Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 357
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Science ; 239(4845): 1269-71, 1988 Mar 11.
Article in English | MEDLINE | ID: mdl-2964085

ABSTRACT

The insulin-like growth factor II (IGF-II) is a polypeptide hormone with structural homologies to insulin and insulin-like growth factor I (IGF-I). In contrast to these other hormones, the in vivo function of IGF-II is not known. Although IGF-II can stimulate a broad range of biological responses in isolated cells, these responses have usually been found to be mediated by the insulin and IGF-I receptors. Recently, the receptor for IGF-II was found to also be the receptor for mannose-6-phosphate. Since this latter receptor has been implicated in targeting of lysosomal enzymes, the question is now raised of whether the same protein can also mediate metabolic responses to IGF-II.


Subject(s)
Carrier Proteins/metabolism , Hexosephosphates/metabolism , Insulin-Like Growth Factor II/metabolism , Mannosephosphates/metabolism , Receptor, Insulin/metabolism , Somatomedins/metabolism , Animals , Humans , Receptor, IGF Type 2 , Receptors, Somatomedin
2.
Science ; 219(4582): 299-301, 1983 Jan 21.
Article in English | MEDLINE | ID: mdl-6849137

ABSTRACT

Highly purified preparations of insulin receptor catalyzed the phosphorylation of the 95,000-dalton subunit of the insulin receptor. This subunit of the insulin receptor was also labeled with [alpha-32P]8-azidoadenosine 5'-triphosphate, a photoaffinity label for adenosine triphosphate binding sites. The identity of the 95,000-dalton band was confirmed in both cases by precipitation with a monoclonal antibody to the insulin receptor. These results suggest that the insulin receptor is itself a protein kinase.


Subject(s)
Protein Kinases/physiology , Receptor, Insulin/physiology , Adenosine Triphosphate/metabolism , Cell Line , Cells, Cultured , Lymphocytes , Molecular Weight , Phosphoproteins/physiology
3.
Science ; 242(4884): 1415-8, 1988 Dec 09.
Article in English | MEDLINE | ID: mdl-3059494

ABSTRACT

A proteinase with high affinity for insulin has been proposed to play a role in the cellular processing of this hormone. A complementary DNA (cDNA) coding for this enzyme has been isolated and sequenced. The deduced amino acid sequence of the enzyme contained the sequences of 13 peptides derived from the isolated protein. The cDNA could be transcribed in vitro to yield a synthetic RNA that in cell-free translations produced a protein that coelectrophoresed with the native proteinase and could be immunoprecipitated with monoclonal antibodies to this enzyme. The deduced sequence of this proteinase did not contain the consensus sequences for any of the known classes of proteinases (that is, metallo, cysteine, aspartic, or serine), but it did show homology to an Escherichia coli proteinase (called protease III), which also cleaves insulin and is present in the periplasmic space. Thus, these two proteins may be members of a family of proteases that are involved in intercellular peptide signaling.


Subject(s)
Endopeptidases/genetics , Escherichia coli/genetics , Insulysin/genetics , Metalloendopeptidases , Peptide Hydrolases/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA/genetics , Escherichia coli/enzymology , Genes , Humans , Molecular Sequence Data , Sequence Homology, Nucleic Acid
4.
Science ; 236(4803): 840-3, 1987 May 15.
Article in English | MEDLINE | ID: mdl-3554510

ABSTRACT

The oncogene protein product (p21) of the ras gene has been implicated in mediating the effects of a variety of growth factors and hormones. Microinjection of monoclonal antibody 6B7, which is directed against a synthetic peptide corresponding to a highly conserved region of p21 (amino acids 29 to 44) required for p21 function, specifically inhibited Xenopus oocyte maturation induced by incubation with insulin. The inhibition was dose-dependent and specific since (i) the same antibody had no effect on progesterone-induced maturation, (ii) immunoprecipitation and Western blotting indicated that the antibody recognized a single protein of molecular weight 21,000 in oocyte extracts, and (iii) inhibition was not observed with identical concentrations of normal immunoglobulin. Thus, p21 appears to be involved in mediating insulin-induced maturation of Xenopus oocytes. Furthermore, the mechanism may involve phosphorylation of p21, as p21 was found to be a substrate of the insulin receptor kinase.


