Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters








Publication year range
1.
Horm Metab Res ; 44(9): 650-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22674476

ABSTRACT

Adenosine monophosphate-activated protein kinase (AMPK), silent mating type information regulation 2 homologue 1 (SIRT 1), and peroxisome proliferator-activated receptor γ co-activator α (PGC1α) constitute an energy sensing cellular network that controls mitochondrial biogenesis. Caloric restriction activates both AMPK and SIRT-1 to increase ATP production from fat oxidation. We characterized AMPK and SIRT 1 expression and activity in human skeletal muscle in response to dietary fat or carbohydrate intake on the background of either overfeeding or caloric restriction. AMPK phosphorylation and acetylation of PGC1α (as a measure of SIRT activity) were determined. Euglycemic-hyperinsulinemic clamp and muscle biopsies were performed in human subjects participating in 2 separate studies. In study 1, 21 lean healthy individuals were overfed for 5 days, while in study 2, 18 obese otherwise healthy individuals consumed a calorie-restricted diet for 5 days. Under both conditions - overfeeding and caloric restriction - high fat/low carbohydrate (HF/LC) diet significantly increased phosphorylation of AMPK and deacetylation of PGC1α in skeletal muscle without affecting total amounts of AMPK, PGC1α, or SIRT 1. In contrast, low fat/high carbohydrate (LF/HC) hypocaloric diet reduced phosphorylation of AMPK and deacetylation of PGC1α. Our data indicate that a relative deficiency in carbohydrate intake or, albeit less likely, a relative excess of fat intake even in the absence of caloric deprivation is sufficient to activate the AMPK-SIRT 1-PGC1α energy-sensing cellular network in human skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/genetics , Dietary Carbohydrates/analysis , Dietary Fats/analysis , Muscle, Skeletal/enzymology , Obesity/diet therapy , Obesity/enzymology , Sirtuin 1/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Caloric Restriction , Dietary Carbohydrates/metabolism , Dietary Fats/metabolism , Female , Gene Expression , Humans , Male , Obesity/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation , Sirtuin 1/metabolism
2.
Horm Metab Res ; 41(10): 757-61, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19598077

ABSTRACT

Phosphoinositide 3-kinase is a key signaling intermediate necessary for the metabolic actions of insulin. In this study, we assessed the effects of in vivo knockdown of the p85alpha subunit of phosphoinositide 3-kinase on insulin sensitivity, using an antisense oligonucleotide, in lean mice, diet-induced obese mice, and obese leptin-deficient Lep (ob/ob) mice. Mice were injected with either p85alpha-targeted antisense oligonucleotide or saline twice weekly for 4 weeks. Fasting levels of glycemia and insulinemia and insulin and glucose tolerance tests were used to determine insulin sensitivity. Western blot analysis and real-time polyacrylamide chain reaction were used to assess p85alpha protein and mRNA expression. IN VIVO administration of antisense oligonucleotide resulted in 50 and 60% knockdown of liver p85alpha protein and mRNA, respectively, in the lean, diet-induced obese and Lep (ob/ob) mice. This was associated with increased phosphoinositide 3-kinase activity and improved insulin sensitivity in diet-induced obese and Lep (ob/ob) mice. Thus, p85alpha could be an important therapeutic target to ameliorate insulin resistance.


Subject(s)
Insulin Resistance/physiology , Obesity/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blood Glucose/analysis , Blotting, Western , Glucose Tolerance Test , Insulin/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinases/genetics , RNA , Reverse Transcriptase Polymerase Chain Reaction
3.
Diabetologia ; 49(4): 748-54, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16491394

ABSTRACT

AIMS/HYPOTHESIS: We sought to define early molecular alterations associated with nutritionally induced insulin resistance in humans. METHODS: Insulin sensitivity was assessed using a hyperinsulinaemic-euglycaemic clamp in eight healthy women while on an isocaloric diet and after 3 days of overfeeding (50% above eucaloric diet). Expression of phosphatidylinositol (PI) 3-kinase subunits p85alpha and p110 was assessed and measurements were made of IRS-1-associated PI 3-kinase activity, tyrosine and serine phosphorylation of IRS-1, and serine and threonine phosphorylation of p70S6 kinase. Measurements were made in skeletal muscle biopsies obtained before and after overfeeding. RESULTS: Three days of overfeeding resulted in a reduction of insulin sensitivity accompanied by: (1) increased expression of skeletal muscle p85alpha; (2) an alteration in the ratio of p85alpha to p110; (3) a decrease in the amount of IRS-1-associated p110; and (4) a decrease in PI 3-kinase activity. Increases in expression of p85alpha and in the p85alpha:p110 ratio demonstrated a highly significant inverse correlation with insulin sensitivity, and changes in PI 3-kinase activity correlated with changes in insulin sensitivity. Tyrosine and serine phosphorylation of IRS-1 and serine and threonine phosphorylation of p70S6 kinase were unaffected by 3 days of overfeeding. CONCLUSIONS/INTERPRETATION: We identified a novel mechanism of nutritionally induced insulin resistance in healthy women of normal weight. We conclude that increased expression of p85alpha may be one of the earliest molecular alterations in the mechanism of the insulin resistance associated with overfeeding.


