Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Cancer Res ; 80(24): 5583-5596, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33093168

ABSTRACT

T-cell exhaustion was initially identified in chronic infection in mice and was subsequently described in humans with cancer. Although the distinct signature of exhausted T (TEX) cells in cancer has been well investigated, the molecular mechanism of T-cell exhaustion in cancer is not fully understood. Using single-cell RNA sequencing, we report here that TEX cells in esophageal cancer are more heterogeneous than previously clarified. Sprouty RTK signaling antagonist 1 (SPRY1) was notably enriched in two subsets of exhausted CD8+ T cells. When overexpressed, SPRY1 impaired T-cell activation by interacting with CBL, a negative regulator of ZAP-70 tyrosine phosphorylation. Data from the Tumor Immune Estimation Resource revealed a strong correlation between FGF2 and SPRY1 expression in esophageal cancer. High expression of FGF2 was evident in fibroblasts from esophageal cancer tissue and correlated with poor overall survival. In vitro administration of FGF2 significantly upregulated expression of SPRY1 in CD8+ T cells and attenuated T-cell receptor-triggered CD8+ T-cell activation. A mouse tumor model confirmed that overexpression of FGF2 in fibroblasts significantly upregulated SPRY1 expression in TEX cells, impaired T-cell cytotoxic activity, and promoted tumor growth. Thus, these findings identify FGF2 as an important regulator of SPRY1 expression involved in establishing the dysfunctional state of CD8+ T cells in esophageal cancer. SIGNIFICANCE: These findings reveal FGF2 as an important regulator of SPRY1 expression involved in establishing the dysfunctional state of CD8+ T cells and suggest that inhibition of FGF2 has potential clinical value in ESCC. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/24/5583/F1.large.jpg.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CD8-Positive T-Lymphocytes/immunology , Cancer-Associated Fibroblasts/metabolism , Esophageal Neoplasms/metabolism , Fibroblast Growth Factor 2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Animals , Disease Models, Animal , Esophageal Neoplasms/pathology , Female , Fibroblast Growth Factor 2/pharmacology , Humans , Jurkat Cells , Lymphocyte Activation , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Oncogene Protein v-cbl/genetics , Oncogene Protein v-cbl/metabolism , Phosphoproteins/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transfection
2.
Pharmacol Res ; 160: 105149, 2020 10.
Article in English | MEDLINE | ID: mdl-32822868

ABSTRACT

Cancer chemotherapy induced neutropenia (CCIN) is one of the most common toxicity caused by cytotoxic anticancer agents. Despite granulocyte colony-stimulating factor (GCSF) is widely used in clinical practice, the infection and infection-related mortality rate is still high for lack of functionally mature neutrophils. Saikosaponin d (SSD) is one of the major bioactive constituents of Radix Bupleuri (RB), which exerts immune-modulatory properties. We explored the function of SSD in CCIN therapy, we found that SSD contributed to generate functional mature neutrophils which capable of fighting infection both in vitro and in vivo. Network pharmacology was employed to explore the mechanism, 61 signal pathways might play an important role in CCIN treatment. Western Blot was employed to further confirm the potential pathway involved. We found CBL-ERK1/2 pathway was activated by SSD, followed by upregulating PU.1 and CEBPß expression and leading to neutrophil differentiation. Our findings suggest a natural regimen SSD which could regenerate microbicidal neutrophils to effectively reduce CCIN-associated infection via activating CBL-ERK1/2, providing a rationale for future therapeutic approaches.


Subject(s)
Antineoplastic Agents/adverse effects , Immunologic Factors/therapeutic use , MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Neutropenia/chemically induced , Neutropenia/drug therapy , Neutrophils/drug effects , Oleanolic Acid/analogs & derivatives , Oncogene Protein v-cbl/drug effects , Saponins/therapeutic use , Animals , Blood Bactericidal Activity , CCAAT-Enhancer-Binding Protein-beta/drug effects , Cell Differentiation/drug effects , Infection Control , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/therapeutic use
3.
Blood Rev ; 43: 100652, 2020 09.
Article in English | MEDLINE | ID: mdl-31980238