Subject(s)
Insulin/pharmacology , Oncogenes , Oocytes/cytology , Proto-Oncogene Proteins/physiology , Animals , Antibodies , Female , Immunoglobulin G , Oocytes/drug effects , Progesterone/pharmacology , Proto-Oncogene Proteins/genetics , Xenopus laevis
5.
J Clin Invest ; 74(4): 1436-43, 1984 Oct.
Article in English | MEDLINE | ID: mdl-6090502

ABSTRACT

Insulin and the insulinlike growth factors (IGF-I and IGF-II) are members of a family of hormones that regulate the metabolism and growth of many tissues. Cultured HEP-G2 cells (a minimal deviation human hepatoma) have insulin receptors and respond to insulin by increasing their glycogen metabolism. In the present study with HEP-G2 cells, we used 125I-labeled insulin, IGF-I, and IGF-II to identify distinct receptors for each hormone by competition-inhibition studies. Unlabeled insulin was able to inhibit 125I-IGF-I binding but not 125I-IGF-II binding. A mouse monoclonal antibody to the human insulin receptor that inhibits insulin binding and blocks insulin action inhibited 75% of 125I-insulin binding, but inhibited neither 125I-IGF-I nor 125I-IGF-II binding. When glycogen metabolism was studied, insulin stimulated [3H]glucose incorporation into glycogen in a biphasic manner; one phase that was 20-30% of the maximal response occurred over 1-100 pM, and the other phase occurred over 100 pM-100 nM. The anti-receptor monoclonal antibody inhibited the first phase of insulin stimulation but not the second. Both IGF-I and IGF-II stimulated [3H]glucose incorporation over the range of 10 pM-10 nM; IGF-I was three to fivefold more potent. The monoclonal antibody, however, was without effect on IGF regulation of glycogen metabolism. Therefore, these studies indicate that insulin as well as the IGFs at physiological concentrations regulate glycogen metabolism in HEP-G2 cells. Moreover, this regulation of glycogen metabolism is mediated by both the insulin receptor and the IGF receptors.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Glycogen/metabolism , Insulin/physiology , Liver Neoplasms/metabolism , Peptides/physiology , Somatomedins/physiology , Animals , Antibodies, Monoclonal/physiology , Binding, Competitive , Cell Line , Glucose/metabolism , Humans , Insulin/metabolism , Mice , Peptides/metabolism , Receptor, Insulin/analysis , Receptor, Insulin/immunology , Receptors, Cell Surface/analysis , Receptors, Somatomedin , Somatomedins/metabolism
6.
J Clin Invest ; 93(3): 966-73, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8132782

ABSTRACT

Insulin-degrading enzyme (IDE) hydrolyzes both insulin and IGFs and has been proposed to play a role in signal termination after binding of these peptides to their receptors. In situ hybridization was used to investigate the cellular distribution of IDE mRNA and to compare it with insulin receptor (IR) and IGF-I receptor (IGFR) gene expression in serial thin sections from a variety of tissues in embryonic and adult rats. IDE mRNA is highly abundant in kidney and liver, tissues known to play a role in insulin degradation. IDE and IR mRNAs are highly coexpressed in brown fat and liver. The highest level IDE gene expression, on a per cell basis, is found in germinal epithelium. IDE and IGFR mRNAs are colocalized in oocytes, while IDE is colocalized with the IGF-II receptor in spermatocytes, suggesting that IDE may be involved with degradation of IGF-II in the testis. In summary, IDE expression demonstrates significant anatomical correlation with insulin/IGF receptors. These data are compatible with a role for IDE in degrading insulin and IGFs after they bind to and are internalized with their respective receptors and may also suggest a novel role for IDE in germ cells.