Subject(s)
Insulin Resistance , Nutritional Physiological Phenomena , Phosphatidylinositol 3-Kinases/metabolism , Up-Regulation , Adult , Female , Glucose/metabolism , Humans , Phosphorylation , Protein Subunits/metabolism , Time Factors
4.
Int J Obes Relat Metab Disord ; 27(1): 40-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12532152

ABSTRACT

OBJECTIVE: To investigate whether the expression of a dominant negative (DN) farnesyl- and geranygeranyl-transferase I (FTase/GGTase I) alpha-subunit in 3T3-L1 pre-adipocytes can inhibit insulin's ability to induce differentiation. DESIGN: 3T3-L1 pre-adipocytes were stably transfected with vector alone or vector expressing a mutated DN FTase/GGTase I alpha-subunit (S60A)(S62A) and incubated in serum-free medium in the absence and presence of insulin. MEASUREMENTS: Various assays were performed to determine the effect of DN FTase/GGTase I alpha-subunit expression in 3T3-L1 pre-adipocyte on insulin-induced DNA synthesis, cell count, phosphorylation of the FTase/GGTase I alpha-subunit, FTase and GGTase I activity, amounts of prenylated p21Ras and RhoA, phosphorylation of MAP kinase and Akt, and differentiation to mature fat cells. RESULTS: Expression of DN FTase/GGTase I alpha-subunit inhibited insulin's ability to increase DNA synthesis, cell count, FTase and GGTase I activity, amounts of prenylated p21Ras and RhoA, and magnitude of phosphorylation of MAP kinase. Expression of DN FTase/GGTase I alpha-subunit in 3T3-L1 pre-adipocytes was without effect on insulin-induced Akt phosphorylation. CONCLUSION: Expression of DN FTase/GGTase I alpha-subunit inhibits insulin-induced differentiation of 3T3-L1 pre-adipocytes to mature adipocytes, and thus could indicate potential therapeutic avenues to assuage the deleterious effects of obesity and type 2 diabetes.


Subject(s)
Adipocytes/drug effects , Alkyl and Aryl Transferases/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , 3T3 Cells/drug effects , Adipocytes/cytology , Adipocytes/enzymology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/physiology , Diabetes Mellitus, Type 2/metabolism , Farnesyltranstransferase , Humans , Mice , Phosphorylation , Proto-Oncogene Proteins p21(ras)/metabolism , rhoA GTP-Binding Protein/metabolism
5.
J Biol Chem ; 276(41): 38023-8, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11500498

ABSTRACT

We recently demonstrated that in MCF-7 breast cancer cells, insulin promoted the phosphorylation and activation of geranylgeranyltransferase I (GGTI-I), increased the amounts of geranylgeranylated Rho-A and potentiated the transactivating activity of lysophosphatidic acid (LPA) (Chappell, J., Golovchenko, I., Wall, K., Stjernholm, R., Leitner, J., Goalstone, M., and Draznin, B. (2000) J. Biol. Chem. 275, 31792-31797). In the present study, we explored the mechanism of this potentiating effect of insulin on LPA. Insulin (10 nm) potentiated the ability of LPA to stimulate cell cycle progression and DNA synthesis in MCF-7 cells. The potentiating effect of insulin appears to involve increases in the expression of cyclin E and decreases in the expression of the cyclin-dependent kinase inhibitor p27Kip1. All potentiating effects of insulin were inhibited in the presence of an inhibitor of GGTase I, GGTI-286 (3 microm) or by an expression of a dominant negative mutant of Rho-A. In contrast to its potentiating action, a direct mitogenic effect of insulin in MCF-7 cells involves activation of phosphatidylinositol 3-kinase and increased expression of cyclin D1. We conclude that the ability of insulin to increase the cellular amounts of geranylgeranylated Rho-A results in potentiation of the LPA effect on cyclin E expression and degradation of p27Kip1 and cell cycle progression in MCF-7 breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle/drug effects , Insulin/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Division/drug effects , Cyclin D , Cyclin E/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , DNA Replication/drug effects , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Lysophospholipids/pharmacology , Phosphorylation , Tumor Cells, Cultured
6.
Am J Physiol Endocrinol Metab ; 281(2): E217-23, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11440896

