Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Arthritis Rheumatol ; 66(12): 3424-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25156507

ABSTRACT

OBJECTIVE: Systemic lupus erythematosus (SLE) is characterized by B cell hyperactivity and autoantibody production. As spleen tyrosine kinase (Syk) is pivotal in B cell activation, these experiments aimed to examine the extent to which Syk was abnormally expressed in SLE B cells and the nature of the B cell subset that differently expressed Syk. METHODS: B cells from healthy donors and SLE patients were analyzed by flow cytometry to assess basal expression of Syk and phosphorylated Syk. B cell subsets expressing higher levels of Syk were found, and their detailed phenotype, in vitro differentiation into plasmablasts/plasma cells, and Syk induction by cytokines were determined. RESULTS: Syk expression was higher in CD27+ memory B cells than in naive B cells from SLE patients. However, a significantly increased frequency of CD27- B cells with bright expression of Syk (Syk++) was found in SLE patients. CD27-Syk++ B cells showed enhanced basal expression of p-Syk and stronger Syk phosphorylation upon B cell receptor (BCR) engagement as compared to CD27-Syk+ B cells. CD27-Syk++ B cells were CD38- as well as CD19++, CD20++, and mainly CD21-, with decreased ABCB1 transporter activity. In contrast to CD27-Syk+ B cells, CD27-Syk++ B cells exhibited enhanced differentiation into CD27++ IgG-secreting cells and expressed somatically mutated BCR gene rearrangements. Syk++ B cells were inducible in vitro by stimulation with interferon-γ, lipopolysaccharide, or tumor necrosis factor α. CONCLUSION: SLE patients exhibit an increased frequency of hitherto unknown CD27-Syk++ memory-like B cells, indicating that intracellular Syk density could distinguish CD27- memory B cells from truly naive B cell subsets. Furthermore, the CD27-Syk++ subset is a candidate for a source of increased plasma cells in SLE.


Subject(s)
B-Lymphocyte Subsets/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Lupus Erythematosus, Systemic/enzymology , Plasma Cells/enzymology , Protein-Tyrosine Kinases/metabolism , Adult , Aged , B-Lymphocyte Subsets/immunology , Case-Control Studies , Cell Differentiation/immunology , Female , Flow Cytometry , Humans , Immunologic Memory/immunology , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Phosphorylation , Plasma Cells/immunology , Syk Kinase , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Young Adult
2.
Exp Gerontol ; 54: 123-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24389059

ABSTRACT

The impairment of humoral immune response in elderly humans has been extensively demonstrated. We have reported the increase of memory B cells (IgG(+)IgD(-)CD27(-), double negative, DN) population in the elderly, in which there is also a typical inflammatory micro-environment. In order to evaluate whether this pro-inflammatory status could influence the trafficking phenotype of naïve/memory B cells, we have assessed the expression of CCR7, CCR6, CXCR3, CXCR4, CXCR5 and CD62L on naïve/memory B cell subpopulations in young and elderly subjects. Moreover, the combination of pro-inflammatory interleukin-21 (IL-21) and B cell receptor (BCR) stimulation enables B cells to produce and secrete granzyme B (GrB), which plays a critical role in early anti-viral immune responses, in the regulation of autoimmune mechanisms and in cancer immunosurveillance. Our data demonstrate that in the elderly, naïve/memory B cell populations present a different expression of the studied receptors that could be discussed in terms of "inflamm-aging". In particular IgG(+)IgD(-)CD27(-) DN B cells show a tissue trafficking phenotype and they can be stimulated to produce GrB.


Subject(s)
Aging/immunology , B-Lymphocyte Subsets/enzymology , Granzymes/biosynthesis , Adult , Aged , Aged, 80 and over , Humans , Immunoglobulin D/metabolism , Immunoglobulin G/metabolism , Interleukins/physiology , L-Selectin/metabolism , Phenotype , Receptors, CXCR/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
3.
Am J Physiol Renal Physiol ; 305(8): F1228-38, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23926186

ABSTRACT

Focal segmental glomerulosclerosis (FSGS) and collapsing glomerulopathy are common causes of nephrotic syndrome. Variants in >20 genes, including genes critical for mitochondrial function, have been associated with these podocyte diseases. One such gene, PDSS2, is required for synthesis of the decaprenyl tail of coenzyme Q10 (Q10) in humans. The mouse gene Pdss2 is mutated in the kd/kd mouse model of collapsing glomerulopathy. We examined the hypothesis that human PDSS2 polymorphisms are associated with podocyte diseases. We genotyped 377 patients with primary FSGS or collapsing glomerulopathy, together with 900 controls, for 9 single-nucleotide polymorphisms in the PDSS2 gene in a case-control study. Subjects included 247 African American (AA) and 130 European American (EA) patients and 641 AA and 259 EA controls. Among EAs, a pair of proxy SNPs was significantly associated with podocyte disease, and patients homozygous for one PDSS2 haplotype had a strongly increased risk for podocyte disease. By contrast, the distribution of PDSS2 genotypes and haplotypes was similar in AA patients and controls. Thus a PDSS2 haplotype, which has a frequency of 13% in the EA control population and a homozygote frequency of 1.2%, is associated with a significantly increased risk for FSGS and collapsing glomerulopathy in EAs. Lymphoblastoid cell lines from FSGS patients had significantly less Q10 than cell lines from controls; contrary to expectation, this finding was independent of PDSS2 haplotype. These results suggest that FSGS patients have Q10 deficiency and that this deficiency is manifested in patient-derived lymphoblastoid cell lines.


