Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Neurochem Res ; 43(8): 1631-1640, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29934689

ABSTRACT

E3 ubiquitin ligase c-Caritas B cell lymphoma (c-cbl) is associated with negative regulation of receptor tyrosine kinases, signal transduction of antigens and cytokine receptors, and immune response. However, the expression and function of c-cbl in the regulation of neuropathic pain after chronic constriction injury (CCI) are unknown. In rat CCI model, c-cbl inhibited the activation of spinal cord microglia and the release of pro-inflammatory factors including tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1ß) and interleukin 6 (IL-6), which alleviated mechanical and heat pain through down-regulating extracellular signal-regulated kinase (ERK) pathway. Additionally, exogenous TNF-α inhibited c-cbl protein level vice versa. In the primary microglia transfected with c-cbl siRNA, when treated with TNF-α or TNF-α inhibitor, the corresponding secretion of IL-1ß and IL-6 did not change. In summary, CCI down-regulated c-cbl expression and induced the activation of microglia, then activated microglia released inflammatory factors via ERK signaling to cause pain. Our data might supply a novel molecular target for the therapy of CCI-induced neuropathic pain.


Subject(s)
Microglia/drug effects , Neuralgia/physiopathology , Peripheral Nerve Injuries/physiopathology , Proto-Oncogene Proteins c-cbl/physiology , Spinal Cord/physiopathology , Animals , Base Sequence , Constriction , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Male , Phosphorylation/physiology , Proto-Oncogene Proteins c-cbl/genetics , Rats, Sprague-Dawley , Sciatic Nerve/injuries , Tumor Necrosis Factor-alpha/pharmacology
2.
Biochim Biophys Acta ; 1853(8): 1879-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25960397

ABSTRACT

Phosphatidylinositol 3-kinaseß (PI3Kß) plays a predominant role in integrin outside-in signaling and in platelet activation by GPVI engagement. We have shown that the tyrosine kinase Pyk2 mediates PI3Kß activation downstream of integrin αIIbß3, and promotes the phosphorylation of the PI3K-associated adaptor protein c-Cbl. In this study, we compared the functional correlation between Pyk2 and PI3Kß upon recruitment of the two main platelet collagen receptors, integrin α2ß1 and GPVI. PI3Kß-mediated phosphorylation of Akt was inhibited in Pyk2-deficient platelets adherent to monomeric collagen through integrin α2ß1, but occurred normally upon GPVI ligation. Integrin α2ß1 engagement led to Pyk2-independent association of c-Cbl with PI3K. However, c-Cbl was not phosphorylated in adherent platelets, and phosphorylation of Akt occurred normally in c-Cbl-deficient platelets, indicating that the c-Cbl is dispensable for Pyk2-mediated PI3Kß activation. Stimulation of platelets with CRP, a selective GPVI ligand, induced c-Cbl phosphorylation in the absence of Pyk2, but failed to promote its association with PI3K. Pyk2 activation was completely abrogated in PI3KßKD, but not in PI3KγKD platelets, and was strongly inhibited by Src kinases and phospholipase C inhibitors, and by BAPTA-AM. The absence of PI3Kß activity also hampered GPVI-induced tyrosine-phosphorylation and activation of PLCγ2, preventing intracellular Ca2+ increase and phosphorylation of pleckstrin. Moreover, GPVI-induced intracellular Ca2+ increase and pleckstrin phosphorylation were also strongly inhibited in human platelets treated with the PI3Kß inhibitor TGX-221. These results outline important differences in the regulation of PI3Kß by GPVI and integrin α2ß1 and suggest that inhibition of Pyk2 may target PI3Kß activation in a selective context of platelet stimulation.


Subject(s)
Focal Adhesion Kinase 2/physiology , Integrin alpha2beta1/physiology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-cbl/physiology , Animals , Cells, Cultured , Enzyme Activation , Humans , Mice , Mice, Knockout , Signal Transduction
3.
Circulation ; 129(20): 2031-43, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24583314

ABSTRACT

BACKGROUND: The proto-oncogene Casitas b-lineage lymphoma (c-Cbl) is an adaptor protein with an intrinsic E3 ubiquitin ligase activity that targets receptor and nonreceptor tyrosine kinases, resulting in their ubiquitination and downregulation. However, the function of c-Cbl in the control of cardiac function is currently unknown. In this study, we examined the role of c-Cbl in myocyte death and cardiac function after myocardial ischemia. METHODS AND RESULTS: We show increased c-Cbl expression in human ischemic and dilated cardiomyopathy hearts and in response to pathological stress stimuli in mice. c-Cbl-deficient mice demonstrated a more robust functional recovery after myocardial ischemia/reperfusion injury and significantly reduced myocyte apoptosis and improved cardiac function. Ubiquitination and downregulation of key survival c-Cbl targets, epidermal growth factor receptors and focal adhesion kinase, were significantly reduced in c-Cbl knockout mice. Inhibition of c-Cbl expression or its ubiquitin ligase activity in cardiac myocytes offered protection against H2O2 stress. Interestingly, c-Cbl deletion reduced the risk of death and increased cardiac functional recovery after chronic myocardial ischemia. This beneficial effect of c-Cbl deletion was associated with enhanced neoangiogenesis and increased expression of vascular endothelial growth factor-a and vascular endothelial growth factor receptor type 2 in the infarcted region. CONCLUSIONS: c-Cbl activation promotes myocyte apoptosis, inhibits angiogenesis, and causes adverse cardiac remodeling after myocardial infarction. These findings point to c-Cbl as a potential therapeutic target for the maintenance of cardiac function and remodeling after myocardial ischemia.


Subject(s)
Cardiomyopathy, Dilated/physiopathology , Heart Failure/physiopathology , Myocardial Ischemia/physiopathology , Proto-Oncogene Proteins c-cbl/physiology , Adult , Aged , Animals , Apoptosis/physiology , Cardiac Catheterization , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Echocardiography , Electrocardiography , Female , Heart Failure/genetics , Heart Failure/pathology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-cbl/genetics , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/metabolism
4.
Biochim Biophys Acta ; 1833(1): 122-39, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23085373

ABSTRACT

Protein tyrosine kinases (PTKs) coordinate a broad spectrum of cellular responses to extracellular stimuli and cell-cell interactions during development, tissue homeostasis, and responses to environmental challenges. Thus, an understanding of the regulatory mechanisms that ensure physiological PTK function and potential aberrations of these regulatory processes during diseases such as cancer are of broad interest in biology and medicine. Aside from the expected role of phospho-tyrosine phosphatases, recent studies have revealed a critical role of covalent modification of activated PTKs with ubiquitin as a critical mechanism of their negative regulation. Members of the Cbl protein family (Cbl, Cbl-b and Cbl-c in mammals) have emerged as dominant "activated PTK-selective" ubiquitin ligases. Structural, biochemical and cell biological studies have established that Cbl protein-dependent ubiquitination targets activated PTKs for degradation either by facilitating their endocytic sorting into lysosomes or by promoting their proteasomal degradation. This mechanism also targets PTK signaling intermediates that become associated with Cbl proteins in a PTK activation-dependent manner. Cellular and animal studies have established that the relatively broadly expressed mammalian Cbl family members Cbl and Cbl-b play key physiological roles, including their critical functions to prevent the transition of normal immune responses into autoimmune disease and as tumor suppressors; the latter function has received validation from human studies linking mutations in Cbl to human leukemia. These newer insights together with embryonic lethality seen in mice with a combined deletion of Cbl and Cbl-b genes suggest an unappreciated role of the Cbl family proteins, and by implication the ubiquitin-dependent control of activated PTKs, in stem/progenitor cell maintenance. Future studies of existing and emerging animal models and their various cell lineages should help test the broader implications of the evolutionarily-conserved Cbl family protein-mediated, ubiquitin-dependent, negative regulation of activated PTKs in physiology and disease.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-cbl/physiology , Ubiquitination/physiology , Amino Acid Sequence , Animals , Humans , Mice , Models, Biological , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Ubiquitin/metabolism
5.
Thorax ; 69(4): 335-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24258832

ABSTRACT

BACKGROUND: T-cell targeted peptide epitope tolerogens from grass pollen allergens may be useful in treating seasonal allergic rhinitis, but there is urgent need for optimisation of approaches from improved understanding of mechanism. OBJECTIVE: We sought to identify human leukocyte antigen (HLA)-DR1-restricted epitopes from the Timothy grass pollen allergen, Phleum pratense, and characterise T-cell immune regulation following intranasal administration of a single, immunodominant epitope. METHODS: T-cell epitopes within P pratense were identified using HLA-DR1 transgenic mice and tetramer-guided epitope mapping (TGEM) in HLA-DR1-positive individuals with grass allergy. An immunodominant epitope was tested in HLA-DR1 transgenics for impact on responses to whole Phl p5 b or peptide. Microarrays and quantitative PCR were used to characterise T-cell immunity. RESULTS: Peptide 26 (p26) was identified in HLA-DR1 transgenic mice and by TGEM analysis of HLA-DR1-positive individuals with grass allergy. p26 shows promiscuous binding to a wide range of HLA class II alleles, making it of relevance across immunogenetically diverse patients. The epitope is conserved in rye and velvet grass, making it applicable across a spectrum of grass pollen allergy. Intranasal pretreatment of mice with p26 results in significantly reduced T-cell responses. Transcriptomic array analysis in mice showed T-cell regulation in the intranasal treatment group associated with increased expression of members of the Cbl-b and Itch E3 ubiquitin ligase pathway. CONCLUSIONS: We defined an immunodominant P pratense epitope, p26, with broad binding across multiple HLA class II alleles. Intranasal treatment of mice with p26 results in T-cell regulation to whole allergen, involving the Cbl-b and Itch regulatory pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , HLA-DR1 Antigen/immunology , Immunodominant Epitopes/immunology , Plant Proteins/immunology , Pollen/immunology , Proto-Oncogene Proteins c-cbl/physiology , Rhinitis, Allergic, Seasonal/immunology , Ubiquitin-Protein Ligases/physiology , Adult , Animals , Female , Humans , Immunity, Cellular , Male , Mice , Mice, Transgenic , Microarray Analysis , Middle Aged , Phleum/immunology , Real-Time Polymerase Chain Reaction , United Kingdom , Young Adult
6.
Biochem Biophys Res Commun ; 453(3): 557-62, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25285640

ABSTRACT

SIRT2 is a member of the sirtuin family of NAD(+)-dependent protein deacetylases. It is involved in metabolic homeostasis and has been linked to the progression of age-related diseases. Casitas B-lineage lymphoma (Cbl) proteins regulate signal transduction through many pathways and, consequently, regulate cell function and development. Cbl proteins are ubiquitin ligases that ubiquitinate and target many signaling molecules for degradation. The function of SIRT2 is modulated by post-translational modifications. However, the precise molecular signaling mechanism of SIRT2 through interactions with Cbl proteins has not yet been established. In this study, we investigated the potential regulation of SIRT2 function by the Cbl mammalian family members Cbl-b and c-Cbl. We found that Cbl-b and c-Cbl increased the protein level and stability of SIRT2 and that Cbl-b and c-Cbl interact with SIRT2. They were also found to regulate the deacetylase activity of SIRT2. Further investigation revealed that Cbl-mediated SIRT2 regulation occurred via ubiquitination of SIRT2.


Subject(s)
Proto-Oncogene Proteins c-cbl/physiology , Sirtuin 2/physiology , Cell Line , Humans , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Ubiquitination
7.
Endocr J ; 61(6): 529-38, 2014.
Article in English | MEDLINE | ID: mdl-24614797

ABSTRACT

Obesity causes type 2 diabetes, atherosclerosis and cardiovascular diseases by inducing systemic insulin resistance. It is now recognized that obesity is related to chronic low-grade inflammation in adipose tissue. Specifically, activated immune cells infiltrate adipose tissue and cause inflammation. There is increasing evidence that activated macrophages accumulate in the hypertrophied adipose tissue of rodents and humans and induce systemic insulin resistance by secreting inflammatory cytokines. Accordingly, a better understanding of the molecular mechanisms underlying macrophage activation in adipose tissue will facilitate the development of new therapeutic strategies. Currently, little is known about the regulation of macrophage activation, although E3 ubiquitin ligase Casitas B-lineage lymphoma (Cbl)-b was identified recently as a novel negative regulator of macrophage activation in adipose tissue. Cbl-b, which is a suppressor of T- and B-cell activation, inhibits intracellular signal transduction by targeting some tyrosine kinases. Notably, preventing Cbl-b-mediated macrophage activation improves obesity-induced insulin resistance in mice. c-Cbl is another member of the Cbl family that is associated with insulin resistance in obesity. These reports suggest that Cbl-b and c-Cbl are potential therapeutic targets for treating obesity-induced insulin resistance. In this review, we focus on the importance of Cbl-b in macrophage activation in aging-induced and high-fat diet-induced obesity.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Insulin Resistance/genetics , Obesity/metabolism , Proto-Oncogene Proteins c-cbl/physiology , Aging/physiology , Animals , Diet, High-Fat , Humans , Immune System/enzymology , Immune System/metabolism , Macrophage Activation/genetics , Mice , Obesity/complications , Obesity/genetics
8.
Proc Natl Acad Sci U S A ; 108(51): 20579-84, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22158902

ABSTRACT

Cbl-b is a RING-type E3 ubiquitin ligase that functions as a negative regulator of T-cell activation and growth factor receptor and nonreceptor-type tyrosine kinase signaling. Cbl-b dysfunction is related to autoimmune diseases and cancers in humans. However, the molecular mechanism regulating its E3 activity is largely unknown. NMR and small-angle X-ray scattering analyses revealed that the unphosphorylated N-terminal region of Cbl-b forms a compact structure by an intramolecular interaction, which masks the interaction surface of the RING domain with an E2 ubiquitin-conjugating enzyme. Phosphorylation of Y363, located in the helix-linker region between the tyrosine kinase binding and the RING domains, disrupts the interdomain interaction to expose the E2 binding surface of the RING domain. Structural analysis revealed that the phosphorylated helix-RING region forms a compact structure in solution. Moreover, the phosphate group of pY363 is located in the vicinity of the interaction surface with UbcH5B to increase affinity by reducing their electrostatic repulsion. Thus, the phosphorylation of Y363 regulates the E3 activity of Cbl-b by two mechanisms: one is to remove the masking of the RING domain from the tyrosine kinase binding domain and the other is to form a surface to enhance binding affinity to E2.


Subject(s)
Autoimmune Diseases/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Proto-Oncogene Proteins c-cbl/chemistry , Proto-Oncogene Proteins c-cbl/physiology , Ubiquitin-Protein Ligases/chemistry , Escherichia coli/metabolism , Humans , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , Scattering, Radiation , X-Rays
9.
J Immunol ; 186(4): 2138-47, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21248250

ABSTRACT

E3 ubiquitin ligases have been placed among the essential molecules involved in the regulation of T cell functions and T cell tolerance. However, it has never been experimentally proven in vivo whether these functions indeed depend on the catalytic E3 ligase activity. The Casitas B-cell lymphoma (Cbl) family protein Cbl-b was the first E3 ubiquitin ligase directly implicated in the activation and tolerance of the peripheral T cell. In this study, we report that selective genetic inactivation of Cbl-b E3 ligase activity phenocopies the T cell responses observed when total Cbl-b is ablated, resulting in T cell hyperactivation, spontaneous autoimmunity, and impaired induction of T cell anergy in vivo. Moreover, mice carrying a Cbl-b E3 ligase-defective mutation spontaneously reject tumor cells that express human papilloma virus Ags. These data demonstrate for the first time, to our knowledge, that the catalytic function of an E3 ligase, Cbl-b, is essential for negative regulation of T cells in vivo. Thus, modulation of the E3 ligase activity of Cbl-b might be a novel modality to control T cell immunity in vaccination, cancer biology, or autoimmunity.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Proto-Oncogene Proteins c-cbl/physiology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Line, Tumor , Clonal Anergy/genetics , Enzyme Activation/genetics , Enzyme Activation/immunology , Female , Gene Knock-In Techniques , Immunophenotyping , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Point Mutation , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/genetics , RING Finger Domains/genetics , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Ubiquitin-Protein Ligases/genetics
10.
J Immunol ; 186(4): 2024-32, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21248249

ABSTRACT

The E3 ubiquitin ligase Cbl-b regulates T cell activation thresholds and has been associated with protecting against type 1 diabetes, but its in vivo role in the process of self-tolerance has not been examined at the level of potentially autoaggressive CD4(+) T cells. In this study, we visualize the consequences of Cbl-b deficiency on self-tolerance to lysozyme Ag expressed in transgenic mice under control of the insulin promoter (insHEL). By tracing the fate of pancreatic islet-reactive CD4(+) T cells in prediabetic 3A9-TCR × insHEL double-transgenic mice, we find that Cbl-b deficiency contrasts with AIRE or IL-2 deficiency, because it does not affect thymic negative selection of islet-reactive CD4(+) cells or the numbers of islet-specific CD4(+) or CD4(+)Foxp3(+) T cells in the periphery, although it decreased differentiation of inducible regulatory T cells from TGF-ß-treated 3A9-TCR cells in vitro. When removed from regulatory T cells and placed in culture, Cblb-deficient islet-reactive CD4(+) cells reveal a capacity to proliferate to HEL Ag that is repressed in wild-type cells. This latent failure of T cell anergy is, nevertheless, controlled in vivo in prediabetic mice so that islet-reactive CD4(+) cells in the spleen and the pancreatic lymph node of Cblb-deficient mice show no evidence of increased activation or proliferation in situ. Cblb deficiency subsequently precipitated diabetes in most TCR:insHEL animals by 15 wk of age. These results reveal a role for peripheral T cell anergy in organ-specific self-tolerance and illuminate the interplay between Cblb-dependent anergy and other mechanisms for preventing organ-specific autoimmunity.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 1/immunology , Genetic Predisposition to Disease , Islets of Langerhans/immunology , Proto-Oncogene Proteins c-cbl/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Autoantibodies/biosynthesis , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Disease Progression , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/physiology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Organ Specificity/immunology , Pancreas/immunology , Pancreas/metabolism , Pancreas/pathology , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
11.
Platelets ; 24(6): 419-27, 2013.
Article in English | MEDLINE | ID: mdl-22931288

ABSTRACT

Platelets play a fundamental role in hemostasis. Their functional responses have to be tightly controlled as any disturbance may lead to bleeding disorders or thrombosis. It is thus important to clearly identify and understand the signaling mechanisms involved in platelet function. An important role of c-Cbl and Cbl-b ubiquitin ligases in platelet functional responses and in hematological malignancies has been recently described. Cbl proteins perform negative and positive regulation of several signaling pathways in platelets. In this review, we explore the role of Cbl proteins in platelet functional responses.


Subject(s)
Platelet Activation/physiology , Proto-Oncogene Proteins c-cbl/physiology , Blood Platelets/physiology , Humans
12.
J Biol Chem ; 286(17): 15073-84, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21372129

ABSTRACT

We previously described a non-classical mechanism that arrests FcγRIIa signaling in human neutrophils once engaged by immune complexes or opsonized pathogens. The engagement of FcγRIIa leads to its ubiquitination by the ubiquitin ligase c-Cbl and degradation by the proteasome. Herein, we further examined some of the events regulating this novel pathway. The adaptor protein CIN85 was described in other systems to be involved in the regulation of the c-Cbl-dependent pathway. We found that CIN85 is expressed in human neutrophils and that it translocates like c-Cbl from the cytosol to the plasma membrane following receptor cross-linking. CIN85 was also recruited to the same subset of high density detergent-resistant membrane fractions in which stimulated FcγRIIa partitioned with c-Cbl. The integrity of these microdomains is essential to the FcγRIIa degradation process because the cholesterol-depleting agent methyl-ß-cyclodextrin inhibits this event. Silencing the expression of CIN85 by siRNA in dibutyryl cyclic AMP-differentiated PLB 985 cells prevented FcγRIIa degradation and increased IgG-mediated phagocytosis. Confocal microscopy revealed that the presence of CIN85 is essential to the proper sorting of FcγRIIa during endocytosis. We also provide direct evidence that CIN85 is a substrate of serine/threonine kinase PKCs. Classical PKCs positively regulate FcγRIIa ubiquitination and degradation because these events were inhibited by Gö6976, a classical PKC inhibitor. We conclude that the ubiquitination and degradation of stimulated FcγRIIa mediated by c-Cbl are positively regulated by the adaptor protein CIN85 in a PKC-dependent manner and that these events contribute to the termination of FcγRIIa signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Neutrophils/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-cbl/physiology , Receptors, IgG/biosynthesis , Adaptor Proteins, Signal Transducing/metabolism , Down-Regulation/genetics , Humans , Protein Stability , Protein Transport , Signal Transduction/immunology , Ubiquitination
13.
J Biol Chem ; 286(35): 30263-30273, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21768087

ABSTRACT

The class III receptor-tyrosine kinase Flt3 regulates normal hematopoiesis. An internal tandem duplication (ITD) in the juxtamembrane domain of Flt3 (Flt3-ITD) contributes to transformation and is associated with poor prognosis in acute myeloid leukemia. Here, we demonstrate that, as compared with wild-type Flt3 (Flt3-WT), Flt3-ITD more rapidly undergoes degradation through the proteasomal and lysosomal pathways in model hematopoietic 32D cells and in human leukemic MV4-11 cells. The Hsp90 inhibitor 17-allylaminodemethoxygeldanamycin (17-AAG) preferentially induced the polyubiquitination and proteasomal degradation of Flt3-ITD autophosphorylated on Tyr-591 in these cells. The E3 ubiquitin ligases c-Cbl and to a lesser extent Cbl-b facilitated at least partly Lys-48-linked polyubiquitination of autophosphorylated Flt3-ITD when coexpressed in 293T cells. Moreover, c-Cbl and Cbl-b facilitated degradation of Flt3-ITD in 293T cells and significantly enhanced the 17-AAG-induced decline in autophosphorylated Flt3-ITD. The enhancement of Flt3-ITD degradation was also observed in 32D cells inducibly overexpressing c-Cbl or Cbl-b. Furthermore, overexpression of loss-of-function mutants of both c-Cbl (c-Cbl-R420Q) and Cbl-b (Cbl-b-C373A) together in 32D cells retarded the degradation of autophosphorylated Flt3-ITD and significantly inhibited the 17-AAG-induced degradation of Flt3-ITD to confer the resistance to cytotoxicity of 17-AAG on these cells. These results suggest that c-Cbl as well as Cbl-b may play important roles in Hsp90 inhibitor-induced degradation of Flt3-ITD through the ubiquitin proteasome system and in regulation of the basal expression level of Flt3-ITD in leukemic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Benzoquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Proto-Oncogene Proteins c-cbl/physiology , fms-Like Tyrosine Kinase 3/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Leukemic , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Kinetics , Leukemia, Myeloid, Acute/drug therapy , Lysosomes/metabolism , Mice , Phosphorylation , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/chemistry
14.
Med Sci (Paris) ; 28(11): 970-5, 2012 Nov.
Article in French | MEDLINE | ID: mdl-23171901

ABSTRACT

Cbl ubiquitin ligases are important molecules that control the process of ubiquitination and degradation of proteins by the proteasome. Because this process regulates several intracellular mechanisms, alterations in Cbl activity lead to several pathologies including cancer. In bone, the c-Cbl ubiquitin ligase is known to control osteoclast activity. Our studies indicate that c-Cbl also regulates osteoblast proliferation, differentiation and survival. We recently showed that inhibition of c-Cbl activity using a c-Cbl mutant leads to promote osteoblast differentiation in mesenchymal stromal cells as a consequence of increased receptor tyrosine kinase expression. Conversely, we found that overexpression of c-Cbl leads to inhibit osteosarcoma cell proliferation and tumorigenesis through downregulation of these receptors. Thus, the use of pharmacological agents capable of modulating c-Cbl activity may be of therapeutic interest for promoting bone formation in normal bone, or to reduce tumorigenesis in primary bone cancer.


Subject(s)
Bone Neoplasms/enzymology , Bone Resorption/enzymology , Cell Transformation, Neoplastic , Neoplasm Proteins/physiology , Osteoblasts/enzymology , Osteoclasts/enzymology , Osteogenesis/physiology , Osteosarcoma/enzymology , Proto-Oncogene Proteins c-cbl/physiology , Animals , Bone Neoplasms/drug therapy , Cell Differentiation , Cell Division , Humans , Mice , Mice, Knockout , Models, Biological , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Osteosarcoma/drug therapy , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology , Ubiquitination
15.
J Biol Chem ; 285(47): 36745-58, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20851882

ABSTRACT

Cbl is an adaptor protein and an E3 ligase that plays both positive and negative roles in several signaling pathways that affect various cellular functions. Tyrosine 737 is unique to Cbl and is phosphorylated by Syk and Src family kinases. Phosphorylated Cbl Tyr(737) creates a binding site for the p85 regulatory subunit of PI3K, which also plays an important role in the regulation of bone resorption by osteoclasts. To investigate the role of Cbl-PI3K interaction in bone homeostasis, we examined the knock-in mice (Cbl(YF/YF)) in which the PI3K binding site in Cbl is ablated due to the mutation in the regulatory tyrosine. We report that in Cbl(YF/YF) mice, despite increased numbers of osteoclasts, bone volume is increased due to defective osteoclast function. Additionally, in ex vivo cultures, mature Cbl(YF/YF) osteoclasts showed an increased ability to survive in the presence of RANKL due to delayed onset of apoptosis. RANKL-mediated signaling is perturbed in Cbl(YF/YF) osteoclasts, and most interestingly, AKT phosphorylation is up-regulated, suggesting that the lack of PI3K sequestration by Cbl results in increased survival and decreased bone resorption. Cumulatively, these in vivo and in vitro results show that, on one hand, binding of Cbl to PI3K negatively regulates osteoclast differentiation, survival, and signaling events (e.g. AKT phosphorylation), whereas on the other hand it positively influences osteoclast function.


Subject(s)
Apoptosis , Bone Resorption/prevention & control , Osteoclasts/cytology , Osteoclasts/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-cbl/physiology , RANK Ligand/metabolism , Animals , Blotting, Western , Bone Resorption/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
16.
J Biol Chem ; 285(14): 10969-81, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20133944

ABSTRACT

The ability of thymocytes to assess T cell receptor (TCR) signaling strength and initiate the appropriate downstream response is crucial for determining their fate. We have previously shown that a c-Cbl RING finger mutant knock-in mouse, in which the E3 ubiquitin ligase activity of c-Cbl is inactivated, is highly sensitive to TCR-induced death signals that cause thymic deletion. This high intensity signal involves the enhanced tyrosine phosphorylation of the mutant c-Cbl protein promoting a marked increase in the activation of Akt. Here we show that this high intensity signal in c-Cbl RING finger mutant thymocytes also promotes the enhanced induction of two mediators of TCR-directed thymocyte apoptosis, Nur77 and the pro-apoptotic Bcl-2 family member, Bim. In contrast, a knock-in mouse harboring a mutation at Tyr-737, the site in c-Cbl that activates phosphatidylinositol 3-kinase, shows reduced TCR-mediated responses including suppression of Akt activation, a reduced induction of Nur77 and Bim, and greater resistance to thymocyte death. These findings identify tyrosine-phosphorylated c-Cbl as a critical sensor of TCR signal strength that regulates the engagement of death-promoting signals.


Subject(s)
Apoptosis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/physiology , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Blotting, Western , Cells, Cultured , Flow Cytometry , Immunoprecipitation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mutation/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thymus Gland/cytology , Tyrosine/genetics
17.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G368-76, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21617115

ABSTRACT

Upon ligand binding, epidermal growth factor (EGF) receptor (R) autophosphorylates on COOH-terminal tyrosines, generating docking sites for signaling partners that stimulate proliferation, restitution, and chemotaxis. Specificity for individual EGFR tyrosines in cellular responses has been hypothesized but not well documented. Here we tested the requirement for particular tyrosines, and associated downstream pathways, in mouse colon epithelial cell chemotactic migration. We compared these requirements to those for the phenotypically distinct restitution (wound healing) migration. Wild-type, Y992/1173F, Y1045F, Y1068F, and Y1086F EGFR constructs were expressed in EGFR(-/-) cells; EGF-induced chemotaxis or restitution were determined by Boyden chamber or modified scratch wound assay, respectively. Pharmacological inhibitors of p38, phospholipase C (PLC), Src, MEK, JNK/SAPK, phosphatidylinositol 3-kinase (PI 3-kinase), and protein kinase C (PKC) were used to block EGF-stimulated signaling. Pathway activation was determined by immunoblot analysis. Unlike wild-type EGFR, Y992/1173F and Y1086F EGFR did not stimulate colon epithelial cell chemotaxis toward EGF; Y1045F and Y1068F EGFR partially stimulated chemotaxis. Only wild-type EGFR promoted colonocyte restitution. Inhibition of p38, PLC, and Src, or Grb2 knockdown, blocked chemotaxis; JNK, PI 3-kinase, and PKC inhibitors or c-Cbl knockdown blocked restitution but not chemotaxis. All four EGFR mutants stimulated downstream signaling in response to EGF, but Y992/1173F EGFR was partially defective in PLCγ activation whereas both Y1068F and Y1086F EGFR failed to activate Src. We conclude that specific EGFR tyrosines play key roles in determining cellular responses to ligand. Chemotaxis and restitution, which have different migration phenotypes and physiological consequences, have overlapping but not identical EGFR signaling requirements.


Subject(s)
Chemotaxis/physiology , Epidermal Growth Factor/physiology , Epithelial Cells/physiology , ErbB Receptors/metabolism , Signal Transduction/physiology , Tyrosine/physiology , Animals , Binding Sites/physiology , Cell Line , Chemotaxis/drug effects , Colon/metabolism , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , ErbB Receptors/deficiency , ErbB Receptors/genetics , ErbB Receptors/physiology , GRB2 Adaptor Protein/deficiency , GRB2 Adaptor Protein/physiology , Mice , Mutation , Phospholipase C gamma/physiology , Phosphorylation , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/physiology , RNA, Small Interfering , Transfection , Wound Healing/drug effects , Wound Healing/physiology , p38 Mitogen-Activated Protein Kinases/physiology , src Homology Domains/physiology , src-Family Kinases
18.
Acta Oncol ; 50(5): 693-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21175263

ABSTRACT

BACKGROUND: Tamoxifen (TAM) is a nonsteroidal antiestrogen that has been widely used in the treatment of breast cancer through its anti-estrogen activity. Recent studies show that TAM is cytotoxic to both estrogen receptor (ER)-positive and ER-negative cells via the induction of apoptosis. However, the molecular mechanisms of this effect are not well understood. In the present study, we investigated the roles of c-Src, ERK, AKT and c-Cbl ubiquitin ligases during TAM-induced apoptosis of MCF-7 cells. MATERIAL AND METHODS: MCF-7 cell proliferation and apoptosis were measured by 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, and flow cytometry, respectively. c-Cbl expression, and the activity of c-Src, ERK, AKT were assayed by Western blotting. Overexpression of the wild and the dominant-negative type of c-Cbl (70Z/Cbl) were achieved by transient transfection of plasmids encoding c-Cbl and 70Z/Cbl, respectively, and were confirmed by Western blotting. Statistical analysis was performed using the t-test, and a p-value <0.05 was considered to be statistically significant. RESULTS: A high concentration of TAM (25 µM) induced a time-dependent apoptosis of MCF-7 cells. ERK1/2 and AKT were activated during TAM-induced apoptosis. The ERK1/2 inhibitor PD98059, the PI3K/Akt inhibitor LY294002, and the c-Src inhibitor PP2 all enhanced TAM action. Moreover, the ubiquitin ligase c-Cbl was up-regulated during this process. Over-expression of c-Cbl significantly enhanced the apoptosis-inducing effects of TAM, while 70Z/Cbl suppressed the apoptosis-inducing effects of TAM. Further investigation revealed that, overexpression of c-Cbl significantly downregulated the c-Src protein levels and TAM-induced AKT activity. But 70Z/Cbl significantly upregulated TAM-induced ERK and AKT activity. CONCLUSIONS: This study demonstrates that c-Src, ERK, and AKT played a protective role during TAM-induced apoptosis, and that c-Cbl sensitized MCF-7 cells to TAM by modulating the expression of c-Src, and TAM-induced ERK and AKT activity.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Carcinoma/pathology , Proto-Oncogene Proteins c-cbl/physiology , Tamoxifen/pharmacology , Apoptosis/genetics , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Estrogen Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology
19.
J Immunol ; 182(9): 5654-62, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380812

ABSTRACT

Fc gamma receptor (Fc gammaR)-mediated phagocytosis is known to require tyrosine kinases (TKs). We identified c-Cbl and Cbl-b as proteins that undergo tyrosine phosphorylation during phagocytosis. Cbl-deficient macrophages displayed enhanced Fc gammaR-mediated signaling and phagocytosis. Surprisingly, binding of IgG-coated targets (EIgG) was also enhanced. c-Cbl-deficient macrophages expressed less Fc gammaRIIb, the inhibitory Fc gamma receptor; however, this did not account for enhanced target binding. We isolated the function of one Fc receptor isoform, Fc gammaRI, using IgG2a-coated targets (EIgG2a). Cbl-deficient macrophages demonstrated a disproportionate increase in binding EIgG2a, suggesting that signal strength regulates binding efficiency toward opsonized targets. In resting cells, Fc gammaRI colocalized with the Src family TK Hck in F-actin-rich structures, which was enhanced in Cbl-deficient macrophages. Target binding was sensitive to TK inhibitors, profoundly inhibited following depletion of cholesterol, and ablated at 4 degrees C or in the presence of inhibitors of actin polymerization. Sensitivity of EIgG binding to cytoskeletal disruption was inversely proportional to opsonin density. These findings challenge the view that Fc gammaR-mediated binding is a passive event. They suggest that dynamic engagement of TKs and the cytoskeleton enables macrophages to serve as cellular "Venus fly traps", with the capacity to capture phagocytic targets under conditions of limiting opsonin density.


Subject(s)
Actins/metabolism , Macrophages/immunology , Macrophages/metabolism , Phagocytosis/immunology , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/genetics , Receptors, IgG/physiology , Signal Transduction/immunology , Actins/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Down-Regulation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Binding/immunology , Proto-Oncogene Proteins c-cbl/physiology , Receptors, IgG/antagonists & inhibitors , Tyrosine/metabolism
20.
Front Immunol ; 12: 757231, 2021.
Article in English | MEDLINE | ID: mdl-34630435

ABSTRACT

Dendritic cells (DCs) are composed of multiple lineages of hematopoietic cells and orchestrate immune responses upon detecting the danger and inflammatory signals associated with pathogen and damaged tissues. Under steady-state, DCs are maintained at limited numbers and the functionally quiescent status. While it is known that a fine balance in the DC homeostasis and activation status is also important to prevent autoimmune diseases and hyperinflammation, mechanisms that control DC development and activation under stead-state remain not fully understood. Here we show that DC-specific ablation of CBL and CBL-B (CBL-/-CBL-B-/-) leads to spontaneous liver inflammation and fibrosis and early death of the mice. The mutant mice have a marked expansion of classic CD8α+/CD103+ DCs (cDC1s) in peripheral lymphoid organs and the liver. These DCs exhibit atypical activation phenotypes characterized by an increased production of inflammatory cytokines and chemokines but not the cell surface MHC-II and costimulatory ligands. While the mutant mice also have massive T cell activation, lymphocytes are not required for the disease development. The CBL-/-CBL-B-/- mutation enhances FLT3-mTOR signaling, due to defective FLT3 ubiquitination and degradation. Blockade of FLT3-mTOR signaling normalizes the homeostasis of cDC1s and attenuates liver inflammation. Our result thus reveals a critical role of CBLs in the maintenance of DC homeostasis and immune quiescence. This regulation could be relevant to liver inflammatory diseases and fibrosis in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Dendritic Cells/immunology , Proto-Oncogene Proteins c-cbl/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigen Presentation , Cell Division , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/immunology , Homeostasis , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Point Mutation , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/genetics , Sirolimus/pharmacology , fms-Like Tyrosine Kinase 3/physiology
SELECTION OF CITATIONS
SEARCH DETAIL