ABSTRACT

Even though the role of fetal hyperinsulinemia in the pathogenesis of fetal macrosomia in patients with overt diabetes and gestational diabetes mellitus seems plausible, the molecular mechanisms of action of hyperinsulinemia remain largely enigmatic. Recent indications that hyperinsulinemia "primes" various tissues to the mitogenic influence of growth factors by increasing the pool of prenylated Ras proteins prompted us to investigate the effect of fetal hyperinsulinemia on the activitiy of farnesyltransferase (FTase) and the amounts of farnesylated p21 Ras in fetal tissues in the ovine experimental model. Induction of fetal hyperinsulinemia by direct infusion of insulin into the fetus and by either fetal or maternal infusions of glucose resulted in significant increases in the activity of FTase and the amounts of farnesylated p21 Ras in fetal liver, skeletal muscle, fat, and white blood cells. An additional infusion of somatostatin into hyperglycemic fetuses blocked fetal hyperinsulinemia and completely prevented these increases, specifying insulin as the causative factor. We conclude that the ability of fetal hyperinsulinemia to increase the size of the pool of farnesylated p21 Ras may prime fetal tissues to the action of other growth factors and thereby constitute one mechanism by which fetal hyperinsulinemia could induce macrosomia in diabetic pregnancies.


Subject(s)
Fetal Diseases/metabolism , Hyperinsulinism/metabolism , Protein Prenylation/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Adipose Tissue/drug effects , Adipose Tissue/embryology , Adipose Tissue/metabolism , Alkyl and Aryl Transferases/metabolism , Animals , Disease Models, Animal , Farnesyltranstransferase , Female , Fetal Diseases/chemically induced , Fetal Weight/drug effects , Fetus , Glucose/administration & dosage , Hyperinsulinism/chemically induced , Infusions, Intravenous , Insulin , Leukocytes/drug effects , Leukocytes/metabolism , Liver/drug effects , Liver/embryology , Liver/metabolism , Maternal-Fetal Exchange , Muscle, Skeletal/drug effects , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Pregnancy , Sheep , Somatostatin/administration & dosage
7.
J Biol Chem ; 276(30): 28430-5, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11375992

ABSTRACT

Insulin is a potent adipogenic hormone that triggers an induction of a series of transcription factors governing differentiation of pre-adipocytes into mature adipocytes. However, the exact link between the insulin signaling cascade and the intrinsic cascade of adipogenesis remains incompletely understood. Herein we demonstrate that inhibition of prenylation of p21ras and Rho-A arrests insulin-stimulated adipogenesis. Inhibition of farnesylation of p21ras also blocked the ability of insulin to activate mitogen-activated protein (MAP) kinase and cyclic AMP response element-binding (CREB) protein. Expression of two structurally different inducible constitutively active CREB constructs rescued insulin-stimulated adipocyte differentiation from the inhibitory influence of prenylation inhibitors. Constitutively active CREB constructs induced expression of PPARgamma2, fatty acid synthase, GLUT-4, and leptin both in control and prenylation inhibitors-treated cells. It appears that insulin-stimulated prenylation of the Ras family GTPases assures normal phosphorylation and activation of CREB that, in turn, triggers the intrinsic cascade of adipogenesis.


Subject(s)
Adipocytes/cytology , Cyclic AMP Response Element-Binding Protein/metabolism , Insulin/metabolism , Muscle Proteins , Proto-Oncogene Proteins p21(ras)/metabolism , Adipocytes/metabolism , Animals , Blotting, Western , Cell Differentiation , Cell Line , Enzyme Activation , Fatty Acid Synthases/biosynthesis , Fibroblasts/metabolism , Glucose Transporter Type 4 , Leptin/biosynthesis , MAP Kinase Signaling System , Mice , Models, Biological , Monosaccharide Transport Proteins/biosynthesis , Phosphorylation , Protein Prenylation , Receptors, Cytoplasmic and Nuclear/biosynthesis , Recombinant Fusion Proteins/metabolism , Transcription Factors/biosynthesis , Transfection
8.
J Biol Chem ; 276(16): 12805-12, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11278505

