Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Rheumatology (Oxford) ; 60(7): 3420-3431, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33230538

ABSTRACT

OBJECTIVE: RA encompasses a complex, heterogeneous and dynamic group of diseases arising from molecular and cellular perturbations of synovial tissues. The aim of this study was to decipher this complexity using an integrative systems approach and provide novel insights for designing stratified treatments. METHODS: An RNA sequencing dataset of synovial tissues from 152 RA patients and 28 normal controls was imported and subjected to filtration of differentially expressed genes, functional enrichment and network analysis, non-negative matrix factorization, and key driver analysis. A naïve Bayes classifier was applied to the independent datasets to investigate the factors associated with treatment outcome. RESULTS: A matrix of 1241 upregulated differentially expressed genes from RA samples was classified into three subtypes (C1-C3) with distinct molecular and cellular signatures. C3 with prominent immune cells and proinflammatory signatures had a stronger association with the presence of ACPA and showed a better therapeutic response than C1 and C2, which were enriched with neutrophil and fibroblast signatures, respectively. C2 was more occupied by synovial fibroblasts of destructive phenotype and carried highly expressed key effector molecules of invasion and osteoclastogenesis. CXCR2, JAK3, FYN and LYN were identified as key driver genes in C1 and C3. HDAC, JUN, NFKB1, TNF and TP53 were key regulators modulating fibroblast aggressiveness in C2. CONCLUSIONS: Deep phenotyping of synovial heterogeneity captured comprehensive and discrete pathophysiological attributes of RA regarding clinical features and treatment response. This result could serve as a template for future studies to design stratified approaches for RA patients.


Subject(s)
Arthritis, Rheumatoid/genetics , Fibroblasts/metabolism , Neutrophils/metabolism , Synovial Membrane/metabolism , Anti-Citrullinated Protein Antibodies/immunology , Arthritis, Rheumatoid/immunology , Bayes Theorem , Databases, Genetic , Fibroblasts/immunology , Gene Expression Profiling , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Humans , Janus Kinase 3/genetics , Janus Kinase 3/immunology , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/immunology , Neutrophils/immunology , Osteogenesis/genetics , Osteogenesis/immunology , Phenotype , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/immunology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Synovial Membrane/immunology , Systems Analysis , Transcriptome , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , src-Family Kinases/genetics , src-Family Kinases/immunology
2.
Toxicol Appl Pharmacol ; 383: 114763, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31526816

ABSTRACT

Mast cells (MCs) play an important role as effector cells that cause allergic responses in allergic diseases. For these reasons, MC is considered an attractive therapeutic target for allergic disease treatment. In this study, we investigated the inhibitory effect of WZ3146, N-[3-[5-chloro-2-[4-(4-methylpiperazin-1-yl)anilino]pyrimidin-4-yl]oxyphenyl]prop-2-enamide, and the mechanisms of its actions on the MC activation and IgE-mediated allergic response by using three types of MCs such as rat basophilic leukemia (RBL)-2H3 cells, mouse bone marrow mast cells (BMMCs), and human Laboratory of Allergic Diseases 2 (LAD2) cells. WZ3146 inhibited antigen-stimulated degranulation in a dose-dependent manner (IC50, ~ 0.35 µM for RBL-2H3 cells; ~ 0.39 µM for BMMCs; ~ 0.41 for LAD2 cells). WZ3146 also suppressed the production of histamine, tumor necrosis factor (TNF)-α and interleukin (IL)-6, which mediate various allergic responses, in a dose-dependent manner. As the mechanism of WZ3146 to inhibit MCs, it inhibited the activation of spleen tyrosine kinase (Syk) and the downstream signaling proteins of Syk such as linker for activation of T cell (LAT) and phospholipase (PL) Cγ1 in the signaling pathway of FcεRI. In addition, WZ3146 inhibited the activation of Akt, extracellular signal-regulated kinase (ERK)1/2, p38, and c-Jun N-terminal kinase (JNK). However, WZ3146 did not inhibit degranulation of MCs by thapsigargin or ionomycin, which increase calcium concentration in cytosol. Notably, WZ3146 inhibited the activity of Lyn and Fyn, but not Syk. In an following animal experiment, WZ3146 inhibited IgE-dependent passive cutaneous anaphylaxis (PCA) in a dose-dependent manner (ED50, ~ 20 mg/kg). Taken together, in this study we show that the pyrimidine derivative, WZ3146, inhibits the IgE-mediated allergic response by inhibiting Lyn and Fyn Src-family kinases, which are initially activated by antigen stimulation in MCs. Therefore, we propose that WZ3146 could be used as a new therapeutic agent for the treatment of allergic diseases.