Subject(s)
Gene Expression Regulation , Insulin/metabolism , Receptor, Insulin/genetics , Receptors, Somatomedin/genetics , Animals , Embryo, Mammalian/metabolism , Female , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Somatomedins/metabolism
7.
Oncogene ; 19(2): 200-9, 2000 Jan 13.
Article in English | MEDLINE | ID: mdl-10644997

ABSTRACT

Loss of the tumor suppressor MMAC1 has been shown to be involved in breast, prostate and brain cancer. Consistent with its identification as a tumor suppressor, expression of MMAC1 has been demonstrated to reduce cell proliferation, tumorigenicity, and motility as well as affect cell-cell and cell-matrix interactions of malignant human glioma cells. Subsequently, MMAC1 was shown to have lipid phosphatase activity towards PIP3 and protein phosphatase activity against focal adhesion kinase (FAK). The lipid phosphatase activity of MMAC1 results in decreased activation of the PIP3-dependent, anti-apoptotic kinase, AKT. It is thought that this inhibition of AKT culminates with reduced glioma cell proliferation. In contrast, MMAC1's effects on cell motility, cell - cell and cell - matrix interactions are thought to be due to its protein phosphatase activity towards FAK. However, recent studies suggest that the lipid phosphatase activity of MMAC1 correlates with its ability to be a tumor suppressor. The high rate of mutation of MMAC1 in late stage metastatic tumors suggests that effects of MMAC1 on motility, cell - cell and cell - matrix interactions are due to its tumor suppressor activity. Therefore the lipid phosphatase activity of MMAC1 may affect PIP3 dependent signaling pathways and result in reduced motility and altered cell - cell and cell - matrix interactions. We demonstrate here that expression of MMAC1 in human glioma cells reduced intracellular levels of inositol trisphosphate and inhibited extracellular Ca2+ influx, suggesting that MMAC1 affects the phospholipase C signaling pathway. In addition, we show that MMAC1 expression inhibits integrin-linked kinase activity. Furthermore, we show that these effects require the catalytic activity of MMAC1. Our data thus provide a link of MMAC1 to PIP3 dependent signaling pathways that regulate cell - matrix and cell - cell interactions as well as motility. Lastly, we demonstrate that AKT3, an isoform of AKT highly expressed in the brain, is also a target for MMAC1 repression. These data suggest an important role for AKT3 in glioblastoma multiforme. We therefore propose that repression of multiple PIP3 dependent signaling pathways may be required for MMAC1 to act as a tumor suppressor.


Subject(s)
Genes, Tumor Suppressor/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Type C Phospholipases/antagonists & inhibitors , Animals , Brain/enzymology , Calcium Signaling/physiology , Enzyme Activation , Glioblastoma/enzymology , Glioma/enzymology , Humans , Isoenzymes/biosynthesis , Isoenzymes/physiology , Mice , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured , Type C Phospholipases/metabolism
8.
Biochim Biophys Acta ; 788(2): 189-92, 1984 Jul 31.
Article in English | MEDLINE | ID: mdl-6743666

ABSTRACT

Rat liver thiol:protein-disulfide oxidoreductase/glutathione-insulin transhydrogenase (glutathione:protein disulfide oxidoreductase, EC 1.8.4.2) was purified and found to give two bands on sodium dodecyl sulfate polyacrylamide gel electrophoresis. A monoclonal antibody was produced against this enzyme preparation and found to remove all the insulin degrading activity of purified preparations of the enzyme. This monoclonal antibody was also found to react with the two different forms of the enzyme observed on gel electrophoresis. These results suggest that glutathione-insulin transhydrogenase can exist in more than one state.


Subject(s)
Oxidoreductases/immunology , Protein Disulfide Reductase (Glutathione)/immunology , Animals , Antibodies, Monoclonal , Antibody Specificity , Liver/enzymology , Molecular Weight , Protein Disulfide Reductase (Glutathione)/isolation & purification , Rats
9.
Biochim Biophys Acta ; 1224(3): 533-40, 1994 Dec 30.
Article in English | MEDLINE | ID: mdl-7803513