ABSTRACT

We assessed the roles of insulin receptor substrate-1 (IRS-1) and Shc in insulin action on farnesyltransferase (FTase) and geranylgeranyltransferase I (GGTase I) using Chinese hamster ovary (CHO) cells that overexpress wild-type human insulin receptors (CHO-hIR-WT) or mutant insulin receptors lacking the NPEY domain (CHO-DeltaNPEY) or 3T3-L1 fibroblasts transfected with adenoviruses that express the PTB or SAIN domain of IRS-1 and Shc, the pleckstrin homology (PH) domain of IRS-1, or the Src homology 2 (SH2) domain of Shc. Insulin promoted phosphorylation of the alpha-subunit of FTase and GGTase I in CHO-hIR-WT cells, but was without effect in CHO-DeltaNPEY cells. Insulin increased FTase and GGTase I activities and the amounts of prenylated Ras and RhoA proteins in CHO-hIR-WT (but not CHO-DeltaNPEY) cells. Overexpression of the PTB or SAIN domain of IRS-1 (which blocked both IRS-1 and Shc signaling) prevented insulin-stimulated phosphorylation of the FTase and GGTase I alpha-subunit activation of FTase and GGTase I and subsequent increases in prenylated Ras and RhoA proteins. In contrast, overexpression of the IRS-1 PH domain, which impairs IRS-1 (but not Shc) signaling, did not alter insulin action on the prenyltransferases, but completely inhibited the insulin effect on the phosphorylation of IRS-1 and on the activation of phosphatidylinositol 3-kinase and Akt. Finally, overexpression of the Shc SH2 domain completely blocked the insulin effect on FTase and GGTase I activities without interfering with insulin signaling to MAPK. These data suggest that insulin signaling from its receptor to the prenyltransferases FTase and GGTase I is mediated by the Shc pathway, but not the IRS-1/phosphatidylinositol 3-kinase pathway. Shc-mediated insulin signaling to MAPK may be necessary (but not sufficient) for activation of prenyltransferase activity. An additional pathway involving the Shc SH2 domain may be necessary to mediate the insulin effect on FTase and GGTase I.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Alkyl and Aryl Transferases/metabolism , Insulin/pharmacology , Proteins/metabolism , Receptor, Insulin/physiology , 3T3 Cells , Adenoviridae , Animals , CHO Cells , Cricetinae , Farnesyltranstransferase , GRB2 Adaptor Protein , Humans , Insulin Receptor Substrate Proteins , Kinetics , Mice , Phosphoproteins/metabolism , Phosphorylation , Protein Prenylation , Protein Subunits , Receptor, Insulin/genetics , Recombinant Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transfection , src Homology Domains
9.
J Biol Chem ; 275(41): 31792-7, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-10930411

ABSTRACT

We have shown previously that insulin promotes phosphorylation and activation of farnesyltransferase and geranylgeranyltransferase (GGTase) II. We have now examined the effect of insulin on geranylgeranyltransferase I in MCF-7 breast cancer cells. Insulin increased GGTase I activity 3-fold and augmented the amounts of geranylgeranylated Rho-A by 18%. Both effects of the insulin were blocked by an inhibitor of GGTase I, GGTI-286. The insulin-induced increases in the amounts of geranylgeranylated Rho-A resulted in potentiation of the Rho-A-mediated effects of lysophosphatidic acid (LPA) on a serum response element-luciferase construct. Preincubation of cells with insulin augmented the LPA-stimulated serum response element-luciferase activation to 12-fold, compared with just 6-fold for LPA alone (p < 0.05). The potentiating effect of insulin was dose-dependent, inhibited by GGTI-286 and not mimicked by insulin-like growth factor-1. We conclude that insulin activates GGTase I, increases the amounts of geranylgeranylated Rho-A protein, and potentiates the Rho-A-dependent nuclear effects of LPA in MCF-7 breast cancer cells.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Insulin/pharmacology , Lysophospholipids/pharmacology , rhoA GTP-Binding Protein/metabolism , Alkyl and Aryl Transferases/antagonists & inhibitors , Breast Neoplasms , DNA-Binding Proteins/physiology , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Genes, Reporter/genetics , Guanosine Triphosphate/metabolism , Humans , Hyperinsulinism/enzymology , Hyperinsulinism/metabolism , Insulin-Like Growth Factor I/pharmacology , Luciferases/genetics , Nuclear Proteins/physiology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Response Elements/genetics , Serum Response Factor , Transcriptional Activation/drug effects , Tumor Cells, Cultured
10.
Diabetologia ; 42(3): 310-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10096783

ABSTRACT

We have recently demonstrated that insulin activates farnesyltransferase (FTase) and thereby increases the amounts of cellular farnesylated p21Ras in 3T3-L1 fibroblasts, adipocytes and vascular smooth muscle cells. We postulated that hyperinsulinaemia might considerably increase the the cellular pool of farnesylated p21Ras available for activation by other growth factors. To examine the role of in vivo hyperinsulinaemia in regulating farnesylated p21Ras, we measured the amounts of farnesylated p21Ras in tissues of hyperinsulinaemic animals. Liver, aorta, and skeletal muscle of ob/ob mice, and mice made obese and hyperinsulinaemic by injection of gold-thioglucose contained greater amounts of farnesylated p21Ras than tissues of their lean normoinsulinaemic counterparts. Similarly, farnesylated p21Ras was increased (67 vs. 35 % in control animals, p<0.01) in the livers of hyperinsulinaemic Zucker rats (fa/fa). Reduction of hyperinsulinaemia by exercise training (2 h/day for 7-8 weeks) resulted in decreases in the amounts of farnesylated p21Ras in these animals. Increased farnesylated p21Ras in hyperinsulinaemic animals reflected increasing increments in the activity of FTase in ob/ob mice (2-fold increase) and fa/fa Zucker rats (3.5-fold increase), while the total amounts of Ras proteins remained unchanged. In contrast to insulin-resistant hyperinsulinaemic animals, denervated insulin-resistant rat soleus muscle (in the presence of normoinsulinaemia) showed normal amounts of farnesylated p21Ras. In summary, these data confirm increased amounts of farnesylated p21Ras in tissues of hyperinsulinaemic animals.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Hyperinsulinism/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Muscle, Smooth, Vascular/metabolism , Obesity/metabolism , Protein Prenylation , Proto-Oncogene Proteins p21(ras)/metabolism , 3T3 Cells , Animals , Aurothioglucose , Blood Glucose/metabolism , Body Weight/drug effects , Body Weight/physiology , Clenbuterol/pharmacology , Farnesyltranstransferase , Female , Hyperinsulinism/chemically induced , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle Denervation , Muscle, Skeletal/innervation , Obesity/genetics , Obesity/physiopathology , Physical Conditioning, Animal , Protein Prenylation/drug effects , Rats , Rats, Zucker
11.
J Biol Chem ; 274(5): 2880-4, 1999 Jan 29.
Article in English | MEDLINE | ID: mdl-9915824

ABSTRACT

Rab proteins play a crucial role in the trafficking of intracellular vesicles. Rab proteins are GTPases that cycle between an inactive GDP-bound form and an active GTP-bound conformation. A prerequisite to Rab activation by GTP loading is its post-translational modification by the addition of geranylgeranyl moieties to highly conserved C-terminal cysteine residues. We examined the effect of insulin on the activity of geranylgeranyltransferase II (GGTase II) in 3T3-L1 fibroblasts and adipocytes. In fibroblasts, insulin increased the enzymatic activity of GGTase II 2.5-fold after 1 h of incubation, an effect that is blocked by perillyl alcohol, an inhibitor of prenyltransferases, but not by the geranylgeranyltransferase I inhibitor, GGTI-298, or the farnesyltransferase inhibitor, alpha-hydroxyfarnesylphosphonic acid. Concomitantly, insulin stimulated the phosphorylation of the GGTase II alpha-subunit without any effect on the GGTase II beta-subunit. At the same time, insulin also increased the amounts of geranylgeranylated Rab-3 in 3T3-L1 fibroblasts from 44 +/- 1.2% in control cells to 63 +/- 3.8 and 64 +/- 6.1% after 1 and 24 h of incubation, respectively. In adipocytes, insulin increased the amounts of geranylgeranylated Rab-4 from 38 +/- 0.6% in control cells to 56 +/- 1.7 and 60 +/- 2.6% after 1 and 24 h of incubation, respectively. In both fibroblasts and adipocytes, the presence of perillyl alcohol blocked the ability of insulin to increase geranylgeranylation of Rab-4, whereas GGTI-298 and alpha-hydroxyfarnesylphosphonic acid were without effect, indicating that insulin activates GGTase II. In summary, insulin promotes phosphorylation and activation of GGTase II in both 3T3 L1 fibroblasts and adipocytes and increases the amounts of geranylgeranylated Rab-3 and Rab-4 proteins.


Subject(s)
Alkyl and Aryl Transferases/metabolism , GTP-Binding Proteins/metabolism , Insulin/pharmacology , 3T3 Cells , Adipocytes/drug effects , Adipocytes/enzymology , Androstadienes/pharmacology , Animals , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/enzymology , Mice , Phosphorylation , Wortmannin , rab3 GTP-Binding Proteins , rab4 GTP-Binding Proteins
12.
Endocrinology ; 139(10): 4067-72, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9751484

ABSTRACT

Correlative studies have indicated that hyperinsulinemia is present in many individuals with atherosclerosis. Insulin resistance has also been linked to cardiovascular disease. It has proved to be difficult to decipher whether hyperinsulinemia or insulin resistance plays the most important role in the pathogenesis of atherosclerosis and coronary artery disease. In this study, we demonstrate that insulin increases the amount of farnesylated p21Ras in vascular smooth muscle cells (VSMC), thereby augmenting the pool of cellular Ras available for activation by platelet-derived growth factor (PDGF). In VSMC incubated with insulin for 24 h, PDGF's influence on GTP-loading of Ras was significantly increased. Furthermore, in cells preincubated with insulin, PDGF increased thymidine incorporation by 96% as compared with a 44% increase in control cells (a 2-fold increment). Similarly, preincubation of VSMC with insulin increased the ability of PDGF to stimulate gene expression of vascular endothelial growth factor 5- to 8-fold. The potentiating influence of insulin on PDGF action was abrogated in the presence of a farnesyltransferase inhibitor. Thus, the detrimental influence of hyperinsulinemia on the arterial wall may be related to the ability of insulin to augment farnesyltransferase activity and provide greater amounts of farnesylated p21Ras for stimulation by various growth promoting agents.


Subject(s)
Insulin/pharmacology , Muscle, Smooth, Vascular/drug effects , Platelet-Derived Growth Factor/pharmacology , Animals , Cells, Cultured , Drug Synergism , Endothelial Growth Factors/genetics , Insulin-Like Growth Factor I/biosynthesis , Lymphokines/genetics , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar , Swine , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
13.
J Biol Chem ; 273(37): 23892-6, 1998 Sep 11.
Article in English | MEDLINE | ID: mdl-9727002

ABSTRACT

We have previously demonstrated that insulin activates farnesyltransferase (FTase) and augments the amounts of farnesylated p21 (Goalstone, M. L., and Draznin, B. (1996) J. Biol. Chem. 271, 27585-27589). We postulated that this aspect of insulin action might explain the "priming effect" of insulin on the cellular response to other growth factors. In the present study, we show the specificity of the effect of insulin on FTase. Insulin, but not insulin-like growth factor-1 (IGF-1), epidermal growth factor (EGF), or platelet-derived growth factor (PDGF), stimulated the phosphorylation of the alpha-subunit of FTase and the amounts of farnesylated p21. Even though all four growth factors utilized the Ras pathway to stimulate DNA synthesis, only insulin used this pathway to influence FTase. Insulin failed to stimulate FTase in cells expressing the chimeric insulin/IGF-1 receptor and in cells derived from the insulin receptor knock-out animals. Insulin potentiated the effects of IGF-1, EGF, and PDGF on DNA synthesis in cells expressing the wild type insulin receptor, but this potentiation was inhibited in the presence of the FTase inhibitor, alpha-hydroxyfarnesylphosphonic acid. We conclude that the effect of insulin on FTase is specific, requires the presence of an intact insulin receptor, and serves as a conduit for the "priming" influence of insulin on the nuclear effects of other growth factors.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Epidermal Growth Factor/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor, IGF Type 1/physiology , Receptor, Insulin/physiology , 3T3 Cells , Animals , Cell Division , DNA/biosynthesis , Epidermal Growth Factor/physiology , Farnesyltranstransferase , Insulin/physiology , Insulin-Like Growth Factor I/physiology , Kinetics , Mice , Platelet-Derived Growth Factor/physiology , Protein Prenylation , Receptor, IGF Type 1/biosynthesis , Receptor, Insulin/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Transfection
14.
Endocrinology ; 138(12): 5119-24, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9389491

ABSTRACT

Farnesylation of p21Ras by farnesyltransferase (FTase) is obligatory for anchoring p21Ras to the plasma membrane, where it can be activated by growth factors. Insulin significantly stimulates the phosphorylation of the alpha-subunit of FTase (4-fold) and the enzymatic activity of FTase in 3T3-L1 fibroblasts and adipocytes. FTase activity was assessed by the amount of [3H] mevalonate (a precursor of farnesyl) incorporated into p21Ras in vivo and by quantitating the amount of farnesylated p21Ras before and after insulin administration. Insulin-stimulated phosphorylation of the alpha-subunit of FTase in 3T3-L1 fibroblasts and adipocytes was blocked by the mitogen-activated protein/extracellular-signal regulated kinase-kinase inhibitor, PD98059, but not by wortmannin or bisindolylmaleimide. Additionally, PD98059 blocked insulin-stimulated [3H]mevalonic incorporation and farnesylation of unprocessed p21Ras in both cell lines. Furthermore, expression of the dominant negative mutant of p21Ras precluded insulin-stimulated phosphorylation of the FTase alpha-subunit and activation of its enzymatic activity. In contrast, 3T3-L1 fibroblasts, expressing the constitutively active Raf-1, exhibited enhanced phosphorylation of the FTase alpha-subunit. It seems that insulin's effect on the phosphorylation and activation of FTase in both fibroblasts and adipocytes is mediated via the Ras pathway, resulting in a positive feedback augmentation of the cellular pool of farnesylated p21Ras.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Insulin/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase , Fibroblasts/metabolism , Flavonoids/pharmacology , Mevalonic Acid/antagonists & inhibitors , Mevalonic Acid/metabolism , Mice , Phosphorylation/drug effects
15.
Biochem Biophys Res Commun ; 239(1): 42-5, 1997 Oct 09.
Article in English | MEDLINE | ID: mdl-9345266

ABSTRACT

Insulin promotes the phosphorylation and activation of farnesyltransferase (FTase) in a time- and a dose-dependent manner. Increased FTase activity results in a larger pool of farnesylated p21Ras and allows for enhanced GTP loading. Insulin significantly increases the pool of farnesylated p21Ras from 20-25% in quiescent 3T3-L1 fibroblasts to approximately 70%, most of which is targeted to the plasma membrane. Furthermore, insulin promotes GTP loading of plasma membrane and not cytosolic p21Ras. The half-life of plasma membrane-associated farnesylated p21Ras is approximately 6 hours, and is identical in control and insulin-treated cells. We have also observed a direct correlation between the amounts of farnesylated p21Ras at the plasma membrane and the magnitude of insulin-induced GTP loading of p21Ras.


