Mertk because mice lacking other family members, Axl or Tyro 3, did not display dysregulation in peritoneal cell numbers or the autoimmune phenotype.


Subject(s)
Autoimmunity/immunology , Cell Movement/immunology , Leukocytes/cytology , Peritoneal Cavity/cytology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Autoantibodies/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Separation , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Receptor Protein-Tyrosine Kinases/deficiency , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , c-Mer Tyrosine Kinase
11.
Immunol Res ; 41(3): 295-309, 2008.
Article in English | MEDLINE | ID: mdl-18704765

ABSTRACT

Systemic lupus erythematosus (SLE) highlights the dangers of dysregulated B cells and the importance of initiating and maintaining tolerance. In addition to central deletion, receptor editing, peripheral deletion, receptor revision, anergy, and indifference, we have described a new mechanism of B cell tolerance wherein dendritic cells (DCs) and macrophages (MPhis) regulate autoreactive B cells during innate immune responses. In part, DCs and MPhis repress autoreactive B cells by releasing IL-6 and soluble CD40L (sCD40L). This mechanism is selective in that IL-6 and sCD40L do not affect Ig secretion by naïve cells during innate immune responses, allowing immunity in the absence of autoimmunity. In lupus-prone mice, DCs and MPhis are defective in secretion of IL-6 and sCD40L and cannot effectively repress autoantibody secretion suggesting that defects in DC/MPhi-mediated tolerance may contribute to the autoimmune phenotype. Further, these studies suggest that reconstituting DCs and MPhis in SLE patients might restore regulation of autoreactive B cells and provide an alternative to immunosuppressive therapies.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , Macrophages/immunology , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , Autoantigens/immunology , Autoantigens/metabolism , Autoimmunity/immunology , B-Lymphocytes/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , Dendritic Cells/metabolism , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Lupus Erythematosus, Systemic/metabolism , Macrophages/metabolism , snRNP Core Proteins/immunology , snRNP Core Proteins/metabolism
12.
Int Immunol ; 19(12): 1403-12, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17981794

ABSTRACT

The B cell antigen receptor (BCR) delivers antigen to the endocytic compartment and transduces signals that regulate the stability of the receptor complex. Previous studies showed that BCR-mediated signal transduction dissociates micro-heavy chain (microm) from Ig-alpha/Ig-beta, facilitating the delivery of antigen to clathrin-coated vesicles (CCVs). Herein, we demonstrate that the dissociation of Ig-alpha/Ig-beta from microm requires tyrosine-587 of the microm transmembrane domain. Receptors expressing a mutation at tyrosine-587 (Y587F) transduced signals that were comparable to wild type, yet they failed to dissociate microm from Ig-alpha/Ig-beta. Further, receptors harboring the Y587F mutation failed to associate with CCVs, resulting in diminished antigen in the lysosome-associated membrane protein-1 (LAMP-1(+)) compartment and severely impaired antigen presentation, indicating that endocytosis through CCVs is required for antigen presentation. Thus, the transmembrane tyrosine of mum mediates destabilization of the BCR complex, facilitating antigen processing by promoting the association of antigen with CCVs.


Subject(s)
Antigen Presentation , B-Lymphocytes/immunology , Clathrin-Coated Vesicles/immunology , Immunoglobulin Heavy Chains/immunology , Receptors, Antigen, B-Cell/immunology , Tyrosine/metabolism , Animals , Antigens/immunology , B-Lymphocytes/cytology , B-Lymphocytes/ultrastructure , Cell Line , Clathrin-Coated Vesicles/genetics , Clathrin-Coated Vesicles/metabolism , Clathrin-Coated Vesicles/ultrastructure , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Lysosomal Membrane Proteins/analysis , Mice , Microscopy, Electron, Transmission , Mutation , Signal Transduction , Tyrosine/genetics , Tyrosine/immunology
13.
Blood ; 110(5): 1595-602, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17712049

