Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Diabetes Obes Metab ; 14(8): 709-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22340428

ABSTRACT

AIM: G-protein coupled receptor agonists are currently under investigation for their potential utility in patients with type 2 diabetes mellitus (T2DM). The objective was to determine the pharmacokinetics, pharmacodynamics, safety and tolerability of GPR119 agonist, JNJ-38431055 in T2DM subjects. METHODS: This was a randomized, double-blind, placebo- and positive-controled, single-dose cross-over study and a randomized, double-blind, placebo-controled multiple-dose parallel design study. The study was conducted at 4 US research centres. Two different experiments involving 25 and 32 different subjects were performed in male and female subjects, aged 25-60 years, mean body mass index between 22 and 39.9 kg/m2 who had T2DM diagnosed 6 months to 10 years before screening. JNJ-38431055 (100 and 500 mg) or sitagliptin (100 mg) as a single-dose or JNJ-38431055 (500 mg) once daily for 14 consecutive days were tested. Effects on stimulated plasma glucose, insulin, C-peptide and incretin concentrations were pre-specified outcomes. RESULTS: JNJ-38431055 was well tolerated and not associated with hypoglycaemia. Plasma systemic exposure of JNJ-38431055 increased as the dose increased, was approximately two-fold greater after multiple-dose administration, and attained steady-state after approximately 8 days. Compared with placebo, single-dose administration of oral JNJ-38431055 decreased glucose excursion during an oral glucose tolerance test, but multiple-dose administration did not alter 24-h weighted mean glucose. Multiple dosing of JNJ-38431055 increased post-meal total glucagon-like peptide 1 and gastric insulinotropic peptide concentrations compared to baseline. CONCLUSIONS: These studies provide evidence of limited glucose lowering and incretin activity for JNJ-38431055 in subjects with T2DM.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/blood , Incretins/blood , Pyrazines/pharmacology , Receptors, G-Protein-Coupled/agonists , Triazoles/pharmacology , Administration, Oral , Adult , Blood Glucose/drug effects , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Double-Blind Method , Female , Glucagon-Like Peptide 1/drug effects , Glucose Tolerance Test , Humans , Male , Middle Aged , Pyrazines/administration & dosage , Sitagliptin Phosphate , Treatment Outcome , Triazoles/administration & dosage
2.
Clin Pharmacol Ther ; 90(5): 685-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21975348

ABSTRACT

The incidence of type 2 diabetes mellitus is increasing worldwide. Several G-protein-coupled receptor agonists are being studied for their efficacy as antidiabetes agents. JNJ-38431055 is a novel, potent, and orally available selective agonist of the glucose-dependent insulinotropic (GPR119) receptor. Double-blind, randomized, placebo-controlled studies were conducted to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of single oral doses of JNJ-38431055 (2.5-800 mg) in healthy male volunteers. The systemic exposure of JNJ-38431055 in plasma increased in proportion to the dose and was not influenced by coadministration of food. The terminal elimination half-life was ~13 h when administered as an oral suspension formulation. JNJ-38431055 was well tolerated and was not associated with hypoglycemia. As compared with placebo, single-dose oral JNJ-38431055 increased postmeal plasma glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and peptide YY (PYY) concentrations but did not significantly decrease glucose excursion or increase insulin secretion. However, in a graded glucose infusion study, JNJ-38431055 was shown to induce a higher insulin secretion rate (ISR) relative to placebo at elevated plasma glucose levels. These studies provide evidence for the potential efficacy of JNJ-38431055 as an antidiabetes agent in humans.


Subject(s)
Blood Glucose/drug effects , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Adult , Double-Blind Method , Gastric Inhibitory Polypeptide/blood , Gastric Inhibitory Polypeptide/drug effects , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/drug effects , Half-Life , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Insulin/metabolism , Insulin Secretion , Male , Middle Aged , Peptide YY/blood , Peptide YY/drug effects
3.
Diabetes ; 47(8): 1243-52, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9703324