Subject(s)
Guanosine Triphosphate/metabolism , Insulin/pharmacology , Protein Prenylation , Proto-Oncogene Proteins p21(ras)/metabolism , 3T3 Cells , Alkyl and Aryl Transferases/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Farnesyltranstransferase , Guanosine Diphosphate/metabolism , Half-Life , Mice , Rats
16.
Endocrinology ; 138(5): 2211-4, 1997 May.
Article in English | MEDLINE | ID: mdl-9112423

ABSTRACT

Incubation of 3T3-L1 fibroblasts with insulin (10 nM or 100 nM) for 24 or 48 hours resulted in a significant increase in the amount of farnesylated p21Ras with a concomitant increase in the amount of GTP-loaded p21Ras. Cells preincubated with 100 nM insulin for 24 or 48 hours exhibited further 5-8 fold increases in p21Ras.GTP loading in response to an acute (10 minute) challenge with either insulin, EGF, or IGF-1. Effects of hyperinsulinemia were completely abolished by the presence of 1 microM alpha-hydroxyfarnesylphosphonic acid, a potent inhibitor of farnesyltransferase. These novel observations indicate that hyperinsulinemia increases the cellular pool of farnesylated p21Ras and thereby potentiates activation of p21Ras by growth factors.


Subject(s)
Alkyl and Aryl Transferases , Growth Substances/pharmacology , Insulin/pharmacology , Organophosphonates , Proto-Oncogene Proteins p21(ras)/metabolism , 3T3 Cells , Animals , Drug Synergism , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Farnesol/analogs & derivatives , Farnesol/pharmacology , Farnesyltranstransferase , Guanosine Triphosphate/metabolism , Insulin-Like Growth Factor I/pharmacology , Mice , Organophosphorus Compounds/pharmacology , Protein Prenylation , Transferases/antagonists & inhibitors
17.
Endocrinology ; 137(6): 2362-6, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8641187

ABSTRACT

Insulin-stimulated activity of Raf-1 kinase was examined in Rat-1 fibroblasts transfected with wild-type and mutant human insulin receptors. Insulin stimulated Raf-1 binding to p21Ras in HIRc (wild-type), delta CT (insulin receptor lacking a 43-amino acid C-terminal domain), and Y/F2 (tyrosine 1316 and 1322 replaced by phenylalanine) cells. Despite equal binding to p21Ras, the activity of Raf-1 kinase (measured by phosphorylation of its downstream substrate, mitogen-activated protein/extracellular receptor kinase (MEK) was significantly reduced in the delta CT cells. As an association of Raf-1 with p21Ras does not activate Raf-1 kinase, but merely targets Raf-1 to the plasma membrane, we examined the binding of Raf-1 to 14-3-3 proteins and to the insulin receptor itself. Raf-1 was detected in both 14-3-3 and insulin receptor immunoprecipitates. Association of Raf-1 with either 14-3-3 protein or insulin receptor was not influenced by insulin and was similar in all control and insulin-treated cell lines. These results indicate that the delta CT cells are deficient in stimulating Raf-1 activity despite normal binding of Raf-1 to p21Ras. Thus, an unidentified mechanism of Raf-1 activation at the plasma membrane must be impaired in these cells.


Subject(s)
Insulin/pharmacology , Protein Kinases/metabolism , Receptor, Insulin/metabolism , 3T3 Cells , Animals , Enzyme Activation/drug effects , Guanosine Triphosphate/metabolism , Humans , Immunosorbent Techniques , Mice , Mitogen-Activated Protein Kinase Kinases , Peptide Fragments/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Proto-Oncogene Proteins p21(ras)/metabolism
18.
Mol Cell Biol ; 16(4): 1450-7, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8657118

ABSTRACT

The role of phosphatidylinositol (PI) 3-kinase in specific aspects of insulin signaling was explored in 3T3-L1 adipocytes. Inhibition of PI 3-kinase activity by LY294002 or wortmannin significantly enhanced basal and insulin-stimulated GTPase-activating protein (GAP) activity in 3T3-L1 adipocytes. Furthermore, removal of the inhibitory influence of PI 3-kinase on GAP resulted in dose-dependent decreases in the ability of insulin to stimulate p21ras. This effect was specific to adipocytes, as inhibition of PI 3-kinase did not influence GAP in either 3T3-L1 fibroblasts, Rat-1 fibroblasts, or CHO cells. Immunodepletion of either of the two subunits of the PI 3-kinase (p85 or p110) yielded similar activation of GAP, suggesting that catalytic activity of p110 plays an important role in controlling GAP activity in 3T3-L1 adipocytes. Inhibition of PI 3-kinase activity in 3T3-L1 adipocytes resulted in abrogation of insulin-stimulated glucose uptake and thymidine incorporation. In contrast, effects of insulin on glycogen synthase and mitogen-activated protein kinase activity were inhibited only at higher concentrations of LY294002. It appears that in adipocytes, P1 3-kinase prevents activation of GAP. Inhibition of PI 3-kinase activity or immunodepletion of either one of its subunits results in activation of GAP and decreases in GTP loading of p21ras.


Subject(s)
Adipocytes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , 3T3 Cells , Animals , Blotting, Western , CHO Cells , Chromones/pharmacology , Cricetinae , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblasts , GTPase-Activating Proteins , Glucose/metabolism , Humans , Immunoblotting , Insulin/pharmacology , Mice , Mitogens/pharmacology , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Rats , ras GTPase-Activating Proteins
19.
J Biol Chem ; 271(14): 8338-44, 1996 Apr 05.
Article in English | MEDLINE | ID: mdl-8626530

ABSTRACT

To investigate the mechanisms underlying the enhanced mitogenic activity of the truncated epidermal growth factor receptor (EGFR) lacking the C-terminal autophosphorylation sites (Delta973-EGFR), we studied the intracellular signaling pathways in NR6 cells expressing human wild type EGFR and Delta973-EGFR. Microinjection of dominant/negative p21ras(N17) completely inhibited EGF-induced DNA synthesis in both cell types. EGF stimulated Shc phosphorylation as well as the formation of wild type EGFR.Shc complexes. In contrast, EGF stimulated Shc phosphorylation without formation of Delta973-EGFR.Shc complexes. Tyrosine-phosphorylated Shc formed complexes with Grb2.Sos, and microinjection of anti-Shc antibody and Shc-SH2 GST fusion protein inhibited EGF stimulation of DNA synthesis in both cell lines. EGF markedly increased ErbB2 tyrosine phosphorylation in wild type EGFR cells. In Delta973-EGFR cells, ErbB2 was tyrosine phosphorylated in the basal state and EGFR stimulated further phosphorylation of ErbB2. In addition to ErbB2, additional proteins were tyrosine phosphorylated in Delta973-EGFR cells, mostly in the molecular mass range of 120 170 kDa. Taken together with our findings indicating coupling of ErbB2 to Shc, these data suggest the importance of an alternative signaling pathway in Delta973-EGFR cells mediated by the formation of heterodimeric structures between the truncated EGFR and ErbB2, followed by coupling through Shc to Grb2.Sos and the p21ras pathway, ultimately leading to mitogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Cycle , ErbB Receptors/physiology , Receptor, ErbB-2/physiology , Animals , Cell Line , Cell Membrane/metabolism , Epidermal Growth Factor/physiology , GRB2 Adaptor Protein , Guanine Nucleotide Exchange Factors , Guanosine Diphosphate/metabolism , Humans , Macromolecular Substances , Membrane Proteins/metabolism , Mice , Phosphorylation , Phosphotyrosine/metabolism , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/physiology , Shc Signaling Adaptor Proteins , Signal Transduction , Son of Sevenless Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Structure-Activity Relationship , ras Guanine Nucleotide Exchange Factors
20.
Endocrinology ; 136(6): 2464-9, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7750468

ABSTRACT

Insulin's interaction with its receptor initiates a multitude of cellular effects on metabolism, growth, and differentiation. We recently described an insulin-mediated inhibition of nuclear protein phosphatase 2A (PP-2A), which is associated with an increase in phosphorylation of the transcription factor cAMP response element-binding protein. To clarify the role of nuclear PP-2A inhibition in the insulin signaling cascade, we examined the regulation of this phosphatase activity by insulin in Rat-1 fibroblasts overexpressing normal (HIRc) or mutant human insulin receptors (delta CT cells, deletion of a 43-amino acid C-terminal domain). The delta CT cells represent an excellent model of impaired metabolic and intact mitogenic action of insulin. Insulin inhibited nuclear PP-2A activity and enhanced cAMP response element-binding protein phosphorylation in HIRc cells, but not in delta CT cells. The delta CT cells exhibited normal ras activation and blunted mitogen-activating protein kinase phosphorylation and activation in response to insulin (16-fold in HIRc cells vs. 3-fold in delta CT cells), indicating that the mitogen-activating protein kinase pathway is important for the regulation of nuclear PP-2A activity by insulin. We conclude that insulin inhibits nuclear PP-2A activity, and that the carboxy-terminal domain of the insulin receptor is important for this effect.


Subject(s)
Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins , Insulin/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Receptor, Insulin/metabolism , Activating Transcription Factor 1 , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Protein Phosphatase 2 , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/genetics , Sequence Deletion , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL