scholarly journals Btk levels set the threshold for B-cell activation and negative selection of autoreactive B cells in mice

Blood ◽  
2012 ◽  
Vol 119 (16) ◽  
pp. 3744-3756 ◽  
Author(s):  
Laurens P. Kil ◽  
Marjolein J. W. de Bruijn ◽  
Menno van Nimwegen ◽  
Odilia B. J. Corneth ◽  
Jan Piet van Hamburg ◽  
...  

Abstract On antigen binding by the B-cell receptor (BCR), B cells up-regulate protein expression of the key downstream signaling molecule Bruton tyrosine kinase (Btk), but the effects of Btk up-regulation on B-cell function are unknown. Here, we show that transgenic mice overexpressing Btk specifically in B cells spontaneously formed germinal centers and manifested increased plasma cell numbers, leading to antinuclear autoantibody production and systemic lupus erythematosus (SLE)–like autoimmune pathology affecting kidneys, lungs, and salivary glands. Autoimmunity was fully dependent on Btk kinase activity, because Btk inhibitor treatment (PCI-32765) could normalize B-cell activation and differentiation, and because autoantibodies were absent in Btk transgenic mice overexpressing a kinase inactive Btk mutant. B cells overexpressing wild-type Btk were selectively hyperresponsive to BCR stimulation and showed enhanced Ca2+ influx, nuclear factor (NF)–κB activation, resistance to Fas-mediated apoptosis, and defective elimination of selfreactive B cells in vivo. These findings unravel a crucial role for Btk in setting the threshold for B-cell activation and counterselection of autoreactive B cells, making Btk an attractive therapeutic target in systemic autoimmune disease such as SLE. The finding of in vivo pathology associated with Btk overexpression may have important implications for the development of gene therapy strategies for X-linked agammaglobulinemia, the immunodeficiency associated with mutations in BTK.

1999 ◽  
Vol 190 (5) ◽  
pp. 639-650 ◽  
Author(s):  
Haowei Wang ◽  
Mark J. Shlomchik

In systemic autoimmune disease, self-tolerance fails, leading to autoantibody production. A central issue in immunology is to understand the origins of activated self-reactive B cells. We have used immunoglobulin (Ig) transgenic mice to investigate the regulation of autoreactive B cells with specificity for self-IgG2a (the rheumatoid factor [RF] specificity) to understand how normal mice regulate RF autoantibodies and how this fails in autoimmune mice. We previously showed that normal mice do not tolerize the AM14 RF clone, nor do they appear to activate it. Here we show that in Fas-deficient autoimmune mice, the picture is quite different. RF B cells are activated to divide and secrete, but only when the autoantigen is present. Thus, B cells that are ignored rather than anergized in normal mice can be stimulated to produce autoantibody in Fas-deficient mice. This demonstrates a novel developmental step at which intact Fas–Fas ligand signaling is required to regulate B cells in order to prevent autoimmunity. These data also establish the relevance of ignorant self-specific B cells to autoantibody production in disease and prove that in the case of the RF specificity, the nominal autoantigen IgG2a is the driving autoantigen in vivo.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. SCI-1-SCI-1
Author(s):  
Ann Marshak-Rothstein

Abstract Despite numerous mechanisms that exist to purge the B-cell repertoire of potentially dangerous autoreactive cells, it is now clear that numerous cells with the capability of binding self determinants survive negative selection and persist peripherally as anergic or ignorant cells. Exactly how these B cells are normally constrained, and then aberrantly activated in the context of autoimmunity, are two major questions in B-cell biology. A better understanding of the mechanisms involved in these processes could provide important insights to the regulation of alloreactivity. One clue to our understanding of autoimmunity comes from the nature of the autoantigens commonly targeted in systemic autoimmune diseases. Autoantigens often consist of macromolecular complexes that incorporate self-nucleic acids, and numerous in vitro studies have now shown that many of these canonical autoantigens are essentially potent endogenous autoadjuvants. Mice expressing a low affinity BCR specific for autologous IgG2a can be potently activated by DNA or RNA-associated immune complexes through a mechanism dependent on both the BCR and either TLR9 or TLR7, and serve as a useful prototype for autoreactive B cells in general. A number of groups have now tested this BCR/TLR paradigm in vivo. As expected, Tlr9-/- autoimmune-prone mice fail to produce autoantibodies directed against chromatin, while Tlr7-/- mice fail to produce autoantibodies directed against numerous RNA-associated proteins. However, the Tlr9-/- mice develop accelerated clinical disease, while the Tlr7-/- mice exhibit remarkably prolonged survival. We have now shown that BCR/TLR9 and BCR/TLR7 induce inherently different functional outcomes in B cells. Quite remarkably, both in vitro and in vivo, BCR/TLR7-dependent activation of autoreactive B cells leads to a more prolonged response and increased numbers of antibody producing cells. This response can be defined by a unique gene-expression profile and associated with proteins known to promote plasmablast differentiation. By contrast, BCR/TLR9 activation appears to initially limit autoreactive B-cell expansion; although in the context of systemic autoimmunity TLR9 is required for the production of DNA-reactive autoantibodies. Together these data indicate that the outcome BCR/TLR9 engagement of autoreactive B cells is highly dependent on environmental cues, and suggest that BCR/TLR7 B-cell activation is a key factor in the initiation of systemic lupus erythematosus and other systemic disorders. Disclosures: Marshak-Rothstein: Idera Pharmaceuticals: Consultancy; Abbvie: Consultancy; Genentech: Honoraria.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2578-2578
Author(s):  
Mrinmoy Sanyal ◽  
Rosemary Fernandez ◽  
Shoshana Levy