ABSTRACT

The insulin receptor (IR) is expressed by insulin-secreting beta-cells, but its cellular function is unknown. We transfected betaTC6-F7 beta-cells with cDNAs encoding either wild-type or mutant kinase-inactive (A/K1018) IRs, and by fluorescence-activated cell sorting generated polyclonal beta-cell lines that overexpressed each receptor type at levels two- to fourfold higher than the parental cells. Beta-cells overexpressing wild-type IRs had a proportional increase in insulin-stimulated tyrosine kinase activity; no change occurred in beta-cells expressing the mutant IR. We observed a threefold increase in cellular insulin content in beta-cells that overexpressed the wild-type IR, as determined by radioimmunoassay. This increase occurred despite a fivefold elevated rate of both basal and 10 mmol/l glucose-induced insulin secretion. Fractional insulin secretion (percentage of total cell insulin releasable at 10 mmol/l glucose) was unchanged in beta-cells overexpressing the wild-type IR compared with the parental beta-cell line. Insulin content and insulin secretion were unaffected by overexpression of kinase-inactive IRs. Steady-state insulin mRNA levels were elevated twofold in the beta-cells overexpressing the wild-type IR and unchanged in the beta-cells expressing the kinase-inactive receptor, as determined by Northern blot analysis. The rate of insulin mRNA degradation measured in the presence of 5 microg/ml actinomycin D was not significantly affected in either cell line. In the absence of glucose, the basal level of (pro)insulin biosynthesis in the beta-cells overexpressing the wild-type IR increased significantly (61%) compared with the beta-cells transfected with the kinase-inactive IR. These data indicate that IR signaling can regulate insulin gene transcription and can modulate the steady-state insulin content of beta-cells.


Subject(s)
Autocrine Communication/physiology , Gene Expression/physiology , Insulin/physiology , Islets of Langerhans/physiology , Receptor, Insulin/physiology , Signal Transduction/physiology , Animals , Cell Line , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Transfection/physiology
4.
Biochem J ; 318 ( Pt 2): 623-9, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8809056

ABSTRACT

The physiological role of the beta-cell insulin receptor is unknown. To evaluate a candidate function, the insulin regulation of fluid-phase pinocytosis was investigated in a clonal insulinoma cell line (beta TC6-F7) and, for comparison, also in Chinese hamster ovary cells transfected with the human insulin receptor (CHO-T cells). In CHO-T cells, the net rate of fluid-phase pinocytosis was rapidly increased 3-4-fold over the basal rate by 100 nM insulin, with half-maximal stimulation at 2 nM insulin, as assayed by cellular uptake of horseradish peroxidase from the medium. Wortmannin, an inhibitor of phosphatidylinositol (PI)-3-kinase, blocked insulin-stimulated pinocytosis with an IC50 of 7.5 nM without affecting the basal rate of pinocytosis. In insulin-secreting beta TC6-F7 cells, the secretagogues glucose and carbachol (at maximally effective concentrations of 15 mM and 0.5 mM respectively) augmented fluid-phase pinocytosis 1.65-fold over the basal rate. Wortmannin also inhibited secretagogue-stimulated pinocytosis in these beta-cells with an IC50 of 7 nM but did not affect the basal rate of pinocytosis measured in the absence of secretagogues. Wortmannin did not influence either basal or secretagogue-induced insulin secretion. Although these beta TC6-F7 cells have cell-surface insulin receptors, adding exogenous insulin or insulin-like growth factor 1 did not affect their rate of fluid-phase pinocytosis, either in the absence or presence of secretagogues. From these observations, we conclude that: (1) in both insulin-secreting beta-cells and in conventional, insulin-responsive CHO-T cells, a common, wortmannin-sensitive reaction, which probably involves PI-3-kinase, regulates fluid-phase pinocytosis; (2) the insulin-receptor signal transduction pathway is dissociated from the regulation of fluid-phase pinocytosis in the insulin-secreting beta-cell line we studied; and (3) the enhancement of fluid-phase pinocytosis associated with secretagogue-induced insulin release in beta TC6-F7 cells is not attributable to autocrine activation of beta-cell surface insulin receptors.


Subject(s)
Insulin/metabolism , Insulin/pharmacology , Islets of Langerhans/physiology , Pinocytosis/physiology , Receptor, Insulin/physiology , Androstadienes/pharmacology , Animals , CHO Cells , Carbachol/pharmacology , Clone Cells , Cricetinae , Glucose/pharmacology , Humans , Insulin Antagonists/pharmacology , Insulin Secretion , Insulinoma , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Pancreatic Neoplasms , Pinocytosis/drug effects , Receptor, Insulin/biosynthesis , Recombinant Proteins/metabolism , Transfection , Wortmannin
5.
Diabetes ; 45(7): 854-62, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8666133

