51
|
Bell KS, Schmitz-Peiffer C, Lim-Fraser M, Biden TJ, Cooney GJ, Kraegen EW. Acute reversal of lipid-induced muscle insulin resistance is associated with rapid alteration in PKC-theta localization. Am J Physiol Endocrinol Metab 2000; 279:E1196-201. [PMID: 11052977 DOI: 10.1152/ajpendo.2000.279.5.e1196] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle insulin resistance in the chronic high-fat-fed rat is associated with increased membrane translocation and activation of the novel, lipid-responsive, protein kinase C (nPKC) isozymes PKC-theta and -epsilon. Surprisingly, fat-induced insulin resistance can be readily reversed by one high-glucose low-fat meal, but the underlying mechanism is unclear. Here, we have used this model to determine whether changes in the translocation of PKC-theta and -epsilon are associated with the acute reversal of insulin resistance. We measured cytosol and particulate PKC-alpha and nPKC-theta and -epsilon in muscle in control chow-fed Wistar rats (C) and 3-wk high-fat-fed rats with (HF-G) or without (HF-F) a single high-glucose meal. PKC-theta and -epsilon were translocated to the membrane in muscle of insulin-resistant HF-F rats. However, only membrane PKC-theta was reduced to the level of chow-fed controls when insulin resistance was reversed in HF-G rats [% PKC-theta at membrane, 23.0 +/- 4.4% (C); 39.7 +/- 3.4% (HF-F, P < 0.01 vs. C); 22.5 +/- 2.7% (HF-G, P < 0.01 vs. HF-F), by ANOVA]. We conclude that, although muscle localization of both PKC-epsilon and PKC-theta are influenced by chronic dietary lipid oversupply, PKC-epsilon and PKC-theta localization are differentially influenced by acute withdrawal of dietary lipid. These results provide further support for an association between PKC-theta muscle cellular localization and lipid-induced muscle insulin resistance and stress the labile nature of high-fat diet-induced insulin resistance in the rat.
Collapse
Affiliation(s)
- K S Bell
- Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | |
Collapse
|
52
|
Spitaler M, Villunger A, Grunicke H, Uberall F. Unique structural and functional properties of the ATP-binding domain of atypical protein kinase C-iota. J Biol Chem 2000; 275:33289-96. [PMID: 10906326 DOI: 10.1074/jbc.m002742200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Atypical protein kinase C-iota (aPKCiota) plays an important role in mitogenic signaling, actin cytoskeleton organization, and cell survival. Apart from the differences in the regulatory domain, the catalytic domain of aPKCiota differs considerably from other known kinases, because it contains a modification within the glycine-rich loop motif (GXGXXG) that is found in the nucleotide-binding fold of virtually all nucleotide-binding proteins including PKCs, Ras, adenylate kinase, and the mitochondrial F1-ATPase. We have used site-directed mutagenesis and kinetic analysis to investigate whether these sequence differences affect the nucleotide binding properties and catalytic activity of aPKCiota. When lysine 274, a residue essential for ATP binding and activity conserved in most protein kinases, was replaced by arginine (K274R mutant), aPKCiota retained its normal kinase activity. This is in sharp contrast to results published for any other PKC or even distantly related kinases like phosphoinositide 3-kinase gamma, where the same mutation completely abrogated the kinase activity. Furthermore, the sensitivity of aPKCiota for inhibition by GF109203X, a substance acting on the ATP-binding site, was not altered in the K274R mutant. In contrast, replacement of Lys-274 by tryptophan (K274W) completely abolished the kinase activity of PKCiota. In accordance with results obtained with other kinase-defective PKC mutants, in cultured cells aPKCiota-K274W acted in a dominant negative fashion on signal transduction pathways involving endogenous aPKCiota, whereas the effect of the catalytically active K274R mutant was identical to the wild type enzyme. In summary, aPKCiota differs from classical and novel PKCs also in the catalytic domain. This information could be of significant value for the development of specific inhibitors of aPKCiota as a key factor in central signaling pathways.
Collapse
Affiliation(s)
- M Spitaler
- Institut für Medizinische Chemie und Biochemie, University of Innsbruck, Fritz-Pregl-Strasse 3, A-6020 Innsbruck, Austria.
| | | | | | | |
Collapse
|
53
|
Schmitz-Peiffer C. Signalling aspects of insulin resistance in skeletal muscle: mechanisms induced by lipid oversupply. Cell Signal 2000; 12:583-94. [PMID: 11080610 DOI: 10.1016/s0898-6568(00)00110-8] [Citation(s) in RCA: 179] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A reduced capacity for insulin to elicit increases in glucose uptake and metabolism in target tissues such as skeletal muscle is a common feature of obesity and diabetes. The association between lipid oversupply and such insulin resistance is well established, and evidence for mechanisms through which lipids could play a causative role in the generation of muscle insulin resistance is reviewed. While the effects of lipids may in part be mediated by substrate competition through the glucose-fatty acid cycle, interference with insulin signal transduction by lipid-activated signalling pathways is also likely to play an important role. Thus, studies of insulin resistance in Type 2 diabetes, obesity, fat-fed animals and lipid-treated cells have identified defects both at the level of insulin receptor-mediated tyrosine phosphorylation and at downstream sites such as protein kinase B (PKB) activation. Lipid signalling molecules can be derived from free fatty acids, and include diacylglycerol, which activates isozymes of the protein kinase C (PKC) family, and ceramide, which has several effectors including PKCs and a protein phosphatase. In addition, elevated lipid availability can increase flux through the hexosamine biosynthesis pathway which can also lead to activation of PKC as well as protein glycosylation and modulation of gene expression. The mechanisms giving rise to decreased insulin signalling include serine/threonine phosphorylation of insulin receptor substrate-1, but also direct inhibition of components such as PKB. Thus lipids can inhibit glucose disposal by causing interference with insulin signal transduction, and most likely by more than one pathway depending on the prevalent species of fatty acids.
Collapse
Affiliation(s)
- C Schmitz-Peiffer
- Garvan Institute of Medical Research, 384 Victoria Street, NSW 2010, Darlinghurst, Australia.
| |
Collapse
|
54
|
Formisano P, Oriente F, Fiory F, Caruso M, Miele C, Maitan MA, Andreozzi F, Vigliotta G, Condorelli G, Beguinot F. Insulin-activated protein kinase Cbeta bypasses Ras and stimulates mitogen-activated protein kinase activity and cell proliferation in muscle cells. Mol Cell Biol 2000; 20:6323-33. [PMID: 10938109 PMCID: PMC86107 DOI: 10.1128/mcb.20.17.6323-6333.2000] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In L6 muscle cells expressing wild-type human insulin receptors (L6hIR), insulin induced protein kinase Calpha (PKCalpha) and beta activities. The expression of kinase-deficient IR mutants abolished insulin stimulation of these PKC isoforms, indicating that receptor kinase is necessary for PKC activation by insulin. In L6hIR cells, inhibition of insulin receptor substrate 1 (IRS-1) expression caused a 90% decrease in insulin-induced PKCalpha and -beta activation and blocked insulin stimulation of mitogen-activated protein kinase (MAPK) and DNA synthesis. Blocking PKCbeta with either antisense oligonucleotide or the specific inhibitor LY379196 decreased the effects of insulin on MAPK activity and DNA synthesis by >80% but did not affect epidermal growth factor (EGF)- and serum-stimulated mitogenesis. In contrast, blocking c-Ras with lovastatin or the use of the L61,S186 dominant negative Ras mutant inhibited insulin-stimulated MAPK activity and DNA synthesis by only about 30% but completely blocked the effect of EGF. PKCbeta block did not affect Ras activity but almost completely inhibited insulin-induced Raf kinase activation and coprecipitation with PKCbeta. Finally, blocking PKCalpha expression by antisense oligonucleotide constitutively increased MAPK activity and DNA synthesis, with little effect on their insulin sensitivity. We make the following conclusions. (i) The tyrosine kinase activity of the IR is necessary for insulin activation of PKCalpha and -beta. (ii) IRS-1 phosphorylation is necessary for insulin activation of these PKCs in the L6 cells. (iii) In these cells, PKCbeta plays a unique Ras-independent role in mediating insulin but not EGF or other growth factor mitogenic signals.
Collapse
Affiliation(s)
- P Formisano
- Dipartimento di Biologia e Patologia Cellulare e Molecolare and Centro di Endocrinologia ed Oncologia Sperimentale del CNR, Università di Napoli Federico II, Naples 80131, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Haruta T, Uno T, Kawahara J, Takano A, Egawa K, Sharma PM, Olefsky JM, Kobayashi M. A rapamycin-sensitive pathway down-regulates insulin signaling via phosphorylation and proteasomal degradation of insulin receptor substrate-1. Mol Endocrinol 2000; 14:783-94. [PMID: 10847581 DOI: 10.1210/mend.14.6.0446] [Citation(s) in RCA: 318] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Insulin receptor substrate-1 (IRS-1) is a major substrate of the insulin receptor and acts as a docking protein for Src homology 2 domain containing signaling molecules that mediate many of the pleiotropic actions of insulin. Insulin stimulation elicits serine/threonine phosphorylation of IRS-1, which produces a mobility shift on SDS-PAGE, followed by degradation of IRS-1 after prolonged stimulation. We investigated the molecular mechanisms and the functional consequences of these phenomena in 3T3-L1 adipocytes. PI 3-kinase inhibitors or rapamycin, but not the MEK inhibitor, blocked both the insulin-induced electrophoretic mobility shift and degradation of IRS-1. Adenovirus-mediated expression of a membrane-targeted form of the p110 subunit of phosphatidylinositol (PI) 3-kinase (p110CAAX) induced a mobility shift and degradation of IRS-1, both of which were inhibited by rapamycin. Lactacystin, a specific proteasome inhibitor, inhibited insulin-induced degradation of IRS-1 without any effect on its electrophoretic mobility. Inhibition of the mobility shift did not significantly affect tyrosine phosphorylation of IRS-1 or downstream insulin signaling. In contrast, blockade of IRS-1 degradation resulted in sustained activation of Akt, p70 S6 kinase, and mitogen-activated protein (MAP) kinase during prolonged insulin treatment. These results indicate that insulin-induced serine/threonine phosphorylation and degradation of IRS-1 are mediated by a rapamycin-sensitive pathway, which is downstream of PI 3-kinase and independent of ras/MAP kinase. The pathway leads to degradation of IRS-1 by the proteasome, which plays a major role in down-regulation of certain insulin actions during prolonged stimulation.