ABSTRACT

Protein(s) which bind polyphosphatidylinositol phosphates (PI 3,4,5-P3 and PI 4,5-P2) were identified in the wheat-germ agglutinin bound fraction of cells and tissues. The binding of this protein(s) to the phospholipid could be demonstrated in two ways, either by a shift in the migration of the lipid by size exclusion column chromatography or directly by binding to the protein after capture on wheat-germ agglutinin-coupled beads. Of the rat tissues tested (muscle, spleen, brain, heart, kidney and liver), the activity was highest in liver. The protein(s) was purified more than 5000-fold by sequential chromatography on columns of wheat-germ agglutinin, phosphocellulose, Blue-Sepharose, Mono Q and Superose 6. The peak of activity appeared to have a molecular weight on this latter column of approx. 240,000. The protein(s) bound PI 3,4,5-P3, PI 3,4-P2, and PI 3-P in the ratio of 4:2:1. The binding of 3-phosphorylated PI phosphates to the protein(s) was not significantly inhibited by 36 micrograms/ml of either phosphatidylinositol or phosphatidylcholine, but was inhibited 10% and 65% by 36 micrograms/ml of PI 4-P and PI 4,5-P2, respectively. Since these results suggested that the binding protein(s) could also bind PI 4,5-P2, binding of this lipid was directly tested and found to be comparable to that of PI 3,4,5-P3. These results suggest that this protein(s) could be involved in the signaling mechanism elicited by these polyphosphoinositides.


Subject(s)
Phosphatidylinositol Phosphates/metabolism , Proteins/metabolism , Animals , Brain/metabolism , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Kidney/metabolism , Liver/metabolism , Muscles/metabolism , Myocardium/metabolism , Phosphatidylinositol 4,5-Diphosphate , Protein Binding , Proteins/isolation & purification , Rats , Spleen/metabolism
10.
Diabetes ; 36(1): 123-6, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3539674

ABSTRACT

Highly purified insulin receptor was shown to be a substrate for cAMP kinase. Approximately 1 phosphate was incorporated per molecule of receptor, and the cAMP kinase's affinity for the receptor was at least as high as its affinity for histone. The sites phosphorylated by cAMP kinase seemed distinct from those phosphorylated by the protein kinase C. Phosphorylation by cAMP kinase had no effect on the ability of several monoclonal antibodies to recognize the receptor or on the insulin-binding activity of the receptor. However, cAMP phosphorylation partially inhibited the tyrosine kinase activity of the receptor (approximately 25%). These results suggest that catecholamine-induced resistance to insulin may be partly due to a direct phosphorylation of the receptor by cAMP kinase and a subsequent inhibition of the ability of the receptor kinase to be activated by insulin.


Subject(s)
Protein Kinases/metabolism , Receptor, Insulin/metabolism , Humans , Insulin/metabolism , Phosphorylation , Receptor, Insulin/isolation & purification
11.
Diabetes ; 43(11): 1297-303, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7926303

ABSTRACT

We have developed and characterized a line of Madin-Darby canine kidney (MDCK) cells overexpressing the human insulin receptor. The expressed receptor was found to be processed normally, and its intrinsic tyrosine kinase was determined to be functional from both in vitro and in vivo phosphorylation studies. The expressed receptor was able to mediate an insulin-stimulated increase in both anti-phosphotyrosine-precipitable and anti-insulin receptor substrate 1-precipitable phosphatidylinositol 3-kinase activity. Moreover, insulin-induced glycogen synthase activity was greater and more sensitive to insulin in the transfected cells than in the parental cells. Interestingly, insulin promoted tubule-like growth in cells overexpressing the insulin receptor but not in the parental cells. Another advantage of this cell system lies in its ability to polarize into distinct basolateral and apical membrane compartments. With the use of biotinylation and Western analysis, the expressed insulin receptor was found to be preferentially expressed in the basolateral membrane (fivefold greater) in comparison with the apical membrane. Therefore, MDCK cells overexpressing the insulin receptor represent a novel system to study not only the pathway of insulin signaling, but also this pathway in the context of cell polarity.


Subject(s)
Kidney/metabolism , Receptor, Insulin/physiology , Signal Transduction/physiology , Animals , Cell Line , Dogs , Humans , Kidney/cytology , Kidney/enzymology , Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/biosynthesis , Recombinant Proteins/biosynthesis , Transfection
12.
Diabetes ; 33(3): 301-4, 1984 Mar.
Article in English | MEDLINE | ID: mdl-6365664

ABSTRACT

The serum from a patient with lupus nephritis, insulin resistance, and hypoglycemia was studied. This serum both inhibits the binding of 125I-insulin to its receptor and has insulin-like activity on fat cells (see refs. 1 and 2). The IgG fraction from this patient's serum one-half maximally inhibited 125I-insulin binding to IM-9 cells at 1 microM, but did not markedly inhibit 125I-monoclonal antibody binding even at concentrations as high as 4 microM. The IgG was then subjected to affinity chromatography on a protein A-Sepharose column. Four protein peaks were eluted from this column by a step pH gradient from 5.5 to 2.3. Three of the four peaks inhibited 125I-insulin binding to its receptors, but none was more potent than the unfractionated IgG itself. One IgG peak, however, was able to inhibit 125I-monoclonal antibody binding at tenfold lower concentrations than the unfractionated IgG. When the ability of the four IgG fractions to stimulate 2-deoxy[3H]-D-glucose transport in rat adipocytes was studied, two fractions showed stimulatory activity. Compared with unfractionated IgG, one had a weak ability to inhibit 125I-insulin binding, but tenfold more potency to mimic insulin action. The other had a strong ability to inhibit 125I-insulin binding but less potency to mimic insulin action. These studies indicate, therefore, that the serum contains multiple populations of antibodies to the insulin receptor, or portions of the plasma membrane adjacent to the receptor, which have different biologic effects.


Subject(s)
Autoantibodies/immunology , Hypoglycemia/blood , Insulin Resistance , Insulin/metabolism , Receptor, Insulin/immunology , Adipose Tissue/cytology , Animals , Antibodies, Monoclonal/immunology , Deoxyglucose/metabolism , Female , Humans , Immunoglobulin G/metabolism , Insulin/immunology , Lymphocytes/metabolism , Mice , Rats
13.
Diabetes ; 35(6): 675-83, 1986 Jun.
Article in English | MEDLINE | ID: mdl-3519322

ABSTRACT

An insulin-degrading enzyme (IDE) was purified from the cytosol of human erythrocytes via the use of ammonium sulfate precipitation and chromatography on columns composed of DEAE-Sephadex, pentylagarose, hydroxylapatite, chromatofocusing resins, and Ultrogel AcA-34. The final preparation was purified greater than 50,000-fold and exhibited a single protein band of Mr = 110,000 on reduced sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis. Cross-linking of 125I-labeled insulin to the enzyme preparation labeled a protein of the same molecular weight, indicating that this band was in fact the enzyme. Intact insulin, insulin B chain, and glucagon inhibited this cross-linking half-maximally at concentrations of 0.1, 1, and 1.5 microM, respectively. Under nondenaturing conditions, the enzyme had an Mr = 300,000, suggesting that the enzyme may exist under physiological conditions as a dimer or timer. The purified enzyme was inhibited by both sulfhydrylmodifying reagents and chelating agents, indicating that a free thiol and metal were both required for the activity of the enzyme. The purified enzyme was found to degrade physiological concentrations of intact insulin more rapidly than insulin B chain, although at high substrate concentrations (greater than 1 microM) the enzyme degraded B chain to a greater extent. Additional characteristics of the enzyme were a pl of 5.2 and a pH optimum of 7.0. These properties of the red blood cell (RBC) enzyme were very similar to those reported for IDEs from other tissues. Moreover, a polyclonal antiserum to the IDE from skeletal muscle was found to recognize the RBC enzyme.


Subject(s)
Erythrocytes/enzymology , Insulin/metabolism , Chemical Precipitation , Chromatography, Gel , Humans , Hydrogen-Ion Concentration , Molecular Weight , Receptor, Insulin/metabolism , Substrate Specificity , Trichloroacetic Acid/pharmacology
14.
Diabetes ; 47(8): 1281-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9703329

ABSTRACT

The serine/threonine kinase Akt (PKB/Rac) has been implicated as playing a role in the insulin-signaling pathway to glucose transport. Little is known regarding the regulation of Akt kinase activity in insulin-sensitive tissues, such as skeletal muscle, or whether this regulation is altered in insulin-resistant states such as NIDDM. We examined the effect of insulin on Akt kinase activity in skeletal muscle from six NIDDM patients and six healthy subjects. Whole-body insulin sensitivity, assessed by the euglycemic-hyperinsulinemic clamp, was significantly lower in NIDDM subjects (P < 0.001), and this was accompanied by impaired in vitro insulin-stimulated glucose transport in skeletal muscle. In both groups, insulin induced a significant increase in Akt kinase activity, but the response to maximal insulin (60 nmol/l) was markedly reduced in skeletal muscle from NIDDM subjects (66% of control levels, P < 0.01). Impaired Akt kinase activity was not accompanied by decreased protein expression of Akt. Instead, a trend toward increased Akt expression was noted in skeletal muscle from NIDDM subjects (P < 0.1). These parallel defects in insulin-stimulated Akt kinase activity and glucose transport in diabetic skeletal muscle suggest that reduced Akt kinase activity may play a role in the development of insulin resistance in NIDDM.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Insulin/pharmacology , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Biological Transport/drug effects , Glucose/metabolism , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-akt , Reference Values
15.
Diabetes ; 33(1): 64-72, 1984 Jan.
Article in English | MEDLINE | ID: mdl-6360769

ABSTRACT

In the present study, we have used isolated mouse pancreatic acini to investigate the relationship between 125I-insulin binding and its degradation in order to probe the nature and cellular localization of the degradative process. In these cells, the proteolysis of 125I-insulin was dependent on time and cell concentration, and was saturated by unlabeled insulin with a Km of 290 nM. Since this value was much higher than the Kd for insulin binding to its receptor (1.1 nM), the data indicated that 125I-insulin degradation by acini occurred primarily via nonreceptor mechanisms. Several lines of evidence suggested that insulin was being degraded by the neutral thiol protease, insulin degrading enzyme (IDE). First, insulin degradation was inhibited by thiolreacting agents such as N-ethylmaleimide and p-chloromercuribenzoate. Second, the Km for degradation in acini was similar to the reported Km for IDE in other tissues. Third, the enzyme activity had a relative mol wt of approximately 130,000 by gel filtration, a value similar to that reported for purified IDE. Fourth, the degrading activity was removed with a specific antibody to IDE. Other lines of evidence suggested that enzymes located on the cell surface played a role in insulin degradation by acini. First, the nonpenetrating sulfhydryl reacting agent 5,5' dithiobis-2-nitrobenzoic acid blocked 125I-insulin degradation. Second, a specific antibody to IDE identified the presence of the enzyme on the cell surface. Third, chloroquine, leupeptin and antipain, agents that inhibit lysosomal function, did not influence 125I-insulin degradation. Fourth, highly purified pancreatic plasma membranes degraded 125I-insulin.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Peptide Hydrolases/metabolism , Receptor, Insulin/metabolism , Animals , Cell Membrane/metabolism , Chloromercuribenzoates/pharmacology , Chromatography, Gel , Ethylmaleimide/pharmacology , In Vitro Techniques , Iodine Radioisotopes , Islets of Langerhans/enzymology , Male , Membrane Proteins/metabolism , Mice , Receptor, Insulin/drug effects
16.
Diabetes ; 46(12): 2110-4, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9392506

ABSTRACT

The serine/threonine kinase Akt (protein kinase B [PKB] or related to A and C protein kinase [RAC]) has recently been implicated to play a role in the signaling pathway to glucose transport. However, little is known concerning the regulation of Akt activity in insulin-sensitive tissues such as skeletal muscle. To explore the role of hyperglycemia on Akt kinase activity in skeletal muscle, normal Wistar rats or Goto-Kakizaki (GK) diabetic rats were treated with phlorizin. Phlorizin treatment normalized fasting blood glucose and significantly improved glucose tolerance (P < 0.001) in GK rats, whereas in Wistar rats, the compound had no effect on glucose homeostasis. In soleus muscle from GK rats, maximal insulin-stimulated (120 nmol/l) Akt kinase activity was reduced by 68% (P < 0.01) and glucose transport was decreased by 39% (P < 0.05), compared with Wistar rats. Importantly, the defects at the level of Akt kinase and glucose transport were completely restored by phlorizin treatment. There was no significant difference in Akt kinase protein expression among the three groups. At a submaximal insulin concentration (2.4 nmol/l), activity of Akt kinase and glucose transport were unaltered. In conclusion, improved glucose tolerance in diabetic GK rats by phlorizin treatment fully restored insulin-stimulated activity of Akt kinase and glucose transport. Thus, hyperglycemia may directly contribute to the development of muscle insulin resistance through alterations in insulin action on Akt kinase and glucose transport.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Glucose Tolerance Test , Glucose/metabolism , Insulin/pharmacology , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Biological Transport/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Insulin Resistance , Male , Muscle, Skeletal/drug effects , Phlorhizin/therapeutic use , Proto-Oncogene Proteins c-akt , Rats , Rats, Mutant Strains
17.
Diabetes ; 48(3): 664-70, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10078575

ABSTRACT

To determine whether defects in the insulin signal transduction pathway to glucose transport occur in a muscle fiber type-specific manner, post-receptor insulin-signaling events were assessed in oxidative (soleus) and glycolytic (extensor digitorum longus [EDL]) skeletal muscle from Wistar or diabetic GK rats. In soleus muscle from GK rats, maximal insulin-stimulated (120 nmol/l) glucose transport was significantly decreased, compared with that of Wistar rats. In EDL muscle from GK rats, maximal insulin-stimulated glucose transport was normal, while the submaximal response was reduced compared with that of Wistar rats. We next treated diabetic GK rats with phlorizin for 4 weeks to determine whether restoration of glycemia would lead to improved insulin signal transduction. Phlorizin treatment of GK rats resulted in full restoration of insulin-stimulated glucose transport in soleus and EDL muscle. In soleus muscle from GK rats, submaximal and maximal insulin-stimulated insulin receptor substrate (IRS)-1 tyrosine phosphorylation and IRS-1-associated phosphatidylinositol (PI) 3-kinase activity were markedly reduced, compared with that of Wistar rats, but only submaximal insulin-stimulated PI 3-kinase was restored after phlorizin treatment. In EDL muscle, insulin-stimulated IRS-1 tyrosine phosphorylation and IRS-1-associated PI-3 kinase were not altered between GK and Wistar rats. Maximal insulin-stimulated Akt (protein kinase B) kinase activity is decreased in soleus muscle from GK rats and restored upon normalization of glycemia (Krook et al., Diabetes 46:2100-2114, 1997). Here, we show that in EDL muscle from GK rats, maximal insulin-stimulated Akt kinase activity is also impaired and restored to Wistar rat levels after phlorizin treatment. In conclusion, functional defects in IRS-1 and PI 3-kinase in skeletal muscle from diabetic GK rats are fiber-type-specific, with alterations observed in oxidative, but not glycolytic, muscle. Furthermore, regardless of muscle fiber type, downstream steps to PI 3-kinase (i.e., Akt and glucose transport) are sensitive to changes in the level of glycemia.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Gene Expression Regulation , Glucose/metabolism , Insulin/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases , Signal Transduction/physiology , 3-O-Methylglucose/pharmacokinetics , Animals , Biological Transport/drug effects , Body Weight , Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation/drug effects , Glucose Transporter Type 4 , Glycogen Synthase/genetics , Insulin Receptor Substrate Proteins , Monosaccharide Transport Proteins/genetics , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Phlorhizin/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Inbred Strains , Rats, Wistar , Species Specificity
18.
Pharmacol Ther ; 48(2): 143-55, 1990.
Article in English | MEDLINE | ID: mdl-2293237

ABSTRACT

The lungs contain enzyme systems that metabolize xenobiotic agents, and the structure and position in the circulation render this organ potentially important in the metabolic removal of substances from the blood. Pulmonary enzyme systems that oxidize xenobiotic agents include cytochrome P450- or flavin-containing monooxygenases. In addition, the lungs accumulate certain agents, notably basic amines, without substantially metabolizing them. Benzo(a)pyrene (B(a)P) is one example of a xenobiotic agents that is eliminated from the circulation largely by oxidative metabolism. We have described the metabolic elimination of B(a)P using a physiologically based pharmacokinetic model applied retrospectively to existing data sets of B(a)P metabolism and disposition in rats. The result suggests that the lungs may, under certain conditions, contribute significantly to xenobiotic disposition and that this contribution is greater than that predicted by the activity of dispositional enzyme in this organ. Thus, the lungs may play a significant role in the metabolic elimination of some xenobiotic agents under certain circumstances.


Subject(s)
Benzo(a)pyrene/pharmacokinetics , Lung/metabolism , Xenobiotics/pharmacokinetics , Animals , Humans , Lung/enzymology , Models, Biological
19.
Mol Endocrinol ; 5(2): 194-200, 1991 Feb.
Article in English | MEDLINE | ID: mdl-1710030

ABSTRACT

In the present studies mutant insulin receptors with regulatory tyrosine residues 1162 and 1163 changed to phenylalanines were tested for tyrosine kinase activity. In agreement with prior studies, this mutant receptor was found to exhibit almost no insulin-stimulated exogenous kinase activity when assayed in vitro. In contrast, this mutant receptor was found in situ to have a significant, albeit reduced, ability to mediate the tyrosine phosphorylation of various endogenous proteins, as assessed by Western blotting with antiphosphotyrosine antibodies. In addition, extracts of insulin-treated cells overexpressing this mutant receptor exhibited increased amounts of tyrosine phosphorylated phosphatidylinositol 3-kinase compared to control cells. Finally, this mutant receptor, like the wild-type receptor, was found to mediate an increase in the activity of a membrane-associated phosphatidylinositol 4,5-biphosphate kinase. These results indicate that 1) in vitro assessments of the tyrosine kinase activity of mutant insulin receptors may not accurately reflect their in vivo activities; and 2) the ability of the mutant receptor lacking tyrosine autophosphorylation sites 1162 and 1163 to mediate insulin-stimulated tyrosine phosphorylation of various endogenous substrates may account for the reported ability of this receptor to mediate various biological responses.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor) , Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/genetics , Tyrosine/analogs & derivatives , Animals , Binding Sites , Blotting, Western , Cell Line , Cricetinae , Humans , Insulin/pharmacology , Kinetics , Mutation , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases/metabolism , Phosphotyrosine , Receptor, Insulin/metabolism , Tyrosine/metabolism
20.
Mol Endocrinol ; 8(1): 51-8, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7512195

ABSTRACT

Chinese hamster ovary (CHO) cells were transfected with a cDNA encoding protein kinase C alpha (PKC) and a cell line (CHO-PKC alpha) expressing approximately 7-fold greater amounts of PKC as the parental cells were isolated. Activation of PKC by 12-O-tetradecanoylphorbol-13-acetate in the CHO-PKC alpha cells inhibited by approximately 75% the: 1) insulin-stimulated increase in antiphosphotyrosine precipitable phosphatidylinositol 3-kinase activity in these cells; 2) insulin-stimulated increase in PI 3-kinase activity associated with insulin receptor substrate-1; and 3) tyrosine phosphorylation of the endogenous substrate, insulin receptor substrate-1. In contrast, 12-O-tetradecanoylphorbol-13-acetate treatment did not inhibit any of these responses in the parental CHO cells. These results indicate that excessive PKC activity can interfere in a very early step in insulin receptor signaling and are consistent with the hypothesis that excessive PKC activity may contribute to some states of insulin resistance.


Subject(s)
Insulin/pharmacology , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Tyrosine/analogs & derivatives , Animals , Blotting, Western , CHO Cells , Cricetinae , DNA, Complementary/genetics , Enzyme Activation , Immunosorbent Techniques , Insulin Receptor Substrate Proteins , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotyrosine , Protein Kinase C/genetics , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL