Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 242: 114635, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-35988448

ABSTRACT

Fluorizoline is a cytotoxic trifluorothiazoline that targets the scaffold proteins prohibitins-1 and -2 (PHB1/2) to inhibit the kinase C-RAF and promote the expression of the cyclin-dependent kinase inhibitor p21 to induce cancer cell death. In melanocytes, fluorizoline also induces the synthesis of melanin. Herein we report the first structural requirement of fluorizoline analogues for these activities. We identified in particular some compounds that display enhanced anti-C-RAF and anti-MEK activities, and a higher cytotoxicity in HeLa cells compared to fluorizoline. These results provide a foundation for further optimization of PHB ligands for the treatment of cancers. We also discovered an analogue of fluorizoline that displays pharmacological effects opposed to those of fluorizoline and that can be used as a chemical tool to explore PHB signaling in cancers and other diseases.


Subject(s)
Apoptosis , Prohibitins , Cyclin-Dependent Kinase Inhibitor p21/metabolism , HeLa Cells , Humans , Ligands , Melanins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/pharmacology , Repressor Proteins , Transcription Factors/metabolism
2.
Eur J Med Chem ; 238: 114434, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35551038

ABSTRACT

This article describes the design, synthesis, and biological screening of a new series of diarylurea and diarylamide derivatives including quinoline core armed with dimethylamino or morpholino side chain. Fifteen target compounds were selected by the National Cancer Institute (NCI, USA) for in vitro antiproliferative screening against a panel of 60 cancer cell lines of nine cancer types. Compounds 1j-l showed the highest mean inhibition percentage values over the 60-cell line panel at 10 µM with broad-spectrum antiproliferative activity. Subsequently, compounds 1j-l were subjected to a dose-response study to measure their GI50 and total growth inhibition (TGI) values against the cell lines. Three of the tested molecules exerted higher potency against most of the cell lines than the reference drug, sorafenib. Compound 1l indicated a higher potency than sorafenib against 53 of tested cancer cell lines. Compounds 1j-l demonstrated promising selectivity against cancer cells than normal cells. Moreover, compound 1l induced apoptosis and necrosis in RPMI-8226 cell line in a dose-dependent manner. In addition, compounds 1j-l were tested against C-RAF kinase as a potential molecular target. The three compounds showed high potency, and the most potent C-RAF kinase inhibitor was compound 1j with an IC50 value of 0.067 µM. In addition, Compounds 1j-l were further tested at 1 µM concentration against a panel of another twelve kinases and they showed a high selectivity for C-RAF kinase. Molecular modeling studies were performed to illuminate on the putative binding interactions of these motifs in the active site of C-RAF kinase. Additional studies were conducted to measure aqueous solubility, partition coefficient, and Caco-2 permeability of the most promising derivatives.


Subject(s)
Antineoplastic Agents , Hydroxyquinolines , Quinolines , Antineoplastic Agents/chemistry , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Humans , Hydroxyquinolines/pharmacology , Molecular Structure , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-raf/pharmacology , Quinolines/chemistry , Sorafenib/pharmacology , Structure-Activity Relationship
3.
Blood ; 108(1): 152-9, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16527894

ABSTRACT

Normal erythropoiesis critically depends on the balance between the renewal of precursor cells and their differentiation. If the renewal phase is shortened, the decrease in the precursor pool results in anemia; conversely, impaired differentiation increases the number of proliferating progenitors and the potential risk of leukemic transformation. Using gene ablation, we have discovered 2 self-sustaining signal transduction loops that antagonize each other and regulate erythroid progenitor proliferation and differentiation, respectively. We identify Raf-1 as the main activator of the MEK/ERK cascade and as the key molecule in maintaining progenitor proliferation. Differentiation, in contrast, is mediated by Fas via the activation of both the ASK1/JNK/p38 module and the caspase cascade. The point of convergence between the 2 cascades is activated ERK, which positively feeds back on the proliferation pathway by maintaining the expression of Raf-1, while inhibiting the expression of Fas and therefore differentiation. In turn, Fas, once expressed, antagonizes proliferation by exerting a negative feedback on ERK activation and Raf-1 expression. Simultaneously, Fas-mediated caspase activation precipitates differentiation. These results identify Raf-1 and Fas as the key molecules whose expression finely tunes erythropoiesis and the extent of ERK activation as the switch that tips the balance between them.


