Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Front Immunol ; 10: 2575, 2019.
Article in English | MEDLINE | ID: mdl-31781099

ABSTRACT

Hyper-inflammation during acute phase and sequential hypo-inflammation during immunosuppressive phase in macrophages/monocytes lead to multiorgan failure syndrome and immune collapse of sepsis, in which toll-like receptor (TLR)-triggered inflammatory responses play a major role. Here, we reported that Siglecg deficiency attenuated TLR4-triggered pro-inflammatory cytokine production and increased anti-inflammatory cytokine [interleukin-10 [IL-10]] production in vivo and in vitro at both acute and immunosuppressive phases. Siglecg deficiency also protected mice from lipopolysaccharide (LPS)-induced sepsis with less inflammation in the lung and less tissue destruction in the spleen. Siglec-G inhibited proto-oncogene tyrosine-protein kinase Src (Src) activation via recruiting and activating tyrosine phosphatase Src homology region 2 domain-containing phosphatase-1 (SHP1) through immunoreceptor tyrosine-based inhibitory motif (ITIM) domain. Src could inhibit TLR4-induced inflammatory cytokines and promote anti-inflammatory cytokine IL-10. Mechanical investigation showed that Src could interact with and phosphorylate STAT3. Src could also promote HIF1α degradation through activating GSK3ß. Our study reveals that Siglec-G orchestrates TLR-induced inflammation, which outlines that blocking Siglec-G or activating Src may be a promising strategy for both acute and chronic inflammatory diseases.


Subject(s)
Inflammation/immunology , Lectins/deficiency , Receptors, Antigen, B-Cell/deficiency , Sepsis/immunology , src-Family Kinases/metabolism , Animals , Cytokines/metabolism , Enzyme Activation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-10/metabolism , Lectins/physiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Receptors, Antigen, B-Cell/physiology , SH2 Domain-Containing Protein Tyrosine Phosphatases/metabolism , STAT3 Transcription Factor/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins , Signal Transduction , Toll-Like Receptors/metabolism
2.
Nature ; 546(7657): 302-306, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28562582

ABSTRACT

Similar to resting mature B cells, where the B-cell antigen receptor (BCR) controls cellular survival, surface BCR expression is conserved in most mature B-cell lymphomas. The identification of activating BCR mutations and the growth disadvantage upon BCR knockdown of cells of certain lymphoma entities has led to the view that BCR signalling is required for tumour cell survival. Consequently, the BCR signalling machinery has become an established target in the therapy of B-cell malignancies. Here we study the effects of BCR ablation on MYC-driven mouse B-cell lymphomas and compare them with observations in human Burkitt lymphoma. Whereas BCR ablation does not, per se, significantly affect lymphoma growth, BCR-negative (BCR-) tumour cells rapidly disappear in the presence of their BCR-expressing (BCR+) counterparts in vitro and in vivo. This requires neither cellular contact nor factors released by BCR+ tumour cells. Instead, BCR loss induces the rewiring of central carbon metabolism, increasing the sensitivity of receptor-less lymphoma cells to nutrient restriction. The BCR attenuates glycogen synthase kinase 3 beta (GSK3ß) activity to support MYC-controlled gene expression. BCR- tumour cells exhibit increased GSK3ß activity and are rescued from their competitive growth disadvantage by GSK3ß inhibition. BCR- lymphoma variants that restore competitive fitness normalize GSK3ß activity after constitutive activation of the MAPK pathway, commonly through Ras mutations. Similarly, in Burkitt lymphoma, activating RAS mutations may propagate immunoglobulin-crippled tumour cells, which usually represent a minority of the tumour bulk. Thus, while BCR expression enhances lymphoma cell fitness, BCR-targeted therapies may profit from combinations with drugs targeting BCR- tumour cells.


Subject(s)
B-Lymphocytes/metabolism , Genes, myc , Genetic Fitness , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Lymphoma/genetics , Lymphoma/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Carbon/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Lymphoma/enzymology , Lymphoma/pathology , MAP Kinase Signaling System , Male , Mice , Mutation , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Tumor Cells, Cultured
3.
Cell Rep ; 14(10): 2348-61, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947073

ABSTRACT

Atherosclerosis is initiated and sustained by hypercholesterolemia, which results in the generation of oxidized LDL (OxLDL) and other metabolic byproducts that trigger inflammation. Specific immune responses have been shown to modulate the inflammatory response during atherogenesis. The sialic acid-binding immunoglobulin-like lectin G (Siglec-G) is a negative regulator of the functions of several immune cells, including myeloid cells and B-1 cells. Here, we show that deficiency of Siglec-G in atherosclerosis-prone mice inhibits plaque formation and diet-induced hepatic inflammation. We further demonstrate that selective deficiency of Siglec-G in B cells alone is sufficient to mediate these effects. Levels of B-1 cell-derived natural IgM with specificity for OxLDL were significantly increased in the plasma and peritoneal cavity of Siglec-G-deficient mice. Consistent with the neutralizing functions of OxLDL-specific IgM, Siglec-G-deficient mice were protected from OxLDL-induced sterile inflammation. Thus, Siglec-G promotes atherosclerosis and hepatic inflammation by suppressing protective anti-inflammatory effector functions of B cells.


Subject(s)
Atherosclerosis/pathology , Lectins/metabolism , Liver/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Atherosclerosis/metabolism , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Chemokines/analysis , Chemokines/blood , Cytokines/analysis , Cytokines/blood , Diet, High-Fat , Immunoassay , Immunoglobulin M/blood , Inflammation/pathology , Lectins/deficiency , Lectins/genetics , Leukocyte Common Antigens/metabolism , Lipoproteins, LDL/blood , Lipoproteins, LDL/immunology , Lipoproteins, LDL/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/prevention & control , Peritonitis/veterinary , RNA, Messenger/metabolism , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Serum Amyloid A Protein/analysis , Sialic Acid Binding Immunoglobulin-like Lectins
4.
J Immunol ; 195(1): 51-60, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25987743

ABSTRACT

Siglec-G, a member of the sialic acid-binding Ig-like lectin (Siglec) family, is expressed on B cell and dendritic cell surfaces. It acts as an inhibitory coreceptor and modulates B cell activation, especially on B1 cells, as Siglec-G-deficient mice show mainly a B1 cell-restricted phenotype resulting in increased B1 cell numbers. Although higher B1 cell numbers are discussed to be associated with autoimmunity, loss of Siglec-G does not result in autoimmune disease in BALB/c mice. However, there is evidence from Siglec-G × CD22 double-deficient mice and Siglec-G(-/-) mice on an autoimmune-prone MRL/lpr background that Siglec-G is important to maintain tolerance in B cells. In this study, we analyzed the role of Siglec-G in induction and maintenance of B cell tolerance on C57BL/6 background and in the FcγRIIb-deficient background. We find that aging Siglec-G-deficient and Siglec-G × FcγRIIb double-deficient mice develop an autoimmune phenotype with elevated autoantibody levels and mild glomerulonephritis. Aging Siglec-G-deficient mice have elevated numbers of plasma cells and germinal center B cells, as well as a higher number of activated CD4 T cells, which likely all contribute to autoantibody production. Additional loss of the inhibitory receptor FcγRIIb in Siglec-G(-/-) mice does not result in exacerbation of disease. These results indicate that Siglec-G is important to maintain tolerance in B cells and prevent autoimmunity.


Subject(s)
Aging/immunology , Autoimmunity , B-Lymphocytes/immunology , Glomerulonephritis/immunology , Lectins/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, IgG/immunology , Aging/genetics , Animals , Autoantibodies/biosynthesis , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Crosses, Genetic , Female , Gene Expression , Germinal Center/immunology , Germinal Center/pathology , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Immune Tolerance , Lectins/deficiency , Lectins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Receptors, IgG/deficiency , Receptors, IgG/genetics , Sialic Acid Binding Immunoglobulin-like Lectins
5.
Nature ; 521(7552): 357-61, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25799995

ABSTRACT

B cells are selected for an intermediate level of B-cell antigen receptor (BCR) signalling strength: attenuation below minimum (for example, non-functional BCR) or hyperactivation above maximum (for example, self-reactive BCR) thresholds of signalling strength causes negative selection. In ∼25% of cases, acute lymphoblastic leukaemia (ALL) cells carry the oncogenic BCR-ABL1 tyrosine kinase (Philadelphia chromosome positive), which mimics constitutively active pre-BCR signalling. Current therapeutic approaches are largely focused on the development of more potent tyrosine kinase inhibitors to suppress oncogenic signalling below a minimum threshold for survival. We tested the hypothesis that targeted hyperactivation--above a maximum threshold--will engage a deletional checkpoint for removal of self-reactive B cells and selectively kill ALL cells. Here we find, by testing various components of proximal pre-BCR signalling in mouse BCR-ABL1 cells, that an incremental increase of Syk tyrosine kinase activity was required and sufficient to induce cell death. Hyperactive Syk was functionally equivalent to acute activation of a self-reactive BCR on ALL cells. Despite oncogenic transformation, this basic mechanism of negative selection was still functional in ALL cells. Unlike normal pre-B cells, patient-derived ALL cells express the inhibitory receptors PECAM1, CD300A and LAIR1 at high levels. Genetic studies revealed that Pecam1, Cd300a and Lair1 are critical to calibrate oncogenic signalling strength through recruitment of the inhibitory phosphatases Ptpn6 (ref. 7) and Inpp5d (ref. 8). Using a novel small-molecule inhibitor of INPP5D (also known as SHIP1), we demonstrated that pharmacological hyperactivation of SYK and engagement of negative B-cell selection represents a promising new strategy to overcome drug resistance in human ALL.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Amino Acid Motifs/genetics , Animals , Antigens, CD/metabolism , B-Lymphocytes/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Female , Fusion Proteins, bcr-abl/genetics , Gene Deletion , Humans , Inositol Polyphosphate 5-Phosphatases , Intracellular Signaling Peptides and Proteins/agonists , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/drug effects , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Syk Kinase , Tyrosine/metabolism , Xenograft Model Antitumor Assays
6.
J Immunol ; 193(2): 909-920, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24929000

ABSTRACT

Signaling through the BCR can drive B cell activation and contribute to B cell differentiation into Ab-secreting plasma cells. The positive BCR signal is counterbalanced by a number of membrane-localized inhibitory receptors that limit B cell activation and plasma cell differentiation. Deficiencies in these negative signaling pathways may cause autoantibody generation and autoimmune disease in both animal models and human patients. We have previously shown that the transcription factor Ets1 can restrain B cell differentiation into plasma cells. In this study, we tested the roles of the BCR and inhibitory receptors in controlling the expression of Ets1 in mouse B cells. We found that Ets1 is downregulated in B cells by BCR or TLR signaling through a pathway dependent on PI3K, Btk, IKK2, and JNK. Deficiencies in inhibitory pathways, such as a loss of the tyrosine kinase Lyn, the phosphatase Src homology region 2 domain-containing phosphatase 1 (SHP1) or membrane receptors CD22 and/or Siglec-G, result in enhanced BCR signaling and decreased Ets1 expression. Restoring Ets1 expression in Lyn- or SHP1-deficient B cells inhibits their enhanced plasma cell differentiation. Our findings indicate that downregulation of Ets1 occurs in response to B cell activation via either BCR or TLR signaling, thereby allowing B cell differentiation and that the maintenance of Ets1 expression is an important function of the inhibitory Lyn → CD22/SiglecG → SHP1 pathway in B cells.


Subject(s)
Cell Differentiation/immunology , Plasma Cells/immunology , Proto-Oncogene Protein c-ets-1/immunology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Line, Tumor , Gene Expression/immunology , Lectins/deficiency , Lectins/genetics , Lectins/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/immunology , Plasma Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Protein c-ets-1/deficiency , Proto-Oncogene Protein c-ets-1/genetics , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialic Acid Binding Ig-like Lectin 2/deficiency , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Ig-like Lectin 2/immunology , Sialic Acid Binding Immunoglobulin-like Lectins , Signal Transduction/genetics , src-Family Kinases/deficiency , src-Family Kinases/genetics , src-Family Kinases/immunology
7.
Haematologica ; 99(8): 1356-64, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24859880

ABSTRACT

The sialic-acid-binding immunoglobulin-like lectin SIGLEC-G is a negative regulator of B-cell receptor-mediated calcium signaling. Its deficiency leads to reduced turnover and increased proliferation and survival of murine B-1a cells. Siglecg(-/-) mice show a premature expansion of polyclonal CD5(+) B cells in the spleen and the peritoneal cavity. Here we studied the fate of B lymphocytes in Siglecg(-/-) mice over time. We demonstrate that in aging animals SIGLEC-G deficiency promotes progressive accumulation of monoclonal B lymphocytes and increases the susceptibility to develop B-cell lymphoproliferative disorders. Lymphoid tumors arising in aged Siglecg(-/-) mice are monoclonal and histologically heterogeneous as they include diffuse large B-cell lymphoma, follicular lymphoma, and medium-to-large B-cell monomorphic lymphoma but surprisingly not chronic lymphocytic leukemia. The tumors express high levels of BCL-2 and are transplantable. In keeping with these findings we have also observed a remarkable down-regulation of the human ortholog SIGLEC10 in human B-cell lymphoma and leukemia cell lines. Taken together, these observations indicate that the down-regulation of negative B-cell receptor regulators such as SIGLEC-G/SIGLEC10 may represent another mechanism relevant to the pathogenesis of B-cell lymphomas.


Subject(s)
B-Lymphocytes/metabolism , Genetic Predisposition to Disease , Lectins/deficiency , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/metabolism , Receptors, Antigen, B-Cell/deficiency , Animals , Genetic Predisposition to Disease/genetics , Humans , Lectins/genetics , Leukemia, B-Cell/genetics , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/metabolism , Lymphoproliferative Disorders/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Immunoglobulin-like Lectins
9.
J Immunol ; 192(7): 2994-3002, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24600033

ABSTRACT

Siglec-G is a member of the sialic acid-binding Ig-like lectin (Siglec) family expressed on all B cells. Siglec-G-deficient mice show a large expansion of the B1 cell compartment, demonstrating the crucial role of Siglec-G as an inhibitory receptor on this cellular subset. Although Siglec-G-deficient mice did not develop spontaneous autoimmunity, mice double-deficient for Siglec-G and the related Siglec protein CD22 did show autoimmunity at an older age. In this study, we addressed the question of whether loss of Siglec G on its own affects disease severity in animal models of rheumatoid arthritis and systemic lupus erythematosus. Siglec-G-deficient mice showed moderately increased clinical severity and higher inflammation of the knee joints following collagen-induced arthritis, when compared with control mice. The Siglec-G-deficient mouse was also backcrossed to the autoimmune prone MLR/lpr background. Although both Siglec-G-deficient and control MRL/lpr mice developed a lupus-like disease, Siglec-G-deficient MRL/lpr mice showed an earlier occurrence of autoantibodies; a higher lymphoproliferation of B and T cells; and an earlier onset of disease, as shown by proteinuria and glomerular damage in the kidney. Moreover, Siglec-G-deficient female mice showed a significantly reduced survival compared with female control MRL/lpr mice. Thus, the loss of the inhibitory receptor Siglec-G led to a moderate exacerbation of disease severity and early onset in both collagen-induced arthritis and spontaneous lupus nephritis in MRL/lpr mice.


Subject(s)
Arthritis, Experimental/immunology , Lectins/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Antibodies, Antinuclear/immunology , Arthritis, Experimental/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Kaplan-Meier Estimate , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lectins/deficiency , Lectins/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lymphocyte Count , Male , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Knockout , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Severity of Illness Index , Sex Factors , Sialic Acid Binding Immunoglobulin-like Lectins , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Time Factors
10.
J Clin Invest ; 123(11): 4781-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24216514

ABSTRACT

Approximately 90% of patients with isolated agammaglobulinemia and failure of B cell development have mutations in genes required for signaling through the pre­B cell and B cell receptors. The nature of the gene defect in the majority of remaining patients is unknown. We recently identified 4 patients with agammaglobulinemia and markedly decreased numbers of peripheral B cells. The B cells that could be detected had an unusual phenotype characterized by the increased expression of CD19 but the absence of a B cell receptor. Genetic studies demonstrated that all 4 patients had the exact same de novo mutation in the broadly expressed transcription factor E47. The mutant protein (E555K) was stable in patient-derived EBV-transformed cell lines and cell lines transfected with expression vectors. E555K in the transfected cells localized normally to the nucleus and resulted in a dominant negative effect when bound to DNA as a homodimer with wild-type E47. Mutant E47 did permit DNA binding by a tissue-specific heterodimeric DNA-binding partner, myogenic differentiation 1 (MYOD). These findings document a mutational hot-spot in E47 and represent an autosomal dominant form of agammaglobulinemia. Further, they indicate that E47 plays a critical role in enforcing the block in development of B cell precursors that lack functional antigen receptors.


Subject(s)
Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Mutation, Missense , Receptors, Antigen, B-Cell/deficiency , Agammaglobulinemia/metabolism , Amino Acid Sequence , Amino Acid Substitution , B-Lymphocytes/metabolism , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Transformed , DNA/genetics , DNA/metabolism , Female , Genes, Dominant , Humans , Male , Molecular Sequence Data , Pedigree , Protein Stability , Sequence Homology, Amino Acid
11.
J Immunol ; 185(6): 3277-84, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20729333

ABSTRACT

Siglec-G is a negative regulator of BCR-mediated signaling in B1a cells. This population of B cells is highly increased in Siglec-G-deficient mice, but the mechanism of this expansion is not known so far. In this study, we demonstrate that Siglecg(-/-) B1a cells show a lower level of spontaneous apoptosis and a prolonged life span. Mechanistically, the lower apoptosis could result from higher expression levels of the transcription factor NFATc1 in Siglec-G-deficient B1a cells. Interestingly, Siglecg(-/-) B1a cells display an altered BCR repertoire compared with wild-type B1a cells. As the BCR repertoire and the VDJ composition of Igs of Siglecg(-/-) B1a cells resembles more the Abs produced by adult bone marrow-derived B cells rather than canonical fetal liver-derived B1a cells, this suggest that the selection into the B1a cell population is altered in Siglec-G-deficient mice.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cell Survival/immunology , Lectins/physiology , Receptors, Antigen, B-Cell/physiology , Adoptive Transfer , Animals , B-Lymphocyte Subsets/transplantation , Cell Compartmentation/genetics , Cell Compartmentation/immunology , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lectins/deficiency , Lectins/genetics , Leukocyte Common Antigens/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , NFATC Transcription Factors/biosynthesis , NFATC Transcription Factors/genetics , Peritoneum/cytology , Peritoneum/immunology , Peritoneum/metabolism , Receptors, Antigen, B-Cell/biosynthesis , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Immunoglobulin-like Lectins , Up-Regulation/genetics , Up-Regulation/immunology
12.
J Immunol ; 184(7): 3618-27, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20200274

ABSTRACT

CD22 and Siglec-G are inhibitory coreceptors for BCR-mediated signaling. Although CD22-deficient mice show increased calcium signaling in their conventional B2 cells and a quite normal B cell maturation, Siglec-G-deficient mice have increased calcium mobilization just in B1 cells and show a large expansion of the B1 cell population. Neither CD22-deficient, nor Siglec-G-deficient mice on a pure C57BL/6 or BALB/c background, respectively, develop autoimmunity. Using Siglec-G x CD22 double-deficient mice, we addressed whether Siglec-G and CD22 have redundant functions. Siglec-G x CD22 double-deficient mice show elevated calcium responses in both B1 cells and B2 cells, increased serum IgM levels and an enlarged population of B1 cells. The enlargement of B1 cell numbers is even higher than in Siglecg(-/-) mice. This expansion seems to happen at the expense of B2 cells, which are reduced in absolute cell numbers, but show an activated phenotype. Furthermore, Siglec-G x CD22 double-deficient mice show a diminished immune response to both thymus-dependent and thymus-independent type II Ags. In contrast, B cells from Siglec-G x CD22 double-deficient mice exhibit a hyperproliferative response to stimulation with several TLR ligands. Aged Siglec-G x CD22 double-deficient mice spontaneously develop anti-DNA and antinuclear autoantibodies. These resulted in a moderate form of immune complex glomerulonephritis. These results show that Siglec-G and CD22 have partly compensatory functions and together are crucial in maintaining the B cell tolerance.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Lectins/immunology , Receptors, Antigen, B-Cell/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Animals , Autoantibodies/blood , Autoantibodies/immunology , Calcium Signaling/immunology , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glomerulonephritis/immunology , Glomerulonephritis/pathology , Immune Tolerance/immunology , Lectins/deficiency , Lectins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Immunoglobulin-like Lectins
13.
Nature ; 463(7277): 88-92, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20054396

ABSTRACT

A role for B-cell-receptor (BCR) signalling in lymphomagenesis has been inferred by studying immunoglobulin genes in human lymphomas and by engineering mouse models, but genetic and functional evidence for its oncogenic role in human lymphomas is needed. Here we describe a form of 'chronic active' BCR signalling that is required for cell survival in the activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL). The signalling adaptor CARD11 is required for constitutive NF-kappaB pathway activity and survival in ABC DLBCL. Roughly 10% of ABC DLBCLs have mutant CARD11 isoforms that activate NF-kappaB, but the mechanism that engages wild-type CARD11 in other ABC DLBCLs was unknown. An RNA interference genetic screen revealed that a BCR signalling component, Bruton's tyrosine kinase, is essential for the survival of ABC DLBCLs with wild-type CARD11. In addition, knockdown of proximal BCR subunits (IgM, Ig-kappa, CD79A and CD79B) killed ABC DLBCLs with wild-type CARD11 but not other lymphomas. The BCRs in these ABC DLBCLs formed prominent clusters in the plasma membrane with low diffusion, similarly to BCRs in antigen-stimulated normal B cells. Somatic mutations affecting the immunoreceptor tyrosine-based activation motif (ITAM) signalling modules of CD79B and CD79A were detected frequently in ABC DLBCL biopsy samples but rarely in other DLBCLs and never in Burkitt's lymphoma or mucosa-associated lymphoid tissue lymphoma. In 18% of ABC DLBCLs, one functionally critical residue of CD79B, the first ITAM tyrosine, was mutated. These mutations increased surface BCR expression and attenuated Lyn kinase, a feedback inhibitor of BCR signalling. These findings establish chronic active BCR signalling as a new pathogenetic mechanism in ABC DLBCL, suggesting several therapeutic strategies.


Subject(s)
B-Lymphocytes/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Motifs , B-Lymphocytes/pathology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , CD79 Antigens/chemistry , CD79 Antigens/genetics , CD79 Antigens/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Mutation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA Interference , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , src-Family Kinases/metabolism
14.
J Immunol ; 183(9): 5630-43, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19843948

ABSTRACT

Generation of mature B lymphocytes from early (T1) and late transitional (T2) precursors requires cooperative signaling through BCR and B cell-activating factor receptor 3 (BR3). Recent studies have shown that BCR signaling positively regulates NF-kappaB2, suggesting BCR regulation of BR3 signaling. To investigate the significance of signal integration from BCR and BR3 in B cell development and function, we crossed Btk-deficient mice (btk(-/-)), which are developmentally blocked between the T2 and the mature follicular B cell stage as a result of a partial defect in BCR signaling, and A/WySnJ mice, which possess a mutant BR3 defective in propagating intracellular signals that results in a severely reduced peripheral B cell compartment, although all B cell subsets are present in relatively normal ratios. A/WySnJ x btk(-/-) mice display a B cell-autonomous defect, resulting in a developmental block at an earlier stage (T1) than either mutation alone, leading to the loss of mature splenic follicular and marginal zone B cells, as well as the loss of peritoneal B1 and B2 cell populations. The competence of the double mutant T1 B cells to respond to TLR4 and CD40 survival and activation signals is further attenuated compared with single mutations as evidenced by severely reduced humoral immune responses in vivo and proliferation in response to anti-IgM, LPS, and anti-CD40 stimulation in vitro. Thus, BCR and BR3 independently and in concert regulate the survival, differentiation, and function of all B cell populations at and beyond T1, earliest transitional stage.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Lymphopenia/immunology , Lymphopenia/pathology , Receptors, Antigen, B-Cell/deficiency , Signal Transduction/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Cell Activation Factor Receptor/deficiency , B-Cell Activation Factor Receptor/physiology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Lymphopenia/genetics , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-bcr/physiology , Receptors, Antigen, B-Cell/physiology , Signal Transduction/genetics
15.
J Immunol ; 183(5): 3237-48, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19667096

ABSTRACT

Somatic hypermutation normally occurs as a consequence of the expression of activation-induced cytidine deaminase (AID) by Ag-activated, mature B cells during T cell-dependent germinal center responses. Nonetheless, despite their inability to express CD154 and initiate GC responses, patients with type 1 hyper-IgM syndrome (HIGM1) support populations of IgM(+)IgD(+)CD27(+) B cells that express mutated Ig genes. The origin of these mutated B cells is unknown; the IgM(+)IgD(+)CD27(+) cells do not express AID and appear to acquire mutations independent of stringent selection by Ag. Here, we demonstrate that immature/transitional 1 B cells from the bone marrow of CD154-deficient mice express AID and acquire Ig mutations that lack the hallmarks of antigenic selection via BCR signaling. Comparable levels of AID expression was found in developmentally immature B cells recovered from murine fetal liver and from human immature/transitional 1 B cells recovered from umbilical cord blood. AID expression in human fetal liver was also robust, approaching that of human tonsil tissue and the human germinal center B cell line, Ramos. These observations led us to conclude that AID expression in developing human B cells is the origin of the mutated IgM(+)IgD(+)CD27(+) B cells present in HIGM1 patients, and we propose that both mice and humans share a latent, AID-dependent pathway for the preimmune diversification of B lymphocytes that is more prominent in chicken, sheep, and rabbits.


Subject(s)
Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Germinal Center/immunology , Germinal Center/pathology , Hyper-IgM Immunodeficiency Syndrome/enzymology , Hyper-IgM Immunodeficiency Syndrome/immunology , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Line, Transformed , Cell Line, Tumor , Cytidine Deaminase/biosynthesis , Female , Gene Expression Regulation, Developmental/immunology , Gene Rearrangement, B-Lymphocyte/genetics , Germinal Center/enzymology , Humans , Hyper-IgM Immunodeficiency Syndrome/genetics , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Somatic Hypermutation, Immunoglobulin/genetics , Stem Cells/enzymology , Stem Cells/immunology , Stem Cells/pathology
16.
Immunol Cell Biol ; 86(3): 261-7, 2008.
Article in English | MEDLINE | ID: mdl-18195726

ABSTRACT

Immunoglobulins are expressed as membrane-bound or secreted forms. Plasma cells produce little or no membrane immunoglobulin but secrete immunoglobulin molecules in large amounts. Immunoglobulin idiotypes of malignant B cells are tumor-specific antigens that may be targeted for immunotherapy. Thus, idiotype vaccination is being evaluated in clinical trials to control residual disease in multiple myeloma and non-Hodgkin's lymphoma. It is traditionally considered that anti-idiotype antibodies are not effective against plasma cell tumors, because the large amounts of immunoglobulin molecules secreted by the tumors block anti-idiotype antibodies, and because the absence of membrane immunoglobulin on the surface of these tumor cells renders them resistant to the effect of anti-idiotype antibodies. While the obstacle of abundant circulating idiotype may be obviated by reducing tumor burden to minimal residual disease, the absence of membrane immunoglobulin has been considered as a limiting factor that prevents tumor eradication by anti-idiotype antibodies. We demonstrate here that murine plasmacytoma cells can produce small amounts of membrane immunoglobulin M (IgM) heavy chains. However, the latter are precursor molecules that do not reach the cell surface. Although membrane-bound IgM is absent, the cells stain positively for surface IgM, reflecting molecules of the secreted form in the process of secretion. In spite of the relatively low levels of secreted immunoglobulin on the cell surface, anti-idiotype antibodies are effective in retardation of tumor growth in vivo. Thus, while there is no doubt that idiotype-specific cell-mediated responses are very important, myeloma patients in complete remission may additionally benefit from idiotype-specific humoral responses.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/therapeutic use , Cell Proliferation , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Receptors, Antigen, B-Cell/immunology , Animals , Antibody-Dependent Cell Cytotoxicity , Cancer Vaccines , Cell Line, Tumor , Cell Proliferation/drug effects , Immunization, Passive , Immunoglobulin Idiotypes/biosynthesis , Immunoglobulin Idiotypes/immunology , Immunoglobulin M/biosynthesis , Immunoglobulin M/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Multiple Myeloma/pathology , Neoplasm Transplantation , Receptors, Antigen, B-Cell/deficiency
17.
Nat Immunol ; 8(7): 695-704, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17572677

ABSTRACT

B1 cells are an important cell population for the production of natural antibodies and for antibacterial immunoglobulin responses. Here we identified the mouse protein Siglec-G as a B1 cell inhibitory receptor. Siglec-G was expressed in a B cell-restricted way, with large amounts present in B1 cells. When overexpressed, Siglec-G inhibited B cell receptor-mediated calcium signaling. Siglec-G-deficient mice had massive expansion of the B1a cell population, which began early in development and was B cell intrinsic. Siglec-G-deficient mice had higher titers of natural IgM antibodies but not a higher penetrance of IgG autoantibodies. Siglec-G-deficient B1 cells showed a strongly enhanced calcium signaling. Our results demonstrate that Siglec-G-dependent negative regulation exists in B1 cells, which may explain the naturally muted signaling response of B1 cells.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Calcium Signaling/immunology , Cell Proliferation , Lectins/physiology , N-Acetylneuraminic Acid/metabolism , Receptors, Antigen, B-Cell/physiology , Animals , Calcium Signaling/genetics , Cell Line , Chickens , Cloning, Molecular , Down-Regulation/genetics , Down-Regulation/immunology , Lectins/deficiency , Lectins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Sialic Acid Binding Immunoglobulin-like Lectins
18.
J Immunol ; 178(4): 2192-203, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277124

ABSTRACT

The terminal stage of B cell differentiation culminates in the formation of plasma cells (PC), which secrete large quantities of Igs. Despite recent progress in understanding the molecular aspect of PC differentiation and maintenance, the requirement for the synthesis of secretory Igs as a contributing factor has not been explored. To address this issue, we generated activation-induced cytidine deaminase (AID)/secretory mu-chain (mus) double-knockout mice, in which a normally diverse repertoire of B cell receptors is retained, yet B cells are unable to synthesize secretory Igs. These mice possess polyclonal B cells but have no serum Igs. Following immunization in vivo, PCs, identified by CD138 expression and loss of the B220 marker, were starkly reduced in number in spleen and bone marrow of AID(-/-)mus(-/-) agammaglobulinemic mice compared with wild-type mice. Upon mitogenic stimulation in vitro, AID(-/-)mus(-/-) B cells differentiated into plasmablasts to some extent, but showed reduced survival compared with wild-type B cells. We found no evidence that this reduced survival was attributable to accumulation of membrane IgM. Our results indicate that the synthesis of secretory Igs is a requirement for maintenance of B220(-)CD138(+) PCs.


Subject(s)
Agammaglobulinemia/immunology , Cytidine Deaminase/immunology , Immunoglobulin mu-Chains/immunology , Leukocyte Common Antigens/immunology , Plasma Cells/immunology , Syndecan-1/immunology , Agammaglobulinemia/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Cell Survival/immunology , Cytidine Deaminase/deficiency , Immunoglobulin mu-Chains/genetics , Mice , Mice, Knockout , Plasma Cells/cytology , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/immunology , Spleen/cytology , Spleen/immunology
19.
J Immunol ; 177(1): 395-400, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785535

ABSTRACT

Ig hypermutation is limited to a region of approximately 2 kb downstream of the transcription start sites of the Ig loci. The process requires transcription and the presence of Ig enhancer sequences, and is initiated by the activation-induced cytidine deaminase (AID)-mediated deamination of cytidine bases. It remains unknown why AID causes mutations selectively in the Ig genes and not in most other transcribed loci of B cells. In this study, we report that the inactivation of the E2A gene strongly reduces the rate of Ig L chain mutations in the chicken B cell line DT40 without affecting the levels of surface Ig or AID expression. The defect is complemented by the expression of cDNAs corresponding to either of the two E2A splice variants E12 or E47. The results suggest that E2A-encoded proteins enhance Ig hypermutation by recruitment of AID to the Ig loci.


Subject(s)
Avian Proteins/biosynthesis , Avian Proteins/genetics , Somatic Hypermutation, Immunoglobulin , TCF Transcription Factors/biosynthesis , TCF Transcription Factors/genetics , Animals , Avian Proteins/deficiency , Avian Proteins/physiology , Base Sequence , Cell Line , Chickens , Clone Cells , Cytidine Deaminase/biosynthesis , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA, Complementary/biosynthesis , Gene Silencing , Genetic Complementation Test , Genetic Markers , Immunoglobulin Light Chains/genetics , Molecular Sequence Data , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/genetics , Sequence Analysis, DNA , TCF Transcription Factors/deficiency , TCF Transcription Factors/physiology
20.
Blood ; 106(13): 4345-50, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16131568

ABSTRACT

In classic Hodgkin lymphoma (HL) and posttransplantation lymphoproliferative disease (PTLD), 2 malignancies frequently associated with Epstein-Barr virus (EBV), the tumor cells often appear to derive from B-cell receptor (BCR)-deficient and therefore preapoptotic germinal center (GC) B cells. To test whether EBV can rescue BCR-less GC B cells, we infected human tonsillar CD77+ GC B cells in vitro with EBV. More than 60 monoclonal lymphoblastoid cell lines (LCLs) were established. Among these, 28 cell lines did not express surface immunoglobulin (sIg). Two of the sIg-negative cell lines carry obviously destructive mutations that have been introduced into originally functional V(H) gene rearrangements during the process of somatic hypermutation. Quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) showed that in most other lines the sIg deficiency was not simply the result of transcriptional down-regulation, but it was rather due to posttranscriptional defects. These findings strongly support the idea that EBV plays a central role in the pathogenesis of classic HL and PTLD by rescuing BCR-deficient, preapoptotic GC B cells from apoptosis, and that EBV infection renders the cells independent from survival signals normally supplied by a BCR. The monoclonal LCLs represent valuable models for early stages of lymphoma development in classic HL and PTLD.


Subject(s)
B-Lymphocytes/metabolism , B-Lymphocytes/virology , Germinal Center/pathology , Herpesvirus 4, Human/physiology , Hodgkin Disease/pathology , Lymphoma/pathology , Receptors, Antigen, B-Cell/deficiency , Amino Acid Sequence , B-Lymphocytes/immunology , Cells, Cultured , Down-Regulation/genetics , Germinal Center/immunology , Hodgkin Disease/genetics , Hodgkin Disease/virology , Humans , Immunoglobulins/genetics , Lymphoma/etiology , Lymphoma/genetics , Lymphoma/virology , Molecular Sequence Data , Phenotype , Receptors, Antigen, B-Cell/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Somatic Hypermutation, Immunoglobulin/genetics , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...