ABSTRACT

Activation of the innate immune system promotes polyclonal antibody secretion to eliminate invading pathogens. Inherent in this process is the potential to activate autoreactive B cells and induce autoimmunity. We showed previously that TLR-stimulated dendritic cells and macrophages regulate B cell tolerance to Smith antigen, in part through the secretion of interleukin-6 (IL-6). In this manuscript, we show that neutralization of IL-6 fails to abrogate macrophage-mediated repression and identify soluble CD40 ligand (CD40L) as a second repressive factor secreted by macrophages. CD40L selectively repressed Ig secretion by chronically antigen-experienced (anergic) immunoglobulin transgenic and nontransgenic B cells but not by transiently stimulated B cells. The importance of macrophages in maintaining B cell tolerance was apparent in lupus-prone MRL/lpr mice. Compared with C57BL/6 mice, macrophages from MRL/lpr mice were significantly less efficient at repressing immunoglobulin secretion coincident with diminished IL-6 and CD40 ligand production. These data indicate that macrophages regulate autoreactive B cells by secreting repressive factors that prohibit terminal differentiation of B cells. The regulation of autoreactive B cells by macrophages is diminished in lupus-prone mice suggesting a role in autoimmunity.


Subject(s)
Autoimmunity , B-Lymphocytes/immunology , CD40 Ligand/immunology , Cell Differentiation/immunology , Clonal Anergy , Interleukin-6/immunology , Macrophages/immunology , Animals , Antibody Formation/drug effects , Antibody Formation/immunology , Autoantigens/immunology , Autoantigens/pharmacology , Autoimmunity/drug effects , Cell Differentiation/drug effects , Clonal Anergy/drug effects , Immunity, Innate/drug effects , Immunoglobulin G/immunology , Mice , Mice, Inbred MRL lpr , Mice, Knockout , Ribonucleoproteins, Small Nuclear/immunology , Ribonucleoproteins, Small Nuclear/pharmacology , snRNP Core Proteins
14.
Nat Rev Immunol ; 7(8): 633-43, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17641666

ABSTRACT

Anergy, a condition in which cells persist in the periphery but are unresponsive to antigen, is responsible for silencing many self-reactive B cells. Loss of anergy is known to contribute to the development of autoimmune diseases, including systemic lupus erythematosus and type 1 diabetes. Multiple transgenic mouse models have enabled the dissection of mechanisms that underlie anergy, and recently, anergic B cells have been identified in the periphery of wild-type mice. Heterogeneity of mechanistic concepts developed using model systems has complicated our understanding of anergy and its biological features. In this Review, we compare and contrast the salient features of anergic B cells with a view to developing unifying mechanistic hypotheses that explain their lifestyles.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Autoimmunity/immunology , Epitopes , Humans , Mice , Mice, Transgenic , Models, Animal , Signal Transduction
15.
J Immunol ; 178(8): 4803-10, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404261

ABSTRACT

Autoimmunity results from a breakdown in tolerance mechanisms that regulate autoreactive lymphocytes. We recently showed that during innate immune responses, secretion of IL-6 by dendritic cells (DCs) maintained autoreactive B cells in an unresponsive state. In this study, we describe that TLR4-activated DCs from lupus-prone mice are defective in repressing autoantibody secretion, coincident with diminished IL-6 secretion. Reduced secretion of IL-6 by MRL/lpr DCs reflected diminished synthesis and failure to sustain IL-6 mRNA production. This occurred coincident with lack of NF-kappaB and AP-1 DNA binding and failure to sustain IkappaBalpha phosphorylation. Analysis of individual mice showed that some animals partially repressed Ig secretion despite reduced levels of IL-6. This suggests that in addition to IL-6, DCs secrete other soluble factor(s) that regulate autoreactive B cells. Collectively, the data show that MRL/lpr mice are defective in DC/IL-6-mediated tolerance, but that some individuals maintain the ability to repress autoantibody secretion by an alternative mechanism.


Subject(s)
Dendritic Cells/physiology , Immunoglobulins/biosynthesis , Interleukin-6/physiology , Lupus Erythematosus, Systemic/immunology , Animals , Autoantibodies/biosynthesis , DNA/metabolism , Immune Tolerance , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Transgenic , NF-kappa B/metabolism , Receptors, Antigen, B-Cell/physiology , Toll-Like Receptor 4/physiology
16.
J Immunol ; 176(2): 790-802, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16393962

ABSTRACT

Ab-secreting plasma cells (PCs) are the effectors of humoral immunity. In this study, we describe regulation of autoreactive B cells specific for the ribonucleoprotein Smith (Sm) at an early pre-PC stage. These cells are defined by the expression of the PC marker CD138 and normal levels of CD19 and B220. They are present at a high frequency in normal mouse spleen and bone marrow, are Ag dependent, and are located predominantly along the T cell-B cell border and near bridging channels. Anti-Sm pre-PCs also occur at a high frequency in nonautoimmune mice and show additional phenotypic characteristics of PC differentiation. However, while some of these pre-PCs are Ab-secreting cells, those specific for Sm are not, indicating regulation. Consistent with this, anti-Sm pre-PCs have a higher turnover rate and higher frequency of cell death than those that do not bind Sm. Regulation of anti-Sm pre-PCs occurs upstream of the transcriptional repressor, B lymphocyte-induced maturation protein-1, expression. Regulation at this stage is overcome in autoimmune MRL/lpr mice and is accompanied by an altered B lymphocyte stimulator receptor profile. These data reveal a new B cell tolerance checkpoint that is overcome in autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Immune Tolerance , Plasma Cells/immunology , Animals , Apoptosis/immunology , Autoantigens , Autoimmunity/genetics , B-Lymphocytes/cytology , Base Sequence , Cell Differentiation , DNA/genetics , Immune Tolerance/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred MRL lpr , Mice, Transgenic , Plasma Cells/cytology , Positive Regulatory Domain I-Binding Factor 1 , Proteoglycans/metabolism , Receptors, Tumor Necrosis Factor/genetics , Repressor Proteins/genetics , Ribonucleoproteins, Small Nuclear/immunology , Spleen/cytology , Spleen/immunology , Syndecan-1 , Syndecans , Transcription Factors/genetics , Transcription, Genetic , snRNP Core Proteins
17.
J Immunol ; 175(1): 37-41, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972629

ABSTRACT

Polyclonal B cell activation promotes immunity without the loss of tolerance. Our data show that during activation of the innate immune system, B cell tolerance to Smith Ag Sm is maintained by dendritic cells (DCs) and macrophages (MPhi). TLR4-activated myeloid DCs and MPhi, but not plasmacytoid or lymphoid DCs, repressed autoreactive B cells through the secretion of soluble mediators, including IL-6. Although IL-6 promotes plasma cell differentiation of B cells acutely stimulated by Ag, we show that it repressed cells that were chronically exposed to self-Ag. This mechanism of tolerance was not limited to Smith Ag-specific B cells as hen egg lysozyme- and p-azophenylarsonate-specific B cells were similarly affected. Our data define a tolerogenic role for MPhi and DCs in regulating autoreactive B cells during activation of the innate immune system.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Macrophages/immunology , Ribonucleoproteins, Small Nuclear/immunology , Animals , Autoantigens/immunology , Autoimmunity , Immune Tolerance , Immunity, Innate , In Vitro Techniques , Interleukin-6/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muramidase/immunology , snRNP Core Proteins
18.
J Immunol ; 175(1): 147-54, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972641

ABSTRACT

The BCR relays extracellular signals and internalizes Ag for processing and presentation. We have previously demonstrated that ligation of the BCR destabilizes Ig-alpha/Ig-beta (Ig-alphabeta) from mu-H chain (mum). In this study we report that receptor destabilization represents a physical separation of mum from Ig-alphabeta. Sucrose gradient fractionation localized Ig-alphabeta to G(M1)-containing lipid microdomains in the absence of mum. Confocal and electron microscopy studies revealed the colocalization of unsheathed mum with clathrin-coated vesicles. Furthermore, mum failed to associate with clathrin-coated vesicles when receptor destabilization was inhibited, suggesting that unsheathing of mum is required for clathrin-mediated endocytosis. In summary, we found that Ag stimulation physically separates Ig-alphabeta from mum, facilitating concomitant signal transduction and Ag delivery to the endocytic compartment.


