Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(20): 2634-44, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26364612

ABSTRACT

The tumor microenvironment (TME) exerts critical pro-tumorigenic effects through cytokines and growth factors that support cancer cell proliferation, survival, motility and invasion. Insulin-like growth factor-1 (IGF-1) and signal transducer and activator of transcription 3 (STAT3) stimulate colorectal cancer development and progression via cell autonomous and microenvironmental effects. Using a unique inhibitor, NT157, which targets both IGF-1 receptor (IGF-1R) and STAT3, we show that these pathways regulate many TME functions associated with sporadic colonic tumorigenesis in CPC-APC mice, in which cancer development is driven by loss of the Apc tumor suppressor gene. NT157 causes a substantial reduction in tumor burden by affecting cancer cells, cancer-associated fibroblasts (CAF) and myeloid cells. Decreased cancer cell proliferation and increased apoptosis were accompanied by inhibition of CAF activation and decreased inflammation. Furthermore, NT157 inhibited expression of pro-tumorigenic cytokines, chemokines and growth factors, including IL-6, IL-11 and IL-23 as well as CCL2, CCL5, CXCL7, CXCL5, ICAM1 and TGFß; decreased cancer cell migratory activity and reduced their proliferation in the liver. NT157 represents a new class of anti-cancer drugs that affect both the malignant cell and its supportive microenvironment.


Subject(s)
Colorectal Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Adenomatous Polyposis Coli Protein/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Tumor Burden/drug effects
2.
Oncogene ; 35(20): 2675-80, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26119932

ABSTRACT

It is well known that specific signal transduction inhibitors rarely suffice as anti-cancer agents. In most cases, tumors possess primary drug resistance due to their inherent heterogeneity, or acquire drug resistance due to genomic instability and acquisition of mutations. Here we expand our previous study of the novel compound, NT157, and show that it acts as a dual-targeting agent that invokes the blockage of two signal transduction pathways that are central to the development and maintenance of multiple human cancers. We show that NT157 targets not only IGF1R-IRS1/2, as previously reported, but also the Stat3 signaling pathway and demonstrates remarkable anti-cancer characteristics in A375 human melanoma cells and in a metastatic melanoma model in mice.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Molecular Targeted Therapy/methods , Pyrogallol/analogs & derivatives , Receptors, Somatomedin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Discovery , Humans , Melanoma/pathology , Neoplasm Invasiveness , Pyrogallol/pharmacology , Pyrogallol/therapeutic use , Receptor, IGF Type 1 , Sulfonamides/therapeutic use
3.
Oncogene ; 29(46): 6149-59, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-20729914

ABSTRACT

DNA damage checkpoints cause cell cycle arrest, allowing DNA repair before resumption of the cell cycle. These checkpoints can be activated through several signaling pathways. Checkpoint activators include p53, checkpoint kinase 1 (CHK1), checkpoint kinase 2 and/or MAPKAP kinase 2 (MK2). Many cancer cells lack p53 activity and, therefore, depend on alternative checkpoint activators to arrest the cell cycle following DNA damage. Inhibition of these pathways is expected to specifically sensitize these p53-deficient cells to DNA damage caused by chemotherapy. Using isogenic p53-proficient and p53-deficient cancer cell lines, we show that inactivation of CHK1, but not MK2, abrogates cell cycle arrest following chemotherapy, specifically in p53-deficient cells. However, we show that CHK1 is required to maintain genome integrity and cell viability, and that p53-proficient cells are no less sensitive than p53-deficient cells to CHK1 inhibition in the presence of DNA damage. Thus, combining CHK1 inhibition with DNA damage does not lead to preferential killing of p53-deficient over p53-proficient cells, and inhibiting CHK1 does not appear to be a promising approach for potentiation of cancer chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genes, p53/physiology , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinases/physiology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival , Checkpoint Kinase 1 , DNA Damage , Humans , Intracellular Signaling Peptides and Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/physiology
4.
Oncogene ; 25(56): 7381-90, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-16785992