Collapse
Affiliation(s)
- T Haruta
- First Department of Medicine, Toyama Medical and Pharmaceutical University Japan.
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Boytim ML, Lilly P, Drouvalakis K, Lyu SC, Jung R, Krensky AM, Clayberger C. A human class II MHC-derived peptide antagonizes phosphatidylinositol 3-kinase to block IL-2 signaling. J Clin Invest 2000; 105:1447-53. [PMID: 10811852 PMCID: PMC315461 DOI: 10.1172/jci8139] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
MHC molecules bind antigenic peptides and present them to T cells. There is a growing body of evidence that MHC molecules also serve other functions. We and others have described synthetic peptides derived from regions of MHC molecules that inhibit T-cell proliferation or cytotoxicity in an allele-nonspecific manner that is independent of interaction with the T-cell receptor. In this report, we describe the mechanism of action of a synthetic MHC class II-derived peptide that blocks T-cell activation induced by IL-2. Both this peptide, corresponding to residues 65-79 of DQA*03011 (DQ 65-79), and rapamycin inhibit p70 S6 kinase activity, but only DQ 65-79 blocks Akt kinase activity, placing the effects of DQ 65-79 upstream of mTOR, a PI kinase family member. DQ 65-79, but not rapamycin, inhibits phosphatidylinositol 3-kinase (PI 3-kinase) activity in vitro. The peptide is taken up by cells, as demonstrated by confocal microscopy. These findings indicate that DQ 65-79 acts as an antagonist with PI 3-kinase, repressing downstream signaling events and inhibiting proliferation. Understanding the mechanism of action of immunomodulatory peptides may provide new insights into T-cell activation and allow the development of novel immunosuppressive agents.
Collapse
Affiliation(s)
- M L Boytim
- Program in Immunology, Division of Immunology and Transplantation Biology, Department of Pediatrics, Stanford University, Stanford, California 94305-5407, USA
| | | | | | | | | | | | | |
Collapse
|
57
|
Zheng WH, Kar S, Quirion R. Stimulation of protein kinase C modulates insulin-like growth factor-1-induced akt activation in PC12 cells. J Biol Chem 2000; 275:13377-85. [PMID: 10788447 DOI: 10.1074/jbc.275.18.13377] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of protein kinase C (PKC) plays an important role in the negative regulation of receptor signaling, but its effect on insulin-like growth factor-1 (IGF-1) receptor signaling remains unclear. In this study, we characterized the intracellular pathways involved in IGF-1-induced activation of Akt and evaluated the effects of the PKC activator phorbol 12-myristate 13-acetate (PMA) on the Akt activation by IGF-1. IGF-1 induced a time- and concentration-dependent activation of Akt. The effect of IGF-1 was blocked by the phosphatidylinositide 3-kinase (PI3K) inhibitors LY294002 (50 micrometer) and wortmannin (0.5 micrometer), but not by the MEK inhibitor PD98059 (50 micrometer) or the p70 S6 kinase pathway inhibitor rapamycin (50 nm), suggesting that the stimulation of Akt by IGF-1 is mediated by the PI3K pathway. Interestingly, cotreatment with PMA (400 nm) attenuated IGF-1-induced activation of Akt. The attenuation was blocked completely by the PKC inhibitor GO6983 (0.5 micrometer), but only partially by the MEK inhibitor PD98059 (50 micrometer), indicating that MAPK-dependent and -independent pathways are involved. PMA induced the activation of PKC in PC12 cells, and this induction was blocked by GO6983. These data further support the role of PKC in the effect of PMA. Moreover, PKCdelta is likely involved in the action of PMA on the basis of data obtained using isoform-specific inhibitors such as rottlerin. PMA also decreased IGF-1-induced tyrosine phosphorylation of insulin receptor substrate-1 and its association with PI3K. Taken together, these results suggest, for the first time, that stimulation of PKC modulates IGF-1-induced activation of Akt.
Collapse
Affiliation(s)
- W H Zheng
- Douglas Hospital Research Center, Departments of Psychiatry and of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H4H 1R3, Canada
| | | | | |
Collapse
|
58
|
Cortright RN, Azevedo JL, Zhou Q, Sinha M, Pories WJ, Itani SI, Dohm GL. Protein kinase C modulates insulin action in human skeletal muscle. Am J Physiol Endocrinol Metab 2000; 278:E553-62. [PMID: 10710511 DOI: 10.1152/ajpendo.2000.278.3.e553] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is good evidence from cell lines and rodents that elevated protein kinase C (PKC) overexpression/activity causes insulin resistance. Therefore, the present study determined the effects of PKC activation/inhibition on insulin-mediated glucose transport in incubated human skeletal muscle and primary adipocytes to discern a potential role for PKC in insulin action. Rectus abdominus muscle strips or adipocytes from obese, insulin-resistant, and insulin-sensitive patients were incubated in vitro under basal and insulin (100 nM)-stimulated conditions in the presence of GF 109203X (GF), a PKC inhibitor, or 12-deoxyphorbol 13-phenylacetate 20-acetate (dPPA), a PKC activator. PKC inhibition had no effect on basal glucose transport. GF increased (P < 0.05) insulin-stimulated 2-deoxyglucose (2-DOG) transport approximately twofold above basal. GF plus insulin also increased (P < 0.05) insulin receptor tyrosine phosphorylation 48% and phosphatidylinositol 3-kinase (PI 3-kinase) activity approximately 50% (P < 0.05) vs. insulin treatment alone. Similar results for GF on glucose uptake were observed in human primary adipocytes. Further support for the hypothesis that elevated PKC activity is related to insulin resistance comes from the finding that PKC activation by dPPA was associated with a 40% decrease (P < 0.05) in insulin-stimulated 2-DOG transport. Incubation of insulin-sensitive muscles with GF also resulted in enhanced insulin action ( approximately 3-fold above basal). These data demonstrate that certain PKC inhibitors augment insulin-mediated glucose uptake and suggest that PKC may modulate insulin action in human skeletal muscle.
Collapse
Affiliation(s)
- R N Cortright
- School of Medicine, East Carolina University, Greenville, North Carolina 27858, USA
| | | | | | | | | | | | | |
Collapse
|
59
|
Miller WL. The molecular basis of premature adrenarche: an hypothesis. ACTA PAEDIATRICA (OSLO, NORWAY : 1992). SUPPLEMENT 1999; 88:60-6. [PMID: 10626547 DOI: 10.1111/j.1651-2227.1999.tb14405.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Adrenarche is characterized by a prepubertal rise in adrenal secretion of dehydroepiandrosterone (DHEA) and DHEA sulfate (DHEAS) that is independent of the gonads or gonadotropins. Adrenopause is the corresponding diminution in DHEA and DHEAS concentrations in later life. The mechanisms by which adrenarche and adrenopause are induced and regulated are unknown. Early work focused on identifying hypothetical adrenal androgen regulatory hormones that would induce DHEA in much the same way that adrenocorticotropin induces cortisol, but no such factors have been found. Current studies of adrenarche focus on intra-adrenal events, particularly those concerning 3beta-hydroxysteroid dehydrogenase (3beta-HSD) and 17alpha-hydroxylase/17,20-lyase (P450c17). Molecular data implicate a decrease in 3beta-HSD specifically in the adrenal zona reticularis. However, a decrease in 3beta-HSD is insufficient to explain why the reticularis catalyzes 17,20-lyase activity and hence makes DHEA, rather than catalyzing only 17alpha-hydroxylase activity, as does the zona fasciculata. P450c17 appears to catalyze 17,20-lyase activity only if P450c17 has undergone serine phosphorylation and has access to cytochrome b5 as an allosteric cofactor. Although these two factors have not yet been investigated in adrenarche, it appears that both a zone-specific diminution in 3beta-HSD and a zone-specific induction of 17,20-lyase activity are required to account for the physiological data. Exaggerated premature adrenarche appears to be an early sign of polycystic ovary syndrome (PCOS). Mechanistic considerations of PCOS suggest a key role for serine phosphorylation of P450c17 in both adrenarche and some forms of heritable PCOS.
