Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Publication year range
1.
Osteoarthritis Cartilage ; 29(10): 1389-1398, 2021 10.
Article in English | MEDLINE | ID: mdl-34284112

ABSTRACT

Osteoarthritis (OA) is a major health problem worldwide that affects the joints and causes severe disability. It is characterized by pain and low-grade inflammation. However, the exact pathogenesis remains unknown and the therapeutic options are limited. In OA articular chondrocytes undergo a phenotypic transition becoming hypertrophic, which leads to cartilage damage, aggravating the disease. Therefore, a therapeutic agent inhibiting hypertrophy would be a promising disease-modifying drug. The therapeutic use of tyrosine kinase inhibitors has been mainly focused on oncology, but the Food and Drug Administration (FDA) approval of the Janus kinase inhibitor Tofacitinib in Rheumatoid Arthritis has broadened the applicability of these compounds to other diseases. Interestingly, tyrosine kinases have been associated with chondrocyte hypertrophy. In this review, we discuss the experimental evidence that implicates specific tyrosine kinases in signaling pathways promoting chondrocyte hypertrophy, highlighting their potential as therapeutic targets for OA.


Subject(s)
Chondrocytes/pathology , Osteoarthritis/drug therapy , Protein Kinase Inhibitors/pharmacology , Discoidin Domain Receptors/physiology , ErbB Receptors/physiology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Humans , Hypertrophy/drug therapy , Janus Kinase 2/physiology , Osteoarthritis/physiopathology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/physiology , Receptor Tyrosine Kinase-like Orphan Receptors/physiology , Receptor, IGF Type 1/physiology , Receptor, trkA/physiology , Receptors, Fibroblast Growth Factor/physiology , Signal Transduction
2.
Immunity ; 36(6): 974-85, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22683124

ABSTRACT

The adaptor SAP, mutated in X-linked lymphoproliferative disease, has critical roles in multiple immune cell types. Among these, SAP is essential for the ability of natural killer (NK) cells to eliminate abnormal hematopoietic cells. Herein, we elucidated the molecular and cellular bases of this activity. SAP enhanced NK cell responsiveness by a dual molecular mechanism. It coupled SLAM family receptors to the kinase Fyn, which triggered the exchange factor Vav-1 and augmented NK cell activation. SAP also prevented the inhibitory function of SLAM family receptors. This effect was Fyn independent and correlated with uncoupling of SLAM family receptors from the lipid phosphatase SHIP-1. Both mechanisms cooperated to enable conjugate formation with target cells and to stimulate cytotoxicity and cytokine secretion by NK cells. These data showed that SAP secures NK cell activation by a dichotomous molecular mechanism, which is required for conjugate formation. These findings may have implications for the role of SAP in other immune cell types.


Subject(s)
Antigens, CD/immunology , Intracellular Signaling Peptides and Proteins/physiology , Killer Cells, Lymphokine-Activated/immunology , Lymphocyte Activation/immunology , Phosphoric Monoester Hydrolases/physiology , Proto-Oncogene Proteins c-fyn/physiology , Proto-Oncogene Proteins c-vav/physiology , Receptors, Cell Surface/immunology , Animals , Antigens, CD/metabolism , Binding Sites , CHO Cells , Calcium Signaling/drug effects , Cell Adhesion , Cell Line, Tumor , Cricetinae , Cricetulus , Cytotoxicity, Immunologic , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Inositol Polyphosphate 5-Phosphatases , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Lymphokine-Activated/enzymology , Lymphoma, T-Cell/pathology , Melanoma, Experimental/pathology , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phospholipase C gamma/physiology , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family Member 1
3.
Cereb Cortex ; 30(3): 929-941, 2020 03 14.
Article in English | MEDLINE | ID: mdl-31609430

ABSTRACT

The coordination of cytoskeletal regulation is a prerequisite for proper neuronal migration during mammalian corticogenesis. Neuronal tyrosine-phosphorylated adaptor for the phosphoinositide 3-kinase 1 (Nyap1) is a member of the Nyap family of phosphoproteins, which has been studied in neuronal morphogenesis and is involved in remodeling of the actin cytoskeleton. However, the precise role of Nyap1 in neuronal migration remains unknown. Here, overexpression and knockdown of Nyap1 in the embryonic neocortex of mouse by in utero electroporation-induced abnormal morphologies and multipolar-bipolar transitions of migrating neurons. The level of phosphorylated Nyap1 was crucial for neuronal migration and morphogenesis in neurons. Furthermore, Nyap1 regulated neuronal migration as a downstream target of Fyn, a nonreceptor protein-tyrosine kinase that is a member of the Src family of kinases. Importantly, Nyap1 mediated the role of Fyn in the multipolar-bipolar transition of migrating neurons. Taken together, these results suggest that cortical radial migration is regulated by a molecular hierarchy of Fyn via Nyap1.