Abstract CD81 is a component of the CD19/CD21 coreceptor complex in B cells. This tetraspanin molecule was previously shown to enable membrane reorganization in B cells responding to complement-bound antigens. Here we stimulated B cells via their B cell receptor (BCR) and demonstrate that Cd81−/− B cells fluxed higher intracellular free calcium ion along with increased phosphorylation of PLCγ2 and Syk. The stimulated Cd81−/− B cells also proliferated faster and secreted higher amounts of antibodies. Moreover, activation of the TLR4 pathway in Cd81−/− B cells induced increased proliferation and antibody secretion. Furthermore, Cd81−/− mice mounted a significantly higher immune response to T-cell independent antigens than their wildtype counterparts. Finally, analysis of Cd81−/− B cells that were generated by bone marrow transplantation into Rag1−/− mice confirmed a cell intrinsic hyperactive phenotype. Taken together, these results indicate that CD81 plays a negative role in B cell activation in vitro and in vivo.


2021 ◽  
Author(s):  
Dillon G Patterson ◽  
Anna K Kania ◽  
Madeline J Price ◽  
James R Rose ◽  
Christopher D Scharer ◽  
...  

Cell division is an essential component of B cell differentiation to antibody-secreting plasma cells, with critical reprogramming occurring during the initial stages of B cell activation. However, a complete understanding of the factors that coordinate early reprogramming events in vivo remain to be determined. In this study, we examined the initial reprogramming by IRF4 in activated B cells using an adoptive transfer system and mice with a B cell-specific deletion of IRF4. IRF4-deficient B cells responding to influenza, NP-Ficoll and LPS divided, but stalled during the proliferative response. Gene expression profiling of IRF4-deficient B cells at discrete divisions revealed IRF4 was critical for inducing MYC target genes, oxidative phosphorylation, and glycolysis. Moreover, IRF4-deficient B cells maintained an inflammatory gene expression signature. Complementary chromatin accessibility analyses established a hierarchy of IRF4 activity and identified networks of dysregulated transcription factor families in IRF4-deficient B cells, including E-box binding bHLH family members. Indeed, B cells lacking IRF4 failed to fully induce Myc after stimulation and displayed aberrant cell cycle distribution. Furthermore, IRF4-deficient B cells showed reduced mTORC1 activity and failed to initiate the B cell-activation unfolded protein response and grow in cell size. Myc overexpression in IRF4-deficient was sufficient to overcome the cell growth defect. Together, these data reveal an IRF4-MYC-mTORC1 relationship critical for controlling cell growth and the proliferative response during B cell differentiation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 707-707 ◽  
Author(s):  
Melania Capasso ◽  
Mandeep K Bhamrah ◽  
Robert S Boyd ◽  
Kelvin Cain ◽  
Karen Pulford ◽  
...  

