Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Leukemia ; 16(4): 683-92, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11960350

ABSTRACT

Activation of the MEK/ERK/MAP kinase signaling pathway promotes the proliferation and survival of hematopoietic cells. The kinases MEK-1, MEK-2, ERK-1/MAPK and ERK-2/MAPK are activated by phosphorylation at specific sites, and these events can be monitored using phospho-specific antibodies. In this report we examined the importance of the MEK/ERK/MAP kinase pathway in the monocytic and granulocytic differentiation of myeloid cell lines. Induction of monocytic differentiation in HL-60 cells by treatment with phorbol 12-myristate 13-acetate (PMA) led to rapid and sustained activation of MEK-1/-2, ERK-1/MAPK and ERK-2/MAPK, while induction of granulocytic differentiation by retinoic acid (RA) caused similar activation of MEK-1/-2 and ERK-2/MAPK, but not ERK-1/MAPK. The total levels of these kinases were not affected during the course of differentiation along either pathway. Pretreatment of cells with 5 microM of the MEK-1/-2-specific inhibitor U0126 abrogated PMA- or RA-induced activation of ERK-1/MAPK and ERK-2/MAPK. Importantly, pretreatment of HL-60 cells with U0126 was found to potently inhibit both monocytic and granulocytic differentiation, as assessed by cytochemical staining for non-specific esterase or nitroblue tetrazolium reduction, flow cytometric analysis of myeloid surface markers, and immunoblotting for the cell cycle inhibitor p21 WAF1/Cip1. Similar results were seen in U937 cells, where U0126 inhibited PMA-induced monocytic differentiation, and in 32D cells, where G-CSF-induced granulocytic differentiation was inhibited by U0126 pretreatment. Additional experiments revealed that inhibition of MEK-1/-2 in HL-60 cells resulted in nearly complete inhibition of differentiation-induced cell death during monocytic differentiation. By contrast, U0126 only partially inhibited cell death resulting from granulocytic differentiation. Taken together, our findings demonstrate that the MEK/ERK/MAP kinase signaling pathway is activated, and plays a critical role, during both monocytic and granulocytic differentiation of myeloid cell lines.


Subject(s)
Granulocytes/pathology , Leukemia, Myeloid/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/pathology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Butadienes/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Granulocytes/enzymology , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Monocytes/enzymology , Nitriles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology
2.
Clin Cancer Res ; 7(12): 4164-73, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751517

ABSTRACT

Previously we have shown that dexamethasone (DEX) enhances the antitumor activity and ligand binding of the active form of vitamin D, 1alpha,25-dihydroxyvitamin D(3) (1,25-D(3)), in the murine squamous cell carcinoma model SCC VII/SF. DEX also reduces the hypercalcemia toxicity of 1,25-D(3) treatment. However, the mechanism of the enhanced antitumor activity has not been defined. Here, we demonstrate that both cell cycle arrest and apoptosis were enhanced by DEX, effects that were inhibited by RU486. We also demonstrate that vitamin D receptor (VDR) protein levels were increased by the combination of 1,25-D(3) and DEX above the level observed with 1,25-D(3) treatment alone, whereas protein levels of the heterodimeric partner of VDR, retinoid X receptor, were lower for the combination than for 1,25-D(3) alone. Glucocorticoid receptor protein levels and ligand binding were increased by 1,25-D(3) but not by the combination. Treatment with the combination of 1,25-D(3) and DEX did not result in greater activation of a vitamin D response element-reporter than 1,25-D(3) alone or of a glucocorticoid response element-reporter than DEX alone. Nevertheless, the levels of phospho-Erk1/2 and phospho-Akt, signaling molecules that are modulated in 1,25-D(3)-treated squamous cell carcinoma cells, were reduced by the combination of 1,25-D(3) and DEX more than by either agent alone. These trends were also observed in vivo. Our results suggest the involvement of the Erk and Akt signaling pathways in the antiproliferative effects of the combination of 1,25-D(3) and DEX and that phospho-Erk1/2 and phospho-Akt may be useful markers of response to this combination.


Subject(s)
Apoptosis/drug effects , Calcitriol/pharmacology , Cell Cycle/drug effects , Dexamethasone/pharmacology , MAP Kinase Signaling System/physiology , Receptor Cross-Talk/physiology , Animals , Dose-Response Relationship, Drug , Drug Synergism , Kinetics , Mice , Mice, Inbred C3H , Mifepristone/pharmacology , Tumor Cells, Cultured
3.
J Biol Chem ; 276(28): 26365-73, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11331275

ABSTRACT

Vitamin D(3) inhibits cell growth and induces apoptosis in several human cancer lines in vitro and in vivo. However, little is known about the molecular events involved in vitamin D(3)-induced apoptosis. Here, we demonstrate that the growth-promoting/pro-survival signaling molecule mitogen-activated protein kinase kinase (MEK) is cleaved in a caspase-dependent manner in murine squamous cell carcinoma (SCC) cells induced to undergo apoptosis by treatment with vitamin D(3). Cleavage resulted in nearly complete loss of full-length MEK and ERK1/2 phosphorylation. ERK1/2 expression was affected only slightly. The phosphorylation and expression of Akt, a kinase regulating a second cell survival pathway, was also inhibited after treatment with vitamin D(3). However, the pro-apoptotic signaling molecule MEKK-1 was up-regulated in both apoptotic and non-apoptotic cells with greater induction and partial N-terminal proteolysis of MEKK-1 observed in apoptotic cells. In contrast to vitamin D(3), cisplatin and etoposide down-regulated Akt levels only modestly, did not promote significant loss of MEK expression, and did not up-regulate MEKK-1. We propose that vitamin D(3) induces apoptosis in SCC cells by a unique mechanism involving selective caspase-dependent MEK cleavage and up-regulation of MEKK-1. Additional evidence is provided that vitamin D(3)-induced apoptosis may be mediated via p38 MAPK.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cholecalciferol/pharmacology , MAP Kinase Kinase Kinase 1 , Animals , Carcinoma, Squamous Cell/metabolism , Caspases/metabolism , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Cells, Cultured , Up-Regulation
4.
J Pharmacol Exp Ther ; 289(2): 781-90, 1999 May.
Article in English | MEDLINE | ID: mdl-10215653

ABSTRACT

The chimeric oncogene bcr-abl is detected in virtually every case of chronic myelogenous leukemia. It has been shown that cells (such as K562) expressing Bcr-Abl/p210, a protein tyrosine kinase, not only undergo cellular transformation but also demonstrate multiple drug resistance. Recent studies also demonstrate that the proteasome is involved in the survival signaling pathway(s). In the current study, we tested the hypothesis that the proteasome might play a role in regulating Bcr-Abl function. We have demonstrated by using a variety of inhibitors that inhibition of the proteasome, but not of the cysteine protease, activity is able to activate the apoptotic cell death program in K562 cells. Proteasome inhibition-induced apoptosis is demonstrated by condensation and fragmentation of nuclei, appearance of an apoptotic population with sub-G1 DNA content, the internucleosomal fragmentation of DNA, and cleavage of poly(ADP-ribose) polymerase, and can be blocked by a specific caspase-3-like tetrapeptide inhibitor. Western blot analysis with specific antibodies to c-Abl and Bcr proteins show that treatment of K562 cells with a proteasome inhibitor results in significant reduction of Bcr-Abl protein expression, which occurs several hours before the onset of apoptotic execution. Levels of c-Abl/p145 and Bcr/p160 proteins, however, remain essentially unaltered at that time. Furthermore, reduced Bcr-Abl expression is reflected in significantly attenuated Bcr-Abl-mediated protein tyrosine phosphorylation. Taken together, these results indicate that proteasome inhibition is sufficient to inactivate Bcr-Abl function and subsequently activate the apoptotic death program in cells that are resistant to apoptosis induced by chemotherapy.


Subject(s)
Apoptosis , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Fusion Proteins, bcr-abl/biosynthesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Multienzyme Complexes/metabolism , Blotting, Western , Caspases/metabolism , Caspases/physiology , Cell Nucleus/ultrastructure , DNA Fragmentation , Enzyme Activation/physiology , Flow Cytometry , Humans , Leupeptins/pharmacology , Proteasome Endopeptidase Complex , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured
5.
J Biol Chem ; 272(21): 13484-8, 1997 May 23.
Article in English | MEDLINE | ID: mdl-9153192

ABSTRACT

Recently, we have designed farnesyltransferase and geranylgeranyltransferase I inhibitors (FTI-277 and GGTI-298) that selectively block protein farnesylation and geranylgeranylation, respectively. In this study, we describe the opposing effects of these inhibitors on interleukin-1beta (IL-1beta)-stimulated induction of nitric-oxide synthase-2 (NOS-2) in rat pulmonary artery smooth muscle cells (RPASMC) and rat hepatocytes. Pretreatment of cells with GGTI-298 caused a superinduction of NOS-2 by IL-1beta. RPASMC treated with GGTI-298 (10 microM) prior to IL-1beta (10 ng/ml) expressed levels of NOS-2 protein five times higher than those exposed to IL-1beta alone. This superinduction of NOS-2 protein by pretreatment with GGTI-298 resulted in nitrite concentrations in the medium that were 5-fold higher at 10 ng/ml IL-1beta and 10-fold higher at 1 ng/ml IL-1beta. Furthermore, NOS-2 mRNA levels in RPASMC were also increased 6- and 14-fold (at 10 and 1 ng/ml IL-1beta, respectively) when the cells were pretreated with GGTI-298. In contrast, treatment of cells with the inhibitor of protein farnesylation, FTI-277 (10 microM), blocked IL-1beta-induced NOS-2 expression at mRNA and protein levels. Pretreatment with lovastatin, an inhibitor of protein prenylation, resulted in superinduction of NOS-2. This superinduction was reversed by geranylgeraniol, but not by farnesol, further confirming that inhibition of geranylgeranylation, not farnesylation, is responsible for enhanced NOS-2 expression. The results demonstrate that a farnesylated protein(s) mediates IL-1beta induction of NOS-2, whereas a geranylgeranylated protein(s) represses this induction.


Subject(s)
Alkyl and Aryl Transferases , Enzyme Inhibitors/pharmacology , Interleukin-1/pharmacology , Muscle, Smooth, Vascular/enzymology , Nitric Oxide Synthase/biosynthesis , Protein Prenylation , Transferases/antagonists & inhibitors , Animals , Benzamides/pharmacology , Enzyme Induction/drug effects , Farnesyltranstransferase , GTP-Binding Proteins/metabolism , Liver/metabolism , Male , Methionine/analogs & derivatives , Methionine/pharmacology , Muscle, Smooth, Vascular/drug effects , Nitrites/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , rap GTP-Binding Proteins , ras Proteins/metabolism
6.
Oncogene ; 14(3): 305-12, 1997 Jan 23.
Article in English | MEDLINE | ID: mdl-9018116

ABSTRACT

Oncogenic H-Ras requires farnesylation for its transforming activity. Lovastatin inhibits both protein farnesylation and geranylgeranylation by decreasing cellular pools of farnesylpyrophosphate (FPP) and geranylgeranylpyrophosphate (GGPP), respectively. Use of lovastatin as a chemotherapeutic agent has been precluded by its significant cytotoxic effects. In this report, we describe a novel approach utilizing a combination of lovastatin and geranylgeraniol (GGOH) to potentiate the ability of lovastatin to block oncogenic H-Ras signaling and concomitantly rescue lovastatin toxicity. GGOH co-treatment with lovastatin enhances inhibition of oncogenic H-Ras processing and constitutive activation of mitogen-activated protein kinase (MAPK), and preserves the processing of geranylgeranyltransferase (GGTase) I and GGTase II protein substrates. Moreover, co-treatment with GGOH significantly (15-fold) attenuates the cytotoxic effects of lovastatin as well as prevents lovastatin-induced cell rounding. These results demonstrate that GGOH potentiates the anti-oncogenic/anti-signaling activity of lovastatin while antagonizing its cytotoxicity. These opposing effects are due to a GGOH metabolite that serves simultaneously as a potent inhibitor for farneslyltransferase as well as a substrate for GGTases I and II.


Subject(s)
Alkyl and Aryl Transferases , Antineoplastic Agents/pharmacology , Diterpenes/pharmacology , Genes, ras/drug effects , Lovastatin/pharmacology , Protein Kinases/biosynthesis , 3T3 Cells , Animals , Diterpenes/metabolism , Drug Synergism , Enzyme Activation/drug effects , Mice , Signal Transduction , Transferases/metabolism
7.
Oncogene ; 13(9): 1991-9, 1996 Nov 07.
Article in English | MEDLINE | ID: mdl-8934546

ABSTRACT

In order to assess the relative contributions of farnesylated and/or geranylgeranylated proteins on cell cycle progression from G1 to S phase we designed potent and selective farnesyltransferase (FTI-277) and geranylgeranyltransferase-I (GGTI-298) inhibitors. Flow cytometry studies showed that treatment of NIH3T3 cells with GGTI-298 or lovastatin, which inhibits both protein farnesylation and geranylgeranylation, arrested cells in G0/G1 whereas cells treated with FTI-277 progressed normally through the cell cycle. [3H]thymidine incorporation studies showed that mevalonate and geranylgeraniol, but not farnesol, released the lovastatin G1 block. Furthermore, mevalonate release of the lovastatin G1 block was inhibited by GGTI-298 but not by FTI-277. These results demonstrate that geranylgeranylated proteins are required for cells to proceed from G1 to S phase, and that farnesylated proteins do not play an essential role in the G1 to S phase transition


Subject(s)
Alkyl and Aryl Transferases , G1 Phase/physiology , Protein Prenylation/physiology , S Phase/physiology , Actins/drug effects , Actins/ultrastructure , Animals , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , DNA/biosynthesis , DNA/drug effects , Diterpenes/metabolism , Enzyme Inhibitors/pharmacology , Farnesol/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , G1 Phase/drug effects , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Genes, ras , Lovastatin/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Mevalonic Acid/pharmacology , Mice , Protein Prenylation/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/physiology , S Phase/drug effects , Transferases/antagonists & inhibitors , rap GTP-Binding Proteins , ras Proteins/drug effects , ras Proteins/metabolism
8.
J Biol Chem ; 271(44): 27402-7, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8910319

ABSTRACT

We have used specific inhibitors for farnesyltransferase (FTase) and geranylgeranyltransferase (GGTase) I as well as combinations of lovastatin with geranylgeraniol (GGOH) or farnesol (FOH) to investigate the role of protein prenylation in platelet-derived growth factor (PDGF)-induced PDGF receptor tyrosine phosphorylation. NIH-3T3 cells treated with the highly specific FTase inhibitor FTI-277 had no effect on PDGF receptor tyrosine phosphorylation or PDGF activation of mitogen-activated protein kinase (MAPK) at doses that completely inhibit FTase-dependent processing. In contrast, treatment of these cells with GGTase I inhibitor GGTI-298 strongly inhibited receptor tyrosine phosphorylation, and co-treatment with FTI-277 had no additional effect. Interestingly, the inhibitory effect of GGTI-298 on PDGF activation of MAPK was only partial. Furthermore, although lovastatin, which inhibits both protein geranylgeranylation and protein farnesylation, blocked PDGF receptor tyrosine phosphorylation, co-treatment with GGOH, but not FOH, reversed the lovastatin block. In addition, although lovastatin was observed to block MAPK activation by PDGF, co-treatment with GGOH, but not FOH, restored its activation. Further investigations indicated that inhibition of receptor tyrosine phosphorylation was not due to decreased expression of the receptor or to inhibition of GGTase II. Thus, these results demonstrate that PDGF receptor tyrosine phosphorylation requires protein geranylgeranylation but not protein farnesylation and that the tyrosine phosphorylation levels of the receptor are modulated by a protein that is a substrate for GGTase I.


Subject(s)
Alkyl and Aryl Transferases , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Transferases/metabolism , 3T3 Cells , Animals , Diterpenes/pharmacology , Enzyme Inhibitors/pharmacology , Farnesol/pharmacology , Farnesyltranstransferase , Kinetics , Lovastatin/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Phosphorylation , Phosphotyrosine , Protein Prenylation , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection , Transferases/antagonists & inhibitors
9.
Arch Biochem Biophys ; 326(2): 233-7, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8611028

ABSTRACT

Lovastatin, a cholesterol biosynthesis inhibitor, has recently been shown to inhibit mitogenesis and tumor growth. We have investigated the effects of lovastatin on the activation of MAP kinase by insulin using as a model HIRcB cells, a rat fibroblast cell line that overexpresses the human insulin receptor. Treatment with lovastatin (1-30 microM) for 24 h decreased the level of activation of MAP kinase by insulin by as much as 60%. Immunoblotting experiments using a specific anti-MAP kinase monoclonal antibody demonstrated that the amount of MAP kinase protein in the cells was not altered by lovastatin treatment. Likewise, lovastatin had no apparent effects on the expression of the insulin receptor. Treatment with lovastatin (20 microM) reduced the percentage of farnesylated Ras by 50%. Immunoprecipitation of tyrosine phosphorylated proteins from HIRcB cell lysates followed by immunodetection of MAP kinase using specific antibodies demonstrated a reduced level of insulin-induced tyrosine phosphorylation levels of MAP kinase in lovastatin-treated cells. Furthermore, immunodetection of the beta-subunit of the insulin receptor in anti-phosphotyrosine immunoprecipitates revealed that treatment with lovastatin reduced the tyrosine phosphorylation levels of the receptor. Lysates obtained from cells treated with increasing concentrations of lovastatin demonstrated a dose-dependent inhibition of the insulin-induced tyrosine phosphorylation of the receptor. Treatment with mevalonic acid prevented the effects of lovastatin demonstrating that the effects of the drug are a consequence of its inhibitory effects on the synthesis of steroids. It is concluded that, in addition to inhibition of Ras farnesylation, lovastatin reduces receptor tyrosine phosphorylation levels which also contributes to the blockade of MAPK activation by the insulin receptor.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Insulin/pharmacology , Lovastatin/pharmacology , Animals , Cell Line , Enzyme Activation/drug effects , Fibroblasts , Humans , Mevalonic Acid/pharmacology , Mitogen-Activated Protein Kinase 1 , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/metabolism , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Tyrosine/metabolism , ras Proteins/metabolism
10.
Biochem Biophys Res Commun ; 214(1): 295-303, 1995 Sep 05.
Article in English | MEDLINE | ID: mdl-7669049

ABSTRACT

Cysteine farnesylation of the Ras carboxyl terminal tetrapeptide CAAX motif (where C = cysteine, A = leucine, isoleucine, or valine, and X = methionine or serine) is required for Ras biological activity. In this report, we describe the effects of inhibitors of farnesyltransferase (FTase), the enzyme responsible for this lipid modification, on platelet-derived growth factor (PDGF) signaling in NIH-3T3 cells. In vitro, the CAAX peptidomimetic FTI-232 exhibits potent inhibition of FTase activity (IC50 = 150 nM) and its carboxyl-methylated counterpart, FTI-244, inhibits Ras processing in vivo. Treatment of NIH-3T3 cells with FTI-244 inhibits PDGF-induced DNA synthesis but not stimulation of mitogen-activated protein kinase (MAPK). However, FTI-244 significantly reduces PDGF-induced tyrosine phosphorylation levels of PDGF receptor (PDGFR) as well as its association with, and activation of, phosphatidylinositol-3-kinase (PI-3-K), a key enzyme in PDGF-induced mitogenesis.


Subject(s)
Alkyl and Aryl Transferases , Oligopeptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Tyrosine/metabolism , 3T3 Cells , Animals , DNA Replication , Enzyme Activation , Farnesyltranstransferase , Mice , Molecular Mimicry , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational , Signal Transduction , Transferases/antagonists & inhibitors , Transferases/metabolism , ras Proteins/metabolism
11.
Biochem Biophys Res Commun ; 204(1): 399-406, 1994 Oct 14.
Article in English | MEDLINE | ID: mdl-7524501

ABSTRACT

The mechanism by which lovastatin lowers cholesterol levels is well characterized but little is known about its anti-mitogenic and anti-tumorigenic mechanism. Here we demonstrate that lovastatin disrupts early events in the mitogenic signaling pathways of insulin. Insulin treatment (200 mM) of quiescent HIR rat-1 fibroblasts results in an 8-fold stimulation of phosphatidylinositol-3-kinase (PI-3-K) activity. Overnight pretreatment of cells with lovastatin (20 microM) inhibits insulin stimulation of PI-3-K activity by 75%. Immunoprecipitation and immunoblotting experiments using antibodies against the regulatory subunit of PI-3-K (p85), phosphotyrosine, and insulin receptor alpha and beta subunits demonstrate that lovastatin inhibits the association of p85 with tyrosine phosphorylated insulin receptor substrate-1 and the beta subunit of the insulin receptor. Furthermore, lovastatin dramatically reduces (70-100%) the level of tyrosine phosphorylated insulin receptor beta subunit following insulin stimulation. These results clearly demonstrate that lovastatin disrupts early events of insulin mitogenic signaling by reducing the levels of tyrosine phosphorylated beta subunit and suggest that this disruption is a potential mechanism for the anti-mitogenic effect of lovastatin.


Subject(s)
Insulin/pharmacology , Lovastatin/pharmacology , Receptor, Insulin/physiology , Signal Transduction/drug effects , Animals , Cell Line , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Insulin Receptor Substrate Proteins , Kinetics , Macromolecular Substances , Phosphatidylinositol 3-Kinases , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotyrosine , Rats , Receptor, Insulin/biosynthesis , Signal Transduction/physiology , Transfection , Tyrosine/analogs & derivatives , Tyrosine/metabolism
12.
J Biol Chem ; 268(30): 22227-30, 1993 Oct 25.
Article in English | MEDLINE | ID: mdl-7693661

ABSTRACT

Lovastatin inhibits mitogenesis in cultured cells and growth of tumors in vivo by unknown mechanism(s). Phosphatidylinositol 3-kinase (PI-3-kinase) is a putative second messenger-generating enzyme whose physical association with the platelet-derived growth factor receptor (PDGFR) has been demonstrated to be required for the mitogenic activity of PDGF in cultured fibroblasts. Here we examine the effect of lovastatin on PDGF- and insulin-stimulated PI-3-kinase activity. In quiescent NIH-3T3 cells, PDGF (25 ng/ml) and insulin (200 nM) stimulate PI-3-kinase activity 10- and 6-fold, respectively. However, overnight pretreatment of cells with 10 microM lovastatin inhibits this stimulation of PI-3-kinase activity by PDGF and insulin. Immunoprecipitation of the PI-3-kinase p85 subunit demonstrates a PDGF-dependent association of PI-3-kinase with the tyrosine-autophosphorylated PDGFR. However, upon exposure of cells to 10 microns lovastatin for 40 h, the level of autophosphorylated PDGFR associating with PI-3-kinase after PDGF stimulation decreases significantly (75% reduction). No change in the expression of the PI-3-kinase p85 subunit was observed after treatment of cells with lovastatin. These results demonstrate that lovastatin disrupts a major growth factor signaling pathway and that inhibition of PDGF-induced association of PI-3-kinase with PDGFR and subsequent inhibition of PI-3-kinase activity is one potential mechanism by which lovastatin inhibits cell growth.


Subject(s)
Lovastatin/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptors, Platelet-Derived Growth Factor/metabolism , Tyrosine/analogs & derivatives , 3T3 Cells , Adenosine Triphosphate/metabolism , Animals , Fibroblasts/drug effects , Fibroblasts/metabolism , Insulin/pharmacology , Kinetics , Macromolecular Substances , Mice , Phosphatidylinositol 3-Kinases , Phosphotyrosine , Platelet-Derived Growth Factor/antagonists & inhibitors , Protein Binding , Tyrosine/metabolism
13.
Arch Biochem Biophys ; 268(1): 161-75, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2783543

ABSTRACT

Many liver processes are sexually dimorphic. In particular, the microsomal content of specific enzymes and the synthesis of specific proteins are under sex steroid hormone control. Because the liver of male rats is strikingly androgen responsive, we sought evidence for an androgen receptor in this tissue. We detected and characterized both cytosolic and nuclear androgen-binding proteins. Both forms bind [3H]R1881 (methyltrienolone, 17 beta-hydroxy-17 alpha-methyl-4,9,11-estratriene-3-one) with the high affinity, low capacity, and specificity for androgens and antiandrogens characteristic of androgen receptors. No high-affinity binding of [3H]DHT could be detected in unfractionated cytosol because of the rapid metabolism of this ligand; however, binding of a DHT metabolite to the high-capacity male-specific estrogen binder (MEB) of cytosol was observed. Both gel filtration and heparin-Sepharose affinity chromatography separate the cytosolic androgen receptor from MEB. Incubation of cytosol in the absence of sodium molybdate resulted in androgen-binding activity which was retained by DNA-cellulose. Castration of male rats results in a time-dependent loss of both cytosolic and nuclear androgen binding, as well as a loss in MEB activity. Androgen-binding activity is low in livers from female rats, but can be induced by testosterone treatment. An intact pituitary is necessary for maintenance of androgen-binding activity, as hypophysectomy results in complete loss of activity.


Subject(s)
Carrier Proteins/metabolism , Liver/metabolism , Receptors, Androgen/metabolism , Receptors, Estrogen , Animals , Carrier Proteins/isolation & purification , Cell Nucleus/metabolism , Cytosol/metabolism , Dihydrotestosterone/metabolism , Estradiol/metabolism , Estrenes/metabolism , Female , Hypophysectomy , Kinetics , Male , Metribolone , Orchiectomy , Ovariectomy , Rats , Rats, Inbred Strains , Receptors, Androgen/isolation & purification , Reference Values , Sex Characteristics , Testosterone Congeners/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...