ABSTRACT

Cisplatin is an effective DNA-damaging antitumor agent employed for the treatment of various human cancers. In this study, we report that Cisplatin activates PKB/Akt in several cancer cell lines and that this activation is mediated by EGFR, Src and PI3-kinase. Inhibition of PI3-kinase activity decreases the survival of the cells exposed to Cisplatin, suggesting that Cisplatin-induced PKB/Akt activation may lead to Cisplatin resistance. While investigating the EGFR-dependent PKB/Akt activation in MDA-MB-468 cells, we found that the EGFR receptor undergoes a gel mobility shift upon Cisplatin treatment, which is mediated by p38(MAPK). An EGFR, in which threonine 669 was mutated to alanine (A669), is phosphorylated by p38(MAPK) to a much lesser extent, suggesting that threonine 669 is a p38 phosphorylation site. We found that Cisplatin induces EGFR internalization, which is mediated by p38(MAPK-)dependent phosphorylation of the receptor on threonine 669. Our results identify the EGFR as a new substrate of p38 and identify threonine 669 as a new phosphorylation site that regulates EGFR internalization. Together, these results suggest that Cisplatin has side effects, which may alter the signaling pattern of cancer cells and modulate the desired effects of Cisplatin treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor , Chromones/pharmacology , Endocytosis/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation
5.
Biochem Pharmacol ; 71(10): 1422-34, 2006 May 14.
Article in English | MEDLINE | ID: mdl-16522318

ABSTRACT

The tyrphostin 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) is a potent and specific inhibitor of EGFR tyrosine kinase whose favourable preclinical profile supports progression towards clinical trials. Microphysiometric evaluation revealed a short (<24 min) effective inhibition of cellular receptor response to EGF challenge in BaF/ERX cells indicating a need to maintain sustained levels of inhibitor. Initial pharmacokinetic evaluation in mice of novel AG1478 formulations in a beta-cyclodextrin (Captisol) showed monoexponential elimination from plasma (half-life 30 min) following subcutaneous administration. A two-fold dose escalation gave a 2.4-fold increase in the total AUC. Bolus i.v. and 6 h continuous infusion were investigated in rats to mimic a more clinically relevant administration regimen. Drug elimination following bolus i.v. administration was biphasic (terminal elimination half-life 30-48 min). The linear relationship between dose and AUC(0-->infinity) (r2=0.979) enabled the prediction of infusion rates and doses for sustained delivery using continuous 6 h infusions, where steady state was reached in 120 min. Plasma levels of AG1478>10 microM were achieved over the duration of the infusion. At the lowest dose, plasma drug levels after the cessation of infusion declined with a half-life of approximately 43 min. EGFR activity, measured both by autophosphorylation and downstream signalling, was inhibited in a dose-dependent manner by injection of AG1478 in mice bearing xenografts of the human glioblastoma cell line U87MG.delta2-7, which expresses a constitutively active variant of the EGF receptor. Taken together, these experiments provide essential data to assess the anti-tumour efficacy of AG1478 and will assist in the rational design of dose regimens for clinical studies.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , ErbB Receptors/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Tyrphostins/pharmacokinetics , Animals , Area Under Curve , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Injections, Intravenous , Injections, Subcutaneous , Mice , Molecular Structure , Quinazolines , Rats , Thymidine/metabolism , Tyrphostins/chemistry , Tyrphostins/pharmacology , Xenograft Model Antitumor Assays
7.
Curr Med Chem ; 11(6): 675-92, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15032723

ABSTRACT

A stochastic algorithm for constructing multiple loops in homology modeling of proteins is presented. The algorithm discards variable values in iterations based on a cost function and on statistical analysis of results. Values that remain are used for constructing an ensemble of best solutions. In test cases, the stochastic algorithm retains all the best solutions, compared to an exhaustive scan of the full set of conformations. Individual loops are constructed by adding dipeptide units. Dipeptide conformations are extracted from a database of proteins and their conformations include bond lengths and all angles. Single loops are constructed from both N- and C- terminals to the center, and loop closure is evaluated by a combination of penalties for the peptide closure and Miyazawa-Jernigan (MJ) [1]. residue-residue interactions with the rest of the protein. Large ensembles of each loop are clustered and re-evaluated with a refined [2]. energy term. The reduced, clustered set of each loop is then employed to construct simultaneously all the loops. The algorithm was applied to construct simultaneously six loops in c-Src kinase family proteins, incorporating a total of 37-40 residues. The best RMSD for reconstructing the loops is 1.45 A for Lck (structure 1QPE in the Protein Data Bank) and 2.54 A for human c-Src (structure 1FMK). The multiple loop conformations with lowest energy have higher RMSD values, of 2.06 A and 3.09 A, respectively. The average RMSD values for the first 1000 conformations are 3.00 A and 3.46 A, respectively. Models for the "open" structures of c-Src and of Jak-2 were constructed on the basis of 1QPE. The Jak-2 model is found to be more flexible in the loops region than its c-Src counterpart.


Subject(s)
Algorithms , Dipeptides/chemistry , Models, Molecular , src-Family Kinases/chemistry , Databases, Protein , Drug Design , Models, Chemical , Models, Statistical , Molecular Sequence Data , Protein Engineering , Structural Homology, Protein
8.
Arterioscler Thromb Vasc Biol ; 21(9): 1434-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11557668

ABSTRACT

Poor drug residence in the arterial wall hinders clinical implementation of local drug delivery strategies for the treatment of restenosis. A rat carotid model of vascular injury and intraluminal delivery of tyrphostin-containing polylactic acid (PLA) nanoparticles (NPs) were used to determine the relationship between residence properties and biological activity of different formulations and administration modes. The effects of delivery modes (denudation and delivery time) and formulation variables (adsorbed vs encapsulated drug, and NP size) on arterial drug/NP retention were examined. Antirestenotic effects of large (160 nm) and small (90 nm) tyrphostin-containing NPs, surface-absorbed tyrphostin, and systemic treatment were compared. Fluorescent NPs were used to study the spatial distribution of the carrier in the arterial wall. The decrease in arterial tyrphostin level over time fitted a biexponential model. Delivery time and pressure, endothelium integrity, particle size, and drug-polymer association affected local pharmacokinetics and the antirestenotic results after 14 days. The PLA-based tyrphostin NP formulation ensured a prolonged drug residence at the angioplasty site after single intraluminal application. Several readily adjustable formulation and procedural factors considerably modified arterial ingress of the drug-loaded NPs and governed their subsequent redistribution, tissue binding, elimination, and ensuing antirestenotic effect.


Subject(s)
Carotid Stenosis/drug therapy , Drug Delivery Systems , Tyrphostins/administration & dosage , Tyrphostins/pharmacology , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Chemistry, Pharmaceutical , Male , Microscopy, Fluorescence , Microspheres , Rats , Tyrphostins/pharmacokinetics
9.
J Neurosurg ; 95(3): 472-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11565870

ABSTRACT

OBJECT: Activation of signaling by the epidermal growth factor receptor (EGFR) through gene amplification or rearrangement is common in human malignancy, especially in a large fraction of de novo glioblastomas multiforme (GBMs). The most common mutant EGFR, (AEGFR, also known as de2-7 EGFR and EGFRvIII) lacks a portion of the extracellular domain, enhances tumorigenicity in vivo, and causes resistance to the chemotherapeutic drug cisplatin (CDDP). This resistance is due to the suppression of CDDP-induced apoptosis by the constitutively active tyrosine kinase activity of the receptor. The authors have investigated whether inhibition of AEGFR signaling by the tyrosine kinase inhibitor, tyrphostin AG1478, could sensitize tumor xenografts to CDDP and, thereby, enhance its therapeutic efficacy in animals. METHODS: Nude mice were inoculated either subcutaneously or intracerebrally with human GBM cells expressing AEGFR and were then systemically treated with CDDP and/or AG1478. Tumor volumes were monitored and tumor sections were analyzed by using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assays or MIB-1 staining. Expression of AEGFR, but not wild-type EGFR, conferred CDDP resistance to the cells in vivo. Inhibition of receptor signaling by the EGFR-specific tyrosine kinase inhibitor, AG1478. sensitized the xenografts to the cytotoxic effects of CDDP. This combined CDDP/AG1478 treatment significantly suppressed growth of subcutaneous xenografts in nude mice in a synergistic manner (p < 0.01 compared with vehicle control) without causing generalized toxicity, whereas treatments with CDDP or AG1478 alone were ineffective. The synergistic growth suppression by the CDDP/AG1478 combination was not observed in xenografts overexpressing wild-type EGFR or kinase-deficient AEGFR. The combined CDDP/ AG1478 treatment induced tumor growth suppression, which correlated with increased apoptosis and reduced proliferation. This treatment also extended the life span of mice bearing intracerebral xenografts (p < 0.01 compared with controls). CONCLUSIONS: The results of this study may provide the basis for the development of a novel and safe therapeutic strategy for the very aggressive AEGFR-expressing GBM.


Subject(s)
Brain Neoplasms/genetics , Cell Survival/genetics , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/genetics , Glioblastoma/genetics , Mutation/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , Tyrphostins/pharmacology , Animals , Brain Neoplasms/pathology , Cell Survival/drug effects , ErbB Receptors/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Quinazolines , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Mol Cell Biol ; 21(20): 6913-26, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564875

ABSTRACT

Many primary tumors as well as transformed cell lines display high sensitivity to chemotherapeutic drugs and radiation. The molecular mechanisms that underlie this sensitivity are largely unknown. Here we show that the sensitization of transformed cells to stress stimuli is due to the potentiation of the c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase pathways. Activation of these pathways by the antitumor drug cis-platin (CDDP) and by other stress agents is markedly enhanced and is induced by lower stress doses in NIH 3T3 cells overexpressing epidermal growth factor receptor, HER1-2 kinase, or oncogenic Ras than in nontransformed NIH 3T3 cells. Inhibition of stress kinase activity by specific inhibitors reduces CDDP-mediated cell death in transformed cells, whereas overactivation of stress kinase pathways augments cells death. Potentiation of stress kinases is a common feature of cells transformed by different oncogenes, including cells derived from human tumors, and is shown here to be independent of the activity of the particular transforming oncoprotein. We further show that the mechanism that underlies potentiation of stress kinases in transformed cells involves reactive oxygen species (ROS), whose production is elevated in these cells. JNK/p38 activation is inhibited by antioxidants and in particular by inhibitors of the mitochondrial respiratory chain and NADPH oxidase. Conversely, by artificially elevating ROS levels in nontransformed NIH 3T3 cells we were able to induce potentiation of JNK/p38 activation. Taken together, our findings suggest that ROS-dependent potentiation of stress kinase pathways accounts for the sensitization of transformed cells to stress and anticancer drugs.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species , 3T3 Cells , Animals , Anisomycin/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line, Transformed , Cell Survival , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , ErbB Receptors/metabolism , Flow Cytometry , Fluoresceins/pharmacology , Fluorescent Dyes/pharmacology , Humans , Immunoblotting , In Situ Nick-End Labeling , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Plasmids/metabolism , Protein Synthesis Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Stress, Physiological , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases
11.
Oncogene ; 20(20): 2499-513, 2001 May 03.
Article in English | MEDLINE | ID: mdl-11420660

ABSTRACT

Constitutive activation of signal transducer and activator of transcription (STAT) proteins has been detected in a wide variety of human primary tumor specimens and tumor cell lines including blood malignancies, head and neck cancer, and breast cancer. We have previously demonstrated a high frequency of Stat3 DNA-binding activity that is constitutively-induced by an unknown mechanism in human breast cancer cell lines possessing elevated EGF receptor (EGF-R) and c-Src kinase activities. Using tyrosine kinase selective inhibitors, we show here that Src and JAK family tyrosine kinases cooperate to mediate constitutive Stat3 activation in the absence of EGF stimulation in model human breast cancer cell lines. Inhibition of Src or JAKs results in dose-dependent suppression of Stat3 DNA-binding activity, which is accompanied by growth inhibition and induction of programmed cell death. In addition, transfection of a dominant-negative form of Stat3 leads to growth inhibition involving apoptosis of breast cancer cells. These results indicate that the biological effects of the Src and JAK tyrosine kinase inhibitors are at least partially mediated by blocking Stat3 signaling. While EGF-R kinase activity is not required for constitutive Stat3 activation in breast cancer cells, EGF stimulation further increases STAT DNA-binding activity, consistent with an important role for EGF-R in STAT signaling and malignant progression. Analysis of primary breast tumor specimens from patients with advanced disease revealed that the majority exhibit elevated STAT DNA-binding activity compared to adjacent non-tumor tissues. Our findings, taken together, suggest that tyrosine kinases transduce signals through Stat3 protein that contribute to the growth and survival of human breast cancer cells in culture and potentially in vivo.


Subject(s)
Breast Neoplasms/pathology , DNA-Binding Proteins/physiology , Drosophila Proteins , Protein-Tyrosine Kinases/physiology , Trans-Activators/physiology , src-Family Kinases/physiology , Animals , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Division/physiology , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/physiology , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/physiology , Humans , Insect Proteins , Janus Kinase 1 , Mice , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyridones/pharmacology , Pyrimidines/pharmacology , STAT3 Transcription Factor , Signal Transduction/physiology , Trans-Activators/metabolism , Tumor Cells, Cultured , Tyrphostins/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
13.
Nucl Med Biol ; 28(4): 359-74, 2001 May.
Article in English | MEDLINE | ID: mdl-11395308

ABSTRACT

As PET candidate tracers for EGFr-TK, five 4-(anilino)quinazoline derivatives, each fluorinated in the aniline moiety, were prepared. Each was tested in vitro for inhibition of EGFr autophosphorylation in A431 cell line. The leading compounds were then radiolabeled with (18)F and cell binding experiments, biodistribution and PET studies in A431 tumor-bearing mice were performed. Metabolic studies were carried out in a mice control group. From our results, we concluded that while in vitro experiments indicates efficacy of 4-(anilino)quinazoline compounds, kinetic factors and rapid blood clearance make them unsuitable as tracers for nuclear medicine imaging of EGFr-TK.


Subject(s)
ErbB Receptors/metabolism , Quinazolines , Radiopharmaceuticals , Animals , Cell Division , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Fluorine Radioisotopes , Magnetic Resonance Spectroscopy , Mice , Phosphorylation , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tomography, Emission-Computed
15.
Cancer J ; 7(3): 191-202, 2001.
Article in English | MEDLINE | ID: mdl-11419027

ABSTRACT

BACKGROUND: Both EGF family ligands and ErbB family receptor kinases act upstream of RAS to induce mitogenesis of normal cells, such as NIH 3T3 fibroblasts. However, oncogenically mutated RAS, such as v-Ha-RAS is constitutively activated and therefore no longer requires these ligands or receptors for its activation. Nevertheless, it up-regulates the expression of these EGF family ligands. To understand the biologic significance of RAS-induced up-regulation of these ligands in both RAS-induced PAK activation and malignant transformation, we have conducted the following studies, based on the previous observations that (1) the N-terminal SH3 domain of PIX selectively binds a Pro-rich domain of 18 amino acids of PAKs, CDC42/Rac-dependent Ser/Thr kinase family, and (2) this specific interaction is essential for both PAK activation and membrane ruffling RESULTS: Using four distinct, cell-permeable, and highly specific inhibitors, namely WR-PAK18, which blocks the PAK-PIX interaction; AG 1478, which inhibits ErbB1 kinase activity; and AG 825 or AG 879, which inhibits ErbB2 kinase activity, we demonstrate that (1) the PAK-PIX interaction is essential for v-Ha-RAS-induced malignant transformation; (2) v-Ha-RAS requires not only ErbB1 but also ErbB2, which are activated through two independent autocrine pathways to induce both the PIX/Rac/CDC42-dependent PAK activation and malignant transformation in vitro; and (3) a combination of AG 879 and the Src family kinase-specific inhibitor PP1 suppresses almost completely the growth of RAS-induced sarcomas in nude mice. CONCLUSION: These findings not only change our conventional view on the role of these RAS-inducible ligands and ErbB family receptors (serving as RAS activators) but also suggest a new avenue for the treatment of RAS-associated cancers by a combination of inhibitors specific for ERbB, Src, or PAK family kinases.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins , Enzyme Inhibitors/therapeutic use , Glycoproteins/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Plant Proteins , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , Receptor, ErbB-2/drug effects , Tyrphostins/therapeutic use , 3T3 Cells/drug effects , 3T3 Cells/metabolism , Animals , Cattle , Culture Media, Serum-Free , Enzyme Activation/drug effects , Female , Mice , Mice, Nude , Peptide Fragments/pharmacology , Quinazolines , Receptor, ErbB-2/physiology , Rho Guanine Nucleotide Exchange Factors , Sarcoma, Experimental/drug therapy , Tyrphostins/pharmacology , Up-Regulation , p21-Activated Kinases , src-Family Kinases
16.
J Biol Chem ; 276(29): 26741-4, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11356824

ABSTRACT

The platelet-derived growth factor (PDGF) receptor (PDGFR) transactivates the epidermal growth factor (EGF) receptor (ErbB1) to stimulate the cell migration of fibroblasts through an unknown mechanism (Li, J., Kim, Y. N. & Bertics, P. (2000) J. Biol. Chem. 275, 2951-2958). In this paper we provide evidence that the transactivation of the EGF receptor (EGFR) by PDGFR is essential for PDGF to activate p21-activated kinase (PAK) family kinases. Fetal calf serum (10%) transiently stimulates the PAK activity in NIH 3T3 fibroblasts. The activation of PAK was completely inhibited by either PDGFR-specific inhibitor (AG1295) or EGFR-specific inhibitor (AG1478), suggesting that serum requires either the PDGF- or EGF-dependent pathway or the combination of both to activate PAK. PDGF-induced activation of PAK is completely inhibited by either AG1295 or AG1478, indicating that PDGF requires both PDGFR and EGFR for PAK activation. In support of this notion, a mouse embryo fibroblast cell line derived from the EGFR -/- mouse (from Dr. Erwin Wagner) doesn't activate PAK in response to PDGF. Expression of human EGFR in this cell line restores the ability of the PDGF to induce PAK activation. Our results indicate that PDGF activates PAK through transactivation of ErbB1.


Subject(s)
ErbB Receptors/physiology , Platelet-Derived Growth Factor/physiology , Protein Serine-Threonine Kinases/metabolism , 3T3 Cells , Animals , Blood , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Enzyme Activation , Fibroblasts/enzymology , Humans , Mice , Platelet-Derived Growth Factor/metabolism , Signal Transduction
17.
J Biol Chem ; 276(27): 25351-8, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11309396

ABSTRACT

Mitogen-activated protein kinases (MAPKs) play pivotal roles in growth, development, differentiation, and apoptosis. The exact role of a given MAPK in these processes is not fully understood. This question could be addressed using active forms of these enzymes that are independent of external stimulation and upstream regulation. Yet, such molecules are not available. MAPK activation requires dual phosphorylation, on neighboring Tyr and Thr residues, catalyzed by MAPK kinases (MAPKKs). It is not known how to force MAPK activation independent of MAPKK phosphorylation. Here we describe a series of nine hyperactive (catalytically and biologically), MAPKK-independent variants of the MAPK Hog1. Each of the active molecules contains just a single point mutation. Six mutations are in the conserved L16 domain of the protein. The active Hog1 mutants were obtained through a novel genetic screen that could be applied for isolation of active MAPKs of other families. Equivalent mutations, introduced to the human p38alpha, rendered the enzyme active even when produced in Escherichia coli, showing that the mutations increased the intrinsic catalytic activity of p38. It implies that the activating mutations could be directly used for production of active forms of MAPKs from yeasts to humans and could open the way to revealing their biological functions.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Enzyme Activation , Humans , MAP Kinase Kinase 1 , Mice , Mitogen-Activated Protein Kinases/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Point Mutation , Rats , Saccharomyces cerevisiae , Structure-Activity Relationship , Threonine/metabolism , Tyrosine/metabolism , p38 Mitogen-Activated Protein Kinases
18.
J Chromatogr B Biomed Sci Appl ; 754(1): 193-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11318415

ABSTRACT

The tyrphostin 4-(3-chloroanilino)-6,7-dimethoxyquinazoline (AG1478) is undergoing evaluation as a potential new anticancer agent. We have developed a specific and sensitive reversed-phase HPLC assay for AG1478 in mouse plasma. The method involves a rapid and simple extraction process followed by separation on a Symmetry C8 stationary phase with a gradient of acetonitrile in ammonium acetate buffer. A linear response was achieved over the concentration range of 0.2-100 microM using multilevel calibration with internal standard method of calculation. Inter- and intra-assay accuracy and precision were better than +/-10%. The limit of quantitation was 0.2 microM. We have used this method to study the preclinical pharmacokinetics of this new agent in mice.


Subject(s)
Chromatography, High Pressure Liquid/methods , ErbB Receptors/antagonists & inhibitors , Tyrphostins/blood , Animals , Enzyme Inhibitors/blood , Enzyme Stability , Freezing , Mice , Quinazolines , Reference Standards , Sensitivity and Specificity , Tyrphostins/pharmacokinetics
20.
Biochemistry ; 39(51): 15705-12, 2000 Dec 26.
Article in English | MEDLINE | ID: mdl-11123895

ABSTRACT

IGF-1 and its receptor play a pivotal role in many cancers, and therefore, IGF-1R is an attractive target for the design of inhibitors. In this communication, we report on a number of lead compounds for inhibitors of the isolated IGF-1R kinase. The search for these compounds utilized two novel in vitro assays and was aided by the knowledge of the three-dimensional structure of the insulin receptor kinase domain, which is 84% homologous to the IGF-1R kinase domain. The most potent inhibitor found in these assays was tyrphostin AG 538, with an IC(50) = 400 nM. In computer modeling, AG 538 was placed in the kinase domain of the insulin receptor and was able to sit in place of tyrosines 1158 and 1162, which undergo autophosphorylation. Experimentally it is indeed found that AG 538 does not compete with ATP but competes with the IGF-1R substrate. We prepared I-OMe AG 538, which is more hydrophobic and less sensitive to oxidation than AG 538. Both AG 538 and I-OMe AG 538 inhibit IGR-1R autophosphorylation in intact cells in a dose-dependent manner but I-OMe-AG 538 is superior, probably because of its enhanced hydrophobic nature. Both compounds inhibit the activation of the downstream targets PKB and Erk2. These findings suggest that AG 538 and I-OMe-AG 538 can serve as a lead compound for the development of substrate competitive inhibitors of the IGF-1R. The possible advantage of substrate competitive inhibitors vis-à-vis ATP competitive inhibitors is discussed.


Subject(s)
Catechols/metabolism , Catechols/pharmacology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Tyrphostins/metabolism , Tyrphostins/pharmacology , 3T3 Cells , Adenosine Triphosphate/metabolism , Animals , Binding, Competitive , Dose-Response Relationship, Drug , Intercellular Signaling Peptides and Proteins , Mice , Peptides/antagonists & inhibitors , Peptides/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Signal Transduction/drug effects , Substrate Specificity/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...