Abstract HVCN1 is a highly-conserved voltage-gated proton channel. Voltage-gated proton currents have been recorded in lymphocytes but their functions in B cells remain unknown. We isolated HVCN1 in a proteomic survey of plasma membrane proteins in mantle cell lymphoma (MCL) in leukemic phase. In normal lymphocytes, HVCN1 expression was restricted to the B-cell lineage; HVCN1 was highly expressed in mantle zone cells but down-regulated in germinal center (GC) cells undergoing receptor affinity maturation and class-switch recombination (CSR). Highest level expression was also observed in Chronic Lymphocytic Leukemia (CLL) cells from the peripheral blood. In MCL tumors, HVCN1 was expressed in circulating cells but absent from involved lymph nodes, whereas in diffuse large B cell lymphoma (DLBCL), its expression correlated with cases with a low proliferation index. Thus, in both primary and neoplastic B cells, HVCN1 expression appears to be associated with a non-proliferative phenotype. In human primary resting B cells and B cell lines, HVCN1 directly interacted with the B cell receptor (BCR) complex, as shown by Igβ and HVCN1 reciprocal immunoprecipitation experiments. We also found by confocal microscopy and subcellular fractionation, that upon BCR engagement the channel was internalized with the antigen receptor and the two proteins co-migrated to the endo-lysosomal, MHC class II (MHC-II) containing compartments (MIICs). When overexpressed in a hen egg lysozyme (HEL)-specific B cell clone, LK35.2, HVCN1 showed a basal phosphorylation which increased with HEL stimulation. The increased phosphorylation corresponded to an increase in proton conductance, termed “enhanced gating mode” and it was PKC dependent. We then asked whether HVCN1 over-expression could influence MHC II antigen presentation and if the effect could be mediated by changes in MIICs pH. Indeed, presentation of HEL peptides to a T cell clone was impaired in LK35.2 and A20 D1.3 cells, where HVCN1 had been re-introduced; effect was stronger for plate-bound antigen than for soluble antigen. The reduced antigen presentation was accompanied by an increase in endo-lysosomal pH, from pH4.9 ± 0.2 to 6.3 ± 0.1 (which may reflect HVCN1 channel-mediated proton flux out of the organelles), as measured with an anti-IgM antibody conjugated to a pH sensitive dye in HVCN1 over-expressing cells. Evidently, the presence of HVCN1 leads to increased endo-lysosomal pH, consistent with H+ current from the lysosomal compartment into the cytosol. Hence, active antigen presenting cells, like GC cells, might down-regulate HVCN1 expression to maximize the effect of antigen presentation. In order to investigate the role of HVCN1 in vivo, we used a HVCN1-deficient mouse line generated by genetrap insertion. These mice showed no obvious changes in numbers or composition of B-cell subpopulations. Immunization of HVCN1-deficient mice with a T-dependent antigen resulted in a defect in CSR to all IgG subclasses, particularly marked for the IgG2b, whereas in contrast, no differences were observed in IgM secretion, suggesting a pivotal role for HVCN1 during antigen-driven B-cell activation and subsequent CSR. HVCN1 may influence B-cell activation through alteration of reactive oxygen species (ROS) as HVCN1-deficient B cells showed reduced ROS production following BCR activation, a sign of suboptimal NADPH oxidase activity. It has been postulated that proton channels are required to counterbalance the electrogenic activity of NADPH oxidase during ROS production. Our data suggest that this mechanism also occurs in vivo and shed new light on the role of ROS in B cell activation and downstream effects.


Cells ◽  
2021 ◽  
Vol 10 (5) ◽  
pp. 1190
Author(s):  
Carlo G. Bonasia ◽  
Wayel H. Abdulahad ◽  
Abraham Rutgers ◽  
Peter Heeringa ◽  
Nicolaas A. Bos

Autoreactive B cells are key drivers of pathogenic processes in autoimmune diseases by the production of autoantibodies, secretion of cytokines, and presentation of autoantigens to T cells. However, the mechanisms that underlie the development of autoreactive B cells are not well understood. Here, we review recent studies leveraging novel techniques to identify and characterize (auto)antigen-specific B cells. The insights gained from such studies pertaining to the mechanisms involved in the escape of tolerance checkpoints and the activation of autoreactive B cells are discussed. In addition, we briefly highlight potential therapeutic strategies to target and eliminate autoreactive B cells in autoimmune diseases.


2004 ◽  
Vol 200 (1) ◽  
pp. 115-122 ◽  
Author(s):  
Ling Lin ◽  
Andrea J. Gerth ◽  
Stanford L. Peng

B cell terminal differentiation involves development into an antibody-secreting plasma cell, reflecting the concerted activation of proplasma cell transcriptional regulators, such as Blimp-1, IRF-4, and Xbp-1. Here, we show that the microphthalmia-associated transcription factor (Mitf) is highly expressed in naive B cells, where it antagonizes the process of terminal differentiation through the repression of IRF-4. Defective Mitf activity results in spontaneous B cell activation, antibody secretion, and autoantibody production. Conversely, ectopic Mitf expression suppresses the expression of IRF-4, the plasma cell marker CD138, and antibody secretion. Thus, Mitf regulates B cell homeostasis by suppressing the antibody-secreting fate.