Collapse
Affiliation(s)
- W L Miller
- Department of Pediatrics, University of California - San Francisco, 94143-0978, USA
| |
Collapse
|
60
|
Standaert ML, Bandyopadhyay G, Galloway L, Soto J, Ono Y, Kikkawa U, Farese RV, Leitges M. Effects of knockout of the protein kinase C beta gene on glucose transport and glucose homeostasis. Endocrinology 1999; 140:4470-7. [PMID: 10499500 DOI: 10.1210/endo.140.10.7073] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The beta-isoform of protein kinase C (PKC) has paradoxically been suggested to be important for both insulin action and insulin resistance as well as for contributing to the pathogenesis of diabetic complications. Presently, we evaluated the effects of knockout of the PKCbeta gene on overall glucose homeostasis and insulin regulation of glucose transport. To evaluate subtle differences in glucose homeostasis in vivo, knockout mice were extensively backcrossed in C57BL/6 mice to diminish genetic differences other than the absence of the PKCbeta gene. PKCbeta-/- knockout offspring obtained through this backcrossing had 10% lower blood glucose levels than those observed in PKCbeta+/+ wild-type offspring in both the fasting state and 30 min after i.p. injection of glucose despite having similar or slightly lower serum insulin levels. Also, compared with commercially obtained C57BL/6-129/SV hybrid control mice, serum glucose levels were similar, and serum insulin levels were similar or slightly lower, in C57BL/6-129/SV hybrid PKCbeta knockout mice in fasting and fed states and after i.p. glucose administration. In keeping with a tendency for slightly lower serum glucose and/or insulin levels in PKCbeta knockout mice, insulin-stimulated 2-deoxyglucose (2-DOG) uptake was enhanced by 50-100% in isolated adipocytes; basal and insulin-stimulated epitope-tagged GLUT4 translocations in adipocytes were increased by 41% and 27%, respectively; and basal 2-DOG uptake was mildly increased by 20-25% in soleus muscles incubated in vitro. The reason for increased 2-DOG uptake and/or GLUT4 translocation in these tissues was uncertain, as there were no significant alterations in phosphatidylinositol 3-kinase activity or activation or in levels of GLUT1 or GLUT4 glucose transporters or other PKC isoforms. On the other hand, increases in 2-DOG uptake may have been partly caused by the loss of PKCbeta1, rather than PKCbeta2, as transient expression of PKCbeta1 selectively inhibited insulin-stimulated translocation of epitope-tagged GLUT4 in adipocytes prepared from PKCbeta knockout mice. Our findings suggest that 1) PKCbeta is not required for insulin-stimulated glucose transport; 2) overall glucose homeostasis in vivo is mildly enhanced by knockout of the PKCbeta gene; 3) glucose transport is increased in some tissues in PKCbeta knockout mice; and 4) increased glucose transport may be partly due to loss of PKCbeta1, which negatively modulates insulin-stimulated GLUT4 translocation.
Collapse
Affiliation(s)
- M L Standaert
- J.A. Haley Veterans Hospital Research Service, and Department of Internal Medicine, University of South Florida College of Medicine, Tampa 33612, USA
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Caruso M, Miele C, Oriente F, Maitan A, Bifulco G, Andreozzi F, Condorelli G, Formisano P, Beguinot F. In L6 skeletal muscle cells, glucose induces cytosolic translocation of protein kinase C-alpha and trans-activates the insulin receptor kinase. J Biol Chem 1999; 274:28637-44. [PMID: 10497232 DOI: 10.1074/jbc.274.40.28637] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In L6 skeletal muscle cells expressing human insulin receptors (L6(hIR)), exposure to 25 mM glucose for 3 min induced a rapid 3-fold increase in GLUT1 and GLUT4 membrane translocation and glucose uptake. The high glucose concentration also activated the insulin receptor kinase toward the endogenous insulin receptor substrates (IRS)-1 and IRS-2. At variance, in L6 cells expressing kinase-deficient insulin receptors, the exposure to 25 mM glucose elicited no effect on glucose disposal. In the L6(hIR) cells, the acute effect of glucose on insulin receptor kinase was paralleled by a 2-fold decrease in both the membrane and the insulin receptor co-precipitated protein kinase C (PKC) activities and a 3-fold decrease in receptor Ser/Thr phosphorylation. Western blotting of the receptor precipitates with isoform-specific PKC antibodies revealed that the glucose-induced decrease in membrane- and receptor-associated PKC activities was accounted for by dissociation of PKCalpha but not of PKCbeta or -delta. This decrease in PKCalpha was paralleled by a similarly sized increase in cytosolic PKCalpha. In intact L6(hIR) cells, inhibition of PKCalpha expression by using a specific antisense oligonucleotide caused a 3-fold increase in IRS phosphorylation by the insulin receptor. This effect was independent of insulin and accompanied by a 2.5-fold increase in glucose disposal by the cells. Thus, in the L6 skeletal muscle cells, glucose acutely regulates its own utilization through the insulin signaling system, independent of insulin. Glucose autoregulation appears to involve PKCalpha dissociation from the insulin receptor and its cytosolic translocation.
Collapse
Affiliation(s)
- M Caruso
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Centro di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche, Federico II University of Naples, Naples 80131, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Jiang ZY, Lin YW, Clemont A, Feener EP, Hein KD, Igarashi M, Yamauchi T, White MF, King GL. Characterization of selective resistance to insulin signaling in the vasculature of obese Zucker (fa/fa) rats. J Clin Invest 1999; 104:447-57. [PMID: 10449437 PMCID: PMC408521 DOI: 10.1172/jci5971] [Citation(s) in RCA: 452] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Both insulin resistance and hyperinsulinemia have been reported to be independent risk factors for cardiovascular diseases. However, little is known regarding insulin signaling in the vascular tissues in insulin-resistant states. In this report, insulin signaling on the phosphatidylinositol 3-kinase (PI 3-kinase) and mitogen-activated protein (MAP) kinase pathways were compared in vascular tissues of lean and obese Zucker (fa/fa) rats in both ex vivo and in vivo studies. Ex vivo, insulin-stimulated tyrosine phosphorylation of insulin receptor beta subunits (IRbeta) in the aorta and microvessels of obese rats was significantly decreased compared with lean rats, although the protein levels of IRbeta in the 2 groups were not different. Insulin-induced tyrosine phosphorylation of insulin receptor substrates 1 and 2 (IRS-1 and IRS-2) and their protein levels were decreased in the aorta of obese rats compared with lean rats. The association of p85 subunit to the IRS proteins and the IRS-associated PI 3-kinase activities stimulated by insulin in the aorta of obese rats were significantly decreased compared with the lean rats. In addition, insulin-stimulated serine phosphorylation of Akt, a downstream kinase of PI 3-kinase pathway, was also reduced significantly in isolated microvessels from obese rats compared with the lean rats. In euglycemic clamp studies, insulin infusion greatly increased tyrosine phosphorylation of IRbeta- and IRS-2-associated PI 3-kinase activity in the aorta of lean rats, but only slight increases were observed in obese rats. In contrast, insulin stimulated tyrosine phosphorylation of MAP kinase (ERK-1/2) equally in isolated microvessels of lean and obese rats, although basal tyrosine phosphorylation of ERK-1/2 was higher in the obese rats. To our knowledge, these data provided the first direct measurements of insulin signaling in the vascular tissues, and documented a selective resistance to PI 3-kinase (but not to MAP kinase pathway) in the vascular tissues of obese Zucker rats.
Collapse
Affiliation(s)
- Z Y Jiang
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Gustafson TA, Moodie SA, Lavan BE. The insulin receptor and metabolic signaling. Rev Physiol Biochem Pharmacol 1999; 137:71-190. [PMID: 10207305 DOI: 10.1007/3-540-65362-7_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- T A Gustafson
- Metabolex, Inc., Section of Signal Transduction, Hayward, CA 94545, USA
| | | | | |
Collapse
|
64
|
Jin L, Dajin Z, Jiaqing Z. Prevention of tumor necrosis factor-a induced insulin resistance by Radix Astragali. Chin J Integr Med 1999. [DOI: 10.1007/bf02934193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
65
|
Antoine PJ, Bertrand F, Auclair M, Magré J, Capeau J, Cherqui G. Insulin induction of protein kinase C alpha expression is independent of insulin receptor Tyr1162/1163 residues and involves mitogen-activated protein kinase kinase 1 and sustained activation of nuclear p44MAPK. Endocrinology 1998; 139:3133-42. [PMID: 9645686 DOI: 10.1210/endo.139.7.6094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We examined the effect of insulin on protein kinase C alpha (PKCalpha) expression and the implication of the mitogen-activated protein kinase kinase 1 mitogen-activated protein kinase (MAPK) pathway in this effect. PKCalpha expression was measured by quantitative RT-PCR and Western blotting using Chinese hamster ovary (CHO) cells overexpressing human insulin receptors of the wild type (CHO-R) or insulin receptors mutated at Tyr1162/1163 autophosphorylation sites (CHO-Y2). In CHO-R cells, insulin caused a time- and concentration-dependent increase in PKCalpha messenger RNA, with a maximum at 6 h and 10-(8)M insulin. This increase involved a transcriptional mechanism, as it was not due to stabilization of PKCalpha messenger RNA and was associated with a similar increase in the immunoreactive PKCalpha level. Insulin induction of PKCalpha expression involved the MEK1MAPK pathway, as it was 1) almost completely suppressed by the potent MEK1 inhibitor PD98059, 2) mimicked by the dominant-active MEK1 (S218D/S222D) mutant, and 3) associated with sustained MAPK activation. In CHO-Y2 cells in which the early phase of MAPK activation by insulin was lost and only the late and sustained phase of activation was observed, insulin signaling of PKCalpha expression was preserved and again involved the MEK1-MAPK pathway. Moreover, we show that in both CHO-R and CHO-Y2 cells, insulin stimulation of PKCalpha gene expression was associated with prolonged activation of nuclear p44MAPK. These results indicate that induction of PKCalpha gene expression by insulin is independent of Tyr1162/1163 autophosphorylation sites and correlates with sustained activation of p44MAPK at the nuclear level.