Subject(s)
Hypersensitivity/metabolism , Immunoglobulin E/metabolism , Mast Cells/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Pyrimidines/pharmacology , src-Family Kinases/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Male , Mast Cells/drug effects , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/immunology , Pyrimidines/chemistry , Rats , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/immunology
3.
Biochem Biophys Res Commun ; 495(1): 1214-1220, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180007

ABSTRACT

The high concentrations of nitric oxide (NO) produced by inducible nitric oxide synthase (iNOS) in activated glial cells in response to neuroinflammatory stimuli have neurotoxic effects on the brain. At basal levels, iNOS expression is low, and proinflammatory stimuli induce iNOS expression in astrocytes, microglia, and oligodendrocytes. Fyn, a non-receptor tyrosine kinase, regulates iNOS expression in several types of immune cells. However, its role in stimulated astrocytes is less clear. In this study, we investigated the role of Fyn in the regulation of lipopolysaccharide (LPS)-induced iNOS expression in astrocytes from mice and rats. Intracerebroventricular LPS injections in cortical regions enhanced iNOS mRNA and protein levels, which were increased in Fyn-deficient mice. Accordingly, LPS-induced nitrite production was enhanced in primary astrocytes cultured from Fyn-deficient mice or rats. Similar results were observed in cultured astrocytes after the siRNA-induced knockdown of Fyn expression. Finally, we observed increased LPS-induced extracellular signal-regulated protein kinase (ERK) activation in Fyn-deficient astrocytes. These results suggested that Fyn has a regulatory role in iNOS expression in astrocytes during neuroinflammatory responses.


Subject(s)
Astrocytes/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Gene Expression Regulation, Enzymologic/immunology , Inflammation Mediators/immunology , Lipopolysaccharides/pharmacology , Nitric Oxide Synthase Type II/immunology , Proto-Oncogene Proteins c-fyn/immunology , Animals , Astrocytes/drug effects , Cells, Cultured , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
4.
Nat Commun ; 8(1): 246, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28811476

ABSTRACT

Immunoreceptors can transduce either inhibitory or activatory signals depending on ligand avidity and phosphorylation status, which is modulated by the protein kinases Lyn and Fyn. Here we show that Lyn and Fyn control immune receptor signaling status. SHP-1 tyrosine 536 phosphorylation by Lyn activates the phosphatase promoting inhibitory signaling through the immunoreceptor. By contrast, Fyn-dependent phosphorylation of SHP-1 serine 591 inactivates the phosphatase, enabling activatory immunoreceptor signaling. These SHP-1 signatures are relevant in vivo, as Lyn deficiency exacerbates nephritis and arthritis in mice, whereas Fyn deficiency is protective. Similarly, Fyn-activating signature is detected in patients with lupus nephritis, underlining the importance of this Lyn-Fyn balance. These data show how receptors discriminate negative from positive signals that respectively result in homeostatic or inflammatory conditions.Src-family kinases Fyn and Lyn are signaling components downstream of ITAM-bearing antigen receptors. Here the authors show that by phosphorylating SHP-1 at different residues, Lyn and Fyn can have opposing regulatory effects on ITAM receptors.


Subject(s)
Inflammation/enzymology , Proto-Oncogene Proteins c-fyn/immunology , src-Family Kinases/immunology , Animals , Female , Homeostasis , Humans , Inflammation/genetics , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-fyn/genetics , Signal Transduction , src-Family Kinases/genetics
5.
Gene ; 579(2): 193-200, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26743129

ABSTRACT

The non-receptor protein tyrosine kinase (nrPTK) Fyn, a member of the avian sarcoma virus transforming gene (Src) kinase family, plays a very significant role in cell growth, survival, apoptosis, tumor formation and immune response. In this study, a homolog of nrPTK Fyn was identified for the first time in the lamprey, Lampetra japonica and was named "Lja-Fyn". The cDNA fragment of lamprey lja-fyn contains a 1611-bp open reading frame, which encodes a protein of 537 amino acids. Multiple sequence alignment analysis showed that it shares four conserved domains (Src homology (SH) 4, SH3, SH2 and protein kinases catalytic domains) and a variable unique domain with vertebrates Fyn molecules. Though Lja-Fyn has high sequence similarity with typical Fyn and Yes molecules of jawed vertebrates, the identities among Lja-Fyn and typical Fyn molecules in unique domain are relatively higher than that among Lja-Fyn and typical Yes molecules. The result indicates that Lja-Fyn is a homolog of Fyn rather than Yes. The phylogenetic analysis showed that Fyn, Yes and Src molecules are grouped into three distinct phylogenetic clusters, and Lja-Fyn is grouped as a single branch in Fyn cluster. The real-time quantitative PCR assay revealed the wide distribution of the lja-fyn mRNA in lamprey immune related tissues. After stimulation with mixed antigens, the levels of lja-fyn mRNA were obviously up-regulated in the gill and lymphocyte-like cells, and the similar results were got by western blot analysis of Lja-Fyn protein expression. These results indicated that nrPTK Lja-Fyn was likely to be involved in immune response. Furthermore, our present findings also provide the necessary information for understanding the distinction between lamprey Lja-Fyn and other members of jawed vertebrates in Src family.


Subject(s)
Immunity, Innate/genetics , Lampreys/genetics , Proto-Oncogene Proteins c-fyn/genetics , Animals , Lampreys/immunology , Proto-Oncogene Proteins c-fyn/immunology , Sequence Homology, Amino Acid
6.
J Leukoc Biol ; 97(6): 1089-99, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25877924

ABSTRACT

Despite progress in understanding enteric inflammation, current therapies, although effective in many patients with inflammatory bowel disease (IBD), have significant side-effects, and, in many patients, it is refractory to treatment. The Src kinase Fyn mediated IFN-γ-induced increased permeability in model epithelia, and so we hypothesized that inhibition of Fyn kinase would be anti-colitic. Mice [B6.129SF2/J wild-type (WT), Fyn KO, or chimeras] received 2.5% dextran sodium sulfate (DSS) or normal water for 10 d and were necropsied immediately or 3 d later. Gut permeability was assessed by FITC-dextran flux, colitis by macroscopic and histologic parameters, and immune cell status by cytokine production and CD4(+) T cell Foxp3 expression. Fyn KO mice consistently displayed significantly worse DSS-induced disease than WT, correlating with decreased IL-10 and increased IL-17 in splenocytes and the gut; Fyn KO mice failed to thrive after removal of the DSS water. Analysis of chimeric mice indicated that the increased sensitivity to DSS was due to the lack of Fyn kinase in hematopoietic, but not stromal, cells, in accordance with Fyn(+) T cell increases in WT mice exposed to DSS and Fyn KO mice having a reduced number of CD4(+)Foxp3(+) cells in baseline or colitic conditions and a reduced capacity to induce Foxp3 expression in vitro. Other experiments suggest that the colonic microbiota in Fyn KO mice is not preferentially colitogenic. Contrary to our expectation, the absence of Fyn kinase resulted in greater DSS-induced disease, and analysis of chimeric mice indicated that leukocyte Fyn kinase is beneficial in limiting colitis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Colon/immunology , Epithelial Cells/immunology , Proto-Oncogene Proteins c-fyn/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Epithelial Cells/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Knockout , Permeability , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/genetics , Signal Transduction
7.
Nat Immunol ; 16(4): 354-65, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25729923

ABSTRACT

Interleukin 37 (IL-37) and IL-1R8 (SIGIRR or TIR8) are anti-inflammatory orphan members of the IL-1 ligand family and IL-1 receptor family, respectively. Here we demonstrate formation and function of the endogenous ligand-receptor complex IL-37-IL-1R8-IL-18Rα. The tripartite complex assembled rapidly on the surface of peripheral blood mononuclear cells upon stimulation with lipopolysaccharide. Silencing of IL-1R8 or IL-18Rα impaired the anti-inflammatory activity of IL-37. Whereas mice with transgenic expression of IL-37 (IL-37tg mice) with intact IL-1R8 were protected from endotoxemia, IL-1R8-deficient IL-37tg mice were not. Proteomic and transcriptomic investigations revealed that IL-37 used IL-1R8 to harness the anti-inflammatory properties of the signaling molecules Mer, PTEN, STAT3 and p62(dok) and to inhibit the kinases Fyn and TAK1 and the transcription factor NF-κB, as well as mitogen-activated protein kinases. Furthermore, IL-37-IL-1R8 exerted a pseudo-starvational effect on the metabolic checkpoint kinase mTOR. IL-37 thus bound to IL-18Rα and exploited IL-1R8 to activate a multifaceted intracellular anti-inflammatory program.


Subject(s)
Interleukin-18 Receptor alpha Subunit/immunology , Interleukin-1/immunology , Leukocytes, Mononuclear/immunology , Receptors, Interleukin-1/immunology , Signal Transduction/immunology , Animals , Cell Line , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1/genetics , Interleukin-18 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-18 Receptor alpha Subunit/genetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/pathology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , c-Mer Tyrosine Kinase
8.
Sci Signal ; 8(368): ra28, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25783157

ABSTRACT

Subsets of B cells inhibit various immune responses through their production of the cytokine interleukin-10 (IL-10). We found that IL-10-producing CD5(+) B cells suppressed the immunoglobulin E (IgE)- and antigen-mediated activation of mast cells in vitro as well as allergic responses in mice in an IL-10-dependent manner. Furthermore, the suppressive effect of these B cells on mast cells in vitro and in vivo depended on direct cell-to-cell contact through the costimulatory receptor CD40 on CD5(+) B cells and the CD40 ligand on mast cells. This contact enhanced the production of IL-10 by the CD5(+) B cells. Through activation of the Janus-activated kinase-signal transducer and activator of transcription 3 pathway, IL-10 decreased the abundance of the kinases Fyn and Fgr and inhibited the activation of the downstream kinase Syk in mast cells. Together, these findings suggest that an important function of IL-10-producing CD5(+) B cells is inhibiting mast cells and IgE-mediated allergic responses.


Subject(s)
B-Lymphocytes/immunology , CD5 Antigens/immunology , Hypersensitivity/immunology , Immunoglobulin E/immunology , Interleukin-10/immunology , Mast Cells/immunology , Animals , B-Lymphocytes/pathology , CD40 Antigens/genetics , CD40 Antigens/immunology , CD5 Antigens/genetics , Hypersensitivity/genetics , Hypersensitivity/pathology , Immunoglobulin E/genetics , Interleukin-10/genetics , Mast Cells/pathology , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction/genetics , Signal Transduction/immunology , src-Family Kinases/genetics , src-Family Kinases/immunology
9.
PLoS One ; 9(10): e109800, 2014.
Article in English | MEDLINE | ID: mdl-25289695

ABSTRACT

Engagement of high-affinity immunoglobulin E receptors (FcεRI) activates two signaling pathways in mast cells. The Lyn pathway leads to recruitment of Syk and to calcium mobilization whereas the Fyn pathway leads to phosphatidylinositol 3-kinase recruitment. Mapping the connections between both pathways remains an important task to be completed. We previously reported that Phospholipid Scramblase 1 (PLSCR1) is phosphorylated on tyrosine after cross-linking FcεRI on RBL-2H3 rat mast cells, amplifies mast cell degranulation, and is associated with both Lyn and Syk tyrosine kinases. Here, analysis of the pathway leading to PLSCR1 tyrosine phosphorylation reveals that it depends on the FcRγ chain. FcεRI aggregation in Fyn-deficient mouse bone marrow-derived mast cells (BMMC) induced a more robust increase in FcεRI-dependent tyrosine phosphorylation of PLSCR1 compared to wild-type cells, whereas PLSCR1 phosphorylation was abolished in Lyn-deficient BMMC. Lyn association with PLSCR1 was not altered in Fyn-deficient BMMC. PLSCR1 phosphorylation was also dependent on the kinase Syk and significantly, but partially, dependent on detectable calcium mobilization. Thus, the Lyn/Syk/calcium axis promotes PLSCR1 phosphorylation in multiple ways. Conversely, the Fyn-dependent pathway negatively regulates it. This study reveals a complex regulation for PLSCR1 tyrosine phosphorylation in FcεRI-activated mast cells and that PLSCR1 sits at a crossroads between Lyn and Fyn pathways.


Subject(s)
Intracellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Phospholipid Transfer Proteins/immunology , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins c-fyn/immunology , Receptors, IgE/immunology , src-Family Kinases/immunology , Animals , Calcium/metabolism , Cell Degranulation/immunology , Cell Line , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Mast Cells/cytology , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phospholipid Transfer Proteins/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-fyn/genetics , Rats , Receptors, IgE/genetics , Signal Transduction , Syk Kinase , Tyrosine/metabolism , src-Family Kinases/genetics
11.
J Biol Chem ; 288(44): 31423-36, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24045941

ABSTRACT

Signaling lymphocytic activation molecule-associated protein (SAP) is an Src homology 2 domain-only adaptor involved in multiple immune cell functions. It has also been linked to immunodeficiencies and autoimmune diseases, such as systemic lupus erythematosus. Here, we examined the role and mechanism of action of SAP in autoimmunity using a mouse model of autoimmune arthritis, collagen-induced arthritis (CIA). We found that SAP was essential for development of CIA in response to collagen immunization. It was also required for production of collagen-specific antibodies, which play a key role in disease pathogenesis. These effects required SAP expression in T cells, not in B cells. In mice immunized with a high dose of collagen, the activity of SAP was nearly independent of its ability to bind the protein tyrosine kinase Fyn and correlated with the capacity of SAP to promote full differentiation of follicular T helper (TFH) cells. However, with a lower dose of collagen, the role of SAP was more dependent on Fyn binding, suggesting that additional mechanisms other than TFH cell differentiation were involved. Further studies suggested that this might be due to a role of the SAP-Fyn interaction in natural killer T cell development through the ability of SAP-Fyn to promote Vav-1 activation. We also found that removal of SAP expression during progression of CIA attenuated disease severity. However, it had no effect on disease when CIA was clinically established. Together, these results indicate that SAP plays an essential role in CIA because of Fyn-independent and Fyn-dependent effects on TFH cells and, possibly, other T cell types.


Subject(s)
Arthritis, Experimental/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Proto-Oncogene Proteins c-fyn/metabolism , T-Lymphocytes/metabolism , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes , Cell Line , Collagen/toxicity , Gene Expression Regulation/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/immunology , Proto-Oncogene Proteins c-vav/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Nat Immunol ; 14(11): 1127-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036998

ABSTRACT

Inflammation is a critical component of the immune response. However, acute or chronic inflammation can be highly destructive. Uncontrolled inflammation forms the basis for allergy, asthma and various autoimmune disorders. Here we identified a signaling pathway that was exclusively responsible for the production of inflammatory cytokines but not for cytotoxicity. Recognition of tumor cells expressing the NK cell-activatory ligands H60 or CD137L by mouse natural killer (NK) cells led to efficient cytotoxicity and the production of inflammatory cytokines. Both of those effector functions required the kinases Lck, Fyn and PI(3)K (subunits p85α and p110δ) and the signaling protein PLC-γ2. However, a complex of Fyn and the adaptor ADAP exclusively regulated the production of inflammatory cytokines but not cytotoxicity in NK cells. That unique function of ADAP required a Carma1-Bcl-10-MAP3K7 signaling axis. Our results have identified molecules that can be targeted to regulate inflammation without compromising NK cell cytotoxicity.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CARD Signaling Adaptor Proteins/immunology , Cytokines/biosynthesis , Killer Cells, Natural/immunology , MAP Kinase Kinase Kinases/immunology , Proto-Oncogene Proteins c-fyn/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , B-Cell CLL-Lymphoma 10 Protein , CARD Signaling Adaptor Proteins/genetics , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation , Killer Cells, Natural/pathology , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , MAP Kinase Kinase Kinases/genetics , Mice , Proto-Oncogene Proteins c-fyn/genetics
13.
J Hematol Oncol ; 6: 56, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23915951

ABSTRACT

BACKGROUND: High concentrations of plasmatic IgE have been related to distinct systemic inflammatory conditions that frequently predispose individuals to hypersensitivity reactions. Although effects of IgE have been suggested to relay on the low-intensity activation of distinct effector elements of the immune system, such as mast cells (MC), experimental evidence on the role of IgE-induced production of inflammatory mediators on specific pathologies is scarce. MC are an important component in tumor microenvironment where they seem to secrete a number of immunomodulatory and angiogenic mediators, such as the Vascular Endothelial Growth Factor (VEGF) by not well-described mechanisms. In this work, we investigated the effect of monomeric IgE (in the absence of antigen) on the production of VEGF in MC, analyzed if monomeric IgE could exacerbate the pro-tumorigenic properties of that cell type and characterized some of the molecular mechanisms behind the effects of IgE on VEGF production and tumor growth. METHODS: For in vitro studies, murine bone marrow-derived mast cells (BMMCs) were used. Pharmacological inhibitors and phosphorylation of key elements controlling VEGF secretion and protein translation were used to characterize the mechanism of VEGF production triggered by IgE.In vivo, the effect of a single i.v. administration of monomeric IgE on B16 melanoma tumor weight, intratumoral blood vessel formation and tumor-associated MC was assessed in four groups of mice: MC-proficient (WT), MC-deficient (Wsh), Wsh reconstituted with MC derived from WT mice (Wsh Rec WT) and Wsh reconstituted with MC derived from Fyn -/- mice (Wsh Rec Fyn -/-). RESULTS: Monomeric IgE induced VEGF secretion through a Fyn kinase-dependent mechanism and modulated de novo protein synthesis modifying the activity of the translational regulator 4E-BP1 in BMMCs. In vivo, monomeric IgE increased melanoma tumor growth, peritumoral MC and blood vessel numbers in WT but not in Wsh mice. The positive effects of IgE on melanoma tumor growth were reproduced after reconstitution of Wsh mice with WT but not with Fyn -/- BMMCs. CONCLUSION: Our data suggest that monomeric IgE, in the absence of antigen, induces VEGF production in MC and in vivo contributes to melanoma tumor growth through a Fyn kinase-dependent mechanism.


Subject(s)
Immunoglobulin E/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Humans , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Male , Mast Cells/enzymology , Mast Cells/immunology , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-fyn/immunology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
14.
J Leukoc Biol ; 94(6): 1253-64, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23990628

ABSTRACT

That cholestatic conditions are accompanied by an enhanced susceptibility to bacterial infection in human and animal models is a known phenomenon. This correlates with the observation that bile acids have suppressive effects on cells of innate and adaptive immunity. The present study provides evidence that in human macrophages, bile acids inhibit the LPS-induced expression of proinflammatory cytokines without affecting the expression of the anti-inflammatory cytokine IL-10. This results in a macrophage phenotype that is characterized by an increased IL-10/IL-12 ratio. Correspondingly, bile acids suppress basal phagocytic activity of human macrophages. These effects of bile acids can be mimicked by cAMP, which is presumably induced TGR5-dependently. The data provided further suggest that in primary human macrophages, modulation of the macrophage response toward LPS by bile acids involves activation of CREB, disturbed nuclear translocation of NF-κB, and PKA-dependent enhancement of LPS-induced cFos expression. The increase in cFos expression is paralleled by an enhanced formation of a protein complex comprising cFos and the p65 subunit of NF-κB. In summary, the data provided suggest that in human macrophages, bile acids induce an anti-inflammatory phenotype characterized by an increased IL-10/IL-12 ratio via activation of PKA and thereby, prevent their activation as classically activated macrophages. This bile acid-induced modulation of macrophage function may also be responsible for the experimentally and clinically observed anti-inflammatory and immunosuppressive effects of bile acids.


Subject(s)
Bile Acids and Salts/immunology , Cyclic AMP-Dependent Protein Kinases/immunology , Interleukin-10/immunology , Interleukin-12/immunology , Macrophage Activation/immunology , Macrophages/immunology , Bile Acids and Salts/metabolism , Cyclic AMP/genetics , Cyclic AMP/immunology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Interleukin-10/metabolism , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/enzymology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
15.
Glycoconj J ; 30(9): 819-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23877649

ABSTRACT

The association of sulfatide with specific proteins in oligodendrocytes was examined by co-immunoprecipitation with an anti-sulfatide antibody. Protein kinase activity was detected in precipitates with a monoclonal antibody to sulfatide (O4) from the rat primary immature oligodendrocytes. We conducted in vitro kinase assay of tyrosine phosphorylated proteins of 80, 59, 56, 53 and 40 kDa by gel electrophoresis. Of these proteins, the proteins of 59 kDa and 53/56 kDa were identified as the Src family tyrosine kinases Fyn and Lyn on the basis of their sequential immunoprecipitation with anti-Fyn and anti-Lyn antibodies, respectively. The 40 kDa protein was identified as the α subunit of the heterotrimeric G protein. These observations suggest that O4 immunoprecipitates sulfatide rafts including Fyn, Lyn and the α subunit of the heterotrimeric G protein.


Subject(s)
GTP-Binding Protein alpha Subunits/immunology , Oligodendroglia/immunology , Proto-Oncogene Proteins c-fyn/immunology , Sulfoglycosphingolipids/immunology , src-Family Kinases/immunology , Animals , Antibodies, Monoclonal/immunology , Cells, Cultured , GTP-Binding Protein alpha Subunits/metabolism , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Oligodendroglia/metabolism , Protein Binding , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Sulfoglycosphingolipids/metabolism , src-Family Kinases/metabolism
16.
J Allergy Clin Immunol ; 132(3): 729-736.e12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23587332

ABSTRACT

BACKGROUND: Aggregation of FcεRI activates a cascade of signaling events leading to mast cell activation, followed by inhibitory signals that turn off the activating signals. However, the overall view of negative signals in mast cells is still incomplete. Although AMP-activated protein kinase (AMPK), which is generally known as a regulator of energy metabolism, is also associated with anti-inflammation, little is known about the role of AMPK in mast cells. OBJECTIVES: We investigated the role of AMPK and its regulatory mechanism in mast cells. METHOD: The roles of AMPK in FcεRI-dependent activation of bone marrow-derived mast cells (BMMCs) were evaluated by using chemical agents, small interfering RNAs (siRNAs), or adenovirus that modulated the activity or expression of AMPK signaling components. In addition, AMPKα2(-/-) mice were used to verify the role of AMPK in anaphylactic models. RESULTS: FcεRI signaling and associated effector functions in BMMCs were suppressed by the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside (AICAR) and were conversely augmented by siRNA knockdown of AMPKα2 or liver kinase B1 (LKB1), an upstream kinase of AMPK. Furthermore, AMPKα2 deficiency led to increased FcεRI-mediated BMMC activation and anaphylaxis that were insensitive to AICAR, whereas enforced expression of AMPKα2 in AMPKα2(-/-) BMMCs reversed the hypersensitive FcεRI signaling to normal levels. Pharmacologic inhibition or siRNA knockdown of Fyn mimicked AMPK activation, suggesting that Fyn counterregulates the LKB1-AMPK axis. Mechanistically, Fyn controlled AMPK activity by regulating LKB1 localization. CONCLUSIONS: The Fyn-regulated LKB1-AMPK axis acts as a novel inhibitory module for mast cell activation, which points to AMPK activators as therapeutic drugs for allergic diseases.


Subject(s)
AMP-Activated Protein Kinases/immunology , Anaphylaxis/immunology , Mast Cells/immunology , Receptors, IgE/immunology , AMP-Activated Protein Kinases/genetics , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology
17.
Cell Immunol ; 276(1-2): 26-34, 2012.
Article in English | MEDLINE | ID: mdl-22663768

ABSTRACT

Memory and naive CD4 T cells have unique regulatory pathways for self/non-self discrimination. A memory cell specific regulatory pathway was revealed using superantigens to trigger the TCR. Upon stimulation by bacterial superantigens, like staphylococcal enterotoxin B (SEB), TCR proximal signaling is impaired leading to clonal tolerance (anergy). In the present report, we show that memory cell anergy results from the sequestration of the protein tyrosine kinase ZAP-70 away from the TCR/CD3ζ chain. During SEB-induced signaling, ZAP-70 is excluded from both detergent-resistant membrane microdomains and the immunological synapse, thus blocking downstream signaling. We also show that the mechanism underlying memory cell anergy must involve Fyn kinase, given that the suppression of Fyn activity restores the movement of ZAP-70 to the immunological synapse, TCR proximal signaling, and cell proliferation. Thus, toleragens, including microbial toxins, may modulate memory responses by targeting the organizational structure of memory cell signaling complexes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Membrane/immunology , Immunologic Memory , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Membrane/metabolism , Cell Movement , Cells, Cultured , Enterotoxins/immunology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/immunology , Receptors, Antigen, T-Cell/metabolism , Staphylococcus/immunology
18.
J Immunol ; 188(9): 4360-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22450804

ABSTRACT

Anaphylaxis is a rapid, life-threatening hypersensitivity reaction. Until recently, it was mainly attributed to histamine released by mast cells activated by allergen crosslinking (XL) of FcεRI-bound allergen-specific IgE. However, recent reports established that anaphylaxis could also be triggered by basophil, macrophage, and neutrophil secretion of platelet-activating factor subsequent to FcγR stimulation by IgG/Ag complexes. We have investigated the contribution of Fyn and Lyn tyrosine kinases to FcγRIIb and FcγRIII signaling in the context of IgG-mediated passive systemic anaphylaxis (PSA). We found that mast cell IgG XL induced Fyn, Lyn, Akt, Erk, p38, and JNK phosphorylation. Additionally, IgG XL of mast cells, basophils, and macrophages resulted in Fyn- and Lyn-regulated mediator release in vitro. FcγR-mediated activation was enhanced in Lyn-deficient (knockout [KO]) cells, but decreased in Fyn KO cells, compared with wild-type cells. More importantly, Lyn KO mice displayed significantly exacerbated PSA features whereas no change was observed for Fyn KO mice, compared with wild-type littermates. Intriguingly, we establish that mast cells account for most serum histamine in IgG-induced PSA. Taken together, our findings establish pivotal roles for Fyn and Lyn in the regulation of PSA and highlight their unsuspected functions in IgG-mediated pathologies.


Subject(s)
Anaphylaxis/immunology , Immunoglobulin G/immunology , Mast Cells/immunology , Proto-Oncogene Proteins c-fyn/immunology , src-Family Kinases/immunology , Allergens/genetics , Allergens/immunology , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Immunoglobulin G/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Mast Cells/pathology , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-fyn/genetics , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction/genetics , Signal Transduction/immunology , src-Family Kinases/genetics
19.
J Immunol ; 187(4): 1807-15, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21746961

ABSTRACT

Mast cells are critical for various allergic disorders. Mast cells express Src family kinases, which relay positive and negative regulatory signals by Ag. Lyn, for example, initiates activating signaling events, but it also induces inhibitory signals. Fyn and Hck are reported to be positive regulators, but little is known about the roles of other Src kinases, including Fgr, in mast cells. In this study, we define the role of Fgr. Endogenous Fgr associates with FcεRI and promotes phosphorylation of Syk, Syk substrates, which include linkers for activation of T cells, SLP76, and Gab2, and downstream targets such as Akt and the MAPKs in Ag-stimulated mast cells. As a consequence, Fgr positively regulates degranulation, production of eicosanoids, and cytokines. Fgr and Fyn appeared to act in concert, as phosphorylation of Syk and degranulation are enhanced by overexpression of Fgr and further augmented by overexpression of Fyn but are suppressed by overexpression of Lyn. Moreover, knockdown of Fgr by small interfering RNAs (siRNAs) further suppressed degranulation in Fyn-deficient bone marrow-derived mast cells. Overexpression of Fyn or Fgr restored phosphorylation of Syk and partially restored degranulation in Fyn-deficient cells. Additionally, knockdown of Fgr by siRNAs inhibited association of Syk with FcεRIγ as well as the tyrosine phosphorylation of FcεRIγ. Of note, the injection of Fgr siRNAs diminished the protein level of Fgr in mice and simultaneously inhibited IgE-mediated anaphylaxis. In conclusion, Fgr positively regulates mast cell through activation of Syk. These findings help clarify the interplay among Src family kinases and identify Fgr as a potential therapeutic target for allergic diseases.


Subject(s)
Anaphylaxis/immunology , Bone Marrow Cells/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Proto-Oncogene Proteins/immunology , src-Family Kinases/immunology , Anaphylaxis/enzymology , Anaphylaxis/genetics , Anaphylaxis/therapy , Animals , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Cell Degranulation/genetics , Cell Degranulation/immunology , Cell Line, Tumor , Enzyme Activation/genetics , Enzyme Activation/immunology , Gene Knockdown Techniques , Immunoglobulin E/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Mast Cells/enzymology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Phosphorylation/genetics , Phosphorylation/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-fyn/metabolism , RNA, Small Interfering , Rats , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, IgE/metabolism , Syk Kinase , src-Family Kinases/genetics , src-Family Kinases/metabolism
20.
J Neuroimmune Pharmacol ; 6(4): 585-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21671006

ABSTRACT

Although a number of inflammatory cytokines are increased during sepsis, the clinical trials aimed at down-regulating these mediators have not improved the outcome. These paradoxical results are attributed to loss of the "tolerance" phase that normally follows the proinflammatory response. Chronic nicotine (NT) suppresses both adaptive and innate immune responses, and the effects are partly mediated by the nicotinic acetylcholine receptors in the brain; however, the mechanism of neuroimmune communication is not clear. Here, we present evidence that, in rats and mice, NT initially increases IL-1ß in the brain, but the expression is downregulated within 1-2 week of chronic exposure, and the animals become resistant to proinflammatory/pyrogenic stimuli. To examine the relationship between NT, IL-1ß, and immunosuppression, we hypothesized that NT induces IL-1ß in the brain, and its constant presence produces immunological "tolerance". Indeed, unlike wild-type C57BL/6 mice, chronic NT failed to induce immunosuppression or downregulation of IL-1ß expression in IL-1ß-receptor knockout mice. Moreover, while acute intracerebroventricular administration of IL-1ß in Lewis (LEW) rats activated Fyn and protein tyrosine kinase activities in the spleen, chronic administration of low levels of IL-1ß progressively diminished the pyrogenic and T cell proliferative responses of treated animals. Thus, IL-1ß may play a critical role in the perception of inflammation by the CNS and the induction of an immunologic "tolerant" state. Moreover, the immunosuppressive effects of NT might be at least partly mediated through its effects on the brain IL-1ß. This represents a novel mechanism for neuroimmune communication.


Subject(s)
Immune Tolerance/drug effects , Interleukin-1beta/immunology , Neuroimmunomodulation/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Brain/drug effects , Brain/immunology , Brain/metabolism , Immune Tolerance/immunology , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation/immunology , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fyn/immunology , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...