2016 ◽  
Vol 22 (5) ◽  
pp. 307-315 ◽  
Author(s):  
Mingfang Lu ◽  
Robert Munford

Gram-negative bacterial LPS induce murine B-cell activation and innate (polyclonal) Ab production. Mouse B cells express the LPS signaling receptor (TLR4), yet how LPS activates B-cell responses in vivo is not known. Can LPS directly stimulate B cells to induce innate Ab production? Is activation of non-B cells also required? To address these questions, we transfused LPS-responsive ( Tlr4+/+) or non-responsive ( Tlr4−/−) B cells into LPS-responsive or non-responsive mice. Increased expression of the early activation markers CD69 and CD86 could be induced on transfused Tlr4−/− B cells by injecting LPS subcutaneously into Tlr4+/+ mice, demonstrating indirect activation of B cells by TLR4-responsive non-B cells in vivo, but the Tlr4−/− B cells did not increase serum IgM levels. In contrast, when Tlr4−/− recipients were transfused with Tlr4+/+ B cells, LPS induced large amounts of serum IgM and LPS could also enhance specific Ab production to a protein that was co-injected with it (adjuvant response). Thus, LPS-exposed non-B cells mediated increased surface expression of early B-cell activation markers, but this response did not predict innate Ab responses or LPS adjuvanticity in vivo. Direct stimulation of B cells by LPS via TLR4 was necessary and sufficient to induce B cells to produce Ab in vivo.


2021 ◽  
Author(s):  
Ashley N. Barlev ◽  
Susan Malkiel ◽  
Annemarie L. Dorjée ◽  
Jolien Suurmond ◽  
Betty Diamond

AbstractFcγRIIB is an inhibitory receptor expressed throughout B cell development. Diminished expression or function is associated with lupus in mice and humans, in particular through an effect on autoantibody production and plasma cell differentiation. Here, we analysed the effect of B cell-intrinsic FcγRIIB expression on B cell activation and plasma cell differentiation.Loss of FcγRIIB on B cells (Fcgr2b cKO mice) led to a spontaneous increase in autoantibody titers. This increase was most striking for IgG3, suggestive of increased extrafollicular responses. Marginal zone (MZ) and IgG3+ B cells had the highest expression of FcγRIIB and the increase in serum IgG3 was linked to increased MZ B cell signaling and activation in the absence of FcγRIIB. Likewise, human circulating MZ-like B cells had the highest expression of FcγRIIB, and their activation was most strongly inhibited by engaging FcγRIIB. Finally, marked increases in IgG3+ plasma cells and B cells were observed during extrafollicular plasma cell responses with both T-dependent and T-independent antigens in Fcgr2b cKO mice. The increased IgG3 response following immunization of Fcgr2b cKO mice was lost in MZ-deficient Notch2/Fcgr2b cKO mice.Thus, we present a model where high FcγRIIB expression in MZ B cells prevents their hyperactivation and ensuing autoimmunity.Graphical abstract


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1719-1719
Author(s):  
Mrinmoy Sanyal ◽  
Tsipi Shoham ◽  
Rosemary Fernandez ◽  
Shoshana Levy

Abstract The tetraspanin CD81 is required for numerous biological functions including fertilization, infection, cell migration and cellular interactions in the nervous and immune systems. In B cells CD81 is a component of the CD19/CD21 signaling complex. CD81 was shown to facilitate the redistribution of the B cell receptor (BCR) complex and CD21 into lipid rafts in response to co-engagement, and to modulate BCR signaling. In addition, CD81-deficient mice express low levels of cell surface CD19, thereby potentially altering signaling by the CD19/CD21 co-receptor complex. Interestingly, the onset of CD81 expression coincides with the onset of CD19 expression during B cell development. The foregoing observations suggest that CD81 might reduce the in vivo response of B cells to antigenic stimulation. To test this hypothesis we compared the response of CD81-deficient and wild type mice to T-independent (TNP-LPS) and T-dependent (TNP-KLH) antigens. Surprisingly, CD81-deficient mice mounted significantly higher IgM responses against both types of antigens. Moreover, the IgG response of CD81-deficient mice was stronger and persistent in response to T-independent antigen. We further found that CD81-deficient mice have an increase in bone marrow perisinusoidal B cells (IgM+IgD+). These cells are primarily responsible for mounting T-independent immune responses against blood-borne pathogens. In addition, CD81-deficient spleenic B cells have an intrinsic ability to produce higher amounts of IgM. These surprising results suggest that CD81 is involved in modulating B cell activation, particularly in response to infection.


Sign in / Sign up

Export Citation Format

Share Document