Subject(s)
Cell Movement , Neocortex/cytology , Neocortex/embryology , Neurons/cytology , Neurons/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Neocortex/metabolism , Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins c-fyn/metabolism
4.
J Neurosci ; 38(19): 4598-4609, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29661967

ABSTRACT

In the rodent olfactory system, neuroblasts produced in the ventricular-subventricular zone of the postnatal brain migrate tangentially in chain-like cell aggregates toward the olfactory bulb (OB) through the rostral migratory stream (RMS). After reaching the OB, the chains are dissociated and the neuroblasts migrate individually and radially toward their final destination. The cellular and molecular mechanisms controlling cell-cell adhesion during this detachment remain unclear. Here we report that Fyn, a nonreceptor tyrosine kinase, regulates the detachment of neuroblasts from chains in the male and female mouse OB. By performing chemical screening and in vivo loss-of-function and gain-of-function experiments, we found that Fyn promotes somal disengagement from the chains and is involved in neuronal migration from the RMS into the granule cell layer of the OB. Fyn knockdown or Dab1 (disabled-1) deficiency caused p120-catenin to accumulate and adherens junction-like structures to be sustained at the contact sites between neuroblasts. Moreover, a Fyn and N-cadherin double-knockdown experiment indicated that Fyn regulates the N-cadherin-mediated cell adhesion between neuroblasts. These results suggest that the Fyn-mediated control of cell-cell adhesion is critical for the detachment of chain-forming neuroblasts in the postnatal OB.SIGNIFICANCE STATEMENT In the postnatal brain, newly born neurons (neuroblasts) migrate in chain-like cell aggregates toward their destination, where they are dissociated into individual cells and mature. The cellular and molecular mechanisms controlling the detachment of neuroblasts from chains are not understood. Here we show that Fyn, a nonreceptor tyrosine kinase, promotes the somal detachment of neuroblasts from chains, and that this regulation is critical for the efficient migration of neuroblasts to their destination. We further show that Fyn and Dab1 (disabled-1) decrease the cell-cell adhesion between chain-forming neuroblasts, which involves adherens junction-like structures. Our results suggest that Fyn-mediated regulation of the cell-cell adhesion of neuroblasts is critical for their detachment from chains in the postnatal brain.


Subject(s)
Brain/physiology , Neural Stem Cells/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Brain/cytology , Brain/growth & development , Cadherins/genetics , Catenins/metabolism , Cell Adhesion/physiology , Cell Movement/genetics , Female , Gene Knockdown Techniques , Male , Mice , Nerve Tissue Proteins/genetics , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , Olfactory Bulb/physiology
5.
Am J Hematol ; 94(1): 10-20, 2019 01.
Article in English | MEDLINE | ID: mdl-30252956

ABSTRACT

The signaling cascade induced by the interaction of erythropoietin (EPO) with its receptor (EPO-R) is a key event of erythropoiesis. We present here data indicating that Fyn, a Src-family-kinase, participates in the EPO signaling-pathway, since Fyn-/- mice exhibit reduced Tyr-phosphorylation of EPO-R and decreased STAT5-activity. The importance of Fyn in erythropoiesis is also supported by the blunted responsiveness of Fyn-/- mice to stress erythropoiesis. Fyn-/- mouse erythroblasts adapt to reactive oxygen species (ROS) by activating the redox-related-transcription-factor Nrf2. However, since Fyn is a physiologic repressor of Nrf2, absence of Fyn resulted in persistent-activation of Nrf2 and accumulation of nonfunctional proteins. ROS-induced over-activation of Jak2-Akt-mTOR-pathway and repression of autophagy with perturbation of lysosomal-clearance were also noted. Treatment with Rapamycin, a mTOR-inhibitor and autophagy activator, ameliorates Fyn-/- mouse baseline erythropoiesis and erythropoietic response to oxidative-stress. These findings identify a novel multimodal action of Fyn in the regulation of normal and stress erythropoiesis.


Subject(s)
Erythropoiesis/physiology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Autophagy , Doxorubicin/toxicity , Erythroblasts/enzymology , Erythropoiesis/drug effects , Erythropoiesis/genetics , Female , Janus Kinase 2/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Phenylhydrazines/toxicity , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/genetics , Reactive Oxygen Species , Receptors, Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
6.
J Cell Sci ; 129(5): 930-42, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26801084

ABSTRACT

Oligodendrocytes are the myelinating glial cells of the central nervous system (CNS). Myelin is formed by extensive wrapping of oligodendroglial processes around axonal segments, which ultimately allows a rapid saltatory conduction of action potentials within the CNS and sustains neuronal health. The non-receptor tyrosine kinase Fyn is an important signaling molecule in oligodendrocytes. It controls the morphological differentiation of oligodendrocytes and is an integrator of axon-glial signaling cascades leading to localized synthesis of myelin basic protein (MBP), which is essential for myelin formation. The abundant myelin-associated oligodendrocytic basic protein (MOBP) resembles MBP in several aspects and has also been reported to be localized as mRNA and translated in the peripheral myelin compartment. The signals initiating local MOBP synthesis are so far unknown and the cellular function of MOBP remains elusive. Here, we show, by several approaches in cultured primary oligodendrocytes, that MOBP synthesis is stimulated by Fyn activity. Moreover, we reveal a new function for MOBP in oligodendroglial morphological differentiation.


Subject(s)
Cell Differentiation , Myelin Proteins/metabolism , Oligodendroglia/physiology , Proto-Oncogene Proteins c-fyn/physiology , Animals , Cell Shape , Cells, Cultured , Gene Expression , Mice, Inbred C57BL , Myelin Proteins/genetics , Protein Biosynthesis
7.
Ann Rheum Dis ; 77(6): 935-943, 2018 06.
Article in English | MEDLINE | ID: mdl-29555825

ABSTRACT

OBJECTIVES: To investigate the role of tyrosine kinase Fyn in the development of osteoarthritis (OA) and the underlying mechanisms, and to define whether targeting Fyn could prevent OA in mice. METHODS: Cartilage samples from normal and aged mice were analysed with proteome-wide screening. Fyn expression was examined with immunofluorescence in human and age-dependent or experimental mouse OA cartilage samples. Experimental OA in Fyn-knockout mice was induced by destabilisation of the medial meniscus. Primary cultured mouse chondrocytes were treated with proinflammatory cytokine interleukin-1ß. The inhibitor of Src kinase family, AZD0530 (saracatinib), and inhibitor of Fyn, PP1, were used to treat experimental OA in mice. RESULTS: Fyn expression was markedly upregulated in human OA cartilage and in cartilage from aged mice and those with post-traumatic OA. Fyn accumulates in articular chondrocytes and interacts directly with and phosphorylates ß-catenin at Tyr142, which stabilises ß-catenin and promotes its nuclear translocation. The deletion of Fyn effectively delayed the development of post-traumatic and age-dependent OA in mice. Fyn inhibitors AZD0530 and PP1 significantly attenuated OA progression by blocking the ß-catenin pathway and reducing the levels of extracellular matrix catabolic enzymes in the articular cartilage. CONCLUSIONS: Fyn accumulates and activates ß-catenin signalling in chondrocytes, accelerating the degradation of the articular cartilage and OA development. Targeting Fyn is a novel and potentially therapeutic approach to the treatment of OA.


Subject(s)
Arthritis, Experimental/enzymology , Osteoarthritis/enzymology , Proto-Oncogene Proteins c-fyn/physiology , beta Catenin/metabolism , Aging/metabolism , Animals , Arthritis, Experimental/prevention & control , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Cartilage, Articular/enzymology , Cells, Cultured , Chondrocytes/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Knockout Techniques , Humans , Mice, Knockout , Molecular Targeted Therapy/methods , Osteoarthritis/prevention & control , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology
8.
FASEB J ; 29(3): 920-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25416548

ABSTRACT

Worldwide obesity rates are at epidemic levels, and new insight into the regulation of obesity and adipogenesis are required. Thy1 (CD90), a cell surface protein with an enigmatic function, is expressed on subsets of fibroblasts and stem cells. We used a diet-induced obesity model to show that Thy1-null mice gain weight at a faster rate and gain 30% more weight than control C57BL/6 mice. During adipogenesis, Thy1 expression is lost in mouse 3T3-L1 cells. Overexpression of Thy1 blocked adipocyte formation and reduced mRNA and protein expression of an adipocyte marker, fatty acid-binding protein 4, by 5-fold. Although preadipocyte fibroblasts expressed Thy1 mRNA and protein, adipocytes from mouse and human fat tissue had almost undetectable Thy1 levels. Thy1 decreases the activity of the adipogenic transcription factor PPARγ by more than 60% as shown by PPARγ-dependent reporter assays. Using both genetic and pharmacologic approaches, we show Thy1 expression dampens PPARγ by inhibiting the activity of the Src-family kinase, Fyn. Thus, these studies reveal Thy1 blocks adipogenesis and PPARγ by inhibiting Fyn and support the idea that Thy1 is a novel therapeutic target in obesity.


Subject(s)
Adipogenesis/physiology , Gene Expression Regulation, Enzymologic , Obesity/physiopathology , Proto-Oncogene Proteins c-fyn/physiology , Thy-1 Antigens/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Diet, High-Fat , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Male , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thy-1 Antigens/genetics
9.
J Neurochem ; 134(4): 629-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25951993

ABSTRACT

The tyrosine kinase Fyn has two regulatory tyrosine residues that when phosphorylated either activate (Tyr(420)) or inhibit (Tyr(531)) Fyn activity. Within the central nervous system, two protein tyrosine phosphatases (PTPs) target these regulatory tyrosines in Fyn. PTPα dephosphorylates Tyr(531) and activates Fyn, while STEP (STriatal-Enriched protein tyrosine Phosphatase) dephosphorylates Tyr(420) and inactivates Fyn. Thus, PTPα and STEP have opposing functions in the regulation of Fyn; however, whether there is cross talk between these two PTPs remains unclear. Here, we used molecular techniques in primary neuronal cultures and in vivo to demonstrate that STEP negatively regulates PTPα by directly dephosphorylating PTPα at its regulatory Tyr(789). Dephosphorylation of Tyr(789) prevents the translocation of PTPα to synaptic membranes, blocking its ability to interact with and activate Fyn. Genetic or pharmacologic reduction in STEP61 activity increased the phosphorylation of PTPα at Tyr(789), as well as increased translocation of PTPα to synaptic membranes. Activation of PTPα and Fyn and trafficking of GluN2B to synaptic membranes are necessary for ethanol (EtOH) intake behaviors in rodents. We tested the functional significance of STEP61 in this signaling pathway by EtOH administration to primary cultures as well as in vivo, and demonstrated that the inactivation of STEP61 by EtOH leads to the activation of PTPα, its translocation to synaptic membranes, and the activation of Fyn. These findings indicate a novel mechanism by which STEP61 regulates PTPα and suggest that STEP and PTPα coordinate the regulation of Fyn. STEP61 , PTPα, Fyn, and NMDA receptor (NMDAR) have been implicated in ethanol intake behaviors in the dorsomedial striatum (DMS) in rodents. Here, we report that PTPα is a novel substrate for STEP61. Upon ethanol exposure, STEP61 is phosphorylated and inactivated by protein kinase A (PKA) signaling in the DMS. As a result of STEP61 inhibition, there is an increase in the phosphorylation of PTPα, which translocates to lipid rafts and activates Fyn and subsequent NMDAR signaling. The results demonstrate a synergistic regulation of Fyn-NMDAR signaling by STEP61 and PTPα, which may contribute to the regulation of ethanol-related behaviors. NMDA, N-methyl-D-aspartate; PTPα, receptor-type protein tyrosine phosphatase alpha; STEP, STriatal-Enriched protein tyrosine Phosphatase.


Subject(s)
Corpus Striatum/enzymology , Proto-Oncogene Proteins c-fyn/physiology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley
10.
Dev Neurosci ; 37(4-5): 311-20, 2015.
Article in English | MEDLINE | ID: mdl-25720756

ABSTRACT

The Src family kinases (SFKs) are nonreceptor protein tyrosine kinases that are implicated in many normal and pathological processes in the nervous system. The SFKs Fyn, Src, Yes, Lyn, and Lck are expressed in the brain. This review will focus on Fyn, as Fyn mutant mice have striking phenotypes in the brain and Fyn has been shown to be involved in ischemic brain injury in adult rodents and, with our work, in neonatal animals. An understanding of Fyn's role in neurodevelopment and disease will allow researchers to target pathological pathways while preserving protective ones.


Subject(s)
Animals, Newborn , Brain Injuries/metabolism , Brain Ischemia/metabolism , Brain/growth & development , Proto-Oncogene Proteins c-fyn/physiology , Animals , Brain/metabolism , Brain Injuries/etiology , Brain Ischemia/complications
11.
Purinergic Signal ; 11(3): 361-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26099359

ABSTRACT

Recruitment of oligodendrocyte precursor cells (OPCs) to the lesions is the most important event for remyelination after central nervous system (CNS) injury or in demyelinating diseases. However, the underlying molecular mechanism is not fully understood. In the present study, we found high concentrations of ATP could increase the number of migrating OPCs in vitro, while after pretreatment with oxidized ATP (a P2X7 receptor antagonist), the promotive effect was attenuated. The promotive effect of 2'(3')-O-(4-benzoylbenzoyl) adenosine 5'-triphosphate (BzATP) (a P2X7 receptor agonist) was more potent than ATP. After incubation with BzATP, the activity of Fyn, one member of the Src family of kinases, was enhanced. Moreover, the interaction between P2X7 and Fyn was identified by co-immunoprecipitation. After blocking the activity of Fyn or down-regulating the expression of Fyn, the migration of OPCs induced by BzATP was inhibited. These data indicate that P2X7 receptors/Fyn may mediate ATP-induced OPC migration under pathological conditions.


Subject(s)
Adenosine Triphosphate/pharmacology , Cell Movement/physiology , Oligodendroglia/drug effects , Proto-Oncogene Proteins c-fyn/physiology , Receptors, Purinergic P2X7/physiology , Stem Cells/physiology , Adenosine Triphosphate/analogs & derivatives , Animals , Cell Separation , Humans , Lentivirus/growth & development , Proto-Oncogene Proteins c-fyn/drug effects , Rats , Receptors, Purinergic P2X7/drug effects
12.
J Immunol ; 191(12): 6208-21, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227778

ABSTRACT

Focal adhesion kinase (FAK) is a critical regulator of signal transduction in multiple cell types. Although this protein is activated upon TCR engagement, the cellular function that FAK plays in mature human T cells is unknown. By suppressing the function of FAK, we revealed that FAK inhibits TCR-mediated signaling by recruiting C-terminal Src kinase to the membrane and/or receptor complex following TCR activation. Thus, in the absence of FAK, the inhibitory phosphorylation of Lck and/or Fyn is impaired. Together, these data highlight a novel role for FAK as a negative regulator TCR function in human T cells. These results also suggest that changes in FAK expression could modulate sensitivity to TCR stimulation and contribute to the progression of T cell malignancies and autoimmune diseases.


Subject(s)
Focal Adhesion Kinase 1/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Adolescent , Adult , Amino Acid Substitution , CD4-Positive T-Lymphocytes/enzymology , CSK Tyrosine-Protein Kinase , Enzyme Activation/physiology , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Male , MicroRNAs/genetics , Middle Aged , Multienzyme Complexes , Phosphorylation , Phosphotyrosine/physiology , Protein Processing, Post-Translational , Protein Transport , Proto-Oncogene Proteins c-fyn/physiology , RNA Interference , Recombinant Fusion Proteins/metabolism , Transfection , Young Adult , src-Family Kinases/metabolism
13.
Mol Cell Biochem ; 387(1-2): 63-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24173619

ABSTRACT

Type II phosphatidylinositol 4-kinases are implicated in FcεRI-mediated signaling cascades leading to release of inflammatory molecules. Cross-linking of FcεRI on RBL 2H3 cells results in protein tyrosine phosphorylation and activation of type II PtdIns 4-kinase activity. Protein tyrosine kinase(s) that phosphorylate type II PtdIns 4-kinase(s) in RBL 2H3 cells remains elusive and is being addressed in this manuscript. Anti-Fyn kinase antibodies co-immunoprecipitated type II PtdIns 4-kinase activity from FcεRI cross-linked RBL 2H3 cells. In reciprocal assays, His-tagged types II PtdIns 4-kinases were shown to pull down Fyn kinase. Further, anti-Fyn immunoprecipitates were shown to phosphorylate type II PtdIns 4-kinase α and ß in in vitro assays. Pull down studies with GST-Fyn-SH2 and GST-Fyn-SH3 domains showed that type II PtdIns 4-kinases associate with Fyn-SH2 domain. Knockdown of Fyn kinase in RBL 2H3 cells abrogated activation of type II PtdIns 4-kinase activity in response to FcεRI cross-linking and type II PtdIns 4-kinase activity in anti-phosphotyrosine immunoprecipitates. Knockdown of Fyn kinase was also strongly correlated with a reduction in ß-hexosaminidase release in response to FcεRI cross-linking. These results suggest that type II PtdIns 4-kinases act downstream of Fyn kinase in FcεRI signaling cascades and are regulated by Fyn kinase.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-fyn/physiology , Animals , Cell Line, Tumor , Enzyme Activation , Humans , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-fyn/chemistry , Rats , Receptors, IgE/metabolism , Signal Transduction , beta-N-Acetylhexosaminidases/metabolism
14.
J Immunol ; 189(2): 646-58, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22711887

ABSTRACT

BCR signaling regulates the activities and fates of B cells. BCR signaling encompasses two feedback loops emanating from Lyn and Fyn, which are Src family protein tyrosine kinases (SFKs). Positive feedback arises from SFK-mediated trans phosphorylation of BCR and receptor-bound Lyn and Fyn, which increases the kinase activities of Lyn and Fyn. Negative feedback arises from SFK-mediated cis phosphorylation of the transmembrane adapter protein PAG1, which recruits the cytosolic protein tyrosine kinase Csk to the plasma membrane, where it acts to decrease the kinase activities of Lyn and Fyn. To study the effects of the positive and negative feedback loops on the dynamical stability of BCR signaling and the relative contributions of Lyn and Fyn to BCR signaling, we consider in this study a rule-based model for early events in BCR signaling that encompasses membrane-proximal interactions of six proteins, as follows: BCR, Lyn, Fyn, Csk, PAG1, and Syk, a cytosolic protein tyrosine kinase that is activated as a result of SFK-mediated phosphorylation of BCR. The model is consistent with known effects of Lyn and Fyn deletions. We find that BCR signaling can generate a single pulse or oscillations of Syk activation depending on the strength of Ag signal and the relative levels of Lyn and Fyn. We also show that bistability can arise in Lyn- or Csk-deficient cells.


Subject(s)
Computer Simulation , Models, Immunological , Proto-Oncogene Proteins c-fyn/physiology , Receptors, Antigen, B-Cell/physiology , Signal Transduction/immunology , src-Family Kinases/physiology , Animals , Calcium Signaling/immunology , Feedback, Physiological , Humans , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, B-Cell/metabolism , src-Family Kinases/deficiency , src-Family Kinases/metabolism
15.
J Immunol ; 188(11): 5247-56, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539787

ABSTRACT

Th17 cells constitute a proinflammatory CD4(+) T cell subset that is important for microbial clearance, but also are implicated as propagators of various autoimmune pathologies. Evidence suggests that Th17 cells share common progenitors with immunosuppressive CD4(+) inducible regulatory T cells (T(REG)) and that the developmental pathways of these two subsets are reciprocally regulated. In this study, we show evidence that the Src family tyrosine kinase Fyn helps regulate this Th17/T(REG) balance. When placed under Th17-skewing conditions, CD4(+) T cells from fyn(-/-) mice had decreased levels of IL-17, but increased expression of the T(REG) transcription factor Foxp3. The defect in IL-17 expression occurred independently of the ectopic Foxp3 expression and correlated with a delay in retinoic acid-related orphan receptor γt upregulation and an inability to maintain normal STAT3 activation. Fyn-deficient Th17 cells also exhibited delayed upregulation of Il23r, Il21, Rora, and Irf4, as well as aberrant expression of Socs3, suggesting that Fyn may function upstream of a variety of molecular pathways that contribute to Th17 polarization. The fyn(-/-) mice had fewer IL-17(+)CD4(+) T cells in the large intestinal lamina propria compared with littermate controls. Furthermore, after transfer of either wild-type or fyn(-/-) naive CD4(+) T cells into Rag1(-/-) hosts, recipients receiving fyn(-/-) cells had fewer IL-17-producing T cells, indicating that Fyn may also regulate Th17 differentiation in vivo. These results identify Fyn as a possible novel regulator of the developmental balance between the Th17 cell and T(REG) subsets.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/biosynthesis , Gene Expression Regulation/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Proto-Oncogene Proteins c-fyn/physiology , Th17 Cells/cytology , Th17 Cells/immunology , Animals , Cells, Cultured , Forkhead Transcription Factors/pharmacokinetics , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/pharmacokinetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Time Factors
16.
Dev Dyn ; 242(5): 469-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23361870

ABSTRACT

BACKGROUND: Src family tyrosine kinases (SFKs) are often coincidently expressed but few studies have dissected their individual functions in the same cell during development. Using the classical embryonic lens as our model, we investigated SFK signaling in the regulation of both differentiation initiation and morphogenesis, and the distinct functions of c-Src and Fyn in these processes. RESULTS: Blocking SFK activity with the highly specific inhibitor PP1 induced initiation of the lens differentiation program but blocked lens fiber cell elongation and organization into mini lens-like structures called lentoids. These dichotomous roles for SFK signaling were discovered to reflect distinct functions of c-Src and Fyn and their differentiation-state-specific recruitment to and action at N-cadherin junctions. c-Src was highly associated with the nascent N-cadherin junctions of undifferentiated lens epithelial cells. Its siRNA knockdown promoted N-cadherin junctional maturation, blocked proliferation, and induced lens cell differentiation. In contrast, Fyn was recruited to mature N-cadherin junctions of differentiating lens cells and siRNA knockdown suppressed differentiation-specific gene expression and blocked morphogenesis. CONCLUSIONS: Through inhibition of N-cadherin junction maturation, c-Src promotes lens epithelial cell proliferation and the maintenance of the lens epithelial cell undifferentiated state, while Fyn, signaling downstream of mature N-cadherin junctions, promotes lens fiber cell morphogenesis.


Subject(s)
Cadherins/metabolism , Lens, Crystalline/embryology , Proto-Oncogene Proteins c-fyn/physiology , src-Family Kinases/physiology , Animals , CSK Tyrosine-Protein Kinase , Cells, Cultured , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Models, Biological , Organogenesis/drug effects , Organogenesis/genetics , Organogenesis/physiology , Protein Binding/physiology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Quail/embryology , Quail/genetics , Quail/metabolism , RNA, Small Interfering/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
17.
J Neurosci ; 32(47): 16857-71a, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23175838

ABSTRACT

Amid controversy, the cellular form of the prion protein PrP(c) has been proposed to mediate oligomeric amyloid-ß (Aß)-induced deficits. In contrast, there is consistent evidence that the Src kinase Fyn is activated by Aß oligomers and leads to synaptic and cognitive impairment in transgenic animals. However, the molecular mechanism by which soluble Aß activates Fyn remains unknown. Combining the use of human and transgenic mouse brain tissue as well as primary cortical neurons, we demonstrate that soluble Aß binds to PrP(c) at neuronal dendritic spines in vivo and in vitro where it forms a complex with Fyn, resulting in the activation of the kinase. Using the antibody 6D11 to prevent oligomeric Aß from binding to PrP(c), we abolished Fyn activation and Fyn-dependent tau hyperphosphorylation induced by endogenous oligomeric Aß in vitro. Finally, we showed that gene dosage of Prnp regulates Aß-induced Fyn/tau alterations. Together, our findings identify a complete signaling cascade linking one specific endogenous Aß oligomer, Fyn alteration, and tau hyperphosphorylation in cellular and animal models modeling aspects of the molecular pathogenesis of Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/physiology , PrPC Proteins/physiology , Proto-Oncogene Proteins c-fyn/physiology , tau Proteins/physiology , Aged, 80 and over , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Blotting, Western , Cells, Cultured , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Female , Gene Deletion , Gene Dosage , Humans , Immunoprecipitation , Male , Mice , Mice, Transgenic , Neurofibrillary Tangles/pathology , Phosphorylation , PrPC Proteins/genetics , PrPC Proteins/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , tau Proteins/genetics , tau Proteins/metabolism
18.
Blood ; 117(21): 5744-50, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21427288

ABSTRACT

Platelet hyperactivity associated with hyperlipidemia contributes to development of a pro-thrombotic state. We previously showed that oxidized LDL (oxLDL) formed in the setting of hyperlipidemia and atherosclerosis initiated a CD36-mediated signaling cascade leading to platelet hyperactivity. We now show that the guanine nucleotide exchange factors Vav1 and Vav3 were tyrosine phosphorylated in platelets exposed to oxLDL. Pharmacologic inhibition of src family kinases abolished Vav1 phosphorylation by oxLDL in vitro. Coimmunoprecipitations revealed the tyrosine phosphorylated form of src kinase Fyn was associated with Vav1 in platelets exposed to oxLDL. Using a platelet aggregation assay, we demonstrated that Vav1 deficiency, Fyn deficiency, or Vav1/Vav3 deficiency protected mice from diet-induced platelet hyperactivity. Furthermore, flow cytometric analysis revealed that Vav1/Vav3 deficiency significantly inhibited oxLDL-mediated integrin αIIbßIII activation of platelets costimulated with ADP. Finally, we showed with an in vivo carotid artery thrombosis model that genetic deletion of Vav1 and Vav3 together may prevent the development of occlusive thrombi in mice fed a high-fat diet. These findings implicate Vav proteins in oxLDL-mediated platelet activation and suggest that Vav family member(s) may act as critical modulators linking a prothrombotic state and hyperlipidemia.


Subject(s)
Blood Platelets/metabolism , Hyperlipidemias/metabolism , Proto-Oncogene Proteins c-fyn/physiology , Proto-Oncogene Proteins c-vav/physiology , Thrombosis/etiology , Animals , Blotting, Western , Carotid Artery Thrombosis/metabolism , Dietary Fats , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Hyperlipidemias/pathology , Immunoprecipitation , Lipoproteins, LDL/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Platelet Activation , Platelet Aggregation , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thrombosis/metabolism , Thrombosis/prevention & control , Tyrosine/metabolism , src-Family Kinases/metabolism
19.
Blood ; 117(1): 108-17, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20876849

ABSTRACT

T-cell development is critically dependent on the activities of the Src-family kinases p56(lck) and p59(fyn). While Lck plays a dominant role in the initiation of T-cell receptor (TCR) signaling and in thymocyte differentiation, Fyn plays a more subtle regulatory role. We sought to determine the role of intracellular localization in the differing functions of Lck and Fyn in T cells. By generating transgenic mice that express chimeric Lck-Fyn proteins, we showed that the N-terminal unique domain determines the intracellular localization and function of Lck in pre-TCR and mature αßTCR signaling in vivo. Furthermore, coexpression of a "domain-swap" Lck protein containing the Fyn unique domain with an inducible Lck transgene resulted in the development of thymomas. In contrast to previous reports of Lck-driven thymomas, tumor development was dependent on either pre-TCR or mature TCR signals, and was completely ablated when mice were crossed to a recombination activating gene 1 (Rag1)-deficient background. These data provide a mechanistic basis for the differing roles of Lck and Fyn in T-cell development, and show that intracellular localization as determined by the N-terminal unique domains is critical for Src-family kinase function in vivo.


Subject(s)
Cell Differentiation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Proto-Oncogene Proteins c-fyn/physiology , Thymoma/pathology , Thymus Gland/cytology , Animals , Blotting, Western , CD2 Antigens/genetics , Female , Flow Cytometry , Humans , Immunoprecipitation , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes/metabolism , Thymoma/metabolism , Thymus Gland/metabolism
20.
J Allergy Clin Immunol ; 130(3): 751-760.e2, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22564682

ABSTRACT

BACKGROUND: The interaction of mast cells (MCs) with regulatory T cells through the OX40 ligand (OX40L):OX40 axis downregulates FcεRI-dependent immediate hypersensitivity responses both in vitro and in vivo. Little is known on OX40L-mediated intracellular signaling or on the mechanism by which OX40L engagement suppresses MC degranulation. OBJECTIVE: We explored the role of OX40L engagement on IgE/antigen-triggered MCs both in vitro and in vivo. METHODS: The soluble form of OX40 molecule was used to selectively trigger OX40L on MCs in vitro and was used to dissect OX40L contribution in an in vivo model of systemic anaphylaxis. RESULTS: OX40L:OX40 interaction led to the recruitment of C-terminal src kinase into lipid rafts, causing a preferential suppression of Fyn kinase activity and subsequent reduction in the phosphorylation of Gab2, the phosphatidylinositol 3-OH kinase regulatory subunit p85, and Akt, without affecting the Lyn pathway. Dampening of Fyn kinase activity also inhibited RhoA activation and microtubule nucleation, key regulators of MC degranulation. The in vivo administration of a blocking antibody to OX40L in wild-type mice caused enhanced immediate hypersensitivity, whereas the administration of soluble OX40 to regulatory T-cell-depleted or OX40-deficient mice reduced MC degranulation. CONCLUSIONS: The engagement of OX40L selectively suppresses Fyn-initiated signals required for MC degranulation and serves to limit immediate hypersensitivity. Our data suggest that soluble OX40 can restore the aberrant or absent regulatory T-cell activity, revealing a previously unappreciated homeostatic role for OX40L in setting the basal threshold of MC response.


Subject(s)
Mast Cells/physiology , Membrane Glycoproteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-fyn/physiology , Receptors, IgE/physiology , Tumor Necrosis Factors/physiology , rho GTP-Binding Proteins/physiology , Adaptor Proteins, Signal Transducing , Anaphylaxis/etiology , Animals , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Microtubules/physiology , OX40 Ligand , Phosphoproteins/physiology , Phosphorylation , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL