Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 26(1): 265-273, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31573955

ABSTRACT

PURPOSE: The identification of novel targets for developing synergistic drug-radiation combinations would pave the way to overcome tumor radioresistance. We conducted cell-based screening of a human kinome siRNA library to identify a radiation-specific kinase that has a synergistic toxic effect with radiation upon inhibition and is not essential for cell survival in the absence of radiation. EXPERIMENTAL DESIGN: Unbiased RNAi screening was performed by transfecting A549 cells with a human kinome siRNA library followed by irradiation. Radiosensitizing effects of a target gene and involved mechanisms were examined. RESULTS: We identified the nonreceptor protein tyrosine kinase FES (FEline Sarcoma oncogene) as a radiosensitizing target. The expression of FES was increased in response to irradiation. Cell viability and clonogenic survival after irradiation were significantly decreased by FES knockdown in lung and pancreatic cancer cell lines. In contrast, FES depletion alone did not significantly affect cell proliferation without irradiation. An inducible RNAi mouse xenograft model verified in vivo radiosensitizing effects. FES-depleted cells showed increased apoptosis, DNA damage, G2-M phase arrest, and mitotic catastrophe after irradiation. FES depletion promoted radiation-induced reactive oxygen species formation, which resulted in phosphorylation of S6K and MDM2. The radiosensitizing effect of FES knockdown was partially reversed by inhibition of S6K activity. Consistent with the increase in phosphorylated MDM2, an increase in nuclear p53 levels was observed, which appears to contribute increased radiosensitivity of FES-depleted cells. CONCLUSIONS: We uncovered that inhibition of FES could be a potential strategy for inducing radiosensitization in cancer. Our results provide the basis for developing novel radiosensitizers.


Subject(s)
Apoptosis , DNA Damage , Lung Neoplasms/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , RNA Interference , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/radiotherapy , Phosphorylation/radiation effects , Proto-Oncogene Proteins c-fes/genetics , Proto-Oncogene Proteins c-fes/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Radiation Tolerance , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
2.
PLoS One ; 12(7): e0181178, 2017.
Article in English | MEDLINE | ID: mdl-28727840

ABSTRACT

Acute myelogenous leukemia (AML) is often associated with activating mutations in the receptor tyrosine kinase, Flt3, including internal tandem duplications (ITDs) within the regulatory juxtamembrane region. Previous studies have linked Flt3-ITD to the activation of the Fes protein tyrosine kinase in AML, and RNAi-knockdown studies suggest that Fes may be required for Flt3 function. In this study, we tested Fes inhibitors from three different chemical classes for their growth-suppressive activity against Flt3-ITD+ myeloid leukemia cell lines (MV4-11, MOLM-13 and MOLM-14) vs. myeloid cells with wild-type Flt3 (THP-1). All Fes inhibitors selectively inhibited the growth of Flt3-ITD+ AML cells, with IC50 values for diaminopyrimidine and pyrrolopyridine inhibitors ranging from 19 to 166 nM. In contrast, a pyrazolopyrimidine inhibitor was less potent in Flt3-ITD+ AML cells, with IC50 values in the 1.0 µM range. In vitro kinase assays showed that the most potent inhibitors of Flt3-ITD+ AML cell proliferation blocked both Fes and Flt3-ITD kinase activity, while the pyrazolopyrimidine was more selective for Fes vs. Flt3-ITD. All three inhibitors induced significant apoptosis in Flt3-ITD+ AML cells, with potency equivalent to or greater than the established Flt3-ITD inhibitor, tandutinib. Transformation of TF-1 cells with Flt3-ITD resulted in constitutive activation of endogenous Fes, and rendered the cells highly sensitive to all three Fes inhibitors with IC50 values in the 30-500 nM range. The pyrrolopyridine compound also induced apoptotic responses in patient-derived Flt3-ITD+ AML bone marrow cells but not in normal bone marrow mononuclear cells. These results demonstrate that Fes kinase activity contributes to Flt3-ITD signaling in AML, and suggests that dual inhibition of both Flt3 and Fes may provide a therapeutic advantage for the treatment of Flt3-ITD+ AML.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Inhibitory Concentration 50 , Leukemia, Myeloid, Acute/enzymology , Male , Middle Aged , Mutation , Signal Transduction , Tandem Repeat Sequences
3.
Chem Biol ; 19(4): 529-40, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22520759

ABSTRACT

The c-Fes protein-tyrosine kinase modulates cellular signaling pathways governing differentiation, the innate immune response, and vasculogenesis. Here, we report the identification of types I and II kinase inhibitors with potent activity against c-Fes both in vitro and in cell-based assays. One of the most potent inhibitors is the previously described anaplastic lymphoma kinase inhibitor TAE684. The crystal structure of TAE684 in complex with the c-Fes SH2-kinase domain showed excellent shape complementarity with the ATP-binding pocket and a key role for the gatekeeper methionine in the inhibitory mechanism. TAE684 and two pyrazolopyrimidines with nanomolar potency against c-Fes in vitro were used to establish a role for this kinase in osteoclastogenesis, illustrating the value of these inhibitors as tool compounds to probe the diverse biological functions associated with this unique kinase.


Subject(s)
Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Binding Sites , COS Cells , Cell Differentiation , Cell Line , Chlorocebus aethiops , Computer Simulation , Crystallography, X-Ray , Mice , Microtubules/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fes/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , src Homology Domains
4.
Methods Mol Biol ; 800: 133-62, 2012.
Article in English | MEDLINE | ID: mdl-21964787

ABSTRACT

Systematic analysis of protein and enzyme function typically requires scale-up of protein expression and purification prior to assay development; this can often be limiting. Miniaturization of assays provides an alternative approach, but simple, generic methods are in short supply. Here we show how custom microarrays can be adapted to this purpose. We discuss the different routes to array fabrication and describe in detail one facile approach in which the purification and immobilization procedures are combined into a single step, significantly simplifying the array fabrication process. We illustrate this approach by reference to the creation of arrays of human protein kinases and of human cytochrome P450s. We discuss methods for both ligand-binding and turnover-based assays, as well as data analysis on such arrays.


Subject(s)
Microtechnology/methods , Protein Array Analysis/methods , Proteomics/methods , Recombinant Proteins/metabolism , Animals , Baculoviridae/genetics , Cell Line , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/isolation & purification , Cytochrome P-450 Enzyme System/metabolism , Genetic Vectors/genetics , Genome, Viral/genetics , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/genetics , Immobilized Proteins/isolation & purification , Immobilized Proteins/metabolism , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/isolation & purification , Mitogen-Activated Protein Kinase 1/metabolism , Models, Molecular , Phosphorylation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , Proto-Oncogene Proteins c-fes/genetics , Proto-Oncogene Proteins c-fes/isolation & purification , Proto-Oncogene Proteins c-fes/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Streptavidin/metabolism
5.
Curr Med Chem ; 18(19): 2913-20, 2011.
Article in English | MEDLINE | ID: mdl-21651488

ABSTRACT

Non receptor protein tyrosine kinases are targets in the treatment of a number of diseases. This review focuses on the role of Fes tyrosine kinase and on the design of inhibitors of this protein. Fes and its homologously related protein Fer are the only two members of a distinct class of non receptor tyrosine kinases and they seem to play a role in cytoskeletal rearrangements and inside-out signaling associated with receptor-ligand, cell-matrix and cell-cell interactions. The knowledge of the three dimensional structure of this protein, in fact, has informed drug design, while at the same time it has helped to shed some light on the molecular mechanism at the basis of kinase activation and functions.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Neoplasms/enzymology , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , Proto-Oncogene Proteins c-fes/metabolism , Animals , Antineoplastic Agents/pharmacology , Humans , Models, Molecular , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fes/chemistry
6.
Leukemia ; 24(4): 721-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20111072

ABSTRACT

The closely related non-receptor tyrosine kinases FEline Sarcoma (FES) and FEs Related (FER) are activated by cell surface receptors in hematopoietic cells. Despite the early description of oncogenic viral forms of fes, v-fes, and v-fps, the implication of FES and FER in human pathology is not known. We have recently shown that FES but not FER is necessary for oncogenic KIT receptor signaling. Here, we report that both FES and FER kinases are activated in primary acute myeloid leukemia (AML) blasts and in AML cell lines. FES and FER activation is dependent on FLT3 in cell lines harboring constitutively active FLT3 mutants. Moreover, both FES and FER proteins are critical for FLT3-internal tandem duplication (ITD) signaling and for cell proliferation in relevant AML cell lines. FER is required for cell cycle transitions, whereas FES seems necessary for cell survival. We concluded that FES and FER kinases mediate essential non-redundant functions downstream of FLT3-ITD.


Subject(s)
Inverted Repeat Sequences/genetics , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-fes/metabolism , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Blotting, Western , Cell Cycle , Cell Proliferation , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation/genetics , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , Proto-Oncogene Proteins c-fes/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
7.
Int J Oncol ; 34(1): 89-96, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082481

ABSTRACT

The c-Fes protein-tyrosine kinase is associated with growth and differentiation of hematopoietic, neuronal, vascular endothelial and epithelial cell types. In this study, we investigated whether small interfering RNA (siRNA)-mediated knockdown of c-Fes expression affected proliferation of the human renal carcinoma cell lines, ACHN and VMRC-RCW. Immunofluorescence microscopy showed that c-Fes was expressed in both the cytosol and nuclei of these cells, and siRNA treatment preferentially downregulated c-Fes expression in the cytosol. Knock-down of c-Fes inhibited cellular proliferation in a dose-dependent manner with minimal increase in cell death. c-Fes siRNA treatment also downregulated the phosphorylation of Akt1 on S473 and IKKalpha on T23, and cyclin D1 expression, enhanced the expression of IkappaBalpha, and prevented the nuclear localization of NFkappaB. Treatment with an NFkappaB inhibitory peptide (SN50) also blocked the proliferation and nuclear localization of NFkappaB in these cells. The effect of SN50 treatment was not enhanced by c-Fes siRNA, suggesting that downregulation of c-Fes expression inhibited cell cycle progression through the Akt1/NFkappaB pathway. In contrast to siRNA-mediated knockdown, ectopic expression of either wild-type or kinase-inactive c-Fes in renal carcinoma cells failed to alter their proliferation in vitro and in vivo. Thus, suppression of proliferation resulting from siRNA-mediated knockdown may depend upon an expression of c-Fes protein rather than its kinase activity. Taken together, our results indicate that downregulation of c-Fes expression may be a potential therapeutic strategy for advanced human renal cell carcinoma and inhibition of its kinase activity as an antiangiogenic therapy does not seem to induce the growth of human renal carcinoma cells.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Proto-Oncogene Proteins c-fes/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Nucleus , Cell Proliferation , Down-Regulation , Fluorescent Antibody Technique, Indirect , Humans , I-kappa B Proteins/metabolism , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fes/antagonists & inhibitors , Proto-Oncogene Proteins c-fes/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...