ABSTRACT

Glucose is the primary stimulus for insulin secretion by pancreatic beta-cells, and it triggers membrane depolarization and influx of extracellular Ca2+. Cholinergic agonists amplify insulin release by several pathways, including activation of phospholipase C, which hydrolyzes membrane polyphosphoinositides. A novel phospholipid, phosphatidylinositol 3,4,5- trisphosphate [PtdIns(3,4,5)P3], a product of phosphatidylinositol 3-kinase (PI 3-kinase), has recently been found in various cell types. We demonstrate by immunoblotting that PI 3-kinase is present in both cytosolic and membrane fractions of insulin-secreting beta-TC3 cells and in rat islets. The catalytic activity of PI 3-kinase in immunoprecipitates of islets and beta-TC3 cells was measured by the production of radioactive phosphatidylinositol 3-monophosphate from phosphatidylinositol (PtdIns) in the presence of [gamma-32P]ATP. Wortmannin, a fungal metabolite, dose dependently inhibited PI 3-kinase activity of both islets and beta-TC3 cells, with an IC50 of 1 nmol/l and a maximally effective concentration of 100 nmol/l, when it was added directly to the kinase assay. However, if intact islets were incubated with wortmannin and PI 3-kinase subsequently was determined in islet immunoprecipitates, approximately 50% inhibition of PI 3-kinase activity (but no inhibition of glucose- and carbachol-stimulated insulin secretion) from intact islets was obtained at wortmannin concentrations of 100 nmol/l. Wortmannin, at higher concentrations (1 and 10 micromol/l), inhibited glucose- and carbachol-induced insulin secretion of Intact rat islets by 58 and 92%, respectively. Wortmannin had no effect on the basal insulin release from rat islets. A similar dose curve of inhibition of glucose- and carbachol-induced insulin secretion by wortmannin was obtained when beta-TC3 cells were used. Cellular metabolism was, not changed by any wortmannin concentrations tested (0.01-10 micromol/l). Both basal cytosolic [Ca2+]i and carbamyl choline-induced increases of [Ca2]i were unaffected by wortmannin in the presence of 2.5 mmol/l Ca2+, while Ca2+ mobilization from intracellular stores was partially decreased by wortmannin. Together, these data suggest that wortmannin at concentrations that inhibit PI 3-kinase does not affect insulin secretion. PI 3-kinase is unlikely to have a major role in insulin secretion induced by glucose and carbachol.


Subject(s)
Androstadienes/pharmacology , Enzyme Inhibitors/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Chloride/pharmacology , Carbachol/pharmacology , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cytosol/metabolism , Dose-Response Relationship, Drug , Insulin Secretion , Islets of Langerhans/drug effects , Kinetics , Male , Phosphatidylinositol 3-Kinases , Rats , Rats, Sprague-Dawley , Wortmannin
6.
Diabetes ; 45(6): 711-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8635642

ABSTRACT

The expression of insulin receptor mRNA was examined in rat pancreatic islet cells by single-cell reverse transcriptase (RT)-polymerase chain reaction (PCR). Single cells from disaggregated islets were individually isolated in a microcapillary pipet, and the beta-cells were identified by amplification of the mRNA for insulin. We found that in single beta-cells, the mRNA for the insulin receptor was also expressed. The fraction of single islet cells expressing both insulin receptor and insulin mRNAs corresponds closely to the fraction of beta-cells in the disaggregated islet cell preparation. These results indicate that normal beta-cells have the potential to express authentic insulin receptors. Immunohistochemical analysis was insufficiently sensitive for assaying insulin receptor protein; however, insulin receptor substrate 1 (IRS-1) was readily immunolocalized in islet beta-cells. Since IRS-1 links several cell surface receptors, including those for insulin and IGF-I, to distal signal transduction pathways, our observations indicate that hormonal regulation of islet beta-cells potentially involves the same signal transduction pathway that mediates insulin and growth factor signaling in peripheral insulin target tissue cell types.


Subject(s)
Islets of Langerhans/metabolism , Phosphoproteins/biosynthesis , Receptor, Insulin/biosynthesis , Transcription, Genetic , Animals , Base Sequence , Cells, Cultured , DNA Primers , Immunohistochemistry , Insulin Receptor Substrate Proteins , Islets of Langerhans/cytology , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction
8.
Diabetes ; 44(7): 802-9, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7540574