Subject(s)
Alkyl and Aryl Transferases/genetics , Glomerulosclerosis, Focal Segmental/enzymology , Glomerulosclerosis, Focal Segmental/genetics , Ubiquinone/analogs & derivatives , Adolescent , Adult , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Case-Control Studies , Glomerulosclerosis, Focal Segmental/ethnology , Haplotypes , Humans , Lymphocyte Activation/genetics , Middle Aged , Polymorphism, Single Nucleotide , Ubiquinone/deficiency , Ubiquinone/metabolism , Young Adult
4.
PLoS One ; 8(4): e61347, 2013.
Article in English | MEDLINE | ID: mdl-23585893

ABSTRACT

Cathepsin L (CTSL) is a ubiquitously expressed lysosomal cysteine peptidase with diverse and highly specific functions. The involvement of CTSL in thymic CD4+ T-cell positive selection has been well documented. Using CTSL(nkt/nkt) mice that lack CTSL activity, we have previously demonstrated that the absence of CTSL activity affects the homeostasis of the T-cell pool by decreasing CD4+ cell thymic production and increasing CD8+ thymocyte production. Herein we investigated the influence of CTSL activity on the homeostasis of peripheral B-cell populations and bone marrow (BM) B-cell maturation. B-cell numbers were increased in lymph nodes (LN), spleen and blood from CTSL (nkt/nkt) mice. Increases in splenic B-cell numbers were restricted to transitional T1 and T2 cells and to the marginal zone (MZ) cell subpopulation. No alterations in the proliferative or apoptosis levels were detected in peripheral B-cell populations from CTSL (nkt/nkt) mice. In the BM, the percentage and the absolute number of pre-pro-B, pro-B, pre-B, immature and mature B cells were not altered. However, in vitro and in vivo experiments showed that BM B-cell production was markedly increased in CTSL (nkt/nkt) mice. Besides, BM B-cell emigration to the spleen was increased in CTSL (nkt/nkt) mice. Colony-forming unit pre-B (CFU pre-B) assays in the presence of BM stromal cells (SC) and reciprocal BM chimeras revealed that both BM B-cell precursors and SC would contribute to sustain the increased B-cell hematopoiesis in CTSL (nkt/nkt) mice. Overall, our data clearly demonstrate that CTSL negatively regulates BM B-cell production and output therefore influencing the homeostasis of peripheral B cells.


Subject(s)
B-Lymphocyte Subsets/cytology , Cathepsin L/immunology , Lymphopoiesis/immunology , Precursor Cells, B-Lymphoid/cytology , Animals , Apoptosis , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cathepsin L/deficiency , Cathepsin L/genetics , Cell Proliferation , Gene Expression Regulation , Homeostasis , Lymph Nodes/cytology , Lymph Nodes/enzymology , Lymph Nodes/immunology , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/enzymology , Precursor Cells, B-Lymphoid/immunology , Spleen/cytology , Spleen/enzymology , Spleen/immunology , Stem Cells/cytology , Stem Cells/enzymology , Stem Cells/immunology
5.
J Immunol ; 190(6): 2527-35, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23396948

ABSTRACT

CD45 is a receptor-like tyrosine phosphatase that positively regulates BCR signaling by dephosphorylating the inhibitory tyrosine of the Src family kinases. We showed previously that a single point mutation, E613R, introduced into the cytoplasmic membrane-proximal "wedge" domain of CD45 is sufficient to drive a lupus-like autoimmune disease on a susceptible genetic background. To clarify the molecular mechanism of this disease, we took advantage of a unique allelic series of mice in which the expression of CD45 is varied across a broad range. Although both E613R B cells and those with supraphysiologic CD45 expression exhibited hyperresponsive BCR signaling, they did so by opposite regulation of the Src family kinase Lyn. We demonstrated that the E613R allele of CD45 does not function as a hyper- or hypomorphic allele but rather alters the substrate specificity of CD45 for Lyn. Despite similarly enhancing BCR signaling, only B cells with supraphysiologic CD45 expression became anergic, whereas only mice harboring the E613R mutation developed frank autoimmunity on a susceptible genetic background. We showed that selective impairment of a Lyn-dependent negative-regulatory circuit in E613R B cells drove autoimmunity in E613R mice. This demonstrates that relaxing negative regulation of BCR signaling, rather than enhancing positive regulation, is critical for driving autoimmunity in this system.


Subject(s)
B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Immune Tolerance , Leukocyte Common Antigens/physiology , Receptor-Like Protein Tyrosine Phosphatases/physiology , Alleles , Animals , B-Lymphocyte Subsets/cytology , Genetic Variation/immunology , Immune Tolerance/genetics , Leukocyte Common Antigens/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Protein Structure, Tertiary/genetics , Receptor-Like Protein Tyrosine Phosphatases/deficiency , Receptor-Like Protein Tyrosine Phosphatases/genetics , Substrate Specificity/genetics , Substrate Specificity/immunology , src-Family Kinases/genetics , src-Family Kinases/metabolism
6.
J Immunol ; 190(6): 2575-84, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23408836

ABSTRACT

Breast cancer cells facilitate distant metastasis through the induction of immunosuppressive regulatory B cells, designated tBregs. We report in this study that, to do this, breast cancer cells produce metabolites of the 5-lipoxygenase pathway such as leukotriene B4 to activate the peroxisome proliferator-activated receptor α (PPARα) in B cells. Inactivation of leukotriene B4 signaling or genetic deficiency of PPARα in B cells blocks the generation of tBregs and thereby abrogates lung metastasis in mice with established breast cancer. Thus, in addition to eliciting fatty acid oxidation and metabolic signals, PPARα initiates programs required for differentiation of tBregs. We propose that PPARα in B cells and/or tumor 5-lipoxygenase pathways represents new targets for pharmacological control of tBreg-mediated cancer escape.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Lipoxygenase/physiology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , PPAR alpha/physiology , Animals , B-Lymphocyte Subsets/enzymology , Cell Line, Tumor , Cells, Cultured , Female , Lipoxygenase/genetics , Lipoxygenase/metabolism , Melanoma, Experimental/genetics , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , PPAR alpha/deficiency , PPAR alpha/genetics , Tumor Escape/genetics , Tumor Escape/immunology
7.
J Immunol ; 190(4): 1827-36, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23325890

ABSTRACT

After undergoing Ig somatic hypermutation and Ag selection, germinal center (GC) B cells terminally differentiate into either memory or plasma cells (PCs). It is known that the CD40L and IL-21/STAT3 signaling pathways play critical roles in this process, yet it is unclear how the B cell transcription program interprets and integrates these two types of T cell-derived signals. In this study, we characterized the role of STAT3 in the GC-associated PC differentiation using purified human tonsillar GC B cells and a GC B cell-like cell line. When primary GC B cells were cultured under PC differentiation condition, STAT3 inhibition by AG490 prevented the transition from GC centrocytes to preplasmablast, suggesting that STAT3 is required for the initiation of PC development. In a GC B cell-like human B cell line, although IL-21 alone can induce low-level Blimp-1 expression, maximum Blimp-1 upregulation and optimal PC differentiation required both IL-21 and CD40L. CD40L, although having no effect on Blimp-1 as a single agent, greatly augmented the amplitude and duration of IL-21-triggered Jak-STAT3 signaling. In the human PRDM1 locus, CD40L treatment enhanced the ability of STAT3 to upregulate Blimp-1 by removing BCL6, a potent inhibitor of Blimp-1 expression, from a shared BCL6/STAT3 site in intron 3. Thus, IL-21 and CD40L collaborate through at least two distinct mechanisms to synergistically promote Blimp-1 activation and PC differentiation.


Subject(s)
Adjuvants, Immunologic/physiology , B-Lymphocyte Subsets/immunology , CD40 Ligand/physiology , Cell Differentiation/immunology , Interleukins/physiology , Plasma Cells/immunology , Repressor Proteins/biosynthesis , Up-Regulation/immunology , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/metabolism , Cell Line, Tumor , Humans , Janus Kinases/physiology , Molecular Sequence Data , Organ Culture Techniques , Palatine Tonsil/enzymology , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Plasma Cells/enzymology , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/physiology , STAT3 Transcription Factor/physiology
8.
J Immunol ; 189(8): 3800-4, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22984083

ABSTRACT

Phagocyte NADPH oxidase plays a key role in pathogen clearance via reactive oxygen species (ROS) production. Defects in oxidase function result in chronic granulomatous disease with hallmark recurrent microbial infections and inflammation. The oxidase's role in the adaptive immune response is not well understood. Class II presentation of cytoplasmic and exogenous Ag to CD4(+) T cells was impaired in human B cells with reduced oxidase p40(phox) subunit expression. Naturally arising mutations, which compromise p40(phox) function in a chronic granulomatous disease patient, also perturbed class II Ag presentation and intracellular ROS production. Reconstitution of patient B cells with a wild-type, but not a mutant, p40(phox) allele restored exogenous Ag presentation and intracellular ROS generation. Remarkably, class II presentation of epitopes from membrane Ag was robust in p40(phox)-deficient B cells. These studies reveal a role for NADPH oxidase and p40(phox) in skewing epitope selection and T cell recognition of self Ag.


Subject(s)
Antigen Presentation/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , HLA-DR Antigens/metabolism , NADPH Oxidases/physiology , Antigen Presentation/genetics , B-Lymphocyte Subsets/enzymology , Cell Line, Transformed , Humans , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Phosphoproteins/biosynthesis , Phosphoproteins/deficiency , Phosphoproteins/genetics , Reactive Oxygen Species/metabolism , Up-Regulation/genetics , Up-Regulation/immunology
9.
J Immunol ; 189(9): 4437-43, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23002441

ABSTRACT

The ubiquitin-editing enzyme A20 (TNFAIP3) and the deubiquitinase CYLD are central negative regulators of NF-κB signaling. Both can act by removing nonproteolytic K63-linked polyubiquitin chains from an overlapping set of signaling molecules. In B cells, A20 deficiency results in hyperactivity, loss of immune homeostasis, inflammation, and autoimmunity. The reported consequences of CYLD deficiency are controversial, ranging from an absence of effects to dramatic B cell hyperplasia. These differences could be due to varying compensation for the loss of CYLD function by A20. Therefore, to explore potential overlapping physiological functions between A20 and CYLD, we generated and characterized A20/CYLD double-deficient B cells. Interestingly, the lack of both A20 and CYLD did not exacerbate the developmental defects and hyperresponsive activity of A20-deficient B cells. In addition, the extent of B cell activation after in vitro stimulation with anti-CD40, LPS, and CpG was comparable in B cells lacking A20/CYLD and A20 alone. However, in response to BCR cross-linking, we observed small but reproducible additive effects of the lack of A20 and CYLD. Taken together, our results demonstrate that A20 and CYLD do not share significant functions during B cell development and activation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Cysteine Endopeptidases/physiology , DNA-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Lymphocyte Activation/immunology , Ubiquitin-Protein Ligases/physiology , Animals , B-Lymphocyte Subsets/enzymology , Cell Culture Techniques , Cell Differentiation/genetics , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Deubiquitinating Enzyme CYLD , Genes, Overlapping/immunology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
10.
J Immunol ; 189(6): 2931-40, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896639

ABSTRACT

The lupus-prone NZM2410 mice present an expanded B1a cell population that we have mapped to the Sle2c1 lupus susceptibility locus. The expression of Cdkn2c, a gene encoding for cyclin-dependent kinase inhibitor p18(Ink4c) and located within Sle2c1, is significantly lower in B6.Sle2c1 B cells than in B6 B cells. To test the hypothesis that the B1a cell expansion in B6.Sle2c1 mice was due to a defective p18 expression, we analyzed the B1a cell phenotypes of p18-deficient C57BL/6 mice. We found a dose-dependent negative correlation between the number of B1a cells and p18 expression in B cells, with p18-deficient mice showing an early expansion of the peritoneal B1a cell pool. p18 deficiency enhanced the homeostatic expansion of B1a cells but not of splenic conventional B cells, and the elevated number of B6.Sle2c1 B1a cells was normalized by cyclin D2 deficiency. These data demonstrated that p18 is a key regulator of the size of the B1a cell pool. B6.p18(-/-) mice produced significant amounts of anti-DNA IgM and IgG, indicating that p18 deficiency contributes to humoral autoimmunity. Finally, we have shown that Sle2c1 increases lpr-associated lymphadenopathy and T cell-mediated pathology. B6.p18(-/-).lpr mice showed a greater lymphadenopathy than B6.Sle2c1.lpr mice, but their renal pathology was intermediate between that of B6.lpr and B6.Sle2c1.lpr mice. This indicated that p18-deficiency synergizes, at least partially, with lpr-mediated pathology. These results show that Cdkn2c contributes to lupus susceptibility by regulating the size of the B1a cell compartment and hence their contribution to autoimmunity.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cell Differentiation/immunology , Cyclin-Dependent Kinase Inhibitor p18/deficiency , Cyclin-Dependent Kinases/antagonists & inhibitors , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Animals , B-Lymphocyte Subsets/enzymology , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p18/genetics , Cyclin-Dependent Kinase Inhibitor p18/physiology , Disease Models, Animal , Immunophenotyping , Lupus Erythematosus, Systemic/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Inbred NZB , Mice, Knockout , Mice, Transgenic
11.
J Immunol ; 189(3): 1193-201, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22753944

