Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Am J Hematol ; 99(1): 48-56, 2024 Jan.
Article En | MEDLINE | ID: mdl-37853951

ZAP70 has a prognostic value in chronic lymphocytic leukemia (CLL), through altered B-cell receptor signaling, which is important in CLL pathogenesis. A good correlation between ZAP70 expression in CLL cells and the occurrence of autoimmune phenomena has been reported. Yet, the great majority of CLL-associated autoimmune cytopenia is due to polyclonal immunoglobulin (Ig) G synthesized by nonmalignant B cells, and this phenomenon is poorly understood. Here, we show, using flow cytometry, that a substantial percentage of CD5- nonmalignant B cells from CLL patients expresses ZAP70 compared with CD5- B cells from healthy subjects. This ZAP70 expression in normal B cells from CLL patients was also evidenced by the detection of ZAP70 mRNA at single-cell level with polyclonal Ig heavy- and light-chain gene transcripts. ZAP70+ normal B cells belong to various B-cell subsets and their presence in the naïve B-cell subset suggests that ZAP70 expression may occur during early B-cell development in CLL patients and potentially before malignant transformation. The presence of ZAP70+ normal B cells is associated with autoimmune cytopenia in CLL patients in our cohort of patients, and recombinant antibodies produced from these ZAP70+ nonmalignant B cells were frequently autoreactive including anti-platelet reactivity. These results provide a better understanding of the implication of ZAP70 in CLL leukemogenesis and the mechanisms of autoimmune complications of CLL.


Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Autoimmunity , B-Lymphocytes , Flow Cytometry , Prognosis , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
2.
J Exp Med ; 220(12)2023 12 04.
Article En | MEDLINE | ID: mdl-37773045

Central B cell tolerance is believed to be regulated by B cell receptor signaling induced by the recognition of self-antigens in immature B cells. Using humanized mice with defective MyD88, TLR7, or TLR9 expression, we demonstrate that TLR9/MYD88 are required for central B cell tolerance and the removal of developing autoreactive clones. We also show that CXCL4, a chemokine involved in systemic sclerosis (SSc), abrogates TLR9 function in B cells by sequestering TLR9 ligands away from the endosomal compartments where this receptor resides. The in vivo production of CXCL4 thereby impedes both TLR9 responses in B cells and the establishment of central B cell tolerance. We conclude that TLR9 plays an essential early tolerogenic function required for the establishment of central B cell tolerance and that correcting defective TLR9 function in B cells from SSc patients may represent a novel therapeutic strategy to restore B cell tolerance.


Platelet Factor 4 , Scleroderma, Systemic , Toll-Like Receptor 9 , Animals , Humans , Mice , B-Lymphocytes , Ligands , Myeloid Differentiation Factor 88/metabolism , Platelet Factor 4/metabolism , Scleroderma, Systemic/metabolism , Toll-Like Receptor 7 , Toll-Like Receptor 9/metabolism
3.
J Clin Invest ; 132(2)2022 01 18.
Article En | MEDLINE | ID: mdl-34813502

Although negative selection of developing B cells in the periphery is well described, yet poorly understood, evidence of naive B cell positive selection remains elusive. Using 2 humanized mouse models, we demonstrate that there was strong skewing of the expressed immunoglobulin repertoire upon transit into the peripheral naive B cell pool. This positive selection of expanded naive B cells in humanized mice resembled that observed in healthy human donors and was independent of autologous thymic tissue. In contrast, negative selection of autoreactive B cells required thymus-derived Tregs and MHC class II-restricted self-antigen presentation by B cells. Indeed, both defective MHC class II expression on B cells of patients with rare bare lymphocyte syndrome and prevention of self-antigen presentation via HLA-DM inhibition in humanized mice resulted in the production of autoreactive naive B cells. These latter observations suggest that Tregs repressed autoreactive naive B cells continuously produced by the bone marrow. Thus, a model emerged, in which both positive and negative selection shaped the human naive B cell repertoire and that each process was mediated by fundamentally different molecular and cellular mechanisms.


Antigen Presentation , B-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Severe Combined Immunodeficiency/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID
4.
Sci Transl Med ; 11(519)2019 11 20.
Article En | MEDLINE | ID: mdl-31748230

Patients with rheumatoid arthritis (RA) may display atypical CD21-/lo B cells in their blood, but the implication of this observation remains unclear. We report here that the group of patients with RA and elevated frequencies of CD21-/lo B cells shows decreased ataxia telangiectasia-mutated (ATM) expression and activation in B cells compared with other patients with RA and healthy donor controls. In agreement with ATM involvement in the regulation of V(D)J recombination, patients with RA who show defective ATM function displayed a skewed B cell receptor (BCR) Igκ repertoire, which resembled that of patients with ataxia telangiectasia (AT). This repertoire was characterized by increased Jκ1 and decreased upstream Vκ gene segment usage, suggesting improper secondary recombination processes and selection. In addition, altered ATM function in B cells was associated with decreased osteoprotegerin and increased receptor activator of nuclear factor κB ligand (RANKL) production. These changes favor bone loss and correlated with a higher prevalence of erosive disease in patients with RA who show impaired ATM function. Using a humanized mouse model, we also show that ATM inhibition in vivo induces an altered Igκ repertoire and RANKL production by immature B cells in the bone marrow, leading to decreased bone density. We conclude that dysregulated ATM function in B cells promotes bone erosion and the emergence of circulating CD21-/lo B cells, thereby contributing to RA pathophysiology.


Arthritis, Rheumatoid/immunology , Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/metabolism , Bone Resorption/immunology , Animals , Arthritis, Rheumatoid/physiopathology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Bone Density , Bone Resorption/physiopathology , Cell Survival/immunology , Humans , Immunoglobulins/immunology , Joints/pathology , Lymphocyte Count , Mice , Middle Aged , Osteogenesis , Osteoprotegerin/metabolism , Phenotype , RANK Ligand/metabolism , Receptors, Complement 3d/metabolism , Recombination, Genetic/genetics
6.
Nat Commun ; 10(1): 3106, 2019 07 15.
Article En | MEDLINE | ID: mdl-31308374

Immune responses need to be controlled tightly to prevent autoimmune diseases, yet underlying molecular mechanisms remain partially understood. Here, we identify biallelic mutations in three patients from two unrelated families in differentially expressed in FDCP6 homolog (DEF6) as the molecular cause of an inborn error of immunity with systemic autoimmunity. Patient T cells exhibit impaired regulation of CTLA-4 surface trafficking associated with reduced functional CTLA-4 availability, which is replicated in DEF6-knockout Jurkat cells. Mechanistically, we identify the small GTPase RAB11 as an interactor of the guanine nucleotide exchange factor DEF6, and find disrupted binding of mutant DEF6 to RAB11 as well as reduced RAB11+CTLA-4+ vesicles in DEF6-mutated cells. One of the patients has been treated with CTLA-4-Ig and achieved sustained remission. Collectively, we uncover DEF6 as player in immune homeostasis ensuring availability of the checkpoint protein CTLA-4 at T-cell surface, identifying a potential target for autoimmune and/or cancer therapy.


CTLA-4 Antigen/metabolism , DNA-Binding Proteins/deficiency , Guanine Nucleotide Exchange Factors/deficiency , Primary Immunodeficiency Diseases/genetics , B7-1 Antigen/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Knockout Techniques , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Homeostasis , Humans , Jurkat Cells , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
7.
Sci Immunol ; 4(34)2019 04 12.
Article En | MEDLINE | ID: mdl-30979797

Autoimmune regulator (AIRE) mutations result in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) syndrome characterized by defective central T cell tolerance and the production of many autoantibodies targeting tissue-specific antigens and cytokines. By studying CD3- and AIRE-deficient patients, we found that lack of either T cells or AIRE function resulted in the peripheral accumulation of autoreactive mature naïve B cells. Proteomic arrays and Biacore affinity measurements revealed that unmutated antibodies expressed by these autoreactive naïve B cells recognized soluble molecules and cytokines including insulin, IL-17A, and IL-17F, which are AIRE-dependent thymic peripheral tissue antigens targeted by autoimmune responses in APECED. AIRE-deficient patients also displayed decreased frequencies of regulatory T cells (Tregs) that lacked common TCRß clones found instead in their conventional T cell compartment, thereby suggesting holes in the Treg TCR repertoire of these patients. Hence, AIRE-mediated T cell/Treg selection normally prevents the expansion of autoreactive naïve B cells recognizing peripheral self-antigens.


Autoantibodies/immunology , Autoimmunity/genetics , B-Lymphocytes/immunology , Polyendocrinopathies, Autoimmune/immunology , Transcription Factors/deficiency , Adolescent , Autoantibodies/blood , Autoantibodies/metabolism , Autoantigens/immunology , B-Lymphocytes/metabolism , CD3 Complex/deficiency , CD3 Complex/genetics , CD3 Complex/immunology , Child , Child, Preschool , Cytokines/immunology , Female , Humans , Immune Tolerance/genetics , Lymphocyte Activation/genetics , Male , Middle Aged , Mutation , Polyendocrinopathies, Autoimmune/blood , Polyendocrinopathies, Autoimmune/genetics , Protein Array Analysis , Proteomics/methods , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/immunology , AIRE Protein
8.
J Allergy Clin Immunol ; 143(1): 258-265, 2019 01.
Article En | MEDLINE | ID: mdl-29935219

BACKGROUND: The lack of pathogen-protective, isotype-switched antibodies in patients with common variable immunodeficiency (CVID) suggests germinal center (GC) hypoplasia, yet a subset of patients with CVID is paradoxically affected by autoantibody-mediated autoimmune cytopenias (AICs) and lymphadenopathy. OBJECTIVE: We sought to compare the physical characteristics and immunologic output of GC responses in patients with CVID with AIC (CVID+AIC) and without AIC (CVID-AIC). METHODS: We analyzed GC size and shape in excisional lymph node biopsy specimens from 14 patients with CVID+AIC and 4 patients with CVID-AIC. Using paired peripheral blood samples, we determined how AICs specifically affected B-and T-cell compartments and antibody responses in patients with CVID. RESULTS: We found that patients with CVID+AIC displayed irregularly shaped hyperplastic GCs, whereas GCs were scarce and small in patients with CVID-AIC. GC hyperplasia was also evidenced by an increase in numbers of circulating follicular helper T cells, which correlated with decreased regulatory T-cell frequencies and function. In addition, patients with CVID+AIC had serum endotoxemia associated with a dearth of isotype-switched memory B cells that displayed significantly lower somatic hypermutation frequencies than their counterparts with CVID-AIC. Moreover, IgG+ B cells from patients with CVID+AIC expressed VH4-34-encoded antibodies with unmutated Ala-Val-Tyr and Asn-His-Ser motifs, which recognize both erythrocyte I/i self-antigens and commensal bacteria. CONCLUSIONS: Patients with CVID+AIC do not contain mucosal microbiota and exhibit hyperplastic yet inefficient GC responses that favor the production of untolerized IgG+ B-cell clones that recognize both commensal bacteria and hematopoietic I/i self-antigens.


Autoantibodies/immunology , B-Lymphocytes/immunology , Common Variable Immunodeficiency/immunology , Germinal Center/immunology , Immunoglobulin G/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , B-Lymphocytes/pathology , Biopsy , Child , Common Variable Immunodeficiency/pathology , Female , Germinal Center/pathology , Humans , Hyperplasia , Male , Middle Aged , T-Lymphocytes/pathology
9.
Nature ; 559(7714): 405-409, 2018 07.
Article En | MEDLINE | ID: mdl-29995861

Decades of work have aimed to genetically reprogram T cells for therapeutic purposes1,2 using recombinant viral vectors, which do not target transgenes to specific genomic sites3,4. The need for viral vectors has slowed down research and clinical use as their manufacturing and testing is lengthy and expensive. Genome editing brought the promise of specific and efficient insertion of large transgenes into target cells using homology-directed repair5,6. Here we developed a CRISPR-Cas9 genome-targeting system that does not require viral vectors, allowing rapid and efficient insertion of large DNA sequences (greater than one kilobase) at specific sites in the genomes of primary human T cells, while preserving cell viability and function. This permits individual or multiplexed modification of endogenous genes. First, we applied this strategy to correct a pathogenic IL2RA mutation in cells from patients with monogenic autoimmune disease, and demonstrate improved signalling function. Second, we replaced the endogenous T cell receptor (TCR) locus with a new TCR that redirected T cells to a cancer antigen. The resulting TCR-engineered T cells specifically recognized tumour antigens and mounted productive anti-tumour cell responses in vitro and in vivo. Together, these studies provide preclinical evidence that non-viral genome targeting can enable rapid and flexible experimental manipulation and therapeutic engineering of primary human immune cells.


Cellular Reprogramming/genetics , Gene Editing , Genome, Human/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Autoimmunity/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Male , Mice , Neoplasm Transplantation , Protein Engineering , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
10.
JCI Insight ; 3(5)2018 03 08.
Article En | MEDLINE | ID: mdl-29515028

B cells play a central role in systemic lupus erythematosus (SLE) pathophysiology but dysregulated pathways leading to a break in B cell tolerance remain unclear. Since Toll-like receptor 9 (TLR9) favors the elimination of autoreactive B cells in the periphery, we assessed TLR9 function in SLE by analyzing the responses of B cells and plasmacytoid dendritic cells (pDCs) isolated from healthy donors and patients after stimulation with CpG, a TLR9 agonist. We found that SLE B cells from patients without hydroxychloroquine treatment displayed defective in vitro TLR9 responses, as illustrated by the impaired upregulation of B cell activation molecules and the diminished production of various cytokines including antiinflammatory IL-10. In agreement with CD19 controlling TLR9 responses in B cells, decreased expression of the CD19/CD21 complex on SLE B cells was detected as early as the transitional B cell stage. In contrast, TLR7 function was preserved in SLE B cells, whereas pDCs from SLE patients properly responded to TLR9 stimulation, thereby revealing that impaired TLR9 function in SLE was restricted to B cells. We conclude that abnormal CD19 expression and TLR9 tolerogenic function in SLE B cells may contribute to the break of B cell tolerance in these patients.


Antigens, CD19/immunology , B-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Complement 3d/immunology , Toll-Like Receptor 9/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD19/metabolism , Autoimmunity , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Humans , Immune Tolerance , Lupus Erythematosus, Systemic/blood , Male , Middle Aged , Oligodeoxyribonucleotides/pharmacology , Primary Cell Culture , Receptors, Complement 3d/metabolism , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism , Up-Regulation , Young Adult
12.
J Exp Med ; 214(7): 1991-2003, 2017 Jul 03.
Article En | MEDLINE | ID: mdl-28500047

The germline immunoglobulin (Ig) variable heavy chain 4-34 (VH4-34) gene segment encodes in humans intrinsically self-reactive antibodies that recognize I/i carbohydrates expressed by erythrocytes with a specific motif in their framework region 1 (FWR1). VH4-34-expressing clones are common in the naive B cell repertoire but are rarely found in IgG memory B cells from healthy individuals. In contrast, CD27+IgG+ B cells from patients genetically deficient for IRAK4 or MYD88, which mediate the function of Toll-like receptors (TLRs) except TLR3, contained VH4-34-expressing clones and showed decreased somatic hypermutation frequencies. In addition, VH4-34-encoded IgGs from IRAK4- and MYD88-deficient patients often displayed an unmutated FWR1 motif, revealing that these antibodies still recognize I/i antigens, whereas their healthy donor counterparts harbored FWR1 mutations abolishing self-reactivity. However, this paradoxical self-reactivity correlated with these VH4-34-encoded IgG clones binding commensal bacteria antigens. Hence, B cells expressing germline-encoded self-reactive VH4-34 antibodies may represent an innate-like B cell population specialized in the containment of commensal bacteria when gut barriers are breached.


B-Lymphocytes/immunology , Bacteria/immunology , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Adolescent , Adult , Amino Acid Sequence , Antigens, Bacterial/immunology , Autoantibodies/genetics , Autoantibodies/immunology , B-Lymphocytes/metabolism , Child , Child, Preschool , Clonal Selection, Antigen-Mediated , Female , Gastrointestinal Microbiome/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/immunology , Male , Mutation , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Sequence Homology, Amino Acid , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Young Adult
13.
Blood ; 129(8): 959-969, 2017 02 23.
Article En | MEDLINE | ID: mdl-28077418

Humanized mice are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, the existing models cannot support robust adaptive immune responses, especially the generation of class-switched, antigen-specific antibody responses. Here we describe a new mouse strain, in which human interleukin 6 (IL-6) gene encoding the cytokine that is important for B- and T-cell differentiation was knocked into its respective mouse locus. The provision of human IL-6 not only enhanced thymopoiesis and periphery T-cell engraftment, but also significantly increased class switched memory B cells and serum immunoglobulin G (IgG). In addition, immunization with ovalbumin (OVA) induced OVA-specific B cells only in human IL-6 knock-in mice. These OVA-specific antibodies displayed the highest frequency of somatic mutation, further suggesting that human IL-6 is important for efficient B-cell activation and selection. We conclude that human IL-6 knock-in mice represent a novel and improved model for human adaptive immunity without relying on complex surgery to transplant human fetal thymus and liver. These mice can therefore be used to exploit or evaluate immunization regimes that would be unethical or untenable in humans.


Adaptive Immunity , Antibody Formation , Gene Knock-In Techniques , Immunoglobulin Class Switching , Interleukin-6/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Chickens , Gene Expression , Gene Knock-In Techniques/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Immunization , Immunoglobulin G/immunology , Interleukin-6/immunology , Mice , Ovalbumin/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
14.
Sci Immunol ; 1(1)2016.
Article En | MEDLINE | ID: mdl-27917411

The 1858T protein tyrosine phosphatase nonreceptor type 22 (PTPN22 T) allele is one of the main risk factors associated with many autoimmune diseases and correlates with a defective removal of developing autoreactive B cells in humans. To determine whether inhibiting PTPN22 favors the elimination of autoreactive B cells, we first demonstrated that the PTPN22 T allele interfered with the establishment of central B cell tolerance using NOD-scid-common γ chain knockout (NSG) mice engrafted with human hematopoietic stem cells expressing this allele. In contrast, the inhibition of either PTPN22 enzymatic activity or its expression by RNA interference restored defective central B cell tolerance in this model. Thus, PTPN22 blockade may represent a therapeutic strategy for the prevention or treatment of autoimmunity.

15.
J Clin Invest ; 126(11): 4289-4302, 2016 11 01.
Article En | MEDLINE | ID: mdl-27701145

Patients with mutations in AICDA, which encodes activation-induced cytidine deaminase (AID), display an impaired peripheral B cell tolerance. AID mediates class-switch recombination (CSR) and somatic hypermutation (SHM) in B cells, but the mechanism by which AID prevents the accumulation of autoreactive B cells in blood is unclear. Here, we analyzed B cell tolerance in AID-deficient patients, patients with autosomal dominant AID mutations (AD-AID), asymptomatic AICDA heterozygotes (AID+/-), and patients with uracil N-glycosylase (UNG) deficiency, which impairs CSR but not SHM. The low frequency of autoreactive mature naive B cells in UNG-deficient patients resembled that of healthy subjects, revealing that impaired CSR does not interfere with the peripheral B cell tolerance checkpoint. In contrast, we observed decreased frequencies of SHM in memory B cells from AD-AID patients and AID+/- subjects, who were unable to prevent the accumulation of autoreactive mature naive B cells. In addition, the individuals with AICDA mutations, but not UNG-deficient patients, displayed Tregs with defective suppressive capacity that correlated with increases in circulating T follicular helper cells and enhanced cytokine production. We conclude that SHM, but not CSR, regulates peripheral B cell tolerance through the production of mutated antibodies that clear antigens and prevent sustained interleukin secretions that interfere with Treg function.


B-Lymphocytes/immunology , Cell Cycle Checkpoints/immunology , Cytidine Deaminase/deficiency , Immune Tolerance , Immunologic Memory , Mutation , Somatic Hypermutation, Immunoglobulin/immunology , B-Lymphocytes/pathology , Cell Cycle Checkpoints/genetics , Cytidine Deaminase/immunology , Female , Humans , Male , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
16.
Eur J Immunol ; 46(1): 131-46, 2016 Jan.
Article En | MEDLINE | ID: mdl-26474536

The links between infections and the development of B-cell-mediated autoimmune diseases are still unclear. In particular, it has been suggested that infection-induced stimulation of innate immune sensors can engage low affinity autoreactive B lymphocytes to mature and produce mutated IgG pathogenic autoantibodies. To test this hypothesis, we established a new knock-in mouse model in which autoreactive B cells could be committed to an affinity maturation process. We show that a chronic bacterial infection allows the activation of such B cells and the production of nonmutated IgM autoantibodies. Moreover, in the constitutive presence of their soluble antigen, some autoreactive clones are able to acquire a germinal center phenotype, to induce Aicda gene expression and to introduce somatic mutations in the IgG heavy chain variable region on amino acids forming direct contacts with the autoantigen. Paradoxically, only lower affinity variants are detected, which strongly suggests that higher affinity autoantibodies secreting B cells are counterselected. For the first time, we demonstrate in vivo that a noncross-reactive infectious agent can activate and induce autoreactive B cells to isotype switching and autoantigen-driven mutations, but on a nonautoimmune background, tolerance mechanisms prevent the formation of consequently dangerous autoimmunity.


Autoantigens/immunology , Autoimmunity/immunology , B-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , Lyme Disease/immunology , Animals , Antibody Affinity/immunology , Borrelia burgdorferi , Chronic Disease , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Knock-In Techniques , Immunoglobulin Isotypes/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Real-Time Polymerase Chain Reaction , Surface Plasmon Resonance
17.
J Allergy Clin Immunol ; 137(3): 889-98.e6, 2016 Mar.
Article En | MEDLINE | ID: mdl-26478008

BACKGROUND: CD19 is a B cell-specific molecule that serves as a major costimulatory molecule for amplifying B-cell receptor (BCR) responses. Biallelic CD19 gene mutations cause common variable immunodeficiency in human subjects. BCR- and Toll-like receptor (TLR) 9-induced B-cell responses are impaired in most patients with common variable immunodeficiency. OBJECTIVE: We sought to analyze whether CD19 is required for TLR9 function in human B cells. METHODS: Expression of surface activation markers was assessed after anti-IgM or CpG stimulation by using flow cytometry on B cells from patients with 1 or 2 defective CD19 alleles, which decrease or abrogate CD19 expression, respectively. The phosphorylation or interaction of signaling molecules was analyzed by using phospho flow cytometry, immunoblotting, or co-immunoprecipitation in CD19-deficient or control B cells and in a B-cell line in which CD19 has been knocked down with lentivirus-transduced short hairpin RNA. RESULTS: B cells from subjects with 1 or 2 defective CD19 alleles showed defective upregulation in vitro of CD86, transmembrane activator and CAML interactor (TACI), and CD23 activation markers after TLR9 stimulation. TLR9 ligands normally induce phosphorylation of CD19 through myeloid differentiation primary response gene-88 (MYD88)/proline-rich tyrosine kinase 2 (PYK2)/LYN complexes, which allows recruitment of phosphoinositide 3-kinase (PI3K) and phosphorylation of Bruton tyrosine kinase (BTK) and AKT in human B cells with a different kinetic than that of BCRs. In addition, inhibition of PI3K, AKT, or BTK, as well as BTK deficiency, also resulted in TLR9 activation defects in B cells similar to those in patients with CD19 deficiency. CONCLUSION: CD19 is required for TLR9-induced B-cell activation. Hence CD19/PI3K/AKT/BTK is an essential axis integrating BCRs and TLR9 signaling in human B cells.


Antigens, CD19/genetics , Antigens, CD19/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Toll-Like Receptor 9/metabolism , Agammaglobulinaemia Tyrosine Kinase , Case-Control Studies , Focal Adhesion Kinase 2/metabolism , Gene Knockdown Techniques , Heterozygote , Homozygote , Humans , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, B-Cell/metabolism , Toll-Like Receptor 9/agonists
18.
Immunity ; 43(5): 884-95, 2015 Nov 17.
Article En | MEDLINE | ID: mdl-26546282

Activation-induced cytidine deaminase (AID), the enzyme-mediating class-switch recombination (CSR) and somatic hypermutation (SHM) of immunoglobulin genes, is essential for the removal of developing autoreactive B cells. How AID mediates central B cell tolerance remains unknown. We report that AID enzymes were produced in a discrete population of immature B cells that expressed recombination-activating gene 2 (RAG2), suggesting that they undergo secondary recombination to edit autoreactive antibodies. However, most AID+ immature B cells lacked anti-apoptotic MCL-1 and were deleted by apoptosis. AID inhibition using lentiviral-encoded short hairpin (sh)RNA in B cells developing in humanized mice resulted in a failure to remove autoreactive clones. Hence, B cell intrinsic AID expression mediates central B cell tolerance potentially through its RAG-coupled genotoxic activity in self-reactive immature B cells.


Central Tolerance/genetics , Central Tolerance/immunology , Cytidine Deaminase/genetics , Lymphocyte Activation/immunology , Precursor Cells, B-Lymphoid/immunology , Adolescent , Adult , Aged , Animals , Apoptosis/genetics , Apoptosis/immunology , Case-Control Studies , Child , Child, Preschool , DNA-Binding Proteins/genetics , Female , Genes, Immunoglobulin/genetics , Genes, Immunoglobulin/immunology , Humans , Lymphocyte Activation/genetics , Male , Mice , Middle Aged , Nuclear Proteins/genetics , Recombination, Genetic/genetics , Recombination, Genetic/immunology , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology , Young Adult
19.
J Clin Invest ; 125(10): 3941-51, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26368308

Wiskott-Aldrich syndrome (WAS) is an X-linked immunodeficiency characterized by microthrombocytopenia, eczema, and high susceptibility to developing tumors and autoimmunity. Recent evidence suggests that B cells may be key players in the pathogenesis of autoimmunity in WAS. Here, we assessed whether WAS protein deficiency (WASp deficiency) affects the establishment of B cell tolerance by testing the reactivity of recombinant antibodies isolated from single B cells from 4 WAS patients before and after gene therapy (GT). We found that pre-GT WASp-deficient B cells were hyperreactive to B cell receptor stimulation (BCR stimulation). This hyperreactivity correlated with decreased frequency of autoreactive new emigrant/transitional B cells exiting the BM, indicating that the BCR signaling threshold plays a major role in the regulation of central B cell tolerance. In contrast, mature naive B cells from WAS patients were enriched in self-reactive clones, revealing that peripheral B cell tolerance checkpoint dysfunction is associated with impaired suppressive function of WAS regulatory T cells. The introduction of functional WASp by GT corrected the alterations of both central and peripheral B cell tolerance checkpoints. We conclude that WASp plays an important role in the establishment and maintenance of B cell tolerance in humans and that restoration of WASp by GT is able to restore B cell tolerance in WAS patients.


B-Lymphocytes/immunology , Genetic Therapy , Genetic Vectors/therapeutic use , Immune Tolerance , Wiskott-Aldrich Syndrome Protein/therapeutic use , Wiskott-Aldrich Syndrome/therapy , Adult , Amino Acid Sequence , Bone Marrow/pathology , Child , Child, Preschool , Clonal Deletion , Clone Cells/immunology , Humans , Lentivirus/genetics , Male , Molecular Sequence Data , Receptors, Antigen, B-Cell/immunology , Recombinant Fusion Proteins , T-Lymphocytes, Regulatory/immunology , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics
20.
Immun Inflamm Dis ; 3(3): 265-79, 2015 Sep.
Article En | MEDLINE | ID: mdl-26417441

Systemic Lupus Erythematosus (SLE) is a severe systemic autoimmune disease, characterized by multi-organ damages, triggered by an autoantibody-mediated inflammation, and with a complex genetic influence. It is today accepted that adult SLE arises from the building up of many subtle gene variations, each one adding a new brick on the SLE susceptibility and contributing to a phenotypic trait to the disease. One of the ways to find these gene variations consists in comprehensive analysis of gene expression variation in a precise cell type, which can constitute a good complementary strategy to genome wide association studies. Using this strategy, and considering the central role of B cells in SLE, we analyzed the B cell transcriptome of quiescent SLE patients, and identified an overexpression of FKBP11, coding for a cytoplasmic putative peptidyl-prolyl cis/trans isomerase and chaperone enzyme. To understand the consequences of FKBP11 overexpression on B cell function and on autoimmunity's development, we created lentiviral transgenic mice reproducing this gene expression variation. We showed that high expression of Fkbp11 reproduces by itself two phenotypic traits of SLE in mice: breakdown of B cell tolerance against DNA and initiation of plasma cell differentiation by acting upstream of Pax5 master regulator gene.

...