Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 533(1-2): 18-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23454348

ABSTRACT

B cell antigen receptor (BCR) signaling is positively and negatively regulated by various cell surface receptors such as CD19 and CD45. Functional analysis of these receptors has been performed using gene targeting technology, which is a valid approach to elucidate their functions. However, this type of analysis is restricted when multiple molecules are evaluated simultaneously. From a different perspective, synthetic biology provides a high degree of freedom for analyzing various molecules. Here we developed a system to reconstruct BCR signaling using the J558L myeloma cell line in combination with the protein-based Ca(2+) indicator YC3.60. BCR-reconstituted J558L cells harboring YC3.60 (J558Lµv11 cells) permitted monitoring of Ca(2+) mobilization. Reconstituting CD19 in J558Lµv11 cells resulted in detectable BCR-induced Ca(2+) mobilization but with kinetics different from that of CD45-expressing cells. Furthermore, we evaluated the validity of the J558L system by proteomic analysis of tyrosine-phosphorylated proteins after antigen stimulation. Identification of more than 100 BCR-induced tyrosine-phosphorylated proteins in J558Lµv11 cells revealed a similarity to that observed in B cells, and a novel member, non-receptor protein tyrosine kinase Fer, was found. Thus, this reconstruction system using J558L cells appeared to be valid for comprehensively investigating BCR signaling.


Subject(s)
Multiple Myeloma/pathology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antigens, CD19/metabolism , Calcium Signaling , Cell Line, Tumor , Down-Regulation , Kinetics , Leukocyte Common Antigens/metabolism , Mice , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphorylation , Tyrosine/metabolism , src-Family Kinases/metabolism
2.
J Biol Chem ; 287(3): 2045-54, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22130676

ABSTRACT

Previous studies indicate that STAT5 expression is required for mast cell development, survival, and IgE-mediated function. STAT5 tyrosine phosphorylation is swiftly and transiently induced by activation of the high affinity IgE receptor, FcεRI. However, the mechanism for this mode of activation remains unknown. In this study we observed that STAT5 co-localizes with FcεRI in antigen-stimulated mast cells. This localization was supported by cholesterol depletion of membranes, which ablated STAT5 tyrosine phosphorylation. Through the use of various pharmacological inhibitors and murine knock-out models, we found that IgE-mediated STAT5 activation is dependent upon Fyn kinase, independent of Syk, PI3K, Akt, Bruton's tyrosine kinase, and JAK2, and enhanced in the context of Lyn kinase deficiency. STAT5 immunoprecipitation revealed that unphosphorylated protein preassociates with Fyn and that this association diminishes significantly during mast cell activation. SHP-1 tyrosine phosphatase deficiency modestly enhanced STAT5 phosphorylation. This effect was more apparent in the absence of Gab2, a scaffolding protein that docks with multiple negative regulators, including SHP-1, SHP-2, and Lyn. Targeting of STAT5A or B with specific siRNA pools revealed that IgE-mediated mast cell cytokine production is selectively dependent upon the STAT5B isoform. Altogether, these data implicate Fyn as the major positive mediator of STAT5 after FcεRI engagement and demonstrate importantly distinct roles for STAT5A and STAT5B in mast cell function.


Subject(s)
Cytokines/biosynthesis , Mast Cells/metabolism , Receptors, IgE/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Agammaglobulinaemia Tyrosine Kinase , Animals , Cells, Cultured , Cholesterol/genetics , Cholesterol/metabolism , Cytokines/genetics , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mast Cells/cytology , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, IgE/genetics , STAT5 Transcription Factor/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
Mol Immunol ; 48(4): 472-80, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21044800

ABSTRACT

SHP-1 plays an important role for the regulation of signaling from various hematopoietic cell receptors. In this study, we examined IL-3-induced cell proliferation and IL-3 depletion-induced apoptosis in bone marrow-derived mast cells (BMMC) established from motheaten (me) that lack SHP-1 expression, viable motheaten (me(v)) expressing phosphatase-deficient SHP-1, and wild-type (WT) mice. When BMMC were stimulated with IL-3, increased ERK activation was evident in resting state and sustained in me-BMMC relative to WT-BMMC. ERK is known to be involved in the regulation of cell proliferation and apoptosis in some cells. In accordance with sustained ERK activation, apoptosis was decreased in me- and me(v)-BMMC compared with WT-BMMC. In contrast to the predicted role of ERK as a pro-survival molecule, IL-3-induced cell proliferation was much lower in me- and me(v)-BMMC than WT-BMMC. Stimulation with lower concentration of IL-3 or addition of PD98059, a MEK inhibitor, to the culture resulted in the suppression of decreased apoptosis and cell proliferation in me- and me(v)-BMMC. Collectively, these results suggest that SHP-1 positively regulates IL-3-dependent mast cell proliferation and apoptosis by inhibiting ERK activity through its phosphatase activity. Furthermore, our results indicate that ERK would act as a negative regulator for cell proliferation and induce apoptosis when its activity is highly increased.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-3/pharmacology , Mast Cells/cytology , Mast Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , Animals , Biocatalysis/drug effects , Bone Marrow Cells/cytology , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Mast Cells/drug effects , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
4.
Mol Immunol ; 47(2-3): 222-32, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19875169

ABSTRACT

Src homology region 2 domain-containing phosphatase-1 (SHP-1) is known to act as a negative signal modulator in mast cells but its roles in cell survival and cell death are poorly understood. We previously reported that SHP-1 also positively regulates mast cell activation signaling by acting as an adaptor protein. In the present study, we examined whether SHP-1 plays a role in antigen (Ag)-induced activation-induced mast cell death. Bone marrow-derived mast cells (BMMCs) from SHP-1-deficient motheaten (me) mice (me-BMMCs) were significantly less susceptible to store-operated Ca(2+) channel (SOC) activation, Ag-induced cell death and DNA fragmentation than BMMCs from their wild-type littermates (WT-BMMCs). Subsequent experiments revealed that the differences in these cellular susceptibilities to SOC activation and cell death resulted from the extent of the mitochondrial permeability transition pore (mPTP) opening. Specifically, mPTP opening was sufficiently persistent in WT-BMMCs to evoke mitochondrial integrity disruption, while mPTP opening was too transient to cause the minimal mitochondrial integrity collapse in me-BMMCs. In addition, pro-survival signaling including activation of mitogen-activated protein kinases (MAPKs) such as the extracellular signal-regulated protein kinases, c-Jun NH(2) terminal kinases and p38 and the expression of Bcl-x(L) were significantly prolonged in me-BMMCs compared with WT-BMMCs. Taken together, these data demonstrate that a lack of SHP-1 prevents the mPTP-mediated mitochondrial integrity collapse and augments anti-apoptotic signaling such as MAPKs and Bcl-x(L). These findings suggest that SHP-1 positively regulates mitochondrial death pathways and negatively regulates pro-survival signaling pathways.


Subject(s)
Antigens/immunology , Apoptosis/immunology , Mast Cells/cytology , Mast Cells/enzymology , Mitochondria/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Signal Transduction/immunology , Animals , Bone Marrow Cells/cytology , Calcium Channels/metabolism , Cell Survival , Ion Channel Gating , Mast Cells/immunology , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency
5.
J Immunol ; 181(8): 5414-24, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832698

ABSTRACT

Src homology region 2 domain-containing phosphatase 1 (SHP-1), a cytoplasmic protein tyrosine phosphatase, plays an important role for the regulation of signaling from various hematopoietic cell receptors. Although SHP-1 is shown to be a negative signal modulator in mast cells, its precise molecular mechanisms are not well defined. To elucidate how SHP-1 regulates mast cell signaling, we established bone marrow-derived mast cells from SHP-1-deficient motheaten and wild-type mice and analyzed downstream signals induced by cross-linking of high affinity IgE receptor, Fc epsilonRI. Upon Fc epsilonRI ligation, motheaten-derived bone marrow-derived mast cells showed enhanced tyrosine phosphorylation of Src homology region 2 domain-containing leukocyte protein of 76 kDa (SLP-76) and linker for activation of T cells, activation of mitogen-activated protein kinases and gene transcription and production of cytokine. Because the activity of Syk, responsible for the phosphorylation of SLP-76 and linker for activation of T cells, is comparable irrespective of SHP-1, both molecules might be substrates of SHP-1 in mast cells. Interestingly, the absence of SHP-1 expression disrupted the association between SLP-76 and phospholipase Cgamma, which resulted in the decreased phospholipase Cgamma phosphorylation, calcium mobilization, and degranulation. Collectively, these results suggest that SHP-1 regulates Fc epsilonRI-induced downstream signaling events both negatively and positively by functioning as a protein tyrosine phosphatase and as an adaptor protein contributing to the formation of signaling complex, respectively.