Subject(s)
Erythroblasts/drug effects , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/pharmacology , fas Receptor/genetics , fas Receptor/pharmacology , Animals , Caspases/metabolism , Cell Differentiation/drug effects , Erythroblasts/cytology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction/methods , Structure-Activity Relationship
4.
FEBS J ; 273(1): 170-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16367757

ABSTRACT

A role for the nuclear receptor peroxisome proliferator-activated receptor-beta (PPARbeta) in oncogenesis has been suggested by a number of observations but its precise role remains elusive. Prostaglandin I2 (PGI2, prostacyclin), a major arachidonic acid (AA) derived cyclooxygenase (Cox) product, has been proposed as a PPARbeta agonist. Here, we show that the 4-hydroxytamoxifen (4-OHT) mediated activation of a C-Raf-estrogen receptor fusion protein leads to the induction of both the PPARbeta and Cox-2 genes, concomitant with a dramatic increase in PGI2 synthesis. Surprisingly, however, 4-OHT failed to activate PPARbeta transcriptional activity, indicating that PGI2 is insufficient for PPARbeta activation. In agreement with this conclusion, the overexpression of ectopic Cox-2 and PGI2 synthase (PGIS) resulted in massive PGI2 synthesis but did not activate the transcriptional activity of PPARbeta. Conversely, inhibition of PGIS blocked PGI2 synthesis but did not affect the AA mediated activation of PPARbeta. Our data obtained with four different cell types and different experimental strategies do not support the prevailing opinion that PGI2 plays a significant role in the regulation of PPARbeta.


Subject(s)
Epoprostenol/biosynthesis , PPAR-beta/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , Autocrine Communication , CHO Cells , Cells, Cultured , Cricetinae , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Epoprostenol/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NIH 3T3 Cells , PPAR-beta/genetics , Prostaglandin-E Synthases , Prostaglandins/biosynthesis , Prostaglandins/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/pharmacology , Signal Transduction , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Tamoxifen/pharmacology , Time Factors , Transcriptional Activation , Transfection
5.
Bioconjug Chem ; 16(4): 1034-7, 2005.
Article in English | MEDLINE | ID: mdl-16029047

ABSTRACT

An antisense oligodeoxynucleotide (ODN) delivery system based on polyelectrolyte complex (PEC) micelles composed of an ODN-poly(ethylene glycol) (PEG) conjugate and polyethylenimine (PEI) was demonstrated. The PEC micelles having a core/shell structure were spontaneously formed in an aqueous solution by ionic interactions between ODN part in the conjugate and PEI. The ODN/PEI polyelectrolyte complex formed an inner core while PEG chains surrounded it as a shell. The morphology of the micelles was visualized as a separate sphere by atomic force microscopy (AFM). When the micelles containing a c-raf antisense ODN were intravenously administered into tumor-bearing nude mice, significant antitumor activities against human lung cancer were observed. The intravenously injected micelles also showed significantly higher accumulation level in the solid tumor region compared to that of naked ODN.


Subject(s)
Electrolytes/chemistry , Lung Neoplasms/pathology , Micelles , Oligonucleotides, Antisense/chemistry , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Proto-Oncogene Proteins c-raf/chemistry , Animals , Base Sequence , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Microscopy, Atomic Force , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacology , Polyethyleneimine/administration & dosage , Polyethyleneimine/pharmacology , Proto-Oncogene Proteins c-raf/administration & dosage , Proto-Oncogene Proteins c-raf/pharmacology
6.
Int J Cancer ; 112(2): 190-9, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15352030

ABSTRACT

Ras expression induces increased expression and altered targeting of lysosomal proteases in multiple cell types, but the specific downstream cytoplasmic signaling pathways mediating these changes have not been identified. In this study, we compared the involvement of 3 major Ras effectors, Raf, phosphatidylinositol 3-kinase (PI3K) and Ral guanine nucleotide exchange factor (RalGEF) in the Ras-mediated alteration of lysosomal protease protein expression and targeting in rat 208F fibroblasts and rat ovarian surface epithelial (ROSE) cells. Effector domain mutants of Ras, constitutively activated variants of Raf, PI3K and RalGEF and pharmacologic inhibitors of MEK and PI3K were utilized to determine the role of these downstream pathways in mediating fibroblast transformation and lysosomal protease regulation in the fibroblasts and epithelial cells. We found that Raf activation of the ERK mitogen-activated protein kinase pathway alone was sufficient to cause morphologic and growth transformation of the fibroblasts and was necessary and sufficient to alter cathepsin L expression and targeting. In contrast, transformation and upregulation of cathepsin L expression in the epithelial cells required the activity of all 3 Ras effectors. Increased protease secretion from the epithelial cells was not observed on ectopic expression of Ras, as it was from the fibroblasts, consistent with the utilization of different signaling pathways in the 2 cell types. In neither cell type did Ras expression increase the expression, processing or secretion of 2 other major lysosomal proteases, cathepsin B and cathepsin D. Thus, Ras utilizes different effectors to mediate transformation and to deregulate cathepsin L expression and secretion in fibroblast and epithelial cells.


Subject(s)
Cathepsins/biosynthesis , Guanine Nucleotide Exchange Factors/pharmacology , Phosphatidylinositol 3-Kinases/pharmacology , Proto-Oncogene Proteins c-raf/pharmacology , Signal Transduction , Animals , Cathepsin L , Cell Transformation, Neoplastic , Cysteine Endopeptidases , Enzyme Precursors , Epithelial Cells , Female , Fibroblasts/physiology , Lysosomes , Mitogens , Ovary/cytology , Rats
7.
Am J Physiol Cell Physiol ; 286(5): C1118-29, 2004 May.
Article in English | MEDLINE | ID: mdl-15075211

ABSTRACT

The epidermal growth factor (EGF) receptor plays an important role in epithelial cells by controlling cell proliferation and survival. Keratinocytes also express another class of receptor tyrosine kinases, the neurotrophin receptors. To analyze the biological role of the neurotrophin brain-derived neurotrophic factor (BDNF) in keratinocytes, we expressed the BDNF receptor TrkB in immortalized human HaCaT keratinocytes. Stimulation of HaCaT-TrkB cells with BDNF induced DNA synthesis and increased mitochondrial reduction capacities, both indications of proliferating cells. An analysis of the signal transduction cascade revealed that the activated TrkB receptor effectively utilized components of the EGF receptor signaling pathway to control cell proliferation. Mitogenic signaling induced by BDNF or EGF was completely abrogated by the MAP kinase kinase inhibitor PD-98059, whereas inhibition of phosphatidylinositol 3-kinase by wortmannin only delayed the proliferative response. The importance of the extracellular signal-regulated kinase signaling pathway for growth of HaCaT keratinocytes was further demonstrated with HaCaT cells engineered to express an inducible A-Raf-estrogen receptor fusion protein (DeltaA-Raf:ER). Despite differences in the amplitude and duration of extracellular signal-regulated kinase activation, HaCaT cells expressing DeltaA-Raf:ER proliferated after activation of mutant A-Raf protein kinase. Proliferation was completely inhibited by PD-98059. Proliferation of HaCaT cells induced by EGF, BDNF, or DeltaA-Raf:ER was also accompanied by biosynthesis of the transcription factors Egr-1 and c-Jun, suggesting that these proteins may be part of the mitogenic signaling cascade.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Epidermal Growth Factor/pharmacology , Keratinocytes/cytology , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-raf/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Enzyme Activation/physiology , ErbB Receptors/metabolism , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Keratinocytes/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins A-raf , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-raf/genetics , Receptor, trkB/metabolism , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptional Activation/drug effects
8.
Prostate ; 59(2): 214-25, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15042621

