Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Proteomics ; 11(9): 651-68, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22617229

ABSTRACT

Targeted drugs are less toxic than traditional chemotherapeutic therapies; however, the proportion of patients that benefit from these drugs is often smaller. A marker that confidently predicts patient response to a specific therapy would allow an individual therapy selection most likely to benefit the patient. Here, we used quantitative mass spectrometry to globally profile the basal phosphoproteome of a panel of non-small cell lung cancer cell lines. The effect of the kinase inhibitor dasatinib on cellular growth was tested against the same panel. From the phosphoproteome profiles, we identified 58 phosphorylation sites, which consistently differ between sensitive and resistant cell lines. Many of the corresponding proteins are involved in cell adhesion and cytoskeleton organization. We showed that a signature of only 12 phosphorylation sites is sufficient to accurately predict dasatinib sensitivity. Four of the phosphorylation sites belong to integrin ß4, a protein that mediates cell-matrix or cell-cell adhesion. The signature was validated in cross-validation and label switch experiments and in six independently profiled breast cancer cell lines. The study supports that the phosphorylation of integrin ß4, as well as eight further proteins comprising the signature, are candidate biomarkers for predicting response to dasatinib in solid tumors. Furthermore, our results show that identifying predictive phosphorylation signatures from global, quantitative phosphoproteomic data is possible and can open a new path to discovering molecular markers for response prediction.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Phosphoproteins/analysis , Pyrimidines/pharmacology , Thiazoles/pharmacology , Biomarkers, Tumor , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Drug Resistance, Neoplasm , Female , Gene Expression Profiling , Humans , Integrin beta4/chemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mass Spectrometry , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proteome/analysis
2.
Cancer Res ; 65(15): 6919-26, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061676

ABSTRACT

Knowledge about molecular drug action is critical for the development of protein kinase inhibitors for cancer therapy. Here, we establish a chemical proteomic approach to profile the anticancer drug SU6668, which was originally designed as a selective inhibitor of receptor tyrosine kinases involved in tumor vascularization. By employing immobilized SU6668 for the affinity capture of cellular drug targets in combination with mass spectrometry, we identified previously unknown targets of SU6668 including Aurora kinases and TANK-binding kinase 1. Importantly, a cell cycle block induced by SU6668 could be attributed to inhibition of Aurora kinase activity. Moreover, SU6668 potently suppressed antiviral and inflammatory responses by interfering with TANK-binding kinase 1-mediated signal transmission. These results show the potential of chemical proteomics to provide rationales for the development of potent kinase inhibitors, which combine rather unexpected biological modes of action by simultaneously targeting defined sets of both serine/threonine and tyrosine kinases involved in cancer progression.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Aurora Kinases , COS Cells , Cell Division/drug effects , Chlorocebus aethiops , HeLa Cells , Humans , Oxindoles , Propionates , Protein Serine-Threonine Kinases/antagonists & inhibitors , Transfection
3.
Cancer Res ; 65(2): 379-82, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15695376

ABSTRACT

Targeted inhibition of protein kinases with small molecule drugs has evolved into a viable approach for anticancer therapy. However, the true selectivity of these therapeutic agents has remained unclear. Here, we used a proteomic method to profile the cellular targets of the clinical epidermal growth factor receptor kinase inhibitor gefitinib. Our data suggest alternative cellular modes of action for gefitinib and provide rationales for the development of related drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Animals , COS Cells , Chlorocebus aethiops , ErbB Receptors/antagonists & inhibitors , Gefitinib , HeLa Cells , Humans , Proteomics/methods , Structure-Activity Relationship
4.
Mol Cell Proteomics ; 3(12): 1181-93, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15475568

ABSTRACT

Small molecule inhibitors belonging to the pyrido[2,3-d]pyrimidine class of compounds were developed as antagonists of protein tyrosine kinases implicated in cancer progression. Derivatives from this compound class are effective against most of the imatinib mesylate-resistant BCR-ABL mutants isolated from advanced chronic myeloid leukemia patients. Here, we established an efficient proteomics method employing an immobilized pyrido[2,3-d]pyrimidine ligand as an affinity probe and identified more than 30 human protein kinases affected by this class of compounds. Remarkably, in vitro kinase assays revealed that the serine/threonine kinases Rip-like interacting caspase-like apoptosis-regulatory protein kinase (RICK) and p38alpha were among the most potently inhibited kinase targets. Thus, pyrido[2,3-d]pyrimidines did not discriminate between tyrosine and serine/threonine kinases. Instead, we found that these inhibitors are quite selective for protein kinases possessing a conserved small amino acid residue such as threonine at a critical site of the ATP binding pocket. We further demonstrated inhibition of both p38 and RICK kinase activities in intact cells upon pyrido[2,3-d]pyrimidine inhibitor treatment. Moreover, the established functions of these two kinases as signal transducers of inflammatory responses could be correlated with a potent in vivo inhibition of cytokine production by a pyrido[2,3-d]pyrimidine compound. Thus, our data demonstrate the utility of proteomic methods employing immobilized kinase inhibitors for identifying new targets linked to previously unrecognized therapeutic applications.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Pyridines/pharmacology , Pyrimidines/pharmacology , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Apoptosis , Binding Sites , COS Cells , Cell Line, Tumor , Cells, Cultured , Chromatography, Liquid , Cytokines/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Inflammation , Inhibitory Concentration 50 , Ligands , Lipopolysaccharides/chemistry , Mass Spectrometry , Models, Chemical , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids/metabolism , Polyethylene Glycols/chemistry , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Sequence Homology, Amino Acid , Signal Transduction , Time Factors
5.
Chem Biol ; 11(5): 691-701, 2004 May.
Article in English | MEDLINE | ID: mdl-15157880

ABSTRACT

Some protein kinases are known to acquire resistance to selective small molecule inhibitors upon mutation of a conserved threonine at the ATP binding site to a larger residue. Here, we performed a comprehensive mutational analysis of this structural element and determined the cellular sensitivities of several disease-relevant tyrosine kinases against various inhibitors. Mutant kinases possessing a larger side chain at the critical site showed resistance to most compounds tested, such as ZD1839, PP1, AG1296, STI571, and a pyrido[2,3-d]pyrimidine inhibitor. In contrast, indolinones affected both wild-type and mutant kinases with similar potencies. Resistant mutants were established for pharmacological analysis of betaPDGF receptor-mediated signaling and allowed the generation of a drug-inducible system of cellular Src kinase activity. Our data establish a conserved structural determinant of protein kinase sensitivity relevant for both signal transduction research and drug development.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Conserved Sequence/genetics , Conserved Sequence/physiology , Epidermal Growth Factor/genetics , Molecular Sequence Data , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1 , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/genetics , Sequence Alignment , Signal Transduction , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry , src-Family Kinases/genetics
6.
Proc Natl Acad Sci U S A ; 100(26): 15434-9, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14668439

ABSTRACT

Small molecule inhibitors of protein kinases are widely used in signal transduction research and are emerging as a major class of drugs. Although interpretation of biological results obtained with these reagents critically depends on their selectivity, efficient methods for proteome-wide assessment of kinase inhibitor selectivity have not yet been reported. Here, we address this important issue and describe a method for identifying targets of the widely used p38 kinase inhibitor SB 203580. Immobilization of a suitable SB 203580 analogue and thoroughly optimized biochemical conditions for affinity chromatography permitted the dramatic enrichment and identification of several previously unknown protein kinase targets of SB 203580. In vitro kinase assays showed that cyclin G-associated kinase (GAK) and CK1 were almost as potently inhibited as p38alpha whereas RICK [Rip-like interacting caspase-like apoptosis-regulatory protein (CLARP) kinase/Rip2/CARDIAK] was even more sensitive to inhibition by SB 203580. The cellular kinase activity of RICK, a known signal transducer of inflammatory responses, was already inhibited by submicromolar concentrations of SB 203580 in intact cells. Therefore, our results warrant a reevaluation of the vast amount of data obtained with SB 203580 and might have significant implications on the development of p38 inhibitors as antiinflammatory drugs. Based on the procedures described here, efficient affinity purification techniques can be developed for other protein kinase inhibitors, providing crucial information about their cellular modes of action.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/chemistry , Protein Kinase Inhibitors , Proteomics , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , HeLa Cells , Humans , Mass Spectrometry , Mitogen-Activated Protein Kinases/isolation & purification , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases
7.
J Gen Virol ; 84(Pt 3): 555-560, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12604805

ABSTRACT

The hepatitis C virus (HCV) NS5A protein is highly phosphorylated by cellular protein kinases. To study how NS5A might be integrated in cellular kinase signalling, we isolated phosphoproteins from HuH-7 hepatoma cells that specifically interacted with recombinant NS5A protein. Subsequent mass spectrometry identified the adaptor protein amphiphysin II as a novel interaction partner of NS5A. Mutational analysis revealed that complex formation is primarily mediated by a proline-rich region in the C-terminal part of NS5A, which interacts with the amphiphysin II Src homology 3 domain. Importantly, we could further demonstrate specific co-precipitation and cellular co-localization of endogenous amphiphysin II with NS5A in HuH-7 cells carrying a persistently replicating subgenomic HCV replicon. Although the NS5A-amphiphysin II interaction appeared to be dispensable for replication of these HCV RNAs in cell culture, our results indicate that NS5A-amphiphysin II complex formation might be of physiological relevance for the HCV life cycle.


Subject(s)
Hepacivirus/physiology , Nerve Tissue Proteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Cytoplasm/chemistry , Fluorescent Antibody Technique, Indirect , Hepacivirus/chemistry , Hepacivirus/genetics , Humans , Immunoblotting , Leucine Zippers , MAP Kinase Kinase Kinases , Mutation , Nerve Tissue Proteins/chemistry , Proline , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Recombinant Proteins/metabolism , Replicon , Tumor Cells, Cultured , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication , Mitogen-Activated Protein Kinase Kinase Kinase 11
8.
J Biol Chem ; 278(17): 15435-40, 2003 Apr 25.
Article in English | MEDLINE | ID: mdl-12594213

ABSTRACT

Small molecule inhibitors of protein tyrosine kinases such as STI571 represent a major new class of therapeutics for target-selective treatment of human cancer. Clinical resistance formation to the BCR-ABL inhibitor STI571 has been observed in patients with advanced chronic myeloid leukemia and was frequently caused by a C to T single nucleotide change in the Abl kinase domain, which substituted Thr-315 with isoleucine and rendered BCR-ABL resistant to STI571 inhibition. The corresponding mutation in the epidermal growth factor receptor (EGFR) tyrosine kinase replaced Thr-766 of the EGFR by methionine and dramatically reduced the sensitivity of EGFR to inhibition by selective 4-anilinoquinazoline inhibitors such as PD153035. Inhibitor-resistant EGFR exhibited the same signaling capacity as wild-type receptor in vivo and provides a useful tool for analyzing EGFR-mediated signal transduction. Our data identify Thr-766 of the EGFR as a structural determinant that bears the potential to become a relevant feature in resistance formation during cancer therapy with EGFR-specific 4-anilinoquinazoline inhibitors.


Subject(s)
Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Point Mutation , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Imatinib Mesylate , MAP Kinase Signaling System , Piperazines/pharmacology , Pyrimidines/pharmacology , Quinazolines/pharmacology , Threonine/genetics , Transfection
9.
J Virol ; 76(16): 8124-37, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12134018

ABSTRACT

Phosphorylation of hepatitis B virus (HBV) core protein has recently been shown to be a prerequisite for pregenomic RNA encapsidation into viral capsids, but the host cell kinases mediating this essential step of the HBV replication cycle have not been identified. We detected two kinases of 95 and 115 kDa in HuH-7 total cell lysates which interacted specifically with the HBV core protein and phosphorylated its arginine-rich C-terminal domain. The 95-kDa kinase was purified and characterized as SR protein-specific kinase 1 (SRPK1) by mass spectrometry. Based on this finding, the 115-kDa kinase could be identified as the related kinase SRPK2 by immunoblot analysis. In vitro, both SRPKs phosphorylated HBV core protein on the same serine residues which are found to be phosphorylated in vivo. Moreover, the major cellular HBV core kinase activity detected in the total cell lysate showed biochemical properties identical to those of SRPK1 and SRPK2, as examined by measuring binding to a panel of chromatography media. We also clearly demonstrate that neither the cyclin-dependent kinases Cdc2 and Cdk2 nor protein kinase C, previously implicated in HBV core protein phosphorylation, can account for the HBV core protein kinase activity. We conclude that both SRPK1 and SRPK2 are most likely the cellular protein kinases mediating HBV core protein phosphorylation during viral infection and therefore represent important host cell targets for therapeutic intervention in HBV infection.


Subject(s)
CDC2-CDC28 Kinases , Hepatitis B Core Antigens/metabolism , Hepatitis B virus/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Core Proteins/metabolism , Animals , CDC2 Protein Kinase/metabolism , COS Cells , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Hepatitis B/drug therapy , Hepatitis B/enzymology , Hepatitis B/virology , Hepatitis B Core Antigens/genetics , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Humans , In Vitro Techniques , Phosphorylation , Protein Kinase C/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Core Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...