ABSTRACT

To investigate signals that control B cell selection, we examined expression of G5PR, a regulatory subunit of the serine/threonine protein phosphatase 2A, which suppresses JNK phosphorylation. G5PR is upregulated in activated B cells, in Ki67-negative centrocytes at germinal centers (GCs), and in purified B220(+)Fas(+)GL7(+) mature GC B cells following Ag immunization. G5PR rescues transformed B cells from BCR-mediated activation-induced cell death by suppression of late-phase JNK activation. In G5PR-transgenic (G5PR(Tg)) mice, G5PR overexpression leads to an augmented generation of GC B cells via an increase in non-Ag-specific B cells and a consequent reduction in the proportion of Ag-specific B cells and high-affinity Ab production after immunization with nitrophenyl-conjugated chicken γ-globulin. G5PR overexpression impaired the affinity-maturation of Ag-specific B cells, presumably by diluting the numbers of high-affinity B cells. However, aged nonimmunized female G5PR(Tg) mice showed an increase in the numbers of peritoneal B-1a cells and the generation of autoantibodies. G5PR overexpression did not affect the proliferation of B-1a and B-2 cells but rescued B-1a cells from activation-induced cell death in vitro. G5PR might play a pivotal role in B cell selection not only for B-2 cells but also for B-1 cells in peripheral lymphoid organs.


Subject(s)
Aging/immunology , Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , Epitopes, B-Lymphocyte/immunology , Germinal Center/immunology , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/genetics , Protein Subunits/genetics , Up-Regulation/immunology , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Chickens , Female , Germinal Center/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Transgenic , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , Peritoneal Cavity/cytology , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/physiology , Protein Subunits/biosynthesis , Protein Subunits/physiology , Rats , Rats, Inbred Lew , Sex Characteristics , Up-Regulation/genetics
12.
J Immunol ; 189(1): 161-73, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22634617

ABSTRACT

Mature B cell differentiation involves a well-established transcription factor cascade. However, the temporal dynamics of cell signaling pathways regulating transcription factor network and coordinating cell proliferation and differentiation remain poorly defined. To gain insight into the molecular processes and extrinsic cues required for B cell differentiation, we set up a controlled primary culture system to differentiate human naive B cells into plasma cells (PCs). We identified T cell-produced IL-2 to be critically involved in ERK1/2-triggered PC differentiation. IL-2 drove activated B cell differentiation toward PC independently of its proliferation and survival functions. Indeed, IL-2 potentiated ERK activation and subsequent BACH2 and IRF8 downregulation, sustaining BLIMP1 expression, the master regulator for PC differentiation. Inhibition of the MAPK-ERK pathway, unlike STAT5 signaling, impaired IL-2-induced PC differentiation and rescued the expression profile of BACH2 and IRF8. These results identify IL-2 as a crucial early input in mature B cell fate commitment.


Subject(s)
B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Cell Proliferation , Interleukin-2/physiology , MAP Kinase Signaling System/immunology , Plasma Cells/immunology , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/enzymology , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Humans , Lymphocyte Cooperation/immunology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Plasma Cells/cytology , Plasma Cells/enzymology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology
13.
Immunol Cell Biol ; 90(6): 620-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21894171

ABSTRACT

Blk was identified two decades ago as a B-cell-specific member of the Src family of tyrosine kinases. Recent studies, however, have discovered that Blk is expressed in many cell types outside of the B lineage, including early thymic precursors, interleukin-17-producing γδ T cells and pancreatic ß-cells. In light of these recent discoveries, we performed a more comprehensive analysis of Blk expression patterns in hematopoietic cells and found that Blk is differentially expressed in mature B-cell subsets, with marginal zone (MZ) B cells expressing high levels, B1 B cells expressing intermediate-to-high levels and follicular (FO) B cells expressing low levels of Blk. To determine whether these differences in Blk expression levels reflected differential requirements for Blk in MZ, B1 and FO B-cell development, we analyzed the effects of reducing and eliminating Blk expression on B-cell development. We report that both Blk haploinsufficiency and Blk deficiency impaired the generation of MZ B cells. Moreover, although there were fewer MZ B cells in Blk(+/-) and Blk(-/-) mice as compared with Blk(+/+) mice, Blk-mutant MZ B cells were hyper-responsive to B-cell receptor stimulation, both in vitro and in vivo. Thus, this study has revealed a previously unappreciated role for Blk in the development and activation of MZ B cells.


Subject(s)
B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Haploinsufficiency , src-Family Kinases/genetics , src-Family Kinases/metabolism , Animals , Antigens, T-Independent/immunology , Autoimmunity , Cell Differentiation , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-17/immunology , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, B-Cell/immunology , Signal Transduction
14.
Cancer Immunol Immunother ; 61(1): 19-29, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21822917

ABSTRACT

MVA-BN®-HER2 is a new candidate immunotherapy designed for the treatment of HER-2-positive breast cancer. Here, we demonstrate that a single treatment with MVA-BN®-HER2 exerts potent anti-tumor efficacy in a murine model of experimental pulmonary metastasis. This anti-tumor efficacy occurred despite a strong tumor-mediated immunosuppressive environment characterized by a high frequency of regulatory T cells (T(reg)) in the lungs of tumor-bearing mice. Immunogenicity studies showed that treatment with MVA-BN®-HER2 induced strongly Th1-dominated HER-2-specific antibody and T-cell responses. MVA-BN®-HER2-induced anti-tumor activity was characterized by an increased infiltration of lungs with highly activated, HER-2-specific, CD8+CD11c+ T cells accompanied by a decrease in the frequency of T(reg) cells in the lung, resulting in a significantly increased ratio of effector T cells to T(reg) cells. In contrast, administration of HER2 protein formulated in Complete Freund's Adjuvant (CFA) induced a strongly Th2-biased immune response to HER-2. However, this did not lead to significant infiltration of the tumor-bearing lungs by CD8+ T cells or the decrease in the frequency of T(reg) cells nor did it result in anti-tumor efficacy. In vivo depletion of CD8+ cells confirmed that CD8 T cells were required for the anti-tumor activity of MVA-BN®-HER2. Furthermore, depletion of CD4+ or CD25+ cells demonstrated that tumor-induced T(reg) cells promoted tumor growth and that CD4 effector cells also contribute to MVA-BN®-HER2-mediated anti-tumor efficacy. Taken together, our data demonstrate that treatment with MVA-BN®-HER2 controls tumor growth through mechanisms including the induction of Th1-biased HER-2-specific immune responses and the control of tumor-mediated immunosuppression.


Subject(s)
Adenocarcinoma/therapy , B-Lymphocyte Subsets/immunology , Cancer Vaccines/pharmacology , Colonic Neoplasms/therapy , Immunotherapy/methods , Receptor, ErbB-2/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/pathology , Cancer Vaccines/immunology , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Humans , Immunophenotyping , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/pathology
15.
Pol J Vet Sci ; 14(3): 393-403, 2011.
Article in English | MEDLINE | ID: mdl-21957733

ABSTRACT

Bestatin, a low-molecular weight dipeptide, is a potent inhibitor of aminopeptidase N which has been demonstrated to have antitumor and immunomodulatory effects. The effects of bestatin (10, 1 and 0.1 mg/kg) administered intraperitoneally once, five or ten times to mice on the total number of lymphocytes in the thymus, spleen and mesenteric lymph nodes and the percentage and the absolute number of T cell subsets (CD4+CD8+, CD4-CD8-, CD4+, CD8+) in the thymus and T (CD3+, CD4+, CD8+) and B (CD19+) lymphocytes in the spleen and mesenteric lymph nodes were studied. It has been found that bestatin administered ten times at doses of 10, 1 and 0.1 mg/kg increased the total number of thymocytes, splenocytes and lymphocytes of mesenteric lymph nodes. Bestatin also changed the percentage and the absolute number of T cell subsets in the thymus and T and B lymphocytes in the peripheral lymphatic organs. Five and ten exposures to bestatin (10, 1 and 0.1 mg/kg) increased the absolute count of both immature CD4+CD8+ and CD4-CD8- thymic cells. Moreover, both a single and multiple administration of bestatin (1 and 0.1 mg/kg) decreased the percentage and absolute count of CD3+ splenocytes and mesenteric lymph node cells with corresponding decreases in the percentage and absolute count of CD4+ and CD8+ cells. Both a single and multiple administration of bestatin at all the doses under investigation augmented the percentage and the absolute count of CD19+ (B lymphocytes) in the peripheral lymphatic organs. The results of the study show that there is a relationship between the effect induced by bestatin and the dose of the drug as well as the number of doses applied. The strongest effect on the T and B lymphocyte subsets was noted after five injections of bestatin at doses of 1 and 0.1 mg/kg.


Subject(s)
Aminopeptidases/antagonists & inhibitors , B-Lymphocyte Subsets/drug effects , Leucine/analogs & derivatives , T-Lymphocyte Subsets/drug effects , Animals , B-Lymphocyte Subsets/enzymology , Dose-Response Relationship, Drug , Female , Leucine/pharmacology , Lymph Nodes , Mice , Spleen , T-Lymphocyte Subsets/enzymology , Thymus Gland
16.
J Immunol ; 187(10): 5114-22, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998451

ABSTRACT

A disintegrin and metalloproteinase 10 (ADAM10) is a zinc-dependent proteinase related to matrix metalloproteinases. ADAM10 has emerged as a key regulator of cellular processes by cleaving and shedding extracellular domains of multiple transmembrane receptors and ligands. We have developed B cell-specific ADAM10-deficient mice (ADAM10(B-/-)). In this study, we show that ADAM10 levels are significantly enhanced on germinal center B cells. Moreover, ADAM10(B-/-) mice had severely diminished primary and secondary responses after T-dependent immunization. ADAM10(B-/-) displayed impaired germinal center formation, had fewer follicular Th cells, decreased follicular dendritic cell networks, and altered chemokine expression in draining lymph nodes (LNs). Interestingly, when spleen and LN structures from immunized mice were analyzed for B and T cell localization, tissues structure was aberrant in ADAM10(B-/-) mice. Importantly, when ADAM10-deficient B cells were stimulated in vitro, they produced comparable Ab as wild type B cells. This result demonstrates that the defects in humoral responses in vivo result from inadequate B cell activation, likely because of the decrease in follicular Th cells and the changes in structure. Thus, ADAM10 is essential for the maintenance of lymphoid structure after Ag challenge.


Subject(s)
ADAM Proteins/physiology , Amyloid Precursor Protein Secretases/physiology , Immunity, Humoral , Membrane Proteins/physiology , ADAM Proteins/biosynthesis , ADAM Proteins/deficiency , ADAM10 Protein , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/deficiency , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , CHO Cells , Cricetinae , Germinal Center/enzymology , Germinal Center/immunology , Germinal Center/pathology , Immunity, Humoral/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Peyer's Patches/enzymology , Peyer's Patches/immunology , Peyer's Patches/pathology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Up-Regulation/genetics , Up-Regulation/immunology
17.
J Immunol ; 187(1): 230-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622861

ABSTRACT

The activation of the BCR, which initiates B cell activation, is triggered by Ag-induced self-aggregation and clustering of receptors at the cell surface. Although Ag-induced actin reorganization is known to be involved in BCR clustering in response to membrane-associated Ag, the underlying mechanism that links actin reorganization to BCR activation remains unknown. In this study, we show that both the stimulatory Bruton's tyrosine kinase (Btk) and the inhibitory SHIP-1 are required for efficient BCR self-aggregation. In Btk-deficient B cells, the magnitude of BCR aggregation into clusters and B cell spreading in response to an Ag-tethered lipid bilayer is drastically reduced, compared with BCR aggregation observed in wild-type B cells. In SHIP-1(-/-) B cells, although surface BCRs aggregate into microclusters, the centripetal movement and growth of BCR clusters are inhibited, and B cell spreading is increased. The persistent BCR microclusters in SHIP-1(-/-) B cells exhibit higher levels of signaling than merged BCR clusters. In contrast to the inhibition of actin remodeling in Btk-deficient B cells, actin polymerization, F-actin accumulation, and Wiskott-Aldrich symptom protein phosphorylation are enhanced in SHIP-1(-/-) B cells in a Btk-dependent manner. Thus, a balance between positive and negative signaling regulates the spatiotemporal organization of the BCR at the cell surface by controlling actin remodeling, which potentially regulates the signal transduction of the BCR. This study suggests a novel feedback loop between BCR signaling and the actin cytoskeleton.


Subject(s)
Actins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Actins/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cell Movement/genetics , Cell Movement/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Inositol Polyphosphate 5-Phosphatases , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Protein-Tyrosine Kinases/physiology , Receptors, Antigen, B-Cell/biosynthesis , Wiskott-Aldrich Syndrome Protein/metabolism
18.
Eur J Immunol ; 41(3): 657-68, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21287550

ABSTRACT

B cells have multiple functions in adaptive immunity, including antibody production, antigen presentation and regulation of T-cell responses. Recent evidences indicate that B cells have more subsets than previously thought and may have non-classical functions, such as involvement in innate immunity and immune regulation; however, how B cells respond to microbial infection and elicit innate defense remain unclear. In this study, we identified a new subset of PDCA-1(+) Siglec-H(-) CD19(+) B cells in mice during the early period of bacterial infection with Listeria monocytogenes. PDCA-1(+) Siglec-H(-) CD19(+) B cells secreted large amounts of IFN-α and thus facilitated IFN-γ production and cytotoxicity function of natural killer (NK) cells via IFN-α. B-cell-deficient Btk(-/-) mice were incapable of producing PDCA-1(+) CD19(+) B cells, and were more sensitive to L. monocytogenes infection. Adoptive transfer of PDCA-1(+) CD19(+) B cells to Btk(-/-) mice normalized their resistance to L. monocytogenes infection. Furthermore, we found that macrophages were essential for the inducible generation of PDCA-1(+) Siglec-H(-) CD19(+) B cells via CD40-CD40L ligation. Therefore, we have identified a new subset of PDCA-1(+) Siglec-H(-) CD19(+) B cells, which enhances innate immune responses against bacterial infection by activating NK cells via secretion of IFN-α.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunity, Innate , Interferon-alpha/biosynthesis , Killer Cells, Natural/immunology , Adoptive Transfer , Agammaglobulinaemia Tyrosine Kinase , Animals , Antigens, CD19/metabolism , Antigens, Differentiation/metabolism , B-Lymphocyte Subsets/enzymology , Base Sequence , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Antigens/metabolism , CD40 Ligand/deficiency , CD40 Ligand/genetics , CD40 Ligand/metabolism , DNA Primers/genetics , In Vitro Techniques , Interferon-alpha/deficiency , Interferon-alpha/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lectins/metabolism , Listeriosis/enzymology , Listeriosis/immunology , Lymphocyte Activation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/metabolism
19.
J Immunol ; 186(4): 1943-50, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21228350

ABSTRACT

B cell development involves rapid cellular proliferation, gene rearrangements, selection, and differentiation, and it provides a powerful model to study DNA repair processes in vivo. Analysis of the contribution of the base excision repair pathway in lymphocyte development has been lacking primarily owing to the essential nature of this repair pathway. However, mice deficient for the base excision repair enzyme, apurinic/apyrimidinic endonuclease 2 (APE2) protein develop relatively normally, but they display defects in lymphopoiesis. In this study, we present an extensive analysis of bone marrow hematopoiesis in mice nullizygous for APE2 and find an inhibition of the pro-B to pre-B cell transition. We find that APE2 is not required for V(D)J recombination and that the turnover rate of APE2-deficient progenitor B cells is nearly normal. However, the production rate of pro- and pre-B cells is reduced due to a p53-dependent DNA damage response. FACS-purified progenitors from APE2-deficient mice differentiate normally in response to IL-7 in in vitro stromal cell cocultures, but pro-B cells show defective expansion. Interestingly, APE2-deficient mice show a delay in recovery of B lymphocyte progenitors following bone marrow depletion by 5-fluorouracil, with the pro-B and pre-B cell pools still markedly decreased 2 wk after a single treatment. Our data demonstrate that APE2 has an important role in providing protection from DNA damage during lymphoid development, which is independent from its ubiquitous and essential homolog APE1.


Subject(s)
B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Endonucleases/physiology , Fluorouracil/administration & dosage , Hematopoietic Stem Cells/enzymology , Lymphocyte Subsets/enzymology , Lymphopoiesis/immunology , Animals , B-Lymphocyte Subsets/drug effects , Cells, Cultured , Coculture Techniques , DNA Damage/drug effects , DNA Damage/genetics , DNA Damage/immunology , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair/immunology , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Endonucleases/deficiency , Endonucleases/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Lymphocyte Depletion , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Lymphopoiesis/drug effects , Lymphopoiesis/genetics , Mice , Mice, Knockout , Multifunctional Enzymes , Myelopoiesis/drug effects , Myelopoiesis/genetics , Myelopoiesis/immunology , Tumor Suppressor Protein p53/physiology
20.
J Immunol ; 186(2): 1279-88, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21160042

ABSTRACT

IL-1R-associated kinases (IRAKs) are important mediators of MyD88-dependent signaling by the TLR/IL-1R superfamily and facilitate inflammatory responses. IRAK4 and IRAK1 function as active kinases and as scaffolds for protein-protein interactions. We report that although IRAK1/4 kinase activity is essential for human plasmacytoid dendritic cell (pDC) activation, it is dispensable in B, T, dendritic, and monocytic cells, which is in contrast with an essential active kinase role in comparable mouse cell types. An IRAK1/4 kinase inhibitor abrogated TLR7/9-induced IFN-α responses in both mouse and human pDCs, but other human immune cell populations activated via TLR7/9 or IL-1R were refractory to IRAK4 kinase inhibition. Gene ablation experiments using small interfering RNA demonstrated an essential scaffolding role for IRAK1 and IRAK4 in MyD88-dependent signaling. Finally, we demonstrate that autoimmune patient (systemic lupus erythematosus and rheumatoid arthritis) serum activates both pDC and B cells, but IRAK1/4 kinase inhibition affects only the pDC response, underscoring the differential IRAK1/4 functional requirements in human immune cells. These data reveal important species differences and elaborate cell type requirements for IRAK1/4 kinase activity.


Subject(s)
Antigen-Antibody Complex/blood , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Interleukin-1 Receptor-Associated Kinases/physiology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Animals , Arthritis, Rheumatoid/enzymology , Autoantibodies/biosynthesis , Autoantibodies/blood , B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Cells, Cultured , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Humans , Interleukin-17/biosynthesis , Interleukin-17/blood , Lupus Erythematosus, Systemic/enzymology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Monocytes/enzymology , Monocytes/immunology , Monocytes/pathology , Signal Transduction/immunology , Th1 Cells/enzymology , Th1 Cells/immunology , Toll-Like Receptor 9/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...