ABSTRACT

BACKGROUND: The androgen receptor (AR) is a ligand-activated transcription factor that mediates the biological responses of androgens in the prostate gland. This study focuses on the chemopreventive agents, resveratrol and genistein, on AR-mediated transcription in prostate cancer cells. RESULTS: We found that resveratrol and genistein activated AR-driven gene expression at low concentrations, whereas they repressed the AR-dependent reporter gene activity at high concentrations. We determined that resveratrol and genistein induced AR-driven gene expression by activating the Raf-MEK-ERK kinase pathway. The ERK1 kinase phosphorylated the AR on multiple sites in vitro, but this phosphorylation event did not contribute to the resveratrol-induced AR transactivation. CONCLUSIONS: In vitro and in vivo studies have indicated that resveratrol and genistein are promising chemopreventive agents. Given the clear evidence that AR pathways are involved in the development and progression of prostate cancer, these data showed that the ability to modulate AR function would contribute the observed chemopreventive activity of resveratrol and genistein.


Subject(s)
Anticarcinogenic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genistein/pharmacology , MAP Kinase Kinase Kinase 1 , Prostatic Neoplasms/genetics , Prostatic Neoplasms/prevention & control , Receptors, Androgen/drug effects , Receptors, Androgen/physiology , Stilbenes/pharmacology , Amino Acid Sequence , Dose-Response Relationship, Drug , Humans , MAP Kinase Kinase Kinases/pharmacology , Male , Mitogen-Activated Protein Kinases/pharmacology , Molecular Sequence Data , Phosphorylation , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-raf/pharmacology , Resveratrol , Signal Transduction , Transfection
9.
Br J Cancer ; 90(2): 283-8, 2004 Jan 26.
Article in English | MEDLINE | ID: mdl-14735164

ABSTRACT

The Raf-MEK-ERK signalling pathway controls fundamental cellular processes including proliferation, differentiation and survival. It remains enigmatic how this pathway can reliably convert a myriad of extracellular stimuli in specific biological responses. Recent results have shown that the Raf family isoforms A-Raf, B-Raf and Raf-1 have different physiological functions. Here we review how Raf isozyme diversity contributes to the specification of functional diversity, in particular regarding the role of Raf isozymes in cancer.


Subject(s)
MAP Kinase Kinase Kinase 1 , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/pharmacology , Neoplasms/physiopathology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/pharmacology , Signal Transduction , Cell Differentiation , Cell Survival , Humans , Isoenzymes
10.
J Control Release ; 93(2): 183-91, 2003 Dec 05.
Article in English | MEDLINE | ID: mdl-14636724

ABSTRACT

The conjugate of antisense c-raf oligonucleotide (ODN) and poly(ethylene glycol) (PEG) was synthesized for intracellular ODN delivery. When combined with polyethylenimine (PEI), the ODN-PEG conjugate self-associated to form polyelectrolyte complex micelles in aqueous solution. The effective hydrodynamic diameter of the micelles was ca. 70 nm with a narrow size distribution. Flow cytometry analysis indicated that the cellular uptake of the micelles by A2780 cells was much higher than that of ODN alone. The micelles also showed a superior antiproliferative activity against ovarian cancer cells in vitro and in vivo.


Subject(s)
Drug Carriers/pharmacology , Micelles , Oligonucleotides, Antisense/pharmacology , Polyethylene Glycols/pharmacology , Animals , Cell Division/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Electrolytes/chemistry , Humans , Injections, Intralesional , Injections, Subcutaneous , Mice , Mice, Nude , Neoplasm Transplantation/methods , Oligonucleotides, Antisense/chemical synthesis , Oligonucleotides, Antisense/chemistry , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyethyleneimine/chemical synthesis , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/pharmacology , Solutions , Time Factors
11.
J Environ Pathol Toxicol Oncol ; 22(2): 75-92, 2003.
Article in English | MEDLINE | ID: mdl-14533871

ABSTRACT

Cholesterol synthesis was demonstrated to be mandatory for cellular growth and serves to supply one of the necessary building blocks for new membranes demanded by dividing cells during growth. The mevalonate pathway, which is regulated through a finely tuned mechanism, is responsible mainly for cholesterol enrichment to cells. Among the various steps, the production of mevalonate from 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) is the most critically regulated step catalyzed by HMG-CoA reductase. The ability of sterols to regulate both the transcriptional rates of the reductase gene and the degradative machinery for the reductase protein provides a multilevel system for controlling the expression of this enzyme. Much convincing evidence indicates that cells manifest a higher flux through the mevalonate pathway when proliferating than when they are in the cell cycle arrest condition; furthermore, tumors undergo deregulated cholesterogenesis mainly at the critical rate-controlling juncture (i.e., the reaction catalyzed by HMG-CoA reductase). The mevalonate component of the cholesterol biosynthesis plays a key role in controlling cell proliferation by generating prenyl intermediates, particularly farnesyl and geranyl-geranyl moieties. These isoprenoids covalently modify and thus modulate the biological activity of signal transducing proteins, such as that of the Ras superfamily. The prenylated Ras-mediated signal transduction pathway provides much of the molecular information needed to trigger cell proliferation. Therefore, depletion of mevalonate can block both the processing and the transforming activities of Ras, indicating that drugs such as lovastatin and compactin, which had previously been exploited for lowering cholesterol levels, may be useful chemotherapeutic agents for treating tumors harboring oncogenic Ras mutation. In addition, Ras prenylation, which provides much of the molecular information needed to trigger cell proliferation, represents an inviting target for the design of chemotherapeutic drugs that would interrupt such signaling events and arrest tumor cell proliferation.


Subject(s)
Acyl Coenzyme A/pharmacology , Cell Division , Cell Transformation, Neoplastic , Cholesterol/biosynthesis , Signal Transduction , Antineoplastic Agents/pharmacology , Cholesterol/metabolism , Humans , Mevalonic Acid/chemistry , Proto-Oncogene Proteins c-raf/pharmacology , ras Proteins/pharmacology
12.
J Biol Chem ; 278(45): 44178-87, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-12947115

ABSTRACT

The development of myoblasts is regulated by various growth factors as well as by intrinsic muscle-specific transcriptional factors. In this study, we analyzed the roles for STAT3 in the growth and differentiation of myoblasts in terms of cell cycle regulation and interaction with MyoD using C2C12 cells. Here we found that STAT3 inhibited myogenic differentiation induced by low serum or MyoD as efficiently as the Ras/mitogen-activated protein kinase cascade. As for this mechanism, we found that STAT3 not only promoted cell cycle progression through the induction of c-myc but also inhibited MyoD activities through direct interaction. STAT3 inhibited not only DNA binding activities of MyoD but also its transcriptional activities. However, the inhibited transcriptional activities were restored by the supplement of p300/CBP and PCAF, suggesting that STAT3 might deprive MyoD of these transcriptional cofactors. In addition, we found that MyoD inhibited DNA binding activities of STAT3, thereby inhibiting STAT3-dependent cell growth and survival of Ba/F3 cells. These results suggest that the development of muscle cells is regulated by the coordination of cytokine signals and intrinsic transcription factors.


Subject(s)
Cell Differentiation , Cell Division , DNA-Binding Proteins/physiology , MyoD Protein/physiology , Myoblasts/cytology , Trans-Activators/physiology , Animals , Antigens, CD/genetics , Antigens, CD/pharmacology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cytokine Receptor gp130 , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Drug Interactions , E1A-Associated p300 Protein , Gene Expression/drug effects , Glutathione Transferase/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Mice , Mitogen-Activated Protein Kinases/pharmacology , MyoD Protein/genetics , MyoD Protein/pharmacology , Myoblasts/drug effects , Myogenin/pharmacology , NIH 3T3 Cells , Nuclear Proteins/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/pharmacology , Proto-Oncogene Proteins c-raf/pharmacology , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Fusion Proteins , STAT3 Transcription Factor , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Trans-Activators/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcriptional Activation , Transfection
13.
Mol Biol Cell ; 12(10): 3074-86, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11598192

ABSTRACT

We investigated the role of H-Ras in chemokine-induced integrin regulation in leukocytes. Stimulation of Jurkat T cells with the CXC chemokine stromal cell-derived factor-1alpha (SDF-1alpha) resulted in a rapid increase in the phosphorylation, i.e., activation of extracellular signal receptor-activated kinase (ERK) but not c-Jun NH(2)-terminal kinase or p38 kinase, and phosphorylation of Akt, reflecting phosphatidylinositol 3-kinase (PI3-K) activation. Phosphorylation of ERK in Jurkat cells was enhanced and attenuated by expression of dominant active (D12) or inactive (N17) forms of H-Ras, respectively, while N17 H-Ras abrogated SDF-1alpha-induced Akt phosphorylation. SDF-1alpha triggered a transient regulation of adhesion to intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 mediated by lymphocyte function antigen-1 (LFA-1) and very late antigen-4 (VLA-4), respectively, and a rapid increase in LFA-1 binding to soluble ICAM-1.Ig, which was inhibited by D12 but not N17 H-Ras. Both D12 and N17 H-Ras abrogated the regulation of LFA-1 but not VLA-4 avidity, and impaired LFA-1-mediated transendothelial chemotaxis but not VLA-4-dependent transmigration induced by SDF-1alpha. Analysis of the mutant Jurkat J19 clone revealed LFA-1 with constitutively high affinity and reduced ERK phosphorylation, which were partially restored by expression of active H-Ras. Inhibition of PI3-K blocked the up-regulation of Jurkat cell adhesion to ICAM-1 by SDF-1alpha, whereas inhibition of mitogen-activated protein kinase kinase impaired the subsequent down-regulation and blocking both pathways abrogated LFA-1 regulation. Our data suggest that inhibition of initial PI3-K activation by inactive H-Ras or sustained activation of an inhibitory ERK pathway by active H-Ras prevail to abolish LFA-1 regulation and transendothelial migration induced by SDF-1alpha in leukocytes, establishing a complex and bimodal involvement of H-Ras.


Subject(s)
Cell Movement/physiology , Chemokines, CXC/metabolism , Genes, ras/physiology , Lymphocyte Function-Associated Antigen-1/metabolism , Protein Serine-Threonine Kinases , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Chemotaxis/drug effects , Chemotaxis/physiology , Endothelium/cytology , Endothelium/metabolism , Genes, ras/genetics , Humans , Integrin alpha4beta1 , Integrins/drug effects , Integrins/metabolism , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Leukocytes/physiology , Lymphocyte Function-Associated Antigen-1/drug effects , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/pharmacology , Receptors, Lymphocyte Homing/drug effects , Receptors, Lymphocyte Homing/metabolism , Signal Transduction/physiology , Up-Regulation , p38 Mitogen-Activated Protein Kinases
14.
Endocrinology ; 142(3): 1073-81, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181521

ABSTRACT

The type 1 insulin-like growth factor receptor (IGF-IR) is effective in protecting cells from a variety of apoptotic injuries. In 32D murine hemopoietic cells, the IGF-IR sends three separate survival signals, through insulin receptor substrate-1, Shc, and mitochondrial Raf translocation. We report here that these three pathways for survival have a limited redundancy. If one of these pathways is blocked, the IGF-IR can still protect 32D cells from apoptosis induced by interleukin-3 withdrawal. However, when two of the three pathways are inactivated, the receptor is no longer capable to protect cells from apoptosis. The survival signal can use any two pathway combinations.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Hematopoietic Stem Cells/physiology , Protein Serine-Threonine Kinases , Receptor, IGF Type 1/physiology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/physiology , Chromones/pharmacology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Insulin Receptor Substrate Proteins , Mice , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-raf/pharmacology , Shc Signaling Adaptor Proteins , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1
15.
J Protein Chem ; 19(7): 545-51, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11233167

ABSTRACT

By comparing the average structures, computed using molecular dynamics, of the ras-binding domain of raf (RBD) bound to activated wild-type ras-p21 and its homologous inhibitory protein, rap-1A, we formerly identified three domains of the RBD that changed conformation between the two complexes, residues 62-76, 97-110, and 111-121. We found that one synthetic peptide, corresponding to RBD residues 97-110, selectively inhibited oncogenic ras-p21-induced oocyte maturation. In this study, we performed molecular dynamics on the Val 12-ras-p21-RBD complex and compared its average structure with that for the wild-type protein. We find that there is a large displacement of a loop involving these residues when the structures of the two complexes are compared. This result corroborates our former finding that the RBD 97-110 peptide inhibits only signal transduction by oncogenic ras-p21 and suggests that oncogenic p21 uses this loop to interact with raf in a unique manner.


Subject(s)
Oncogene Protein p21(ras)/chemistry , Proto-Oncogene Proteins c-raf/chemistry , Amino Acid Substitution , Binding Sites , Humans , Models, Molecular , Nonlinear Dynamics , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins c-raf/pharmacology , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/metabolism
16.
Genes Dev ; 12(19): 2997-3007, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9765202

ABSTRACT

The oncogenes RAS and RAF came to view as agents of neoplastic transformation. However, in normal cells, these genes can have effects that run counter to oncogenic transformation, such as arrest of the cell division cycle, induction of cell differentiation, and apoptosis. Recent work has demonstrated that RAS elicits proliferative arrest and senescence in normal mouse and human fibroblasts. Because the Raf/MEK/MAP kinase signaling cascade is a key effector of signaling from Ras proteins, we examined the ability of conditionally active forms of Raf-1 to elicit cell cycle arrest and senescence in human cells. Activation of Raf-1 in nonimmortalized human lung fibroblasts (IMR-90) led to the prompt and irreversible arrest of cellular proliferation and the premature onset of senescence. Concomitant with the onset of cell cycle arrest, we observed the induction of the cyclin-dependent kinase (CDK) inhibitors p21(Cip1) and p16(Ink4a). Ablation of p53 and p21(Cip1) expression by use of the E6 oncoprotein of HPV16 demonstrated that expression of these proteins was not required for Raf-induced cell cycle arrest or senescence. Furthermore, cell cycle arrest and senescence were elicited in IMR-90 cells by the ectopic expression of p16(Ink4a) alone. Pharmacological inhibition of the Raf/MEK/MAP kinase cascade prevented Raf from inducing p16(Ink4a) and also prevented Raf-induced senescence. We conclude that the kinase cascade initiated by Raf can regulate the expression of p16(Ink4a) and the proliferative arrest and senescence that follows. Induction of senescence may provide a defense against neoplastic transformation when the MAP kinase signaling cascade is inappropriately active.


Subject(s)
Cellular Senescence/drug effects , Fibroblasts/drug effects , Proto-Oncogene Proteins c-raf/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Humans , Mice , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase , ras Proteins/metabolism
17.
Am J Respir Cell Mol Biol ; 18(4): 562-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9533945

ABSTRACT

We have previously demonstrated that hydrogen peroxide (H2O2) treatment of bovine tracheal myocytes increases the activity of extracellular signal-regulated kinases (ERK), serine/threonine kinases of the mitogen-activated protein (MAP) kinase superfamily thought to play a key role in the transduction of mitogenic signals to the cell nucleus. Moreover, H2O2-induced ERK activation was partially reduced by pretreatment with phorbol 12,13-dibutyrate, which depletes protein kinase C (PKC). In this study, we further examined the signaling intermediates responsible for ERK activation by H2O2 in airway smooth muscle, focusing on MAP kinase/ERK kinase (MEK), a dual-function kinase which is required and sufficient for ERK activation in bovine tracheal myocytes; Raf-1, a serine/threonine kinase known to activate MEK; and PKC. Pretreatment of cells with inhibitors of MEK (PD98059), Raf-1 (forskolin), and PKC (chelerythrine) each reduced H2O2-induced ERK activity. In addition, H2O2 treatment significantly increased both MEK1 and Raf-1 activity. No activation of MEK2 was detected. Together these data suggest that H2O2 may stimulate ERK via successive activation of PKC, Raf-1, and MEK1.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Hydrogen Peroxide/pharmacology , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Oxidants/pharmacology , Protein Kinases/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cattle , Enzyme Activation/drug effects , HeLa Cells , Humans , Hydroxyl Radical/metabolism , JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase 1 , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Protein Kinase C/pharmacology , Protein Serine-Threonine Kinases/pharmacology , Protein-Tyrosine Kinases/pharmacology , Proto-Oncogene Proteins c-raf/pharmacology , Trachea/cytology , Trachea/drug effects
18.
Mol Biol Cell ; 8(10): 1889-99, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9348531

ABSTRACT

Myosin II heavy chain (MHC) specific protein kinase C (MHC-PKC), isolated from Dictyostelium discoideum, regulates myosin II assembly and localization in response to the chemoattractant cyclic AMP. Immunoprecipitation of MHC-PKC revealed that it resides as a complex with several proteins. We show herein that one of these proteins is a homologue of the 14-3-3 protein (Dd14-3-3). This protein has recently been implicated in the regulation of intracellular signaling pathways via its interaction with several signaling proteins, such as PKC and Raf-1 kinase. We demonstrate that the mammalian 14-3-3 zeta isoform inhibits the MHC-PKC activity in vitro and that this inhibition is carried out by a direct interaction between the two proteins. Furthermore, we found that the cytosolic MHC-PKC, which is inactive, formed a complex with Dd14-3-3 in the cytosol in a cyclic AMP-dependent manner, whereas the membrane-bound active MHC-PKC was not found in a complex with Dd14-3-3. This suggests that Dd14-3-3 inhibits the MHC-PKC in vivo. We further show that MHC-PKC binds Dd14-3-3 as well as 14-3-3 zeta through its C1 domain, and the interaction between these two proteins does not involve a peptide containing phosphoserine as was found for Raf-1 kinase. Our experiments thus show an in vivo function for a member of the 14-3-3 family and demonstrate that MHC-PKC interacts directly with Dd14-3-3 and 14-3-3 zeta through its C1 domain both in vitro and in vivo, resulting in the inhibition of the kinase.


Subject(s)
Dictyostelium/drug effects , Dictyostelium/enzymology , Enzyme Inhibitors/pharmacology , Myosin Heavy Chains , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proteins/pharmacology , Tyrosine 3-Monooxygenase , 14-3-3 Proteins , Animals , Binding Sites , Cyclic AMP/metabolism , Cytosol/enzymology , Drug Interactions , Membrane Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Proteins/analysis , Proteins/chemistry , Proto-Oncogene Proteins c-raf/pharmacology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...