Subject(s)
Cell Degranulation/immunology , Gene Expression Regulation, Enzymologic/immunology , Mast Cells/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Receptors, IgE/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium/immunology , Calcium/metabolism , Cell Degranulation/genetics , Extracellular Signal-Regulated MAP Kinases , Gene Expression Regulation, Enzymologic/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mast Cells/enzymology , Mice , Mice, Mutant Strains , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Receptors, IgE/genetics , Receptors, IgE/metabolism , Signal Transduction/genetics , Syk Kinase , T-Lymphocytes/enzymology , T-Lymphocytes/immunology
6.
Int Immunol ; 19(9): 1049-61, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17675340

ABSTRACT

Src homology region 2-domain-containing phosphatase-1 (SHP-1) plays an important role in the regulation of signaling from various receptors in hematopoietic cells. In mast cells, SHP-1 has been shown to negatively regulate the initial signaling triggered by high-affinity receptor for IgE (FcepsilonRI) and positively regulate downstream outputs. To clarify the molecular mechanisms of SHP-1 in mast cells, we determined substrates for SHP-1 by using the substrate-trapping approach. When phosphatase-inactive SHP-1 was over-expressed in rat basophilic leukemia (RBL)-2H3 cells, tyrosine phosphorylation of a 68-kDa protein was enhanced before and after FcepsilonRI aggregation. Immunoprecipitation and western blot analyses revealed that this protein is SHP-1, either endogenous or ectopically expressed. FcepsilonRI-induced activation of Lyn and Syk was comparable between cells expressing wild-type (wt) and phosphatase-inactive SHP-1. In vitro phosphatase assay and combined transfection, immunoprecipitation and immunoblot analyses showed that tyrosine 536 of SHP-1 was potent phosphorylation site and that SHP-1 could dephosphorylate this site that had been phosphorylated by Lyn. Furthermore, the phosphatase activity of SHP-1 immunoprecipitated from cells expressing a phosphatase-inactive SHP-1 was increased compared with that from vector-transfected or wt SHP-1-expressing cells. Finally, expression of phosphatase-inactive SHP-1 resulted in decreased activation of mitogen-activated protein kinases and suppressed transcription of cytokine genes, whereas wt SHP-1 enhanced these processes. Taken collectively, these results suggest that SHP-1 may be a physiological substrate of SHP-1 in RBL-2H3 cells and that dephosphorylation of SHP-1 leads to a decrease in its catalytic activity and an enhancement of downstream signaling. A negative autoregulatory circuit of SHP-1 may contribute to mast cell regulation.


Subject(s)
Leukemia, Basophilic, Acute/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Down-Regulation/immunology , Gene Expression Profiling , Intracellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Mitogen-Activated Protein Kinases/immunology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein-Tyrosine Kinases/immunology , Rats , Receptors, IgE/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/immunology , Syk Kinase , Transcription, Genetic , src-Family Kinases/immunology
8.
Eur J Immunol ; 35(2): 644-54, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15668918

ABSTRACT

Despite the important role in the development and activation of T cells, NK cells, mast cells, and macrophages, the expression and function of SLP-76 in B cells have been largely unknown. Here we demonstrate that SLP-76 is expressed in all mouse B cell lines tested and in normal splenic B cells, and serves as an SHP-1 substrate. Dephosphorylation of SLP-76 by SHP-1 inhibits its association with Nck, down-regulating c-Jun N-terminal kinase (JNK) activation and exerting a positive effect on apoptosis. Knockdown of SLP-76 in WEHI-231 cells by small interfering RNA attenuated JNK activation, but showed little effects on extracellular signal-regulated kinase (ERK) or p38 activation. Although WEHI-231 does not express linker for activation of T cells (LAT), SLP-76 localizes in membrane fraction, which increases following B cell receptor (BCR) cross-linking. Further analyses revealed that SLP-76 complexed with Gads is associated with tyrosine-phosphorylated CD22 through the SH2 domains of SLP-76 and Gads. Given that SHP-1 binds to CD22 upon BCR ligation, our findings suggest that dephosphorylation of SLP-76 recruited to CD22 by SHP-1 inhibits BCR-induced JNK activation, dictating apoptosis.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/metabolism , Cell Adhesion Molecules/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lectins/metabolism , Lymphocyte Activation/physiology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Intracellular Signaling Peptides and Proteins , Mice , Oncogene Proteins/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases , Receptors, Antigen, B-Cell/metabolism , Sialic Acid Binding Ig-like Lectin 2 , Signal Transduction/physiology , Tyrosine/metabolism
9.
Int Immunol ; 16(7): 971-82, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15148289

ABSTRACT

CD72 is a 45 kDa B cell-specific type II transmembrane protein of the C-type lectin superfamily. It was originally defined as a receptor-like molecule that regulates B cell activation and differentiation; however, its precise function remains unclear since more recent functional analyses, including a gene targeting study, suggest that CD72 may serve as a negative or a positive regulator of B cell signaling. In the present study, we analyzed the cell-autonomous function of CD72 in B cell receptor (BCR) signaling using CD72-deficient cells generated from mature BAL-17 cells. We found that BCR-mediated phosphorylation of CD19, Btk, Vav and phospholipase Cgamma2 and association of CD19 with phosphatidylinositol-3 kinase were impaired in CD72-deficient cells. Inositol trisphosphate synthesis was normally induced initially but ablated at 1 min of stimulation in CD72-deficient cells. In the event, Ca(2+) release from intracellular stores remained intact, though influx of extracellular Ca(2+) was severely impaired in CD72-deficient cells. Furthermore, BCR-evoked activation of mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinase and c-Jun NH(2)-terminal kinase, and growth inhibition in BAL-17 cells were blocked in the absence of CD72. Significantly, these effects were largely reversed by re-expression of CD72. Thus, CD72 appears to exert a positive effect on BCR signaling pathways leading to Ca(2+) influx and MAPK activation, which in turn may determine the fate of BAL-17 cells.


Subject(s)
Calcium Signaling/immunology , Cell Growth Processes/immunology , Lectins, C-Type/immunology , Mitogen-Activated Protein Kinases/immunology , Receptors, Antigen, B-Cell/deficiency , Receptors, Antigen, B-Cell/immunology , Animals , Cell Cycle Proteins/metabolism , Cell Line , Mice , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-vav
10.
Blood ; 103(4): 1425-32, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14563648

ABSTRACT

CD45 is a key protein tyrosine phosphatase regulating Src-family protein tyrosine kinases (Src-PTKs) in lymphocytes; precisely how it exerts its effect remains controversial, however. We previously demonstrated that CD45 negatively regulates Lyn in the WEHI-231 B-cell line. Here we show that negative regulation by CD45 is physiologically significant in B cells and that some CD45 is constitutively associated with glycolipid-enriched microdomains (GEMs), where it inhibits Src-PTKs by dephosphorylating both the negative and the positive regulatory sites. Upon B-cell receptor (BCR) ligation, however, CD45 dissociates from GEMs within 30 seconds, inducing phosphorylation of 2 regulatory sites and activation of Src-PTKs, but subsequently reassociates with the GEMs within 15 minutes. Disruption of GEMs with methyl-beta-cyclodextrin results in abrogation of BCR-induced apoptosis in WEHI-231 cells, suggesting GEMs are critical to signals leading to the fate determination. We propose that the primary function of CD45 is inhibition of Src-PTKs and that the level of Src-PTK activation and the B-cell fate are determined in part by dynamic behavior of CD45 with respect to GEMs.


Subject(s)
B-Lymphocytes/enzymology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , src-Family Kinases/metabolism , Animals , Apoptosis/immunology , B-Lymphocytes/cytology , Cell Line , Membrane Microdomains , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Spleen/cytology , Spleen/immunology
11.
J Immunol ; 169(2): 778-86, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12097380

ABSTRACT

Src homology region 2 domain-containing phosphatase 1 (SHP-1) is a key mediator in lymphocyte differentiation, proliferation, and activation. We previously showed that B cell linker protein (BLNK) is a physiological substrate of SHP-1 and that B cell receptor (BCR)-induced activation of c-Jun NH(2)-terminal kinase (JNK) is significantly enhanced in cells expressing a form of SHP-1 lacking phosphatase activity (SHP-1-C/S). In this study, we confirmed that SHP-1 also exerts negative regulatory effects on JNK activation in splenic B cells. To further clarify the role of SHP-1 in B cells, we examined how dephosphorylation of BLNK by SHP-1 affects downstream signaling events. When a BLNK mutant (BLNK Delta N) lacking the NH(2)-terminal region, which contains four tyrosine residues, was introduced in SHP-1-C/S-expressing WEHI-231 cells, the enhanced JNK activation was inhibited. Among candidate proteins likely to regulate JNK activation through BLNK, Nck adaptor protein was found to associate with tyrosine-phosphorylated BLNK and this association was more pronounced in SHP-1-C/S-expressing cells. Furthermore, expression of dominant-negative forms of Nck inhibited BCR-induced JNK activation. Finally, BCR-induced apoptosis was suppressed in SHP-1-C/S-expressing cells and coexpression of Nck SH2 mutants or a dominant-negative form of SEK1 reversed this phenotype. Collectively, these results suggest that SHP-1 acts on BLNK, modulating its association with Nck, which in turn negatively regulates JNK activation but exerts a positive effect on apoptosis.


Subject(s)
Apoptosis/immunology , Carrier Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Protein Tyrosine Phosphatases/physiology , Receptors, Antigen, B-Cell/physiology , Up-Regulation/immunology , src Homology Domains/immunology , Adaptor Proteins, Signal Transducing , Adjuvants, Immunologic/physiology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Carrier Proteins/physiology , Down-Regulation/immunology , Enzyme Activation/immunology , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/physiology , Oncogene Proteins/physiology , Peptide Fragments/physiology , Phosphoproteins/physiology , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Spleen/cytology , Spleen/enzymology , Spleen/immunology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...