ABSTRACT

Juvenile myelomonocytic leukaemia (JMML) is a rare clonal disorder of early childhood. Constitutive activation of the RAS pathway is the initial event in JMML. Around 90% of patients diagnosed with JMML carry a mutation in the PTPN11, NRAS, KRAS, NF1 or CBL genes. It has been demonstrated that after this first genetic event, an additional somatic mutation or epigenetic modification is involved in disease progression. The available genetic and clinical data have enabled researchers to establish relationships between JMML and several clinical conditions, including Noonan syndrome, Ras-associated lymphoproliferative disease, and Moyamoya disease. Despite scientific progress and the development of more effective treatments, JMML is still a deadly disease: the 5-year survival rate is ~50%. Here, we report on recent research having led to a better understanding of the genetic and molecular mechanisms involved in JMML.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myelomonocytic, Juvenile/genetics , Mutation , Animals , Epigenesis, Genetic , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Oncogene Protein v-cbl/genetics , Oncogene Protein v-cbl/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism
4.
J Int Med Res ; 47(5): 1856-1867, 2019 May.
Article in English | MEDLINE | ID: mdl-30614340

ABSTRACT

OBJECTIVE: To investigate the effect of ascorbic acid (AA) on hemostatic function during living donor liver transplantation (LDLT). METHODS: Blood samples from 21 LDLT recipients were taken within 30 minutes after induction and at 120 minutes after reperfusion. Rotational thromboelastography (TEG) and western blot analysis were used to analyze for fibrinolysis and functional changes in c-Cbl and Cbl-b, respectively. TEG test samples were prepared as one of three groups: C group (0.36 mL of blood), N group (0.324 mL of blood + 0.036 mL of 0.9% normal saline), and A group (0.324 mL of blood + 0.036 mL of 200 µmol/L-AA dissolved in 0.9% normal saline). RESULTS: AA decreased fibrinolysis and increased clot rigidity at baseline and 120 minutes after reperfusion. Cbl-b expression was significantly increased at baseline and 120 minutes after reperfusion in the A group compared with the C and N groups. However, c-Cbl phosphorylation was most significantly decreased in the A group at baseline and 120 minutes after reperfusion. CONCLUSION: AA can significantly decrease fibrinolysis and improve clot rigidity in LT recipients during LDLT, and functional changes in Cbl-b and c-Cbl might represent the underlying mechanism. AA may be considered for use during LDLT to decrease hyperfibrinolysis.


Subject(s)
Ascorbic Acid/therapeutic use , Blood Platelets/drug effects , Gene Expression Regulation/drug effects , Liver Transplantation/adverse effects , Living Donors/statistics & numerical data , Oncogene Protein v-cbl/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Thrombosis/drug therapy , Antioxidants/therapeutic use , Blood Platelets/metabolism , Blood Platelets/pathology , Female , Humans , Male , Middle Aged , Oncogene Protein v-cbl/genetics , Phosphorylation , Prospective Studies , Proto-Oncogene Proteins c-cbl/genetics , Thrombosis/etiology , Thrombosis/metabolism
5.
Eur Heart J ; 40(4): 372-382, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30452556

ABSTRACT

Aims: The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results: The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb-/-Apoe-/- mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb-/-Apoe-/- macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb-/-Apoe-/- CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb-/-Apoe-/- bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion: Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques.


Subject(s)
Atherosclerosis/etiology , CD8-Positive T-Lymphocytes/immunology , Lymphoma, B-Cell/complications , Macrophages/pathology , Oncogene Protein v-cbl/metabolism , Plaque, Atherosclerotic/etiology , Animals , Apoptosis , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
6.
Int J Cancer ; 140(11): 2596-2607, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28198003

