Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Immunol ; 203(1): 58-75, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31109957

ABSTRACT

CD40 is a costimulatory receptor on APCs that is critical for the induction and maintenance of humoral and cell-mediated immunity. Accordingly, CD40 and its ligand, CD40L, have long been considered targets for the treatment of autoimmune diseases. We developed a rat/mouse chimeric anti-mouse CD40 antagonist mAb, 201A3, and evaluated its ability to alleviate murine lupus. Treatment of NZB/W-F1 mice with 201A3 after the onset of severe proteinuria rapidly reversed established severe proteinuria and nephritis and largely restored normal glomerular and tubular morphology. This coincided with a normalization of the expression of genes associated with proteinuria and injury by kidney parenchymal cells. Anti-CD40 treatment also prevented and reversed loss of saliva production and sialadenitis. These effects on kidney and salivary gland function were confirmed using mice of a second strain, MRL/Mp-lpr/lpr, and extended to alleviating joint inflammation. Immunologically, anti-CD40 treatment disrupted multiple processes that contribute to the pathogenesis of systemic lupus erythematosus (SLE), including autoreactive B cell activation, T effector cell function in target tissues, and type I IFN production. This ability to disrupt disease-critical immunological mechanisms, to reverse glomerular and tubular injury at the cellular and gene expression levels, and to confer exceptional therapeutic efficacy suggests that CD40 is a central disease pathway in murine SLE. Thus, a CD40 antagonist Ab could be an effective therapeutic in the treatment of SLE.


Subject(s)
Antibodies, Blocking/therapeutic use , B-Lymphocytes/immunology , CD40 Antigens/immunology , Immunotherapy/methods , Kidney Glomerulus/pathology , Lupus Erythematosus, Systemic/therapy , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/immunology , Animals , Autoantigens/immunology , Cells, Cultured , Disease Models, Animal , Humans , Interferon Type I/metabolism , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred MRL lpr , Mice, Inbred NZB , Proteinuria , Rats , Salivary Elimination
2.
J Immunol ; 198(3): 1263-1273, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28031339

ABSTRACT

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


Subject(s)
Plasma Cells/drug effects , Staphylococcal Protein A/pharmacology , Animals , Antibody-Producing Cells/immunology , Immunoglobulin G/biosynthesis , Immunologic Memory , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Spleen/immunology , Staphylococcus aureus/immunology
3.
J Immunol ; 196(10): 4030-9, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27059595

ABSTRACT

Apoptotic debris, autoantibody, and IgG-immune complexes (ICs) have long been implicated in the inflammation associated with systemic lupus erythematosus (SLE); however, it remains unclear whether they initiate immune-mediated events that promote disease. In this study, we show that PBMCs from SLE patients experiencing active disease, and hematopoietic cells from lupus-prone MRL/lpr and NZM2410 mice accumulate markedly elevated levels of surface-bound nuclear self-antigens. On dendritic cells (DCs) and macrophages (MFs), the self-antigens are part of IgG-ICs that promote FcƎĀ³RI-mediated signal transduction. Accumulation of IgG-ICs is evident on ex vivo myeloid cells from MRL/lpr mice by 10 wk of age and steadily increases prior to lupus nephritis. IgG and FcƎĀ³RI play a critical role in disease pathology. Passive transfer of pathogenic IgG into IgG-deficient MRL/lpr mice promotes the accumulation of IgG-ICs prior to significant B cell expansion, BAFF secretion, and lupus nephritis. In contrast, diminishing the burden IgG-ICs in MRL/lpr mice through deficiency in FcƎĀ³RI markedly improves these lupus pathologies. Taken together, our findings reveal a previously unappreciated role for the cell surface accumulation of IgG-ICs in human and murine lupus.