ABSTRACT

In the beta TC3 insulin-secreting beta-cell line, glucose rapidly induces the tyrosine phosphorylation of the 97-kDa insulin receptor beta-subunit. Phosphorylation is transient, with fourfold stimulation by 2 min and subsequent dephosphorylation to basal levels by 10-15 min. Elevating the extracellular KCl concentration equipotently initiates receptor phosphorylation. Preventing insulin secretion with 1 mumol/l epinephrine or by removing extracellular Ca2+ blocks the effect. In the absence of glucose-induced secretion, exogenous insulin also stimulated insulin receptor autophosphorylation transiently and with an ED50 of 4 x 10(-9) mol/l. In addition, functional insulin-like growth factor I (IGF-I) receptors are also expressed by these beta-cells, as indicated by IGF-I-induced receptor tyrosine phosphorylation (ED50 = 5 x 10(-9) mol/l) and also by detection of hybrid insulin/IGF-I receptor autophosphorylation at 10(-7) mol/l IGF-I. Both glucose and insulin stimulate the tyrosine phosphorylation of the insulin receptor substrate (IRS) IRS-1 and increase by two- to fivefold the rapid association of IRS-1 with the 85-kDa alpha-subunit of the phosphatidylinositol-3-kinase, as determined by co-immunoprecipitation assays. These results demonstrate that in these beta-cells, glucose-induced insulin secretion activates the beta-cell surface insulin receptor tyrosine kinase and its intracellular signal transduction pathway, suggesting a new autocrine mechanism for the regulation of beta-cell function.


Subject(s)
Glucose/pharmacology , Insulin/metabolism , Insulin/pharmacology , Islets of Langerhans/metabolism , Receptor, Insulin/metabolism , Tyrosine/analogs & derivatives , Animals , Calcium/pharmacology , Cell Line , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Insulin Receptor Substrate Proteins , Insulin Secretion , Insulin-Like Growth Factor I/pharmacology , Islets of Langerhans/drug effects , Kinetics , Phosphatidylinositol 3-Kinases , Phosphoproteins/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotyrosine , Potassium Chloride/pharmacology , Receptor, Insulin/drug effects , Tyrosine/analysis , Tyrosine/metabolism
9.
J Clin Invest ; 90(5): 1839-49, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1331176

ABSTRACT

Insulin rapidly stimulates tyrosine phosphorylation of a protein of approximately 185 kD in most cell types. This protein, termed insulin receptor substrate-1 (IRS-1), has been implicated in insulin signal transmission based on studies with insulin receptor mutants. In the present study we have examined the levels of IRS-1 and the phosphorylation state of insulin receptor and IRS-1 in liver and muscle after insulin stimulation in vivo in two rat models of insulin resistance, i.e., insulinopenic diabetes and fasting, and a mouse model of non-insulin-dependent diabetes mellitus (ob/ob) by immunoblotting with anti-peptide antibodies to IRS-1 and anti-phosphotyrosine antibodies. As previously described, there was an increase in insulin binding and a parallel increase in insulin-stimulated receptor phosphorylation in muscle of fasting and streptozotocin-induced (STZ) diabetic rats. There was also a modest increase in overall receptor phosphorylation in liver in these two models, but when normalized for the increase in binding, receptor phosphorylation was decreased, in liver and muscle of STZ diabetes and in liver of 72 h fasted rats. In the hyperinsulinemic ob/ob mouse there was a decrease in insulin binding and receptor phosphorylation in both liver and muscle. The tyrosyl phosphorylation of IRS-1 after insulin stimulation reflected an amplification of the receptor phosphorylation in liver and muscle of hypoinsulinemic animals (fasting and STZ diabetes) with a twofold increase, and showed a significant reduction (approximately 50%) in liver and muscle of ob/ob mouse. By contrast, the levels of IRS-1 protein showed a tissue specific regulation with a decreased level in muscle and an increased level in liver in hypoinsulinemic states of insulin resistance, and decreased levels in liver in the hyperinsulinemic ob/ob mouse. These data indicate that: (a) IRS-1 protein levels are differentially regulated in liver and muscle; (b) insulin levels may play a role in this differential regulation of IRS-1; (c) IRS-1 phosphorylation depends more on insulin receptor kinase activity than IRS-1 protein levels; and (d) reduced IRS-1 phosphorylation in liver and muscle may play a role in insulin-resistant states, especially of the ob/ob mice.