ABSTRACT

Activating mutation of epidermal growth factor receptor (EGFR) is correlated with malignant lung tumor. In our study, we demonstrated that recombinant LZ-8 (rLZ-8), a medicinal mushroom Ganoderma lucidum protein, induced cell cycle arrest and apoptosis by downregulating the expression of wild-type and mutated EGFR and inhibiting EGFR downstream effectors, AKT and ERK1/2 in lung cancer cells. We showed that rLZ-8 effectively inhibited lung cancer progression and suppressed EGFR expression of lung tumor lesions in mouse model. Functional studies revealed that rLZ-8 reduced the amount of EGFR in cell membranes by altering EGFR localization to enhance the EGF-induced degradation of EGFR. Mechanistically, we demonstrated that rLZ-8 bound to EGFR to induce EGFR autophosphorylation at tyrosine1045 and trigger ubiquitination by inducing the formation of EGFR/Cbl complexes, resulting in the degradation of EGFR; however, Cbl-shRNA abolished rLZ-8-induced EGFR degradation. We provide the first evidence showing that rLZ-8 inhibits growth and induces apoptosis of lung cancer cells by promoting EGFR degradation. The current findings therefore suggest a novel anti-cancer function of rLZ-8 that targeting EGFR overexpression or mutation as well as EGFR-dependent processes in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/genetics , Fungal Proteins/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Oncogene Protein v-cbl/genetics , Recombinant Proteins/pharmacology , A549 Cells , Agaricales/genetics , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Proliferation/drug effects , Down-Regulation/drug effects , Fungal Proteins/economics , Humans , Male , Mice , Mice, Inbred C57BL , Mutation/drug effects , Mutation/genetics , Reishi/genetics , Signal Transduction/drug effects , Ubiquitination/drug effects
7.
BMC Biol ; 14: 76, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27609087

ABSTRACT

BACKGROUND: Casitas B-lineage lymphoma (Cbl or c-Cbl) is a RING ubiquitin ligase that negatively regulates protein tyrosine kinase (PTK) signalling. Phosphorylation of a conserved residue (Tyr371) on the linker helix region (LHR) between the substrate-binding and RING domains is required to ubiquitinate PTKs, thereby flagging them for degradation. This conserved Tyr is a mutational hotspot in myeloproliferative neoplasms. Previous studies have revealed that select point mutations in Tyr371 can potentiate transformation in cells and mice but not all possible mutations do so. To trigger oncogenic potential, Cbl Tyr371 mutants must perturb the LHR-substrate-binding domain interaction and eliminate PTK ubiquitination. Although structures of native and pTyr371-Cbl are available, they do not reveal how Tyr371 mutations affect Cbl's conformation. Here, we investigate how Tyr371 mutations affect Cbl's conformation in solution and how this relates to Cbl's ability to potentiate transformation in cells. RESULTS: To explore how Tyr371 mutations affect Cbl's properties, we used surface plasmon resonance to measure Cbl mutant binding affinities for E2 conjugated with ubiquitin (E2-Ub), small angle X-ray scattering studies to investigate Cbl mutant conformation in solution and focus formation assays to assay Cbl mutant transformation potential in cells. Cbl Tyr371 mutants enhance E2-Ub binding and cause Cbl to adopt extended conformations in solution. LHR flexibility, RING domain accessibility and transformation potential are associated with the extent of LHR-substrate-binding domain perturbation affected by the chemical nature of the mutation. More disruptive mutants like Cbl Y371D or Y371S are more extended and the RING domain is more accessible, whereas Cbl Y371F mimics native Cbl in solution. Correspondingly, the only Tyr371 mutants that potentiate transformation in cells are those that perturb the LHR-substrate-binding domain interaction. CONCLUSIONS: c-Cbl's LHR mutations are only oncogenic when they disrupt the native state and fail to ubiquitinate PTKs. These findings provide new insights into how LHR mutations deregulate c-Cbl.


Subject(s)
Cell Proliferation , Myeloproliferative Disorders/genetics , Neoplasms/genetics , Oncogene Protein v-cbl/genetics , Point Mutation , Protein Conformation , 3T3 Cells , Animals , Mice , Oncogene Protein v-cbl/chemistry , Phosphorylation
8.
J Immunol ; 193(4): 1672-80, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25000980

ABSTRACT

The generation of T cell anergy is associated with upregulation of ubiquitin E3 ligases including Casitas B-lineage lymphoma (Cbl-b), Itch, gene related to anergy in lymphocyte, and deltex1 (DTX1). These E3 ligases attenuate T cell activation by targeting to signaling molecules. For example, Cbl-b and Itch promote the degradation of protein kinase Cθ (PKCθ) and phospholipase C-γ1 (PLC-γ1) in anergic Th1 cells. How these anergy-associated E3 ligases coordinate during T cell anergy remains largely unknown. In the current study, we found that PKCθ and PLC-γ1 are also downregulated by DTX1. DTX1 interacted with PKCθ and PLC-γ1 and stimulated the degradation of PKCθ and PLC-γ1. T cell anergy-induced proteolysis of PKCθ was prevented in Dtx1(-/-) T cells, supporting the essential role of DTX1 in PKCθ downregulation. Similar to Cbl-b and Itch, DTX1 promoted monoubiquitination of PKCθ. Proteasome inhibitor did not inhibit DTX1-directed PKCθ degradation, but instead DTX1 directed the relocalization of PKCθ into the lysosomal pathway. In addition, DTX1 interacted with Cbl-b and increased the protein levels of Cbl-b. We further demonstrated the possibility that, through the downregulation of PKCθ, DTX1 prevented PKCθ-induced Cbl-b degradation and increased Cbl-b protein stability. Our results suggest the coordination between E3 ligases during T cell anergy; DTX1 acts with Cbl-b to assure a more extensive silencing of PKCθ, whereas DTX1-mediated PKCθ degradation further stabilizes Cbl-b.


Subject(s)
DNA-Binding Proteins/genetics , Isoenzymes/metabolism , Oncogene Protein v-cbl/biosynthesis , Protein Kinase C/metabolism , Proteolysis , Th1 Cells/immunology , Animals , Calcimycin/pharmacology , Cell Line , Clonal Anergy , DNA-Binding Proteins/biosynthesis , Down-Regulation , HEK293 Cells , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Jurkat Cells , Lymphocyte Activation/immunology , Lysosomes/immunology , Mice , Mice, Knockout , Oncogene Protein v-cbl/genetics , Phospholipase C gamma/biosynthesis , Phospholipase C gamma/metabolism , Proteasome Inhibitors/pharmacology , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Protein Kinase C-theta , RNA Interference , RNA, Small Interfering , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , Ubiquitination , ZAP-70 Protein-Tyrosine Kinase/biosynthesis
9.
Leuk Res ; 36(8): 1009-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22591685

ABSTRACT

Somatic CBL mutations have been reported in a variety of myeloid neoplasms but are rare in acute lymphoblastic leukemia (ALL). We analyzed 77 samples from hematologic malignancies, identifying a somatic mutation in CBL (p.C381R) in one patient with T-ALL that was associated with a uniparental disomy at the CBL locus and a germline heterozygous mutation in one patient with JMML. Two NOTCH1 mutations and homozygous deletions in LEF1 and CDKN2A were identified in T-ALL cells. The activation of the RAS pathway was enhanced, and activation of the NOTCH1 pathway was inhibited in NIH 3T3 cells that expressed p.C381R. This study appears to be the first to identify a CBL mutation in T-ALL.


Subject(s)
Mutation , Oncogene Protein v-cbl/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Base Sequence , Cells, Cultured , Child , Cohort Studies , DNA Mutational Analysis , Female , Gene Dosage , Humans , Infant , Male , Mice , Mutation/physiology , NIH 3T3 Cells , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
10.
J Med Chem ; 55(7): 3583-7, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22394513

ABSTRACT

We describe truncation and SAR studies to identify a pentapeptide that binds Cbl tyrosine kinase binding domain with a higher affinity than the parental peptide. The pentapeptide has an alternative binding mode that allows occupancy of a previously uncharacterized groove. A peptide library was used to map the binding site and define the interface landscape. Our results suggest that the pentapeptide is an ideal starting point for the development of inhibitors against Cbl driven diseases.


Subject(s)
Models, Molecular , Oligopeptides/chemistry , Oncogene Protein v-cbl/chemistry , Protein-Tyrosine Kinases/chemistry , Binding Sites , Oncogene Protein v-cbl/metabolism , Peptide Library , Protein Binding , Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Thermodynamics
11.
Genes Chromosomes Cancer ; 51(3): 250-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22072526

ABSTRACT

Casitas B-lineage lymphoma (CBL) proteins are RING finger ubiquitin E3 ligases that attenuate the signaling of receptor tyrosine kinases and are mutated in a number of myeloid disorders. In this study, mutational screening of the linker-RING domains of CBL and CBLB was performed by denaturing high performance liquid chromatography in a cohort of diagnostic (n = 180) or relapse (n = 46) samples from children with acute lymphoblastic leukemia. Somatic mutations were identified in three children, giving an overall incidence of 1.7% and involved small deletions affecting the intron/exon boundaries of exon 8, leading to skipping of exon 8 and abolishing E3 ligase function. Mutated primary samples were associated with constitutive activation of the RAS pathway and sensitivity to MEK inhibitors was shown. Thus, mutation of CBL is an alternative route to activate the RAS pathway and may identify children who are candidates for MEK inhibitor clinical trials.


Subject(s)
Mutation , Oncogene Protein v-cbl/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction/genetics , Adolescent , Base Sequence , Child , Child, Preschool , Chromatography, Liquid , Cohort Studies , DNA Mutational Analysis , Exons , Female , Humans , Introns , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Molecular Sequence Data , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/pharmacology , RING Finger Domains , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Recurrence , United Kingdom
12.
J Biol Chem ; 286(27): 24443-50, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21596750

ABSTRACT

Human bone marrow-derived mesenchymal stromal cells (hMSCs) have the capacity to differentiate into several cell types including osteoblasts and are therefore an important cell source for bone tissue regeneration. A crucial issue is to identify mechanisms that trigger hMSC osteoblast differentiation to promote osteogenic potential. Casitas B lineage lymphoma (Cbl) is an E3 ubiquitin ligase that ubiquitinates and targets several molecules for degradation. We hypothesized that attenuation of Cbl-mediated degradation of receptor tyrosine kinases (RTKs) may promote osteogenic differentiation in hMSCs. We show here that specific inhibition of Cbl interaction with RTKs using a Cbl mutant (G306E) promotes expression of osteoblast markers (Runx2, alkaline phosphatase, type 1 collagen, osteocalcin) and increases osteogenic differentiation in clonal bone marrow-derived hMSCs and primary hMSCs. Analysis of molecular mechanisms revealed that the Cbl mutant increased PDGF receptor α and FGF receptor 2 but not EGF receptor expression in hMSCs, resulting in increased ERK1/2 and PI3K signaling. Pharmacological inhibition of FGFR or PDGFR abrogated in vitro osteogenesis induced by the Cbl mutant. The data reveal that specific inhibition of Cbl interaction with RTKs promotes the osteogenic differentiation program in hMSCs in part by decreased Cbl-mediated PDGFRα and FGFR2 ubiquitination, providing a novel mechanistic approach targeting Cbl to promote the osteogenic capacity of hMSCs.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Mutation, Missense , Oncogene Protein v-cbl/metabolism , Osteogenesis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Substitution , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Bone Marrow Cells/cytology , Cell Line, Transformed , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Protein v-cbl/antagonists & inhibitors , Oncogene Protein v-cbl/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Stromal Cells/cytology , Stromal Cells/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
13.
J Periodontol ; 82(7): 1018-24, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21189087

ABSTRACT

BACKGROUND: Periodontitis is a complex multifactorial disease and is typically polygenic in origin. Genes play a fundamental part in each biologic process forming complex networks of interactions. However, only some genes have a high number of interactions with other genes in the network and may, therefore, be considered to play an important role. In a preliminary bioinformatic analysis, five genes that showed a higher number of interactions were identified and termed leader genes. In the present study, we use real-time quantitative polymerase chain reaction (PCR) technology to evaluate the expression levels of leader genes in the leukocytes of 10 patients with refractory chronic periodontitis and compare the expression levels with those of the same genes in 24 healthy patients. METHODS: Blood was collected from 24 healthy human subjects and 10 patients with refractory chronic periodontitis and placed into heparinized blood collection tubes by personnel trained in phlebotomy using a sterile technique. Blood leukocyte cells were immediately lysed by using a kit for total RNA purification from human whole blood. Complementary DNA (cDNA) synthesis was obtained from total RNA and then real-time quantitative PCR was performed. PCR efficiencies were calculated with a relative standard curve derived from a five cDNA dilution series in triplicate that gave regression coefficients >0.98 and efficiencies >96%. The standard curves were obtained using glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and growth factor receptor binding protein 2 (GRB2), casitas B-lineage lymphoma (CBL), nuclear factor-KB1 (NFKB1), and REL-A (gene for transcription factor p65) gene primers and amplified with 1.6, 8, 40, 200, and 1,000 ng/µL total cDNA. Curves obtained for each sample showed a linear relationship between RNA concentrations and the cycle threshold value of real-time quantitative PCR for all genes. Data were expressed as mean ± SE (SEM). The groups were compared to the analysis of variance. A probability value <0.01 was considered statistically significant. RESULTS: The present study agrees with the preliminary bioinformatics analysis. In our experiments, the association of pathology with the genes was statistically significant for GRB2 and CBL (P <0.01), and it was not statistically significant for REL-A and NFKB1. CONCLUSION: This article lends support to our preliminary hypothesis that assigned an important role in refractory aggressive periodontitis to leader genes.


Subject(s)
Aggressive Periodontitis/genetics , Chronic Periodontitis/genetics , Polymerase Chain Reaction/methods , Adult , Computational Biology , Disease Progression , Female , GRB2 Adaptor Protein/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Male , Middle Aged , NF-kappa B p50 Subunit/genetics , Oncogene Protein v-cbl/genetics , Transcription Factor RelA/genetics
14.
J Clin Pathol ; 63(10): 942-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20671051

ABSTRACT

A loss-of-function mutation of TET2, CBL and CEBPA has been implicated in the pathogenesis or leukaemic transformation of myeloproliferative neoplasm. As tumour suppressor genes may potentially be inactivated by promoter hypermethylation, the authors studied the methylation status of these genes in three cell lines and diagnostic marrow samples from 45 patients with myeloproliferative neoplasm (MPN) (essential thrombocythaemia, N=34; polycythaemia vera, N=7 and primary myelofibrosis, N=4) by methylation-specific PCR. TET2 was heterozygously methylated in MEG-01 and K562 but completely unmethylated in HEL. On the other hand, both CBL and CEBPA were completely unmethylated in all three cell lines. In the primary marrow samples, methylation of TET2 occurred in two (5.9%) patients with essential thrombocythaemia (4.4% of all patients), both without JAK2 V617 mutation, but not in polycythaemia vera or primary myelofibrosis. There was no association between TET2 methylation with the type of MPN (p=0.713). Hypermethylation of CBL or CEBPA was not detected in any patients. In summary, methylation of TET2, CBL and CEBPA is infrequent in MPN at diagnosis. The role of methylation of these genes at the time of leukaemic transformation warrants further study.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Myeloproliferative Disorders/genetics , Oncogene Protein v-cbl/genetics , Proto-Oncogene Proteins/genetics , Adult , Aged , Aged, 80 and over , Base Sequence , CpG Islands/genetics , Dioxygenases , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Polycythemia Vera/genetics , Polymerase Chain Reaction/methods , Primary Myelofibrosis/genetics , Thrombocythemia, Essential/genetics
15.
Ann N Y Acad Sci ; 1192: 376-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392263

ABSTRACT

The Cbl protein is a key player in macrophage colony-stimulating factor (M-CSF)-induced signaling. To examine the role of Cbl in M-CSF-mediated cellular events, we used Cbl(YF/YF) knockin mice in which the regulatory tyrosine 737, which when phosphorylated binds to the p85 subunit of phosphatidylinositol 3 kinase (PI3K), is substituted to phenylalanine. In ex vivo cultures, M-CSF and receptor activator of nuclear factor-kappaB ligand-mediated differentiation of bone marrow precursors from Cbl(YF/YF) mice generated increased number of osteoclasts; however, osteoclast numbers in Cbl(YF/YF) cultures were unchanged with increasing doses of M-CSF. We found that Cbl(YF/YF) osteoclasts have enhanced intrinsic ability to survive, and this response was further augmented upon exposure to M-CSF. Treatment of osteoclasts with M-CSF-induced actin reorganization and lamellipodia formation in wild-type osteoclasts; however, in Cbl(YF/YF) osteoclasts lamellipodia formation was compromised. Collectively, these results indicate that abrogation of the Cbl-PI3K interaction, although not affecting M-CSF-induced proliferation and differentiation of precursors, is required for regulation of survival and actin cytoskeletal reorganization of mature osteoclasts.


Subject(s)
Colony-Stimulating Factors/pharmacology , Cytoskeleton/drug effects , Oncogene Protein v-cbl/metabolism , Osteoclasts/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Animals , Bone Remodeling/genetics , Bone Remodeling/physiology , Cell Survival/drug effects , Cytoskeleton/metabolism , Cytoskeleton/physiology , Humans , Mice , Mice, Transgenic , Oncogene Protein v-cbl/genetics , Oncogene Protein v-cbl/physiology , Osteoclasts/cytology , Osteoclasts/metabolism , Osteoclasts/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Binding/physiology , Signal Transduction/physiology
16.
Biochem J ; 423(3): 375-80, 2009 Oct 12.
Article in English | MEDLINE | ID: mdl-19689429

