Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Front Oncol ; 13: 1096499, 2023.
Article in English | MEDLINE | ID: mdl-36969004

ABSTRACT

Patients with pancreatic ductal adenocarcinoma (PDAC) have a dismal 5-year survival rate of less than 10%, predominantly due to delayed diagnosis and a lack of effective treatment options. In the PDAC tumor microenvironment (TME), neutrophils are among the immune cell types that are most prevalent and are linked to a poor clinical prognosis. However, treatments that target tumor-associated neutrophils are limited despite recent developments in our understanding of neutrophil function in cancer. The feline sarcoma oncogene (FES) is a nonreceptor tyrosine kinase previously associated with leukemia and hematopoietic homeostasis. Here we describe a newly derived FES null mouse with no distinct phenotype and no defects in hematopoietic homeostasis including neutrophil viability. The immune cell composition and neutrophil population were analyzed with flow cytometry, colony-forming unit (CFU) assay, and a neutrophil viability assay, while the response to PDAC was examined with an in vivo cancer model. In an experimental metastasis model, the FES null model displayed a reduced PDAC hepatic metastatic burden and a reduction in neutrophils granulocytes. Accordingly, our results indicate FES as a potential target for PDAC TME modulation.

3.
Bull Cancer ; 110(3): 331-335, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36775700

ABSTRACT

This article highlights the presentations from the 2021 scientific meeting of the Club Hematopoiesis and Oncogenesis. This annual meeting focuses on hematopoiesis and oncogenic mechanisms. Various topics were presented: expansion of hematopoietic stem cells with in vivo and ex vivo strategies, the role of the hematopoietic stem cell niches in aging and leukemic resistance, the crossroad between hematology and immunology, the importance of the metabolism in normal hematopoiesis and hematopoietic defects, solid tumors and oncogenesis, the noncoding genome, inflammation in monocyte differentiation and leukemia, and importantly, the recent advances in myeloid malignancies, lymphoid leukemia and lymphoma.


Subject(s)
Leukemia , Lymphoma , Humans , Hematopoiesis/genetics , Hematopoietic Stem Cells , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology
4.
JCI Insight ; 7(7)2022 04 08.
Article in English | MEDLINE | ID: mdl-35393954

ABSTRACT

Mutation of the TET2 DNA-hydroxymethylase has been associated with a number of immune pathologies. The disparity in phenotype and clinical presentation among these pathologies leads to questions regarding the role of TET2 mutation in promoting disease evolution in different immune cell types. Here we show that, in primary mast cells, Tet2 expression is induced in response to chronic and acute activation signals. In TET2-deficient mast cells, chronic activation via the oncogenic KITD816V allele associated with mastocytosis, selects for a specific epigenetic signature characterized by hypermethylated DNA regions (HMR) at immune response genes. H3K27ac and transcription factor binding is consistent with priming or more open chromatin at both HMR and non-HMR in proximity to immune genes in these cells, and this signature coincides with increased pathological inflammation signals. HMR are also associated with a subset of immune genes that are direct targets of TET2 and repressed in TET2-deficient cells. Repression of these genes results in immune tolerance to acute stimulation that can be rescued with vitamin C treatment or reiterated with a Tet inhibitor. Overall, our data support a model where TET2 plays a direct role in preventing immune tolerance in chronically activated mast cells, supporting TET2 as a viable target to reprogram the innate immune response for innovative therapies.


Subject(s)
DNA-Binding Proteins , Dioxygenases , Immune Tolerance , Mast Cells , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Mast Cells/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
5.
PLoS One ; 16(12): e0260852, 2021.
Article in English | MEDLINE | ID: mdl-34855882

ABSTRACT

Establishing a universally applicable protocol to assess the impact of BRCA1 variants of uncertain significance (VUS) expression is a problem which has yet to be resolved despite major progresses have been made. The numerous difficulties which must be overcome include the choices of cellular models and functional assays. We hypothesised that the use of induced pluripotent stem (iPS) cells might facilitate the standardisation of protocols for classification, and could better model the disease process. We generated eight iPS cell lines from patient samples expressing either BRCA1 pathogenic variants, non-pathogenic variants, or BRCA1 VUSs. The impact of these variants on DNA damage repair was examined using a ɣH2AX foci formation assay, a Homologous Repair (HR) reporter assay, and a chromosome abnormality assay. Finally, all lines were tested for their ability to differentiate into mammary lineages in vitro. While the results obtained from the two BRCA1 pathogenic variants were consistent with published data, some other variants exhibited differences. The most striking of these was the BRCA1 variant Y856H (classified as benign), which was unexpectedly found to present a faulty HR repair pathway, a finding linked to the presence of an additional variant in the ATM gene. Finally, all lines were able to differentiate first into mammospheres, and then into more advanced mammary lineages expressing luminal- or basal-specific markers. This study stresses that BRCA1 genetic analysis alone is insufficient to establish a reliable and functional classification for assessment of clinical risk, and that it cannot be performed without considering the other genetic aberrations which may be present in patients. The study also provides promising opportunities for elucidating the physiopathology and clinical evolution of breast cancer, by using iPS cells.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/pathology , DNA Damage , DNA Repair , Genetic Predisposition to Disease , Induced Pluripotent Stem Cells/pathology , Mutation , Breast Neoplasms/classification , Breast Neoplasms/genetics , Female , Genetic Testing , Humans , Induced Pluripotent Stem Cells/metabolism
6.
Cancers (Basel) ; 13(11)2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34206000

ABSTRACT

Protein kinases (PK) make up around 2% of the human genome and their expression profile varies depending on the organ and tissue [...].

7.
Blood ; 138(17): 1590-1602, 2021 10 28.
Article in English | MEDLINE | ID: mdl-33974006

ABSTRACT

Systemic mastocytosis (SM) is a KIT-driven hematopoietic neoplasm characterized by the excessive accumulation of neoplastic mast cells (MCs) in various organs and, mainly, the bone marrow (BM). Multiple genetic and epigenetic mechanisms contribute to the onset and severity of SM. However, little is known to date about the metabolic underpinnings underlying SM aggressiveness, which has thus far impeded the development of strategies to leverage metabolic dependencies when existing KIT-targeted treatments fail. Here, we show that plasma metabolomic profiles were able to discriminate indolent from advanced forms of the disease. We identified N-acetyl-d-glucosamine (GlcNAc) as the most predictive metabolite of SM severity. High plasma levels of GlcNAc in patients with advanced SM correlated with the activation of the GlcNAc-fed hexosamine biosynthesis pathway in patients BM aspirates and purified BM MCs. At the functional level, GlcNAc enhanced human neoplastic MCs proliferation and promoted rapid health deterioration in a humanized mouse model of SM. In addition, in the presence of GlcNAc, immunoglobulin E-stimulated MCs triggered enhanced release of proinflammatory cytokines and a stronger acute response in a mouse model of passive cutaneous anaphylaxis. Mechanistically, elevated GlcNAc levels promoted the transcriptional accessibility of chromatin regions that contain genes encoding mediators of receptor tyrosine kinases cascades and inflammatory responses, thus leading to a more aggressive phenotype. Therefore, GlcNAc is an oncometabolite driver of SM aggressiveness. This study suggests the therapeutic potential for targeting metabolic pathways in MC-related diseases to manipulate MCs effector functions.


Subject(s)
Acetylglucosamine/analysis , Chromatin Assembly and Disassembly , Mast Cells/pathology , Mastocytosis, Systemic/pathology , Acetylglucosamine/metabolism , Adult , Animals , Disease Progression , Humans , Mast Cells/metabolism , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/metabolism , Metabolome , Mice, SCID , Prospective Studies
8.
Cancers (Basel) ; 12(7)2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32708273

ABSTRACT

Protein tyrosine kinases have been recognized as important actors of cell transformation and cancer progression, since their discovery as products of viral oncogenes. SRC-family kinases (SFKs) play crucial roles in normal hematopoiesis. Not surprisingly, they are hyperactivated and are essential for membrane receptor downstream signaling in hematological malignancies such as acute myeloid leukemia (AML) and mastocytosis. The precise roles of SFKs are difficult to delineate due to the number of substrates, the functional redundancy among members, and the use of tools that are not selective. Yet, a large num ber of studies have accumulated evidence to support that SFKs are rational therapeutic targets in AML and mastocytosis. These two pathologies are regulated by two related receptor tyrosine kinases, which are well known in the field of hematology: FLT3 and KIT. FLT3 is one of the most frequently mutated genes in AML, while KIT oncogenic mutations occur in 80-90% of mastocytosis. Studies on oncogenic FLT3 and KIT signaling have shed light on specific roles for members of the SFK family. This review highlights the central roles of SFKs in AML and mastocytosis, and their interconnection with FLT3 and KIT oncoproteins.

9.
Nat Commun ; 8(1): 1420, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127277

ABSTRACT

Masitinib, a highly selective protein kinase inhibitor, can sensitise gemcitabine-refractory cancer cell lines when used in combination with gemcitabine. Here we report a reverse proteomic approach that identifies the target responsible for this sensitisation: the deoxycytidine kinase (dCK). Masitinib, as well as other protein kinase inhibitors, such as imatinib, interact with dCK and provoke an unforeseen conformational-dependent activation of this nucleoside kinase, modulating phosphorylation of nucleoside analogue drugs. This phenomenon leads to an increase of prodrug phosphorylation of most of the chemotherapeutic drugs activated by this nucleoside kinase. The unforeseen dual activity of protein kinase inhibition/nucleoside kinase activation could be of great therapeutic benefit, through either reducing toxicity of therapeutic agents by maintaining effectiveness at lower doses or by counteracting drug resistance initiated via down modulation of dCK target.


Subject(s)
Deoxycytidine Kinase/metabolism , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , A549 Cells , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Crystallography, X-Ray , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine Kinase/chemistry , Drug Design , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Humans , Imatinib Mesylate/chemistry , Imatinib Mesylate/pharmacology , Models, Biological , Models, Molecular , Phosphorylation , Piperidines , Polypharmacology , Protein Kinase Inhibitors/chemistry , Proteomics , Pyridines , Thiazoles/chemistry , Gemcitabine
10.
J Clin Invest ; 127(6): 2310-2325, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28463229

ABSTRACT

Identification and functional validation of oncogenic drivers are essential steps toward advancing cancer precision medicine. Here, we have presented a comprehensive analysis of the somatic genomic landscape of the widely used BRAFV600E- and NRASQ61K-driven mouse models of melanoma. By integrating the data with publically available genomic, epigenomic, and transcriptomic information from human clinical samples, we confirmed the importance of several genes and pathways previously implicated in human melanoma, including the tumor-suppressor genes phosphatase and tensin homolog (PTEN), cyclin dependent kinase inhibitor 2A (CDKN2A), LKB1, and others. Importantly, this approach also identified additional putative melanoma drivers with prognostic and therapeutic relevance. Surprisingly, one of these genes encodes the tyrosine kinase FES. Whereas FES is highly expressed in normal human melanocytes, FES expression is strongly decreased in over 30% of human melanomas. This downregulation correlates with poor overall survival. Correspondingly, engineered deletion of Fes accelerated tumor progression in a BRAFV600E-driven mouse model of melanoma. Together, these data implicate FES as a driver of melanoma progression and demonstrate the potential of cross-species oncogenomic approaches combined with mouse modeling to uncover impactful mutations and oncogenic driver alleles with clinical importance in the treatment of human cancer.


Subject(s)
Melanoma/genetics , Proto-Oncogene Proteins c-fes/genetics , Skin Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Proliferation , DNA Copy Number Variations , Genes, Tumor Suppressor , Genomics , Humans , Melanoma/metabolism , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Oncogenes , Proto-Oncogene Proteins c-fes/metabolism , Skin Neoplasms/metabolism , Wnt Signaling Pathway
11.
PLoS One ; 11(7): e0160165, 2016.
Article in English | MEDLINE | ID: mdl-27467080

ABSTRACT

The receptors tyrosine kinases (RTKs) for the colony stimulating factor-1, CSF-1R, and for the stem cell factor, SCFR or KIT, are important mediators of signal transduction. The abnormal function of these receptors, promoted by gain-of-function mutations, leads to their constitutive activation, associated with cancer or other proliferative diseases. A secondary effect of the mutations is the alteration of receptors' sensitivity to tyrosine kinase inhibitors, compromising effectiveness of these molecules in clinical treatment. In particular, the mutation V560G in KIT increases its sensitivity to Imatinib, while the D816V in KIT, and D802V in CSF-1R, triggers resistance to the drug. We analyzed the Imatinib binding affinity to the native and mutated KIT (mutations V560G, S628N and D816V) and CSF-1R (mutation D802V) by using molecular dynamics simulations and energy calculations of Imatinib•target complexes. Further, we evaluated the sensitivity of the studied KIT receptors to Imatinib by measuring the inhibition of KIT phosphorylation. Our study showed that (i) the binding free energy of Imatinib to the targets is highly correlated with their experimentally measured sensitivity; (ii) the electrostatic interactions are a decisive factor affecting the binding energy; (iii) the most deleterious impact to the Imatinib sensitivity is promoted by D802V (CSF-1R) and D816V (KIT) mutations; (iv) the role of the juxtamembrane region, JMR, in the imatinib binding is accessory. These findings contribute to a better description of the mutation-induced effects alternating the targets sensitivity to Imatinib.


Subject(s)
Mutation , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Stem Cell Factor/metabolism , Animals , COS Cells , Chlorocebus aethiops , Hydrogen Bonding , Imatinib Mesylate/chemistry , Imatinib Mesylate/metabolism , Molecular Dynamics Simulation , Protein Binding , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Stem Cell Factor/chemistry , Stem Cell Factor/genetics
12.
Oncotarget ; 7(32): 51163-51173, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27323399

ABSTRACT

CDK4/CDK6 and RB proteins drive the progression through the G1 phase of the cell cycle. In acute myeloid leukemia (AML), the activity of the CDK/Cyclin D complex is increased. The mechanism involved is unknown, as are the respective roles played by CDK4 or CDK6 in this process. Here, we report that AML cells carrying FLT3-ITD mutations are dependent on CDK6 for cell proliferation while CDK4 is not essential. We showed that FLT3-ITD signaling is responsible for CDK6 overexpression, through a pathway involving the SRC-family kinase HCK. Accordingly, FLT3-ITD failed to transform primary hematopoietic progenitor cells from Cdk6-/- mice. Our results demonstrate that CDK6 is the primary target of CDK4/CDK6 inhibitors in FLT3-ITD positive AML. Furthermore, we delineate an essential protein kinase pathway -FLT3/HCK/CDK6- in the context of AML with FLT3-ITD mutations.


Subject(s)
Cyclin-Dependent Kinase 6/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-hck/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Proto-Oncogene Proteins c-hck/metabolism , Signal Transduction/genetics , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3/metabolism
13.
JAMA Dermatol ; 150(12): 1345-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25317746

ABSTRACT

IMPORTANCE The KIT receptor is mutated in approximately 15%of acral, mucosal, and chronic, sun-damaged melanomas. The status of KIT mutations is of interest because they usually are mutually exclusive with N-RAS and B-RAF mutations and because of the availability of KIT kinase inhibitors in the clinic. Some recurrent KIT mutations are well characterized; others are poorly described.OBSERVATIONS We describe a novel KIT mutation in a patient with metastatic melanoma. The mutation, located in exon 13, resulted in S628N substitution in the KIT receptor. Using all-atom molecular dynamics simulations, biochemical assays, and cell-based assays, we showed that the mutation is a bona fide gain-of-function oncogenic mutation. Furthermore,we evaluated the sensitivity of the mutant to imatinib and dasatinib.CONCLUSIONS AND RELEVANCE We report a novel KIT gain-of-function mutation with S628N substitution (exon 13) and show that it is sensitive to imatinib in vitro. Therefore, patients with this mutation may be eligible for KIT kinase inhibitor­based therapy. Further studies are needed to evaluate the clinical benefit of such therapy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Lung Neoplasms/genetics , Melanoma/genetics , Proto-Oncogene Proteins c-kit/genetics , Skin Neoplasms/genetics , Spinal Neoplasms/genetics , Aged, 80 and over , Animals , Benzamides/pharmacology , Cells, Cultured , Colony-Forming Units Assay , Dasatinib , Fatal Outcome , Female , Fibroblasts/cytology , Humans , Imatinib Mesylate , Lung Neoplasms/secondary , Melanoma/secondary , Mutation, Missense , Phosphorylation/drug effects , Piperazines/pharmacology , Point Mutation , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Rats , Skin Neoplasms/pathology , Spinal Neoplasms/secondary , Thiazoles/pharmacology
14.
Haematologica ; 99(5): 830-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24389310

ABSTRACT

Mastocytosis is a rare and chronic disease with phenotypes ranging from indolent to severe. Prognosis for this disease is variable and very few biomarkers to predict disease evolution or outcome are currently known. We have performed comprehensive screening in our large cohort of mastocytosis patients for mutations previously found in other myeloid diseases and that could serve as prognostic indicators. KIT, SRSF2-P95 and TET2 mutations were by far the most frequent, detected in 81%, 24% and 21% of patients, respectively. Where TET2 and SRSF2-P95 mutation both correlated with advanced disease phenotypes, SRSF2-P95 hotspot mutation was found almost exclusively in patients diagnosed with associated clonal hematologic non-mast cell disease. Statistically, TET2 and SRSF2-P95 mutations were highly associated, suggesting a mechanistic link between these two factors. Finally, analysis of both clonal and sorted cell populations from patients confirms the presence of these mutations in the mast cell component of the disease, suggests an ontological mutation hierarchy and provides evidence for the expansion of multiple clones. This highlights the prognostic potential of such approaches, if applied systematically, for delineating the roles of specific mutations in predisposing and/or driving distinct disease phenotypes.


Subject(s)
Epigenesis, Genetic , Mastocytosis/genetics , Mastocytosis/pathology , Mutation , Nuclear Proteins/genetics , Ribonucleoproteins/genetics , Clonal Evolution/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Female , Humans , Male , Mast Cells/metabolism , Mast Cells/pathology , Mastocytosis/mortality , Neoplasm Staging , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit/genetics , Serine-Arginine Splicing Factors
15.
Blood ; 120(24): 4846-9, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23074272

ABSTRACT

Although a role for oncogenic KIT in driving mast cell disease is clear, the mechanisms driving the multiple phenotypic and clinical manifestations of this disorder are not well elucidated. We now show, using a large cohort of mastocytosis patients, including an almost equal number of aggressive and nonaggressive cases of systemic mastocytosis, that in contrast to the oncogenic KITD816V, TET2 mutation statistically associates with aggressive forms of the disease. By infecting primary murine bone marrow-derived mast cells with KITD816V, we also observe a significant and competitive growth advantage for KITD816V in Tet2-nullizygous compared with wild-type cells. TET2-deficient cells display increased proliferation and can survive in the absence of cytokines. Taken together, these data demonstrate a oncogenic cooperation in mast cells and reveal TET2 mutation as a potential marker to diagnose and predict severe forms of mastocytosis.


Subject(s)
DNA-Binding Proteins/genetics , Mast Cells/metabolism , Mastocytosis/genetics , Mutation , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Substitution , Animals , Bone Marrow Cells/metabolism , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cohort Studies , Dioxygenases , Female , Humans , Male , Mast Cells/pathology , Mastocytosis/pathology , Mice , Mice, Knockout , Middle Aged , Time Factors , Transfection
16.
J Biol Chem ; 286(8): 5956-66, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21135090

ABSTRACT

Mutations in the c-kit gene occur in the vast majority of mastocytosis. In adult patients as well as in the cell line derived from mast cell neoplasms, the mutations occur almost exclusively at amino acid 816 within the kinase domain of KIT. Among the downstream effectors of KIT signaling, STAT3 and STAT5 have been shown to be critical for cell proliferation elicited by the KIT-Asp(816) mutant protein. However, little is known about the mechanisms of activation of STAT proteins. In this study, we identify and clarify the contribution of various STAT kinases in two widely used neoplastic mast cell lines, P815 and HMC-1. We show that STAT1, -3, and -5 proteins are activated downstream of the KIT-Asp(816) mutant. All three STAT proteins are located in the nucleus and are phosphorylated on serine residues. KIT-Asp(816) mutant can directly phosphorylate STATs on the activation-specific tyrosine residues in vitro. However, within cells, SRC family kinases and JAKs diversely contribute to tyrosine phosphorylation of STAT proteins downstream of the KIT mutant. Using a panel of inhibitors, we provide evidence for the implication or exclusion of serine/threonine kinases as responsible for serine phosphorylation of STAT1, -3, and -5 in the two cell lines. Finally, we show that only STAT5 is transcriptionally active in these cells. This suggests that the contribution of STAT1 and STAT3 downstream of KIT mutant is independent of their transcription factor function.


Subject(s)
Cell Proliferation , Mast Cells/metabolism , Mastocytosis/metabolism , Proto-Oncogene Proteins c-kit/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Adult , Animals , COS Cells , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Chlorocebus aethiops , Humans , Mast Cells/pathology , Mastocytosis/genetics , Mastocytosis/pathology , Mice , Mutation , Phosphorylation/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/genetics , STAT Transcription Factors/genetics , Transcription, Genetic/genetics
17.
BMC Biochem ; 11: 48, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21122136

ABSTRACT

BACKGROUND: RhoGDI proteins are important regulators of the small GTPase Rac, because they shuttle Rac from the cytoplasm to membranes and also protect Rac from activation, deactivation and degradation. How the binding and release of Rac from RhoGDI is regulated is not precisely understood. RESULTS: We report that the non-receptor tyrosine kinase Fer is able to phosphorylate RhoGDIα and form a direct protein complex with it. This interaction is mediated by the C-terminal end of RhoGDIα. Activation of Fer by reactive oxygen species caused increased phosphorylation of RhoGDIα and pervanadate treatment further augmented this. Tyrosine phosphorylation of RhoGDIα by Fer prevented subsequent binding of Rac to RhoGDIα, but once a RhoGDIα-Rac complex was formed, the Fer kinase was not able to cause Rac release through tyrosine phosphorylation of preformed RhoGDIα-Rac complexes. CONCLUSIONS: These results identify tyrosine phosphorylation of RhoGDIα by Fer as a mechanism to regulate binding of RhoGDIα to Rac.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Protein-Tyrosine Kinases/metabolism , rac GTP-Binding Proteins/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Guanine Nucleotide Dissociation Inhibitors/chemistry , Humans , Phosphorylation , Protein Binding , Rats , Substrate Specificity , Tyrosine/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho-Specific Guanine Nucleotide Dissociation Inhibitors
18.
Blood ; 116(7): 1114-23, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20484085

ABSTRACT

Compared with adults, pediatric mastocytosis has a relatively favorable prognosis. Interestingly, a difference was also observed in the status of c-kit mutations according to the age of onset. Although most adult patients have a D(816)V mutation in phosphotransferase domain (PTD), we have described that half of the children carry mutations in extracellular domain (ECD). KIT-ECD versus KIT-PTD mutants were introduced into rodent Ba/F3, EML, Rat2, and human TF1 cells to investigate their biologic effect. Both ECD and PTD mutations induced constitutive receptor autophosphorylation and ligand-independent proliferation of the 3 hematopoietic cells. Unlike ECD mutants, PTD mutants enhanced cluster formation and up-regulated several mast cell-related antigens in Ba/F3 cells. PTD mutants failed to support colony formation and erythropoietin-mediated erythroid differentiation. ECD and PTD mutants also displayed distinct whole-genome transcriptional profiles in EML cells. We observed differences in their signaling properties: they both activated STAT, whereas AKT was only activated by ECD mutants. Consistently, AKT inhibitor suppressed ECD mutant-dependent proliferation, clonogenicity, and erythroid differentiation. Expression of myristoylated AKT restored erythroid differentiation in EML-PTD cells, suggesting the differential role of AKT in those mutants. Overall, our study implied different pathogenesis of pediatric versus adult mastocytosis, which might explain their diverse phenotypes.


Subject(s)
Mastocytosis/genetics , Mutation/genetics , Phosphotransferases/genetics , Proto-Oncogene Proteins c-kit/genetics , Adult , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Child , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/pathology , Lymphocytes/metabolism , Mast Cells/metabolism , Mastocytosis/metabolism , Mastocytosis/pathology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction
19.
Biochem Biophys Res Commun ; 393(1): 174-8, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20117079

ABSTRACT

FES is a cytoplasmic tyrosine kinase activated by several membrane receptors, originally identified as a viral oncogene product. We have recently identified FES as a crucial effector of oncogenic KIT mutant receptor. However, FES implication in wild-type KIT receptor function was not addressed. We report here that FES interacts with KIT and is phosphorylated following activation by its ligand SCF. Unlike in the context of oncogenic KIT mutant, FES is not involved in wild-type KIT proliferation signal, or in cell adhesion. Instead, FES is required for SCF-induced chemotaxis. In conclusion, FES kinase is a mediator of wild-type KIT signalling implicated in cell migration.


Subject(s)
Chemotaxis , Proto-Oncogene Proteins c-fes/metabolism , Stem Cell Factor/metabolism , src Homology Domains , Cell Adhesion , Cell Line, Tumor , Humans , Phosphorylation , Proto-Oncogene Proteins c-fes/genetics , Two-Hybrid System Techniques , Tyrosine
20.
Blood ; 112(10): 4039-47, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18753636

ABSTRACT

Stem cell factor (SCF) plays critical roles in proliferation, survival, migration, and function of hematopoietic progenitor and mast cells through binding to Kit receptor. Previous studies have implicated the adaptor protein Lnk as an important negative regulator of SCF signaling. However, the molecular mechanism underlying this regulation is unclear. Here, we showed that the Src homology 2 domain (SH2) of Lnk binds directly and preferentially to phosphorylated tyrosine 567 in Kit juxtamembrane domain. Using Lnk(-/-) bone marrow mast cells (BMMCs) transduced with different Lnk proteins, we demonstrated that Lnk down-regulates SCF-induced proliferation with attenuation of mitogen-activated protein kinase (MAPK) and c-jun N-terminal kinase signaling. Furthermore, we showed that Lnk(-/-) BMMCs displayed increased SCF-dependent migration compared with wild-type cells, revealing a novel Lnk-mediated inhibitory function. This correlated with enhanced Rac and p38 MAPK activation. Finally, we found that Lnk domains and carboxy-terminal tyrosine contribute differently to inhibition of in vitro expansion of hematopoietic progenitors. Altogether, our results demonstrate that Lnk, through its binding to Kit tyrosine 567, negatively modulates specific SCF-dependent signaling pathways involved in the proliferation and migration of primary hematopoietic cells.


Subject(s)
Bone Marrow Cells/metabolism , MAP Kinase Signaling System/physiology , Mast Cells/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/cytology , Cell Movement/physiology , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Down-Regulation/physiology , Enzyme Activation/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins , Mast Cells/cytology , Membrane Proteins , Mice , Mice, Knockout , Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/genetics , Stem Cell Factor/genetics , Transduction, Genetic , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...