Subject(s)
Insulin Resistance , Liver/metabolism , Muscles/metabolism , Protein-Tyrosine Kinases/physiology , Proteins/metabolism , Receptor, Insulin/metabolism , Animals , CHO Cells , Cricetinae , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Fasting , Insulin/pharmacology , Male , Mice , Mice, Obese , Phosphorylation , Rats
10.
J Biol Chem ; 267(6): 3946-51, 1992 Feb 25.
Article in English | MEDLINE | ID: mdl-1310987

ABSTRACT

Genistein, an isoflavone putative tyrosine kinase inhibitor, was used to investigate the coupling of insulin receptor tyrosine kinase activation to four metabolic effects of insulin in the isolated rat adipocyte. Genistein inhibited insulin-stimulated glucose oxidation in a concentration-dependent manner with an ID50 of 25 micrograms/ml and complete inhibition at 100 micrograms/ml. Genistein also prevented insulin's (10(-9) M) inhibition of isoproterenol-stimulated lipolysis with an ID50 of 15 micrograms/ml and a complete effect at 50 micrograms/ml. The effect of genistein (25 micrograms/ml) was not reversed by supraphysiological (10(-7) M) insulin levels. In contrast, genistein up to 100 micrograms/ml had no effect on insulin's (10(-9) M) stimulation of either pyruvate dehydrogenase or glycogen synthase activity. We determined whether genistein influenced insulin receptor beta-subunit autophosphorylation or tyrosine kinase substrate phosphorylation either in vivo or in vitro by anti-phosphotyrosine immunoblotting. Genistein at 100 micrograms/ml did not inhibit insulin's (10(-7) M) stimulation of insulin receptor tyrosine autophosphorylation or tyrosine phosphorylation of the cellular substrates pp185 and pp60. Also, genistein did not prevent insulin-stimulated autophosphorylation of partially purified human insulin receptors from NIH 3T3/HIR 3.5 cells or the phosphorylation of histones by the activated receptor tyrosine kinase. In control experiments using either NIH 3T3 fibroblasts or partially purified membranes from these cells, genistein did inhibit platelet-derived growth factor's stimulation of its receptor autophosphorylation. These findings indicate the following: (a) Genistein can inhibit certain responses to insulin without blocking insulin's stimulation of its receptor tyrosine autophosphorylation or of the receptor kinase substrate tyrosine phosphorylation. (b) In adipocytes genistein must block the stimulation of glucose oxidation and the antilipolytic effects of insulin at site(s) downstream from the insulin receptor tyrosine kinase. (c) The inhibitory effects of genistein on hormonal signal transduction cannot necessarily be attributed to inhibition of tyrosine kinase activity, unless specifically demonstrated.


Subject(s)
Adipose Tissue/drug effects , Insulin Antagonists/pharmacology , Insulin/pharmacology , Isoflavones/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Insulin/drug effects , Adipose Tissue/cytology , Animals , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Genistein , Glucose/metabolism , Glycogen Synthase/metabolism , Isoproterenol/pharmacology , Lipolysis/drug effects , Male , Oxidation-Reduction , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Inbred Strains , Signal Transduction/drug effects
11.
J Biol Chem ; 266(13): 8302-11, 1991 May 05.
Article in English | MEDLINE | ID: mdl-2022647

ABSTRACT

Insulin stimulates the tyrosine phosphorylation of a 185-kDa putative cytosolic substrate protein (pp185) in diverse cell types. After intravenous insulin infusion into the live intact rat, pp185 and the 95-kDa insulin receptor beta-subunit were the major proteins that tyrosine phosphorylated in liver, skeletal muscle, and adipose tissue. Both proteins were maximally phosphorylated within 30 s, and both increased in phosphotyrosine content in parallel with increasing insulin dose. However, pp185 tyrosine phosphorylation was transient, with almost complete dephosphorylation within 2-3 min despite continued insulin stimulation. To identify pp185 directly, we purified pp185 from insulin-stimulated rat liver, using a denaturation-based extraction procedure that blocks endogenous protein phosphatases and thus allows a high yield, single step isolation of phosphotyrosyl proteins by anti-phosphotyrosine antibody immunoaffinity absorption. From 50 rat livers, 50-100 pmol of pp185 was isolated. Edman degradation of seven internal tryptic peptide fragments of pp185 yielded novel amino acid sequences, indicating that pp185 is a new protein. Antipeptide antibodies were raised which specifically recognize a single, 185-kDa insulin-stimulated phosphotyrosyl protein in liver, skeletal muscle, adipose tissue, and several cultured cell lines. These results indicate that pp185 is expressed in a variety of insulin-responsive tissues, is the major protein rapidly tyrosine phosphorylated under physiological conditions in the intact animal, and also provide a route for cloning the pp185 gene and elucidating the function of pp185 in insulin signal transduction.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Receptor, Insulin/metabolism , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Insulin/metabolism , Male , Molecular Sequence Data , Peptide Mapping , Phosphorylation , Precipitin Tests , Proteins/chemistry , Proteins/isolation & purification , Rats , Rats, Inbred Strains
12.
J Biol Chem ; 265(18): 10226-31, 1990 Jun 25.
Article in English | MEDLINE | ID: mdl-2354998

ABSTRACT

Phosphorylation of the insulin receptor beta-subunit on serine/threonine residues by protein kinase C reduces both receptor kinase activity and insulin action in cultured cells. Whether this mechanism regulates insulin action in intact animals was investigated in rats rendered insulin-resistant by 3 days of starvation. Insulin-stimulated autophosphorylation of the partially purified hepatic insulin receptor beta-subunit was decreased by 45% in starved animals compared to fed controls. This autophosphorylation defect was entirely reversed by removal of pre-existing phosphate from the receptor with alkaline phosphatase, suggesting that increased basal phosphorylation on serine/threonine residues may cause the decreased receptor tyrosine kinase activity. Tryptic removal of a C-terminal region of the receptor beta-subunit containing the Ser/Thr phosphorylation sites similarly normalized receptor autophosphorylation. To investigate which kinase(s) may be responsible for such increased Ser/Thr phosphorylation in vivo, protein kinase C and cAMP-dependent protein kinase A in liver were studied. A 2-fold increase in protein kinase C activity was found in both cytosol and membrane extracts from starved rats as compared to controls, while protein kinase A activity was diminished in the cytosol of starved rats. A parallel increase in protein kinase C was demonstrated by immunoblotting with a polyclonal antibody which recognizes several protein kinase C isoforms. These findings suggest that in starved, insulin-resistant animals, an increase in hepatic protein kinase C activity is associated with increased Ser/Thr phosphorylation which in turn decreases autophosphorylation and function of the insulin receptor kinase.


Subject(s)
Liver/metabolism , Protein Kinase C/metabolism , Receptor, Insulin/metabolism , Animals , Cell Membrane/enzymology , Cytosol/metabolism , Kinetics , Phosphorylation , Protein Kinases/metabolism , Rats , Receptor, Insulin/isolation & purification , Reference Values , Starvation
13.
J Biol Chem ; 263(30): 15546-52, 1988 Oct 25.
Article in English | MEDLINE | ID: mdl-3139671

ABSTRACT

Insulin modifies cellular responsiveness to some hormones which operate via guanine nucleotide binding proteins (G-proteins); also, G-proteins have been implicated in some actions of insulin. Using pertussis toxin-catalyzed [32P]ADP-ribosylation of Gi as an index of G-protein conformation, we evaluated interaction of insulin receptors with G-proteins. In isolated rat liver plasma membranes, insulin treatment for 10 min inhibited [32P]ADP-ribosylation of Gi by 50%. This effect was half-maximal at 2 x 10(-8) M. A similar effect was observed with rat adipocyte plasma membranes with half-maximal effect at 1 x 10(-8) M. Pertussis toxin activity itself was uninfluenced by insulin, as ribosylation of tubulin or heat-treated bovine serum albumin was unaltered. Elevated Mg2+ diminished basal ADP-ribosylation, but insulin inhibition occurred at all Mg2+ levels between 0 and 1 mM. Insulin inhibition was independent of ATP (20 microM to 10 mM), and GTP (0-100 microM) concentrations. Because both protein kinase C and purified insulin receptor phosphorylate purified Gi in vitro, we examined Gi as a substrate for the insulin receptor tyrosine kinase in vivo. Triton-extracts of isolated rat hepatocytes which had been 32Pi labeled and treated with insulin were immunoprecipitated with a polyclonal anti-Gi antiserum. The dominant labeled phosphoprotein had a molecular weight of 42 kDa, consistent with the alpha-subunit of Gi, contained only phosphoserine, and was unaffected in its phosphorylation by insulin. These results indicate the existence of a novel pathway for physiological "cross-talk" between insulin and other hormones and further suggests that the insulin receptor may interact with regulatory G-proteins via biochemical mechanisms not directly involving the tyrosine kinase activity of the insulin receptor.


Subject(s)
GTP-Binding Proteins/metabolism , Insulin/pharmacology , Pertussis Toxin , Poly(ADP-ribose) Polymerases/metabolism , Receptor, Insulin/metabolism , Virulence Factors, Bordetella/pharmacology , Adenosine Triphosphate/pharmacology , Adipose Tissue/metabolism , Animals , Cell Membrane/metabolism , Liver/metabolism , Magnesium/pharmacology , Magnesium Chloride , Male , Molecular Weight , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...