ABSTRACT

Tie2 [where 'Tie' is an acronym from tyrosine kinase with Ig and EGF (epidermal growth factor) homology domains] is a receptor tyrosine kinase expressed predominantly on the surface of endothelial cells. Activated by its ligands, the angiopoietins, Tie2 initiates signalling pathways that modulate vascular stability and angiogenesis. Deletion of either the Tie2 or Ang1 (angiopoietin-1) gene in mice results in lethal vascular defects, signifying their importance in vascular development. The mechanism employed by the Tie2 signalling machinery to attenuate or cause receptor trafficking is not well defined. Stimulation of Tie2-expressing cells with Ang1 results in its ubiquitylation, suggesting that this may provide the necessary signal for receptor turnover. Using a candidate molecule approach, we demonstrate that Tie2 co-immunoprecipitates with c-Cbl in an Ang1-dependent manner and its ubiquitylation can be inhibited by the dominant-interfering molecule v-Cbl (a viral form of c-Cbl that contains only the tyrosine kinase-binding domain region). Inhibition of the Tie2-Cbl interaction by overexpression of v-Cbl blocks ligand-induced Tie2 internalization and degradation. In summary, our results illustrate that c-Cbl interacts with the Tie2 signalling complex in a stimulation-dependent manner, and that this interaction is required for Tie2 ubiquitylation, internalization and degradation.


Subject(s)
Angiopoietin-1/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Receptor, TIE-2/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Angiopoietin-1/genetics , Angiopoietin-1/pharmacology , Cell Line , Humans , Oncogene Protein v-cbl/genetics , Oncogene Protein v-cbl/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Proto-Oncogene Proteins c-cbl/genetics , Receptor, TIE-2/genetics , Signal Transduction/drug effects , Ubiquitination/drug effects
17.
Eur J Pharmacol ; 585(1): 76-87, 2008 May 06.
Article in English | MEDLINE | ID: mdl-18402935

ABSTRACT

Protein kinase C (PKC) is involved in synaptic remodeling, induction of protein synthesis, and many other processes important in learning and memory. Activation of neuronal protein kinase C correlates with, and may be essential for, all phases of learning, including acquisition, consolidation, and reconsolidation. Protein kinase C activation is closely tied to hydrolysis of membrane lipids. Phospholipases C and A2 produce 1,2-diacylglycerol and arachidonic acid, which are direct activators of protein kinase C. Phospholipase C also produces inositol triphosphate, which releases calcium from internal stores. Protein kinase C interacts with many of the same pathways as insulin; therefore, it should not be surprising that insulin signaling and protein kinase C activation can both have powerful effects on memory storage and synaptic remodeling. However, investigating the possible roles of insulin in memory storage can be challenging, due to the powerful peripheral effects of insulin on glucose and the low concentration of insulin in the brain. Although peripheral for insulin, synthesized in the beta-cells of the pancreas, is primarily involved in regulating glucose, small amounts of insulin are also present in the brain. The functions of this brain insulin are inadequately understood. Protein kinase C may also contribute to insulin resistance by phosphorylating the insulin receptor substrates required for insulin signaling. Insulin is also responsible insulin-long term depression, a type of synaptic plasticity that is also dependent on protein kinase C. However, insulin can also activate PKC signaling pathways via PLC gamma, Erk 1/2 MAP kinase, and src stimulation. Taken together, the available evidence suggests that the major impact of protein kinase C and its interaction with insulin in the mature, fully differentiated nervous system appears to be to induce synaptogenesis, enhance memory, reduce Alzheimer's pathophysiology, and stimulate neurorepair.


Subject(s)
Insulin/physiology , Memory/physiology , Neurons/physiology , Protein Kinase C/physiology , Synapses/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Humans , Insulin-Like Growth Factor I/physiology , Mitogen-Activated Protein Kinases/physiology , Nerve Regeneration , Oncogene Protein v-cbl/physiology , Receptor, Insulin/physiology , Signal Transduction , Stroke/metabolism , Stroke/physiopathology
18.
Blood ; 111(7): 3607-14, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18239087

ABSTRACT

Inside-out signaling regulation of the beta2-integrin leukocyte function-associated antigen-1 (LFA-1) by different cytoplasmic proteins, including 14-3-3 proteins, is essential for adhesion and migration of immune cells. Here, we identify a new pathway for the regulation of LFA-1 activity by Cbl-b, an adapter molecule and ubiquitin ligase that modulates several signaling pathways. Cbl-b-/- mice displayed increased macrophage recruitment in thioglycollate-induced peritonitis, which was attributed to Cbl-b deficiency in macrophages, as assessed by bone marrow chimera experiments. In vitro, Cbl-b-/- bone marrow-derived mononuclear phagocytes (BMDMs) displayed increased adhesion to endothelial cells. Activation of LFA-1 in Cbl-b-deficient cells was responsible for their increased endothelial adhesion in vitro and peritoneal recruitment in vivo, as the phenotype of Cbl-b deficiency was reversed in Cbl-b-/-LFA-1-/- mice. Consistently, LFA-1-mediated adhesion of BMDM to ICAM-1 but not VLA-4-mediated adhesion to VCAM-1 was enhanced by Cbl-b deficiency. Cbl-b deficiency resulted in increased phosphorylation of T758 in the beta2-chain of LFA-1 and thereby in enhanced association of 14-3-3beta protein with the beta2-chain, leading to activation of LFA-1. Consistently, disruption of the 14-3-3/beta2-integrin interaction abrogated the enhanced ICAM-1 adhesion of Cbl-b-/- BMDMs. In conclusion, Cbl-b deficiency activates LFA-1 and LFA-1-mediated inflammatory cell recruitment by stimulating the interaction between the LFA-1 beta-chain and 14-3-3 proteins.


Subject(s)
14-3-3 Proteins/immunology , Cell Movement/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Macrophages, Peritoneal/immunology , Oncogene Protein v-cbl/immunology , Signal Transduction/immunology , 14-3-3 Proteins/genetics , Animals , CD18 Antigens/genetics , CD18 Antigens/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Line, Tumor , Cell Movement/genetics , Endothelium, Vascular/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Integrin alpha4beta1/genetics , Integrin alpha4beta1/immunology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Mice , Mice, Knockout , Oncogene Protein v-cbl/genetics , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
19.
Proc Natl Acad Sci U S A ; 104(44): 17311-6, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17954916

ABSTRACT

The structure of intrinsic factor (IF) in complex with cobalamin (Cbl) was determined at 2.6-A resolution. The overall fold of the molecule is that of an alpha(6)/alpha(6) barrel. It is a two-domain protein, and the Cbl is bound at the interface of the domains in a base-on conformation. Surprisingly, two full-length molecules, each comprising an alpha- and a beta-domain and one Cbl, and two truncated molecules with only an alpha- domain are present in the same asymmetric unit. The environment around Cbl is dominated by uncharged residues, and the sixth coordinate position of Co(2+) is empty. A detailed comparison between the IF-B12 complex and another Cbl transport protein complex, trans-Cbl-B12, has been made. The pH effect on the binding of Cbl analogues in transport proteins is analyzed. A possible basis for the lack of interchangeability of human and rat IF receptors is presented.


Subject(s)
Intrinsic Factor/chemistry , Intrinsic Factor/metabolism , Vitamin B 12/chemistry , Vitamin B 12/metabolism , Crystallography, X-Ray , Humans , Intrinsic Factor/genetics , Models, Molecular , Oncogene Protein v-cbl/chemistry , Oncogene Protein v-cbl/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Static Electricity , Structural Homology, Protein
20.
FEBS Lett ; 580(20): 4889-94, 2006 Sep 04.
Article in English | MEDLINE | ID: mdl-16919274

ABSTRACT

A high-fat diet (HFD) is associated with reduced glucose uptake in muscle, but not in adipose tissue. In the present study, we investigated whether a HFD can modulate glucose uptake in adipose tissue by increasing signal transduction through the CAP/Cbl pathway, independently of the PI3-K/Akt pathway. Our results suggest that, in HFD, the differential regulation of insulin-induced glucose uptake between skeletal muscle and adipose tissue may, in part, be a consequence of the CAP/Cbl/C3G pathway, since the expression of CAP and Cbl, and also the activation of this pathway were increased in adipose tissue but not in muscle.


Subject(s)
Adipose Tissue/metabolism , Cytoskeletal Proteins/metabolism , Dietary Fats/administration & dosage , Oncogene Protein v-cbl/metabolism , Signal Transduction/physiology , Animals , Cytoskeletal Proteins/genetics , Glucose/metabolism , Insulin/metabolism , Male , Muscle, Skeletal/metabolism , Oncogene Protein v-cbl/genetics , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...