Subject(s)
Apoptosis , Blood Cells/immunology , Dendritic Cells/immunology , Lupus Erythematosus, Systemic/immunology , Macrophages/immunology , Adult , Animals , Autoantigens/immunology , Autoantigens/metabolism , B-Cell Activating Factor/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Middle Aged , Receptors, IgG/genetics , Young Adult
4.
J Immunol ; 196(1): 196-206, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26621863

ABSTRACT

Memory B cell responses are vital for protection against infections but must also be regulated to prevent autoimmunity. Cognate T cell help, somatic hypermutation, and affinity maturation within germinal centers (GCs) are required for high-affinity memory B cell formation; however, the signals that commit GC B cells to the memory pool remain unclear. In this study, we identify a role for IgG-immune complexes (ICs), FcƎĀ³Rs, and BAFF during the formation of memory B cells in mice. We found that early secretion of IgG in response to immunization with a T-dependent Ag leads to IC-FcƎĀ³R interactions that induce dendritic cells to secrete BAFF, which acts at or upstream of Bcl-6 in activated B cells. Loss of CD16, hematopoietic cell-derived BAFF, or blocking IC:FcƎĀ³R regions in vivo diminished the expression of Bcl-6, the frequency of GC and memory B cells, and secondary Ab responses. BAFF also contributed to the maintenance and/or expansion of the follicular helper T cell population, although it was dispensable for their formation. Thus, early Ab responses contribute to the optimal formation of B cell memory through IgG-ICs and BAFF. Our work defines a new role for FcƎĀ³Rs in GC and memory B cell responses.


Subject(s)
Antigen-Antibody Complex/immunology , B-Cell Activating Factor/biosynthesis , Immunoglobulin G/immunology , Immunologic Memory/immunology , Receptors, IgG/immunology , Adoptive Transfer , Animals , B-Cell Activating Factor/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/immunology , Germinal Center/cytology , Germinal Center/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/biosynthesis , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Receptors, IgG/genetics , T-Lymphocytes, Helper-Inducer/immunology
5.
Blood ; 125(10): 1578-88, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25564405

ABSTRACT

Chronic lymphocytic leukemia (CLL) is a common B-cell malignancy characterized by a highly variable course and outcome. The disease is believed to be driven by B-cell receptor (BCR) signals generated by external antigens and/or cell-autonomous BCR interactions, but direct in vivo evidence for this is still lacking. To further define the role of the BCR pathway in the development and progression of CLL, we evaluated the capacity of different types of antigen/BCR interactions to induce leukemia in the EĀµ-TCL1 transgenic mouse model. We show that cell autonomous signaling capacity is a uniform characteristic of the leukemia-derived BCRs and represents a prerequisite for CLL development. Low-affinity BCR interactions with autoantigens generated during apoptosis are also positively selected, suggesting that they contribute to the pathogenesis of the disease. In contrast, high-affinity BCR interactions are not selected, regardless of antigen form or presentation. We also show that the capacity of the leukemic cells to respond to cognate antigen correlates inversely with time to leukemia development, suggesting that signals induced by external antigen increase the aggressiveness of the disease. Collectively, these findings provide in vivo evidence that the BCR pathway drives the development and can influence the clinical course of CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/etiology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Amino Acid Sequence , Animals , Antigen Presentation , Autoantigens/genetics , Disease Progression , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Gene Rearrangement, B-Lymphocyte, Light Chain , Humans , Leukemia, Experimental/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Muramidase/genetics , Muramidase/immunology , Proto-Oncogene Proteins/genetics , Receptors, Antigen, B-Cell/genetics , Signal Transduction/immunology , snRNP Core Proteins/genetics , snRNP Core Proteins/immunology
6.
Clin Immunol ; 168: 16-24, 2016 07.
Article in English | MEDLINE | ID: mdl-27102764

ABSTRACT

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


Subject(s)
Immune System/cytology , Interferon Type I/immunology , Kidney/immunology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Urine/cytology , Adult , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Female , Flow Cytometry , Humans , Immune System/immunology , Immunologic Memory/immunology , Interferon Type I/blood , Interferon Type I/urine , Interferon-alpha/blood , Interferon-alpha/immunology , Interferon-alpha/urine , Interferon-beta/blood , Interferon-beta/immunology , Interferon-beta/urine , Kidney/pathology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/urine , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/urine , Lupus Nephritis/blood , Lupus Nephritis/urine , Male , Plasma Cells/immunology
7.
J Immunol ; 189(8): 3859-68, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22984080

ABSTRACT

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


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lupus Nephritis/metabolism , Receptor Cross-Talk/immunology , Toll-Like Receptor 4/physiology , Animals , B-Lymphocyte Subsets/pathology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cells, Cultured , Coculture Techniques , Female , Immune Tolerance/genetics , Lupus Nephritis/enzymology , Lupus Nephritis/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Transgenic , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Transport/genetics , Protein Transport/immunology , Receptors, Antigen, B-Cell/physiology , Receptors, Interleukin-6/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
8.
ACR Open Rheumatol ; 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39364807

ABSTRACT

OBJECTIVE: Janus kinase family members are essential for signaling by multiple cytokines, including many implicated in systemic lupus erythematosus (SLE) pathogenesis. To test whether inhibition of JAK1 signaling can be efficacious in SLE, we used a JAK1-selective inhibitor (ABT-317) and evaluated its ability to ameliorate disease in murine SLE. METHODS: Efficacy of ABT-317 was evaluated using NZB/W-F1 mice treated prophylactically and therapeutically. Primary endpoints were proteinuria, survival, and saliva production. Other endpoints included histological analysis of kidneys and salivary glands, flow cytometric analysis of splenic cell populations, and gene expression analysis by RNA sequencing in the kidneys, salivary glands, and blood. Publicly available human kidney gene transcription data were used to assess the translatability of the mouse findings. RESULTS: ABT-317 was efficacious when dosed prophylactically and prevented disease for up to two months after treatment cessation. When dosed therapeutically, ABT-317 quickly reversed severe proteinuria and restored saliva production, as well as diminished kidney and salivary gland inflammation. ABT-317-induced changes in glomerular morphology coincided with normalization of a human nephrotic gene signature, suggesting translatability to human lupus nephritis (LN). CONCLUSION: JAK1 inhibition prevented and reversed kidney and salivary gland manifestations of murine lupus with long-lasting effects after treatment cessation. These data, along with the presence of JAK1 and nephrotic gene signatures in human LN glomeruli, suggest that a JAK1-selective inhibitor may be an effective therapeutic in the treatment of human SLE and LN.

9.
J Exp Med ; 201(8): 1333-46, 2005 Apr 18.
Article in English | MEDLINE | ID: mdl-15837817

ABSTRACT

Natural CD4(+)CD25(+) regulatory T (CD4(+)CD25(+) T reg) cells play a key role in the immunoregulation of autoimmunity. However, little is known about the interactions between CD4(+)CD25(+) T reg cells and autoreactive T cells. This is due, in part, to the difficulty of using cell surface markers to identify CD4(+)CD25(+) T reg cells accurately. Using a novel real-time PCR assay, mRNA copy number of FoxP3, TGFbeta1, and interleukin (IL)-10 was measured in single cells to characterize and quantify CD4(+)CD25(+) T reg cells in the nonobese diabetic (NOD) mouse, a murine model for type 1 diabetes (T1D). The suppressor function of CD4(+)CD25(+)CD62L(hi) T cells, mediated by TGFbeta, declined in an age-dependent manner. This loss of function coincided with a temporal decrease in the percentage of FoxP3 and TGFbeta1 coexpressing T cells within pancreatic lymph node and islet infiltrating CD4(+)CD25(+)CD62L(hi) T cells, and was detected in female NOD mice but not in NOD male mice, or NOR or C57BL/6 female mice. These results demonstrate that the majority of FoxP3-positive CD4(+)CD25(+) T reg cells in NOD mice express TGFbeta1 but not IL-10, and that a defect in the maintenance and/or expansion of this pool of immunoregulatory effectors is associated with the progression of T1D.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 1/metabolism , T-Lymphocyte Subsets/metabolism , Transforming Growth Factor beta/metabolism , Age Factors , Animals , CD4 Lymphocyte Count , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 1/immunology , Female , Forkhead Transcription Factors , Gene Expression , Interleukin-10/biosynthesis , Interleukin-10/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , RNA, Messenger/biosynthesis , Sex Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
10.
J Immunol ; 181(6): 3834-40, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768837

ABSTRACT

In this study we show that BCR affinity and subset identity make unique contributions to anergy. Analysis of anti-Smith (Sm) B cells of different affinities indicates that increasing affinity improves anergy's effectiveness while paradoxically increasing the likelihood of marginal zone (MZ) and B-1 B cell differentiation rather than just follicular (FO) B cell differentiation. Subset identity in turn determines the affinity threshold and mechanism of anergy. Subset-specific affinity thresholds for anergy induction allow discordant regulation of low-affinity anti-Sm FO and MZ B cells and could account for the higher frequency of autoreactive MZ B cells than that of FO B cells in normal mice. The mechanism of anergy changes during differentiation and differs between subsets. This is strikingly illustrated by the observation that blockade of BCR-mediated activation of FO and MZ B cells occurs at different levels in the signaling cascade. Thus, attributes unique to B cells of each subset integrate with signals from the BCR to determine the effectiveness, affinity threshold, and mechanism of anergy.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Clonal Anergy/immunology , Receptors, Antigen, B-Cell/metabolism , Animals , Autoantibodies/biosynthesis , Autoantigens/immunology , Autoantigens/metabolism , B-Lymphocyte Subsets/cytology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Clonal Anergy/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , Receptors, Antigen, B-Cell/physiology , Ribonucleoproteins, Small Nuclear/immunology , Ribonucleoproteins, Small Nuclear/metabolism , Self Tolerance/genetics , Self Tolerance/immunology , snRNP Core Proteins
11.
J Am Soc Nephrol ; 19(9): 1763-73, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18650487

ABSTRACT

Anti-neutrophil cytoplasmic autoantibodies directed toward myeloperoxidase or proteinase 3 are detected in sera of patients with small vessel vasculitis and participate in the pathogenesis of this disease. Autoantibodies develop when self-reactive B cells escape the regulation that ensures self-tolerance. In this study, regulation of anti-myeloperoxidase B cells was examined in mice that express an anti-myeloperoxidase Vkappa1C-Jkappa5 light-chain transgene, which confers anti-myeloperoxidase specificity when combined with a variety of heavy chains. Vkappa1C-Jkappa5 transgenic mice have splenic anti-myeloperoxidase B cells but do not produce circulating anti-myeloperoxidase antibodies. Two groups of transgenic mice that differed by their relative dosage of the transgene were compared; high-copy mice had a mean relative transgene dosage of 1.92 compared with 1.02 in the low-copy mice. These mice exhibited a 90 and 60% decrease in mature follicular B cells, respectively. High-copy mice were characterized by a large population of anti-myeloperoxidase B cells, a preponderance of B-1 cells, and an increased percentage of apoptotic myeloperoxidase-binding B cells. Low-copy mice had similar changes in B cell phenotype with the exception of an expanded marginal zone population. B cells from low-copy mice but not high-copy mice produced anti-myeloperoxidase antibodies after stimulation with lipopolysaccharide. These results indicate that tolerance to myeloperoxidase is maintained by central and peripheral deletion and that some myeloperoxidase-binding B cells are positively selected into the marginal zone and B-1 B cell subsets. A defect in these regulatory pathways could result in autoimmune disease.


Subject(s)
B-Lymphocytes/immunology , Immune Tolerance , Immunoglobulin kappa-Chains/metabolism , Peroxidase/immunology , Animals , Antibodies/metabolism , Apoptosis , Autoimmune Diseases/metabolism , B-Lymphocytes/metabolism , Bone Marrow Cells/cytology , Cell Count , Immunoglobulin kappa-Chains/genetics , Mice , Mice, Transgenic , Spleen/cytology
12.
J Extracell Vesicles ; 8(1): 1684425, 2019.
Article in English | MEDLINE | ID: mdl-31741724

ABSTRACT

Extracellular vesicles (EVs) have great potential as a source for clinically relevant biomarkers since they can be readily isolated from biofluids and carry microRNA (miRNA), mRNA, and proteins that can reflect disease status. However, the biological and technical variability of EV content is unknown making comparisons between healthy subjects and patients difficult to interpret. In this study, we sought to establish a laboratory and bioinformatics analysis pipeline to analyse the small RNA content within EVs from patient serum that could serve as biomarkers and to assess the biological and technical variability of EV RNA content in healthy individuals. We sequenced EV small RNA from multiple individuals (biological replicates) and sequenced multiple replicates per individual (technical replicates) using the Illumina Truseq protocol. We observed that the replicates of samples clustered by subject indicating that the biological variability (~95%) was greater than the technical variability (~0.50%). We observed that ~30% of the sequencing reads were miRNAs. We evaluated the technical parameters of sequencing by spiking the EV RNA preparation with a mix of synthetic small RNA and demonstrated a disconnect between input concentration of the spike-in RNA and sequencing read frequencies indicating that bias was introduced during library preparation. To determine whether there are differences between library preparation platforms, we compared the Truseq with the Nextflex protocol that had been designed to reduce bias in library preparation. While both methods were technically robust, the Nextflex protocol reduced the bias and exhibited a linear range across input concentrations of the synthetic spike-ins. Altogether, our results indicate that technical variability is much smaller than biological variability supporting the use of EV small RNAs as potential biomarkers. Our findings also indicate that the choice of library preparation method leads to artificial differences in the datasets generated invalidating the comparability of sequencing data across library preparation platforms.

13.
Cell Rep ; 28(2): 472-485.e5, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31291582

ABSTRACT

The NuRD complex contains both chromatin remodeling and histone deacetylase activities. Mice lacking the MTA2 subunit of NuRD show developmental defects in pro-B, pre-B, immature B, and marginal zone B cells, and abnormal germinal center B cell differentiation during immune responses. Mta2 inactivation also causes a derepression of Igll1 and VpreB1 genes in pre-B cells. Furthermore, MTA2/NuRD interacts directly with AIOLOS/IKAROS and shows a striking overlap with AIOLOS/IKAROS target genes in human pre-B cells, suggesting a functional inter-dependence between MTA2/NuRD and AIOLOS. Mechanistically, MTA2 deficiency in mice leads to increased H3K27 acetylation at both Igll1 and VpreB1 promoters. Gene profiling analyses also identify distinct MTA2-dependent transcription programs in pro-B and pre-B cells. In addition, we find a strong synergy between MTA2 and OCA-B in repressing Igll1 and VpreB1 at the pre-B cell stage, and in regulating both the pre-B to immature B transition and splenic B cell development.


Subject(s)
B-Lymphocytes/immunology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Precursor Cells, B-Lymphoid/immunology , Repressor Proteins/genetics , Trans-Activators/genetics , Animals , Humans , Mice
14.
Immunol Res ; 41(3): 203-16, 2008.
Article in English | MEDLINE | ID: mdl-18592142

ABSTRACT

Autoimmunity is a serious health problem and understanding the maintenance and loss of tolerance to self-antigens are key issues in developing new therapeutic strategies to treat these diseases. Despite considerable progress toward understanding B cell tolerance and tolerance loss, much remains unknown, particularly regarding B cells specific for antigens targeted in disease. Our interest in systemic lupus erythematosus (SLE), a B cell-mediated autoimmune disease characterized by the production of autoantibodies to numerous nuclear antigens, is focused on understanding B cell tolerance and tolerance loss to the SLE-specific autoantigen Sm in mice and humans. Our work aims to provide the cellular and molecular underpinnings for the development of rational therapies to target autoreactive B cells in human SLE.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/immunology , snRNP Core Proteins/immunology , Animals , Apoptosis/immunology , Autoantibodies/blood , Autoimmunity/immunology , B-Lymphocytes/metabolism , Dendritic Cells/metabolism , Humans , Lupus Erythematosus, Systemic/metabolism , Mice , snRNP Core Proteins/metabolism
15.
Mol Immunol ; 44(7): 1765-74, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17007932

ABSTRACT

The expression of a pre-B cell receptor (pre-BCR) is required for allelic exclusion and pre-BII cell differentiation. V(H)12 microH chains are unusual in that they form pre-BCRs and mediate allelic exclusion, but most cannot drive pre-BII cell differentiation. To explain this paradox, we examined pre-BCR functions and pre-BII cell differentiation in mice expressing microH chain transgenes encoding a B cell-permissible V(H)12 microH chain (designated 10/G4(6-1)), and a non-permissible V(H)12 microH chain (designated 8/G0). Compared with 10/G4 pre-BCRs, 8/G0 pre-BCRs are expressed at low levels on the cell surface. 8/G0 pre-BCRs mediate allelic exclusion, but 8/G0 pre-BII cells are defective in proliferation and expression of survival factors Bcl-2, Bcl-X(L) and hemokinin 1 (HK1). Increasing 8/G0 microH chain production restores HK1 transcription and improves proliferation of pre-BII cells as well as later stage B cell development. These data reveal a hierarchy of pre-BCR function that determines the development and plasticity of early B cells.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation , Membrane Glycoproteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocytes/chemistry , B-Lymphocytes/drug effects , Bone Marrow/immunology , Cell Differentiation , Cell Membrane/chemistry , Homozygote , Interleukin-7/pharmacology , Lymphocyte Activation/genetics , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Pre-B Cell Receptors , Protein Precursors/analysis , Protein Precursors/genetics , Protein Precursors/metabolism , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptors, Antigen, B-Cell/analysis , Receptors, Antigen, B-Cell/genetics , Spleen/immunology , Tachykinins/analysis , Tachykinins/genetics , Tachykinins/metabolism , bcl-X Protein/analysis , bcl-X Protein/genetics , bcl-X Protein/metabolism
16.
Mol Immunol ; 44(10): 2719-28, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17207856

ABSTRACT

B-1 cells are important players in the first line of defense against pathogens. According to current models for the origin of B-1 cells, they either represent a separate lineage from conventional B-2 cells or differentiate from conventional B-2 cells via an intermediate, B-1(int), in response to positive selection by antigen. Here we show that Btk, a Tec family kinase that mediates B cell antigen receptor (BCR) signaling, is required at multiple stages of B-1 cell development. VH12 anti-phosphatidylcholine (PtC) IgH transgenic mice provide a model for the induced differentiation of B-1 cells. This transgene selects for PtC-reactive cells and induces them to adopt a B-1 phenotype. Both processes have been shown to depend on Btk. To determine whether this is secondary to a requirement for Btk in the development of mature B-2 cells, we crossed VH12 transgenic mice to mice expressing low levels of Btk. B-2 cell development occurs normally in Btk(lo) mice despite reduced responsiveness to BCR crosslinking. Analysis of VH12.Btk(lo) mice reveals that Btk regulates the B-1(int) to B-1 transition and/or the survival of splenic B-1 cells, in part via a mechanism independent of its role in BCR signaling. We also show that Btk mediates the survival of, and expression of IL-10 by, those B-1 cells that do develop and migrate to the peritoneum. Multiple roles for Btk in B-1 cell development and maintenance may explain the particular sensitivity of this population to mutations in components of Btk signaling pathways.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Lineage , Protein-Tyrosine Kinases/physiology , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/enzymology , Cell Lineage/genetics , Cell Survival , Mice , Mice, Inbred Strains , Mice, Transgenic , Peritoneum/immunology , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, IgE/immunology
17.
Curr Opin Immunol ; 16(2): 246-50, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15023420

ABSTRACT

Although autoantigen-induced negative selection plays an important role in shaping the mature B-cell repertoire, studies in recent years have suggested that differentiation into any of the three mature B-cell subsets (marginal zone B cells, follicular B cells and B-1 B cells) is not a passive product of autoreactive B-cell elimination. Instead, evidence suggests that entry into a mature subset involves active B-cell receptor signaling and self-antigen-mediated positive selection.


Subject(s)
Antibody Specificity , B-Lymphocytes/physiology , B-Lymphocyte Subsets/immunology , Cell Differentiation , Gene Expression Regulation , Receptors, Antigen, B-Cell/immunology , Signal Transduction
18.
Mol Immunol ; 46(13): 2655-65, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19541367

ABSTRACT

B-1 cells reside predominantly within the coelomic cavities, tonsils, Peyer's patches, spleen (a minor fraction - approximately 5%) and are absent in the lymph nodes. They are the primary sources of natural IgM in the body. B-1 cells express polyreactive B cell receptors (BCRs) that cross react with self-antigens and are thus implicated in auto-immune disorders. Previously, we reported that peritoneal B-1 cells are deficient in CD19-mediated intracellular signals leading to Ca(2+) mobilization. Here, we find that splenic B-1 cells, like peritoneal B-1 cells, are defective in Ca(2+) release upon B cell activation by co-cross-linking BCR and CD19. In the absence of extracellular sources of Ca(2+), intracellular Ca(2+) flux is similar between B-1 and B-2 cells. Moreover, the intracellular component of Ca(2+) release in both subsets of B cells is mostly PI3K dependent. BCR and CD19 co-cross-linking activates Akt, a key mediator of survival and proliferation signals downstream of PI3K in splenic B-2 cells. Splenic B-1 cells, on the other hand, do not phosphorylate Akt (S473) upon similar treatment. Furthermore, BCR+CD19 cross-linking induced phosphorylation of JNK is much reduced in splenic B-1 cells. In contrast, B-1 cells exhibited increased levels of constitutively active pLyn which appears to have an inhibitory role. The CD19 induced Ca(2+) response and BCR induced proliferation response were restored by a partial inhibition of pLyn with Src kinase specific inhibitors. These findings suggest a defect in CD19-mediated signals in both peritoneal and splenic B-1 B lymphocytes, which is in part, due to higher levels of constitutively active Lyn.


Subject(s)
Antigens, CD19/immunology , B-Lymphocytes/immunology , Signal Transduction/immunology , Animals , Antibodies/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CD40 Antigens/immunology , CD5 Antigens/immunology , Calcium/metabolism , Cell Proliferation/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Immunoblotting , Leukocyte Common Antigens/immunology , MAP Kinase Kinase 4/metabolism , Macrophage-1 Antigen/immunology , Mice , Mice, Inbred C57BL , Peritoneum/cytology , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Spleen/cytology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
19.
J Invest Dermatol ; 129(12): 2823-34, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19571823

ABSTRACT

Fogo selvagem (FS), the endemic form of pemphigus foliaceus (PF), is characterized by pathogenic anti-desmoglein 1 (DSG1) autoantibodies. To study the etiology of FS, hybridomas that secrete either IgM or IgG (predominantly IgG1 subclass) autoantibodies were generated from the B cells of eight FS patients and one individual 4 years before FS onset, and the H and L chain V genes of anti-DSG1 autoantibodies were analyzed. Multiple lines of evidence suggest that these anti-DSG1 autoantibodies are antigen selected. First, clonally related sets of anti-DSG1 hybridomas characterize the response in individual FS patients. Second, H and L chain V gene use seems to be biased, particularly among IgG hybridomas, and third, most hybridomas are mutants and exhibit a bias in favor of CDR (complementary determining region) amino acid replacement (R) mutations. Strikingly, pre-FS hybridomas also exhibit evidence of antigen selection, including an overlap in V(H) gene use and shared multiple R mutations with anti-DSG1 FS hybridomas, suggesting selection by the same or a similar antigen. We conclude that the anti-DSG1 response in FS is antigen driven and that selection for mutant anti-DSG1 B cells begins well before the onset of disease.


Subject(s)
Autoantibodies/genetics , Autoantibodies/immunology , Desmoglein 1/immunology , Pemphigus/genetics , Pemphigus/immunology , Adolescent , Adult , Amino Acid Sequence , Cell Line, Transformed , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Endemic Diseases , Female , Humans , Hybridomas , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Leukocytes, Mononuclear/immunology , Male , Molecular Sequence Data , Pemphigus/epidemiology , Young Adult
20.
Clin Immunol ; 126(2): 189-201, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18077220

ABSTRACT

We previously reported that some systemic lupus erythematosus (SLE) patients have a population of circulating memory B cells with >2-fold higher levels of CD19. We show here that the presence of CD19(hi) B cells correlates with long-term adverse outcomes. These B cells do not appear anergic, as they exhibit high basal levels of phosphorylated Syk and ERK1/2, signal transduce in response to BCR crosslinking, and can become plasma cells (PCs) in vitro. Autoreactive anti-Smith (Sm) B cells are enriched in this population and the degree of enrichment correlates with the log of the serum anti-Sm titer, arguing that they undergo clonal expansion before PC differentiation. PC differentiation may occur at sites of inflammation, as CD19(hi) B cells have elevated CXCR3 levels and chemotax in response to its ligand CXCL9. Thus, CD19(hi) B cells are precursors to anti-self PCs, and identify an SLE patient subset likely to experience poor clinical outcomes.


Subject(s)
Antigens, CD19/blood , B-Lymphocyte Subsets/immunology , Immunologic Memory , Lupus Erythematosus, Systemic/immunology , Adult , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Antigens, CD19/immunology , Autoantigens/blood , Autoantigens/immunology , Autoimmunity , Chemokine CXCL9/blood , Chemokine CXCL9/immunology , Female , Humans , Immunologic Factors/therapeutic use , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/physiopathology , Male , Middle Aged , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, CXCR3/blood , Receptors, CXCR3/immunology , Ribonucleoproteins, Small Nuclear/blood , Ribonucleoproteins, Small Nuclear/immunology , Rituximab , snRNP Core Proteins
SELECTION OF CITATIONS
SEARCH DETAIL