Subject(s)
Immunoglobulin Heavy Chains/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Antigen Presentation , Biological Transport, Active , Cell Line , Clathrin-Coated Vesicles/immunology , Clathrin-Coated Vesicles/metabolism , Endocytosis , G(M1) Ganglioside/metabolism , Immunoglobulin alpha-Chains/metabolism , Immunoglobulin mu-Chains/metabolism , Kinetics , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Microscopy, Electron , Signal Transduction
19.
J Immunol ; 171(9): 4493-503, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14568922

ABSTRACT

CD8 engagement is believed to be a critical event in the activation of naive T cells. In this communication, we address the effects of peptide-MHC (pMHC)/TCR affinity on the necessity of CD8 engagement in T cell activation of primary naive cells. Using two peptides with different measured avidities for the same pMHC-TCR complex, we compared biochemical affinity of pMHC/TCR and the cell surface binding avidity of pMHC/TCR with and without CD8 engagement. We compared early signaling events and later functional activity of naive T cells in the same manner. Although early signaling events are altered, we find that high-affinity pMHC/TCR interactions can overcome the need for CD8 engagement for proliferation and CTL function. An integrated signal over time allows T cell activation with a high-affinity ligand in the absence of CD8 engagement.


Subject(s)
CD8 Antigens/metabolism , H-2 Antigens/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Receptor Cross-Talk/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , Antigens, Viral/immunology , Aspartic Acid/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COS Cells , Chlorocebus aethiops , Cytokines/metabolism , Cytotoxicity, Immunologic , Glycoproteins/immunology , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Ligands , Lymphocyte Activation/genetics , Lymphocytic choriomeningitis virus/immunology , Lysine/genetics , Membrane Microdomains/genetics , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Receptors, Antigen, T-Cell/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Viral Proteins/immunology
20.
J Immunol ; 168(9): 4344-51, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11970976

ABSTRACT

B cell tolerance can be maintained by functional inactivation, or anergy, wherein B cell Ag receptors (BCR) remain capable of binding Ag, but are unable to transduce signals. Although the molecular mechanisms underlying this unresponsiveness are unknown, some models of B cell anergy are characterized by disruption of proximal BCR signaling events, and by destabilization of the BCR complex. Receptor destabilization is manifest by a reduced ability to coimmunoprecipitate membrane Ig with the Ig-alpha/Ig-beta signal-transducing complex. To begin to explore the possibility that anergy is the consequence of receptor destabilization, we analyzed a panel of B lymphoma transfectants expressing constant amounts of signal-competent Ag receptors and varied amounts of a receptor with identical specificity, but bearing mutations that render it incapable of interacting with Ig-alpha/Ig-beta. This analysis revealed that coaggregation of signal-incompetent receptors prevented Ag-induced Ig-alpha and Syk phosphorylation, mobilization of Ca(2+), and the up-regulation of CD69 mediated by competent receptors. In contrast, Ag-induced Cbl and Erk phosphorylation were unaffected. Data indicate that coaggregation of destabilized receptors (as few as approximately 15% of total) with signal-competent receptors significantly affects the ability of competent receptors to transduce signals. Thus, BCR destabilization may underlie the Ag unresponsiveness of anergic B cells.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy , Receptors, Antigen, B-Cell/immunology , Signal Transduction , Ubiquitin-Protein Ligases , Animals , Antigens/immunology , Antigens, CD/biosynthesis , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/biosynthesis , CD79 Antigens , Calcium/metabolism , Cells, Cultured , Enzyme Precursors/metabolism , Immunoglobulin D/immunology , Immunoglobulin M/immunology , Intracellular Signaling Peptides and Proteins , Lectins, C-Type , Lymphoma, B-Cell , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl , Receptor Aggregation , Receptors, Antigen, B-Cell/metabolism , Syk Kinase , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...