Collapse
Affiliation(s)
- P J Antoine
- INSERM U-402, Institut Federatif de Recherche 65, Laboratoire de Biologie Cellulaire, Faculté de Médecine Saint-Antoine, Paris, France
| | | | | | | | | | | |
Collapse
|
66
|
|
67
|
Formisano P, Oriente F, Miele C, Caruso M, Auricchio R, Vigliotta G, Condorelli G, Beguinot F. In NIH-3T3 fibroblasts, insulin receptor interaction with specific protein kinase C isoforms controls receptor intracellular routing. J Biol Chem 1998; 273:13197-202. [PMID: 9582362 DOI: 10.1074/jbc.273.21.13197] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin increased protein kinase C (PKC) activity by 2-fold in both membrane preparations and insulin receptor (IR) antibody precipitates from NIH-3T3 cells expressing human IRs (3T3hIR). PKC-alpha, -delta, and -zeta were barely detectable in IR antibody precipitates of unstimulated cells, while increasing by 7-, 3.5-, and 3-fold, respectively, after insulin addition. Preexposure of 3T3hIR cells to staurosporine reduced insulin-induced receptor coprecipitation with PKC-alpha, -delta, and -zeta by 3-, 4-, and 10-fold, respectively, accompanied by a 1.5-fold decrease in insulin degradation and a similar increase in insulin retroendocytosis. Selective depletion of cellular PKC-alpha and -delta, by 24 h of 12-O-tetradecanoylphorbol-13-acetate (TPA) exposure, reduced insulin degradation by 3-fold and similarly increased insulin retroendocytosis, with no change in PKC-zeta. In lysates of NIH-3T3 cells expressing the R1152Q/K1153A IRs (3T3Mut), insulin-induced coprecipitation of PKC-alpha, -delta, and -zeta with the IR was reduced by 10-, 7-, and 3-fold, respectively. Similar to the 3T3hIR cells chronically exposed to TPA, untreated 3T3Mut featured a 3-fold decrease in insulin degradation, with a 3-fold increase in intact insulin retroendocytosis. Thus, in NIH-3T3 cells, insulin elicits receptor interaction with multiple PKC isoforms. Interaction of PKC-alpha and/or -delta with the IR appears to control its intracellular routing.
Collapse
Affiliation(s)
- P Formisano
- Dipartimento di Biologia e Patologia Cellulare e Molecolare "L. Califano" and Centro di Endocrinologia ed Oncolgia Sperimentale del Consiglio Nazionale delle Ricerche (CNR), "Federico II" University of Naples Medical School, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Kohn AD, Barthel A, Kovacina KS, Boge A, Wallach B, Summers SA, Birnbaum MJ, Scott PH, Lawrence JC, Roth RA. Construction and characterization of a conditionally active version of the serine/threonine kinase Akt. J Biol Chem 1998; 273:11937-43. [PMID: 9565622 DOI: 10.1074/jbc.273.19.11937] [Citation(s) in RCA: 242] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Akt is a serine/threonine kinase that requires a functional phosphatidylinositol 3-kinase to be stimulated by insulin and other growth factors. When directed to membranes by the addition of a src myristoylation sequence, Akt becomes constitutively active. In the present study, a conditionally active version of Akt was constructed by fusing the Akt containing the myristoylation sequence to the hormone binding domain of a mutant murine estrogen receptor that selectively binds 4-hydroxytamoxifen. The chimeric protein was expressed in NIH3T3 cells and was shown to be stimulated by hormone treatment 17-fold after only a 20-min treatment. This hormone treatment also stimulated an approximate 3-fold increase in the phosphorylation of the chimeric protein and a shift in its migration on SDS gels. Activation of this conditionally active Akt resulted in the rapid stimulation of the 70-kDa S6 kinase. This conditionally active Akt was also found to rapidly stimulate in these cells the phosphorylation of properties of PHAS-I, a key protein in the regulation of protein synthesis. The conditionally active Akt, when expressed in 3T3-L1 adipocytes, was also stimulated, although its rate and extent of activation was less then in the NIH3T3 cells. Its stimulation was shown to be capable of inducing glucose uptake into adipocytes by stimulating translocation of the insulin-responsive glucose transporter GLUT4 to the plasma membrane.
Collapse
Affiliation(s)
- A D Kohn
- Department of Molecular Pharmacology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Miller WL. Early steps in androgen biosynthesis: from cholesterol to DHEA. BAILLIERE'S CLINICAL ENDOCRINOLOGY AND METABOLISM 1998; 12:67-81. [PMID: 9890062 DOI: 10.1016/s0950-351x(98)80461-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Sex steroids, both androgens and oestrogens, are made from dehydroepiandrosterone (DHEA). The biosynthesis of DHEA from cholesterol entails four steps. First, cholesterol enters the mitochondria with the assistance of a recently described factor called the steroidogenic acute regulatory protein (StAR). Mutations in the StAR gene cause congenital lipoid adrenal hyperplasia. Next, cholesterol is converted to pregnenolone by the cholesterol side chain cleavage enzyme, P450scc. Mutations in the gene for P450scc and for its electron transfer partners, ferredoxin reductase and ferredoxin, have not been described and are probably incompatible with term gestation. Third, pregnenolone undergoes 17 alpha-hydroxylation by microsomal P450c17. Finally, 17-OH pregnenolone is converted to DHEA by the 17,20 lyase activity of P450c17. Isolated 17,20 lyase deficiency is rare, but the identification of its genetic basis and the study of P450c17 enzymology have recently clarified the mechanisms by which DHEA synthesis may be regulated in adrenarche, and have suggested that the lesion underlying polycystic ovary syndrome might involve a serine kinase.
Collapse
Affiliation(s)
- W L Miller
- Department of Paediatrics, University of California, San Francisco 94143-0978, USA
| |
Collapse
|
70
|
Palmer RM, Thompson MG, Meallet C, Thom A, Aitken RP, Wallace JM. Growth and metabolism of fetal and maternal muscles of adolescent sheep on adequate or high feed intake: possible role of protein kinase C-alpha in fetal muscle growth. Br J Nutr 1998; 79:351-7. [PMID: 9624226 DOI: 10.1079/bjn19980059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
From days 4-104 of pregnancy, adolescent sheep, weighing 43.7 (SE 0.87) kg were offered a complete diet at two different intakes (approximately 5 or 15 kg/week) designed to meet slightly, or well above, maternal maintenance requirements. The fetal and maternal muscles were taken on day 104 of pregnancy and analysed for total DNA, RNA and protein. Ewes offered a high intake to promote rapid maternal weight gain, weighed more (76.5 (SE 4.5) v 50.0 (SE 1.7) kg) and had muscles with a greater fresh weight, whilst their fetuses had smaller muscles, than those fed at a lower intake. Plantaris muscle of the ewes fed at the high intake contained more RNA and protein; again the opposite situation was found in the fetal muscle. On the higher maternal intakes, the DNA, RNA and protein contents of the fetal plantaris muscle were less than in fetuses of ewes fed at the lower intake. To investigate the possible mechanisms involved in this decrease in fetal muscle mass, cytosolic and membrane-associated muscle proteins were subjected to Western immunoblotting with antibodies to nine isoforms of protein kinase C (PKC), a family of enzymes known to play an important role in cell growth. Five PKC isoforms (alpha, epsilon, theta, mu, zeta) were identified in fetal muscle. One of these, PKC-alpha was located predominantly in the cytosolic compartment in the smaller fetuses of the ewes fed at a high plane of nutrition, but was present to a greater extent in the membranes of the more rapidly growing fetuses of the ewes fed at the lower intake. This was the only isoform to demonstrate nutritionally related changes in it subcellular compartmentation suggesting that it may mediate some aspects of the change in fetal growth rate.
Collapse
Affiliation(s)
- R M Palmer
- Rowett Research Institute, Bucksburn, Aberdeen, UK.
| | | | | | | | | | | |
Collapse
|
71
|
Huang C, Ma WY, Dong Z. Potentiation of insulin-induced phosphatidylinositol-3 kinase activity by phorbol ester is mediated by protein kinase C epsilon. Cell Signal 1998; 10:185-90. [PMID: 9607141 DOI: 10.1016/s0898-6568(97)00098-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our previous results have demonstrated that phorbol 12-myristate 13-acetate (TPA) and insulin synergistically stimulate the activity of phosphatidylinositol-3 kinase (PI-3 kinase) and PI-3 kinase plays an important role in both of TPA-induced AP-1 activation and cell transformation in tumour promotion sensitive (P+) JB6 cells. In the present study, we investigated the role of PKC and its isozymes in the synergistic induction of PI-3 kinase by TPA and insulin. Bisindolylmaleimide inhibits TPA- and TPA+ insulin-induced PI-3 kinase activity. Pretreatment of cells for 24 h with TPA has significant inhibitory effects on TPA-induced PI-3 kinase activity and abolishes the synergistic effect of TPA and insulin-stimulated PI-3 kinase activity. Furthermore, overexpression of a dominant negative PKC epsilon, but not dominant negative PKC alpha, blocks the synergistic effect of TPA and insulin-induced PI-3 kinase activity. These results indicate that the potentiation effect of TPA on insulin-induced PI-3 kinase activity is specific through PKC epsilon in JB6 cells.
Collapse
Affiliation(s)
- C Huang
- Hormel Institute, University of Minnesota, Austin 55912, USA
| | | | | |
Collapse
|
72
|
Donnelly R, Qu X. Mechanisms of insulin resistance and new pharmacological approaches to metabolism and diabetic complications. Clin Exp Pharmacol Physiol 1998; 25:79-87. [PMID: 9493493 DOI: 10.1111/j.1440-1681.1998.tb02181.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. Resistance to insulin-mediated glucose transport and metabolism has been identified as a primary mechanism in the pathogenesis of non-insulin-dependent diabetes mellitus (NIDDM) and as a target for drug development. The aetiology of insulin resistance is likely to be multifactorial, but the present review focuses on candidate post-receptor mechanisms of insulin resistance, particularly protein kinase C (PKC), and the metabolic and genetic significance of beta3-adrenoceptors (beta3-AR) in adipose tissue. 2. Multiple lines of evidence suggest that isoform-selective activation of PKC phosphorylates and down-regulates one or more substrates involved in glucose transport and metabolism (e.g. glycogen synthase and the insulin receptor) and recent studies have shown increased expression of calcium-independent isozymes (PKC-epsilon and PKC-theta) in the membrane fraction of skeletal muscle in fructose- and fat-fed rat models of insulin resistance. In addition, there is separate evidence that glucose-induced PKC activation plays an important role in the micro- and macrovascular complications of diabetes. 3. New pharmacological approaches to NIDDM and obesity have focused on insulin-sensitizing agents (e.g. troglitazone), beta3-AR agonists, anti-lipolytic drugs (e.g. the adenosine A1 receptor agonist GR79236) and selective inhibitors of PKC isoforms (e.g. the inhibitor of PKC-beta LY333531). Experimental studies with GR79236 show that this drug ameliorates the hypertriglyceridaemia induced by fructose feeding and that the reduction in fatty acid levels is associated with secondary improvements in glucose tolerance. 4. Recent insights into the pathogenesis of NIDDM and its associated complications have been used to develop a range of new therapeutic agents that are currently showing promise in clinical and preclinical development.
Collapse
Affiliation(s)
- R Donnelly
- Department of Pharmacology, University of Sydney, New South Wales, Australia.
| | | |
Collapse
|
73
|
Li S, Termini J, Hayward A, Siddle K, Zick Y, Koval A, LeRoith D, Fujita-Yamaguchi Y. The carboxyl-terminal domain of insulin-like growth factor-I receptor interacts with the insulin receptor and activates its protein tyrosine kinase. FEBS Lett 1998; 421:45-9. [PMID: 9462837 DOI: 10.1016/s0014-5793(97)01523-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Receptors for insulin and insulin-like growth factor-I (IR and IGFIR) consisting of the alpha2beta2 structure are protein tyrosine kinases (PTKs). Carboxyl-terminal (CT) domains of their beta subunits are structurally diverse while the PTK domains share the highest homology. Interactions between CT and PTK domains of IR and IGFIR were studied by means of PTK activity, fluorescence energy transfer or surface plasmon resonance using BIAcore. We present evidence that IGFIR CT directly interacts with both IGFIR and IR. Although binding to both receptors, stimulation of PTK activity only occurs with IR but not IGFIR.
Collapse
Affiliation(s)
- S Li
- Department of Molecular Biology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | |
Collapse
|
74
|
De Fea K, Roth RA. Modulation of insulin receptor substrate-1 tyrosine phosphorylation and function by mitogen-activated protein kinase. J Biol Chem 1997; 272:31400-6. [PMID: 9395471 DOI: 10.1074/jbc.272.50.31400] [Citation(s) in RCA: 239] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Increased serine phosphorylation of insulin receptor substrate-1 (IRS-1) has been observed in several systems to correlate with a decreased ability of the insulin receptor to tyrosine-phosphorylate this endogenous substrate and to inhibit its subsequent association with phosphatidylinositol 3-kinase. In the present studies we have examined the potential role of the mitogen-activated protein (MAP) kinase in the increased serine phosphorylation of IRS-1 observed in human embryonic kidney cells treated with an activator of protein kinase C, phorbol 12-myristate 13-acetate. First, recombinantly produced kinase was shown to phosphorylate intact IRS-1 in a way that decreased the ability of isolated insulin receptor to phosphorylate the tyrosines recognized by the SH2 domains of the phosphatidylinositol 3-kinase. Second, an inhibitor of MAP kinase activation, PD98059, blocked the phorbol 12-myristate 13-acetate-induced inhibition of the insulin-stimulated increase in IRS-1 associated phosphatidylinositol 3-kinase. Third, activation of MAP kinase in intact cells via a regulatable upstream kinase, a RAF:estrogen receptor construct, could also inhibit the insulin-stimulated increase in IRS-1-associated phosphatidylinositol 3-kinase. Fourth, an in gel kinase assay showed that MAP kinase was the primary renaturable kinase in cell extracts capable of phosphorylating an IRS-1 fusion protein. Finally, IRS-1 was found to associate in coprecipitation studies with endogenous MAP kinase. These studies implicate MAP kinase as one of the kinases capable of phosphorylating and regulating IRS-1 tyrosine phosphorylation.
Collapse
Affiliation(s)
- K De Fea
- Department of Molecular Pharmacology, Stanford University School of Medicine, Stanford, California 94305, USA
| | | |
Collapse
|
75
|
Kellerer M, Mushack J, Mischak H, Häring HU. Protein kinase C (PKC) epsilon enhances the inhibitory effect of TNF alpha on insulin signaling in HEK293 cells. FEBS Lett 1997; 418:119-22. [PMID: 9414108 DOI: 10.1016/s0014-5793(97)01357-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recently we have shown that PKC beta1 and beta2 are able to inhibit the tyrosine kinase activity of the human insulin receptor (HIR). Now we have investigated whether a distinct PKC isoform might be involved in the inhibitory effect of TNF alpha on insulin signaling in HEK293 cells. TNF alpha induces a rapid translocation of the PKC isoform epsilon (TNF alpha 10(-9) M, maximal effect within 5-10 min) in rat-1 fibroblasts, while no effect occurred on other isoforms. Cotransfection of HIR with PKC epsilon did not significantly reduce the insulin stimulated receptor kinase activity; however, when cells were incubated with TNF alpha for 10 min (10(-9) M) a 62 +/- 17% (n = 5) inhibition of the insulin receptor kinase activity was observed which was significantly (P<0.01) higher than that observed in cells which were not transfected with PKC (32 +/- 11.5%, n = 5). The data suggest that translocation of PKC epsilon induced by TNF alpha enables this PKC isoform to interact with insulin signaling and to inhibit the insulin receptor kinase activity.
Collapse
Affiliation(s)
- M Kellerer
- Eberhard-Karls-Universität, Med. Klinik u. Poliklinik, Abt. IV, Innere Medizin, Tübingen, Germany
| | | | | | | |
Collapse
|
76
|
Schmitz-Peiffer C, Oakes ND, Browne CL, Kraegen EW, Biden TJ. Reversal of chronic alterations of skeletal muscle protein kinase C from fat-fed rats by BRL-49653. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:E915-21. [PMID: 9374677 DOI: 10.1152/ajpendo.1997.273.5.e915] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have recently shown that the reduction in insulin sensitivity of rats fed a high-fat diet is associated with the translocation of the novel protein kinase C epsilon (nPKC epsilon) from cytosolic to particulate fractions in red skeletal muscle and also the downregulation of cytosolic nPKC theta. Here we have further investigated the link between insulin resistance and PKC by assessing the effects of the thiazolidinedione insulin-sensitizer BRL-49653 on PKC isoenzymes in muscle. BRL-49653 increased the recovery of nPKC isoenzymes in cytosolic fractions of red muscle from fat-fed rats, reducing their apparent activation and/or downregulation, whereas PKC in control rats was unaffected. Because BRL-49653 also improves insulin-stimulated glucose uptake in fat-fed rats and reduces muscle lipid storage, especially diglyceride content, these results strengthen the association between lipid availability, nPKC activation, and skeletal muscle insulin resistance and support the hypothesis that chronic activation of nPKC isoenzymes is involved in the generation of muscle insulin resistance in fat-fed rats.
Collapse
Affiliation(s)
- C Schmitz-Peiffer
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | | | | | | |
Collapse
|
77
|
Vielmetter J, Chen XN, Miskevich F, Lane RP, Yamakawa K, Korenberg JR, Dreyer WJ. Molecular characterization of human neogenin, a DCC-related protein, and the mapping of its gene (NEO1) to chromosomal position 15q22.3-q23. Genomics 1997; 41:414-21. [PMID: 9169140 DOI: 10.1006/geno.1997.4688] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Neogenin was first identified in the chick embryo, and like a number of cell surface proteins of the immunoglobulin (Ig) superfamily, including N-CAM and L1 (generally called cell adhesion molecules or CAMs), it is expressed on growing nerve cells in the developing nervous system of vertebrate embryos. Neogenin is also expressed in other embryonic tissues, suggesting a more general role in developmental processes such as tissue growth regulation, cell-cell recognition, and cell migration. Neogenin, unlike the CAMs, is closely related to a unique tumor suppressor candidate molecule, deleted in colorectal carcinoma (DCC). Like DCC, the neogenin protein consists of four immunoglobulin-like (Ig-like) domains followed by six fibronectin type III domains, a transmembrane domain, and an intracellular domain. We now report the cloning and sequencing of cDNA clones coding for the human neogenin protein. Human neogenin shares 87% identity with its chicken homolog, and like its chicken counterpart it is expressed in at least two different isoforms derived from alternative splicing in the intracellular domain. Northern blot analysis revealed two mRNA species of about 5 and 7 kb. The chromosomal location of the human neogenin gene (HGMW-approved symbol NEO1) was determined as 15q22.3-q23, using fluorescence in situ hybridization. The gene therefore maps in the vicinity of a locus associated with Bardet-Biedl syndrome. The identification of human neogenin and its chromosomal location provides a basis for studying its involvement in genetic disorders or diseases.
Collapse
MESH Headings
- Alternative Splicing
- Amino Acid Sequence
- Animals
- Base Sequence
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/physiology
- Cell Differentiation/genetics
- Cell Differentiation/physiology
- Chickens
- Chromosome Mapping
- Chromosomes, Human, Pair 15/genetics
- Cloning, Molecular
- DCC Receptor
- DNA Primers/genetics
- DNA, Complementary/genetics
- Humans
- In Situ Hybridization, Fluorescence
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Molecular Sequence Data
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Sequence Homology, Amino Acid
- Species Specificity
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- J Vielmetter
- Division of Biology 156-29, California Institute of Technology, Pasadena 91125, USA.
| | | | | | | | | | | | | |
Collapse
|
78
|
Abstract
Presenting symptoms of hirsutism and virilism often signal a disorder of androgen biosynthesis, especially one of the forms of adrenal hyperplasia. The genetics and physiology of the various disorders are reviewed, emphasizing those that results in increased adrenal androgen production. All of these disorders can be diagnosed genetically, permitting family counseling, and all can be treated successfully with appropriate hormonal replacement therapy. Premature adrenarche is not caused by an enzymatic disorder; its origins remain obscure but may be an early harbinger of the polycystic ovary syndrome.
Collapse
Affiliation(s)
- W L Miller
- Department of Pediatrics, University of California, San Francisco, USA
| |
Collapse
|
79
|
Abstract
1. Protein Kinase C represents a family of Ca(2+)- and phospholipid-dependent enzymes which catalyzes the covalent transfer of phosphate from ATP to serine and threonine residues on proteins. Phosphorylation of the substrate protein induces a conformational change and thereby a modification of its functional properties. 2. PKC family consists of at least twelve members, divided in three subgroups:classical PKCs, (alpha, beta I, beta II, gamma), new PKCs, (delta, epsilon, eta, theta, mu) and atypical PKCs, (zeta, lambda, iota). The three subgroups differ in cofactors requirements and tissue expression; these differences in co-activators dependency and regional distribution account for the differential activation profile of the various PKC isoenzymes. 3. Different molecules involved in the intracellular signaling network are phosphorylated "in vitro" and "in vivo" by PKC. Many target proteins show a preferential pattern of phosphorylation by the different PKC isotypes. 4. Through phosphorylation PKC modulates the functional activity of many different intracellular signaling systems which transport extracellular messages from the membrane to the nucleus. 5. The induction of apoptotic processes by the protein kinase inhibitor staurosporine indicates a possible role for PKC in the modulation of the intracellular mechanisms leading to Programmed Cell Death. 6. Abnormalities in both levels and activity of PKC, recently found in some chronic neurodegenerative syndromes, lead to the possibility that PKC dysfunction could be involved in the pathogenetic mechanisms of disease.
Collapse
|
80
|
Huang C, Schmid PC, Ma WY, Schmid HH, Dong Z. Phosphatidylinositol-3 kinase is necessary for 12-O-tetradecanoylphorbol-13-acetate-induced cell transformation and activated protein 1 activation. J Biol Chem 1997; 272:4187-94. [PMID: 9020132 DOI: 10.1074/jbc.272.7.4187] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Phorbol esters, which activate isoforms of protein kinase C, are general activators of the transcription factor activated protein 1 (AP-1). The pathway involved in this signal transduction is not very clear. Currently, little is known about whether phosphatidylinositol-3 (PI-3) kinase plays any role in phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced signal transduction. We demonstrate here that TPA not only has markedly synergistic effects on insulin-induced PI-3 kinase activity, but it also can induce PI-3 kinase activity and the PI-3 phosphates by itself. We also found that insulin, a PI-3 kinase activator, enhanced TPA-induced AP-1 trans-activation and transformation in JB6 promotion-sensitive cells. Furthermore, wortmannin and LY294002, two PI-3 kinase inhibitors, markedly decreased AP-1 activity induced by insulin, TPA, or TPA and insulin and inhibited JB6 promotion-sensitive cell transformation induced by TPA or TPA and insulin. Most importantly, constitutive overexpression of the dominant negative PI-3 kinase P85 mutants completely blocked insulin- or TPA-induced AP-1 trans-activation and TPA-induced cell transformation. All evidence from present studies suggests that PI-3 kinase acts as a mediator in TPA-induced AP-1 activation and transformation in JB6 cells.
Collapse
Affiliation(s)
- C Huang
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | | | | | | | | |
Collapse
|
81
|
Kole HK, Liotta AS, Kole S, Roth J, Montrose-Rafizadeh C, Bernier M. A synthetic peptide derived from a COOH-terminal domain of the insulin receptor specifically enhances insulin receptor signaling. J Biol Chem 1996; 271:31619-26. [PMID: 8940181 DOI: 10.1074/jbc.271.49.31619] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The role of the insulin receptor COOH-terminal domain in the regulation of insulin signal transduction was explored with a variety of synthetic peptides. One of the peptides, termed peptide HC, whose structure corresponds to residues 1293-1307 of the insulin proreceptor sequence, enhanced insulin-stimulated autophosphorylation of the insulin receptor in cell-free systems and in semipermeabilized Chinese hamster ovary (CHO) cells that had been transfected with an expression plasmid encoding the human insulin receptor (CHO/HIRc) at concentrations where there was no detectable effect on basal autophosphorylation levels or on receptor dephosphorylation. A lipophilic analogue of peptide HC, stearyl peptide HC, added to intact CHO/HIRc cells enhanced significantly insulin-stimulated insulin receptor autophosphorylation while having no effect on ligand-stimulated receptor phosphorylation in CHO cells overexpressing either the IGF-1 receptor or epidermal growth factor receptor. Addition of stearyl peptide HC to CHO/HIRc cells resulted in a 2.4 +/- 0.3-fold increase in the amount of insulin-stimulated phosphatidylinositol 3-kinase detected in anti-IRS-1 immunoprecipitates and a 2.1 +/- 0.6-fold increase in the levels of tyrosine phosphorylation of mitogen-activated protein kinase in response to insulin. Finally, a derivative of peptide HC coupled to a biotin moiety was prepared and showed to bind with the beta-subunit of the wild-type insulin receptor and a truncated receptor that lacks 43 amino acids from its carboxyl terminus. However, there was little binding, if any, of the peptide with the IGF-1 receptors or the epidermal growth factor receptors. Taken together, our data demonstrate that a pentadecapeptide related to the carboxyl terminus of the insulin receptor binds to the insulin receptor beta-subunit and that this interaction may contribute to the increased receptor's intrinsic activity and signal transduction.
Collapse
Affiliation(s)
- H K Kole
- Diabetes Section, Laboratory of Clinical Physiology, NIA, National Institutes of Health, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | |
Collapse
|
82
|
Schubert C, Carel K, DePaolo D, Leitner W, Draznin B. Interactions of protein kinase C with insulin signaling. Influence on GAP and Sos activities. J Biol Chem 1996; 271:15311-4. [PMID: 8663173 DOI: 10.1074/jbc.271.26.15311] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In this study, we investigated the influence of the protein kinase C (PKC)-dependent system upon the ability of insulin to stimulate p21(ras).GTP loading in 3T3-L1 adipocytes. Activation of PKC by 12-0-tetradecanoylphorbol-13-acetate (TPA) did not affect the basal amount of p21(ras).GTP but significantly reduced insulin-induced increases in p21(ras).GTP. This reduction was due to inhibition of the insulin's ability to stimulate guanine nucleotide exchange activity of Sos in cells incubated with 100 nM TPA for either 30 min or 3 h. TPA had no effect on basal activity of Sos. Depletion of PKC by an 18-h incubation with TPA or inhibition by bisindolylmaleimide resulted in profound inhibition of the insulin-induced p21(ras).GTP loading. In contrast to PKC activation, removal of PKC did not influence Sos activity but resulted in a 2-fold stimulation of GTPase activating protein (GAP). This effect of PKC depletion is unique to 3T3-L1 adipocytes and was not observed in either 3T3-L1 fibroblasts or Rat-1 fibroblasts. Thus, it appears that in 3T3-L1 adipocytes, PKC has a constitutive inhibitory effect on GAP that permits insulin to activate Sos and p21(ras). Removal of this inhibitory influence activates GAP and reduces insulin-stimulated p21(ras).GTP loading.
Collapse
Affiliation(s)
- C Schubert
- Medical Research Service, Veterans Affairs Medical Center and the University of Colorado Health Sciences Center, Denver, Colorado 80220, USA
| | | | | | | | | |
Collapse
|
83
|
Standaert ML, Bandyopadhyay G, Galloway L, Farese RV. Effects of phorbol esters on insulin-induced activation of phosphatidylinositol 3-kinase, glucose transport, and glycogen synthase in rat adipocytes. FEBS Lett 1996; 388:26-8. [PMID: 8654582 DOI: 10.1016/0014-5793(96)00492-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In rat adipocytes, phorbol ester-induced activation of PKC did not inhibit insulin signalling through IRS-1-dependent phosphatidylinositol (PI) 3-kinase activation. Moreover, phorbol esters alone provoked an increase in membrane PI 3-kinase activity. These findings may be relevant to the failure of phorbol esters to inhibit insulin effects on glucose transport and glycogen synthesis in rat adipocytes.
Collapse
Affiliation(s)
- M L Standaert
- Research Service, J.A. Haley Veterans' Hospital, Tampa, FL 33612, USA
| | | | | | | |
Collapse
|
84
|
Abstract
In nearly all mammalian cells and tissues examined, protein kinase C (PKC) has been shown to serve as a major regulator of a phosphatidylcholine-specific phospholipase D (PLD) activity. At least 12 distinct isoforms of PKC have been described so far; of these enzymes only the alpha- and beta-isoforms were found to regulate PLD activity. While the mechanism of this regulation has remained unknown, available evidence suggests that both phosphorylating and non-phosphorylating mechanisms may be involved. A phosphatidylcholine-specific PLD activity was recently purified from pig lung, but its possible regulation by PKC has not been reported yet. Several cell types and tissues appear to express additional forms of PLD which can hydrolyze either phosphatidylethanolamine or phosphatidylinositol. It has also been reported that at least one form of PLD can be activated by oncogenes, but not by PKC activators. Similar to activated PKC, some of the primary and secondary products of PLD-mediated phospholipid hydrolysis, including phosphatidic acid, 1,2-diacylglycerol, choline phosphate and ethanolamine, also exhibit mitogenic/co-mitogenic effects in cultured cells. Furthermore, both the PLD and PKC systems have been implicated in the regulation of vesicle transport and exocytosis. Recently the PLD enzyme has been cloned and the tools of molecular biology to study its biological roles will soon be available. Using specific inhibitors of growth regulating signals and vesicle transport, so far no convincing evidence has been reported to support the role of PLD in the mediation of any of the above cellular effects of activated PKC.
Collapse
Affiliation(s)
- Z Kiss
- Hormel Institute, University of Minnesota, Austin 55912, USA
| |
Collapse
|
85
|
Young SW, Dickens M, Tavaré JM. Activation of mitogen-activated protein kinase by protein kinase C isotypes alpha, beta I and gamma, but not epsilon. FEBS Lett 1996; 384:181-4. [PMID: 8612819 DOI: 10.1016/0014-5793(96)00287-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Treatment of CHO.T cells with either PMA or insulin led to the activation of MAP kinase by approximately 3-fold, and p90rsk by approximately 4-fold. Over-expression of the alpha, beta I or gamma isoforms of protein kinase C caused a substantial enhancement of the effect of PMA on the activation of MAP kinase and p90rsk, however, the effect of insulin was unchanged. Over-expression of the epsilon isoform of protein kinase C did not alter the effect of either PMA or insulin on the activation of MAP kinase and p90rsk. The results suggest that protein kinase C isotypes, alpha, beta I and gamma, but not epsilon, can mediate MAP kinase activation by PMA, and strongly support the hypothesis that protein kinase C isoforms can initiate distinct signalling pathways.
Collapse
Affiliation(s)
- S W Young
- Department of Biochemistry, School of Medical Sciences, University of Bristol, UK
| | | | | |
Collapse
|
86
|
Flynn A, Proud CG. Insulin and phorbol ester stimulate initiation factor eIF-4E phosphorylation by distinct pathways in Chinese hamster ovary cells overexpressing the insulin receptor. EUROPEAN JOURNAL OF BIOCHEMISTRY 1996; 236:40-7. [PMID: 8617284 DOI: 10.1111/j.1432-1033.1996.00040.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have developed a one-dimensional isoelectric focusing technique to measure changes in the steady-state phosphorylation of the cap-binding initiation factor, eIF-4E. We have used a Chinese hamster ovary cell line transfected with the human insulin receptor (CHO.T cells) to study the regulation of eIF-4E phosphorylation by insulin and other stimuli. Exposure of CHO.T cells to insulin, phorbol ester or serum resulted in a rapid increase (up to twofold) in eIF-4E phosphorylation. As a control, we have also performed experiments with the parental cell line, CHO.K1 cells, in which both serum and phorbol ester, but not nanomolar concentrations of insulin, produce similar changes in eIF-4E phosphorylation. We have used two complementary approaches to study the role of protein kinase C (PKC) in these responses: a highly specific inhibitor of PKC and down-regulation of PKC by prior treatment of the cells with phorbol ester. In CHO.T cells, both approaches indicate that PKC is required for the response to phorbol ester but that insulin and serum each increase eIF-4E phosphorylation by a mechanism(s) independent of this protein kinase. Similarly, PKC is necessary for the effects of phorbol ester, but not of serum, on eIF-4E phosphorylation in CHO.K1 cells. These data indicate that multiple signal transduction mechanisms are involved in the modulation of eIF-4E phosphorylation and the implications of these findings are discussed.
Collapse
Affiliation(s)
- A Flynn
- Department of Biosciences, University of Kent at Canterbury, United Kingdom
| | | |
Collapse
|
87
|
Brand IA, Kleineke J. Intracellular zinc movement and its effect on the carbohydrate metabolism of isolated rat hepatocytes. J Biol Chem 1996; 271:1941-9. [PMID: 8567642 DOI: 10.1074/jbc.271.4.1941] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The effect of zinc ions on carbohydrate metabolism and intracellular Zn2+ was studied in hepatocytes from fed rats. The addition of ZnCl2 to the medium led to an almost 3-fold increase in lactate production and an increase in net glucose production of about 50%. Half-maximal rates occurred at about 40 microM ZnCl2. These effects were not seen with Mn2+, Co2+, or Ni2+ up to 80 microM, whereas Cu2+ at 80 microM and Cd2+ or Pb2+ at 8 microM exhibited similar effects as 80 microM ZnCl2. Changes in intracellular Zn2+ were followed by single cell epifluorescence using zinquin as a specific probe. Intracellular free Zn2+ in isolated hepatocytes was 1.26 +/- 0.27 microM, and the addition of ZnCl2 led to a concentration-dependent increase in epifluorescence. CdCl2 or PbCl2 at 8 microM was as potent as ZnCl2 at 20-80 microM, whereas NiCl2 at 80 microM was without effect. ZnCl2 completely abolished the inhibition of glycolysis by glucagon (cAMP). Glucagon led to a pronounced drop in cytosolic Zn2+. Both glucagon and zinc stimulated glycogenolysis by increasing the phosphorylation of glycogen phosphorylase but acted oppositely on glycolysis. Zinc overcame the inactivation of pyruvate kinase by glucagon without changing the hormone-induced protein phosphorylation. The antagonistic action of zinc and cAMP on glycolysis together with the rapid and marked decrease in free zinc concentration induced by glucagon (cAMP) may indicate an as yet unknown role of zinc as an important mediator of regulation of carbohydrate metabolism.
Collapse
Affiliation(s)
- I A Brand
- Abteilung Klinische Biochemie, Universitt Göttingen, Federal Republic of Germany
| | | |
Collapse
|
88
|
Manganiello VC, Degerman E, Taira M, Kono T, Belfrage P. Type III cyclic nucleotide phosphodiesterases and insulin action. CURRENT TOPICS IN CELLULAR REGULATION 1996; 34:63-100. [PMID: 8646851 DOI: 10.1016/s0070-2137(96)80003-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- V C Manganiello
- Laboratory of Cellular Metabolism, NHLBI, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
89
|
Issad T, Combettes M, Ferre P. Isoproterenol inhibits insulin-stimulated tyrosine phosphorylation of the insulin receptor without increasing its serine/threonine phosphorylation. EUROPEAN JOURNAL OF BIOCHEMISTRY 1995; 234:108-15. [PMID: 8529629 DOI: 10.1111/j.1432-1033.1995.108_c.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The effect of a beta-adrenergic agonist (isoproterenol) on the tyrosine kinase activity of the insulin receptor was studied in intact adipocytes. Isoproterenol treatment rapidly (5 min) inhibited the insulin-induced autophosphorylation of the insulin receptor on tyrosine residues in intact adipocytes. The effect of insulin on the phosphorylation of cellular proteins on tyrosine residues was also inhibited by isoproterenol. In order to understand the mechanism responsible for this inhibition, two-dimensional phosphopeptide mapping of the insulin receptor was performed. The pattern of phosphorylation of the insulin receptor in freshly isolated adipocytes showed marked differences from that previously observed in cultured cells overexpressing insulin receptors. These differences include a larger proportion of receptors being phosphorylated on the three tyrosines from the kinase domain and no apparent phosphorylation of the two tyrosines close to the C-terminus after insulin stimulation. Isoproterenol markedly inhibited the effect of insulin on the phosphorylation of the three tyrosines from the kinase domain. However, this inhibition was not associated with an increase in the phosphorylation of serine/threonine peptides. Thus, this direct analysis of insulin receptor phosphorylation sites in intact adipocytes does no support the idea that beta-adrenegic agents inhibit the tyrosine kinase activity of the receptor through a serine/threonine phosphorylation-dependent mechanism.
Collapse
Affiliation(s)
- T Issad
- Institut National de la Santé et de la Recherche Médicale Unité 342, Université René Descartes, Paris, France
| | | | | |
Collapse
|
90
|
Abstract
Growth factors are involved in a variety of cellular responses such as growth, differentiation, migration, metabolism, and transformation. Binding of the growth factor to its corresponding cell surface receptor results in activation of the receptor's intrinsic tyrosine kinase activity, and subsequently in activation of complex multistep signal transduction cascades. Activation of these interconnected signaling pathways eventually leads to a biological response, which involves changes in gene expression and protein synthesis. The biological response has been shown to be receptor-specific and also cell-type (tissue)-specific, indicating that various receptors activate distinct signal transduction pathways in one tissue and that one receptor activates different pathways in various tissues. What determines receptor specificity and tissue specificity? In this context, this article will focus on certain receptors with intrinsic tyrosine kinase activity, including receptors for platelet-derived growth factor (PDGF), epidermal growth factor (EGF), insulin, and nerve growth factor (NGF).
Collapse
Affiliation(s)
- K Seedorf
- Department of Molecular Signaling, Hagedorn Research Institute, Gentofte, Denmark
| |
Collapse
|
91
|
Danielsen AG, Liu F, Hosomi Y, Shii K, Roth RA. Activation of protein kinase C alpha inhibits signaling by members of the insulin receptor family. J Biol Chem 1995; 270:21600-5. [PMID: 7545165 DOI: 10.1074/jbc.270.37.21600] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Stimulation of the activity of protein kinase C by pretreatment of cells with phorbol esters was tested for its ability to inhibit signaling by four members of the insulin receptor family, including the human insulin and insulin-like growth factor-I receptors, the human insulin receptor-related receptor, and the Drosophila insulin receptor. Activation of overexpressed protein kinase C alpha resulted in a subsequent inhibition of the ligand-stimulated increase in antiphosphotyrosine-precipitable phosphatidylinositol 3-kinase mediated by the kinase domains of all four receptors. This inhibition varied from 97% for the insulin receptor-related receptor to 65% for the Drosophila insulin receptor. In addition, the activation of protein kinase C alpha inhibited the in situ ligand-stimulated increase in tyrosine phosphorylation of the GTPase-activating protein-associated p60 protein as well as Shc mediated by these receptors. The mechanism for this inhibition was further studied in the case of the insulin-like growth factor-I receptor. Although the in situ phosphorylation of insulin-receptor substrate-1 and p60 by this receptor was inhibited by prior stimulation of protein kinase C alpha, the in vitro tyrosine phosphorylation of these two substrates by this receptor was not decreased by prior stimulation of the protein kinase C alpha in the cells that served as a source of the substrates. Finally, the insulin-like growth factor-I-stimulated increase in cell proliferation was found to be inhibited by prior activation of protein kinase C alpha.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- A G Danielsen
- Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305, USA
| | | | | | | | | |
Collapse
|
92
|
Seedorf K, Shearman M, Ullrich A. Rapid and long-term effects on protein kinase C on receptor tyrosine kinase phosphorylation and degradation. J Biol Chem 1995; 270:18953-60. [PMID: 7642554 DOI: 10.1074/jbc.270.32.18953] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Rapid and long term effects of protein kinase C alpha activation on receptor tyrosine kinase signaling parameters were investigated in human 293 embryonic fibroblasts and mouse NIH 3T3 cells. Within minutes of phorbol 12-myristate 13-acetate treatment, epidermal growth factor receptor and HER2 tyrosine phosphorylation was decreased, while platelet-derived growth factor receptor and insulin receptor autophosphorylation was upregulated. These effects are not mediated by protein kinase C-dependent receptor tyrosine kinase phosphorylation but apparently by activation or inactivation of receptor tyrosine kinase-specific phosphatases, as indicated by neutralization of these phenomena upon treatment of cells with sodium orthovanadate. In contrast to these short term effects, sustained activation of protein kinase C alpha by phorbol 12-myristate 13-acetate results in translocation of protein kinase C from the cytosol to the membrane fraction where it forms stable complexes with all receptor tyrosine kinases investigated. Ligand-induced receptor tyrosine kinase/protein kinase C association in NIH 3T3 fibroblasts is accompanied by a mobility shift of the receptor, indicating phosphorylation by activated protein kinase C. This phenomenon correlates with the disappearance of receptor tyrosine kinases from the cell surface, implying that this interaction plays a role in the process of receptor internalization and degradation. Interestingly, ligand-stimulated receptor down-regulation is also enhanced by overexpression of phospholipase C gamma, which strongly indicates a role for this common receptor tyrosine kinase substrate in negative regulation of growth factor signals.
Collapse
Affiliation(s)
- K Seedorf
- Department of Molecular Biology, Max-Planck-Institut für Biochemie, Martinsried, Germany
| | | | | |
Collapse
|
93
|
Yamaguchi T, Fernandez R, Roth RA. Comparison of the signaling abilities of the Drosophila and human insulin receptors in mammalian cells. Biochemistry 1995; 34:4962-8. [PMID: 7711018 DOI: 10.1021/bi00015a007] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Chimeric receptors encoding either the whole or a portion of the cytoplasmic domain of the drosophila insulin receptor (IR) with the extracellular domain of the human IR were expressed either transiently in COS cells or stably in Chinese hamster ovary cells and compared with the wild-type human IR. All three receptors bound insulin equally and exhibited an insulin-activated tyrosine kinase activity. The ability of the drosophila cytoplasmic domain to mediate the tyrosine phosphorylation of insulin receptor substrate 1, stimulate cell proliferation, and activate MAP kinase was found to be indistinguishable from that of the human IR. The chimeric drosophila receptors did not bind more phosphatidylinositol 3-kinase than the human IR, despite containing a C-terminal extension with potential tyrosine phosphorylation sites in the motif recognized by the SH2 domain of this enzyme. Thus, the essential signal-transducing abilities of the IR appear to have been conserved from invertebrates to mammals, despite the considerable differences in the sequences of these receptors.
Collapse
Affiliation(s)
- T Yamaguchi
- Department of Molecular Pharmacology, Stanford University School of Medicine, California 94305, USA
| | | | | |
Collapse
|
94
|
Abstract
Insulin treatment increases the SN-1,2-diacylglycerol (DAG) concentration in skeletal muscle. Because DAG may participate in transmission or modulation of the insulin receptor signal, we examined the effect of insulin on total DAG and on different DAG species in isolated rat hemidiaphragms incubated with 5 mmol/L glucose. Five DAG species (16:0-18:1 omega 9, 16:0-18:1 omega 7, 18:0-18:1 omega 9, 18:0-18:2 omega 6, and 18:1-18:2) were identified and quantified. After a 5-minute incubation with 60 nmol/L insulin, neither total DAG nor a DAG species increased; exposure to insulin for 10 or 20 minutes increased the concentration of total DAG and of several DAG species. Insulin did not increase DAG in muscles incubated without glucose. Two sources for the insulin-mediated DAG increase were considered: phosphatidylcholine (PC) hydrolysis and de novo DAG synthesis from glucose. Concentrations of choline and phosphocholine in muscle were not increased after 10-minute incubations with insulin. However, insulin increased 14C incorporation from [U-14C]glucose into DAG, triacylglycerol (TAG), and total lipids approximately threefold. Okadaic acid (OKA), an inhibitor of phosphoprotein phosphatases 1 and 2A, increased muscle DAG content and synthesis from glucose, similar to the effect of insulin. Doses of OKA or insulin that increased DAG mass greatly exceeded those required for stimulation of glucose transport. The insulin-mediated, relatively slow increase in muscle DAG observed here likely reflects primarily de novo synthesis from glucose. This effect would be downstream of insulin stimulation of glucose transport. However, a possible insulin-mediated, rapid transient increase in muscle DAG content and PC hydrolysis cannot be ruled out by our studies.
Collapse
Affiliation(s)
- K P Boggs
- Department of Medicine, Medical University of South Carolina, Charleston 29425
| | | |
Collapse
|
95
|
Russ M, Eckel J. Insulin action on cardiac glucose transport: studies on the role of protein kinase C. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1265:73-8. [PMID: 7857987 DOI: 10.1016/0167-4889(94)00200-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Isolated ventricular cardiomyocytes from adult rat have been used to elucidate a possible relationship between protein kinase C (PKC) and the stimulatory action of insulin on cardiac glucose transport. Cells were incubated in the presence of either insulin or phospholipase C from Clostridium perfringens (PLC-Cp) and intracellular sn-1,2-diacylglycerol (DAG) levels and initial rates of 3-O-methylglucose transport were determined. Insulin had no effect on the DAG mass level, whereas it was elevated by PLC-Cp to 200% of control. Under these conditions the hormone produced a 2.7-fold stimulation of glucose transport with no significant effect of PLC-Cp. Insulin was unable to produce a redistribution of PKC, whereas phorbol 12-myristate 13-acetate (PMA) increased membrane associated PKC twofold. The PKC inhibitors tamoxifen and staurosporine did not interfere with glucose transport stimulation by insulin. Furthermore, cells treated with PMA exhibited unaltered basal and maximally insulin stimulated rates of glucose transport. In contrast, at physiological concentrations of insulin the stimulatory action of the hormone was significantly reduced. We conclude from our data that PKC is not involved in insulin action on cardiac glucose transport. However, activation of this enzyme may lead to a modified insulin sensitivity of the cardiac cell.
Collapse
Affiliation(s)
- M Russ
- Laboratory of Molecular Cardiology, Diabetes Research Institute, Düsseldorf, Germany
| | | |
Collapse
|
96
|
Dunaif A. Hyperandrogenic anovulation (PCOS): a unique disorder of insulin action associated with an increased risk of non-insulin-dependent diabetes mellitus. Am J Med 1995; 98:33S-39S. [PMID: 7825639 DOI: 10.1016/s0002-9343(99)80057-6] [Citation(s) in RCA: 141] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Polycystic ovary syndrome is the most common endocrine disorder in women of reproductive age. Recent prevalence estimates suggest that 5-10% of premenopausal women have the full-blown syndrome of hyperandrogenism, chronic anovulation, and polycystic ovaries. Evidence suggests that women with polycystic ovary syndrome have a unique disorder of insulin action and are at increased risk to develop non-insulin-dependent diabetes mellitus. Further, non-insulin-dependent diabetes mellitus in women with polycystic ovary syndrome has a substantially earlier age of onset (third to fourth decades) than it does in the general population (sixth to seventh decades). Studies assessing whether abnormalities in insulin action are intrinsic or secondary to the hormonal milieu have found that insulin-induced receptor autophosphorylation is markedly diminished in approximately 50% of polycystic ovary syndrome women. This defect is unique to women with polycystic ovary syndrome and is not seen in other common insulin-resistant states of obesity and non-insulin-dependent diabetes mellitus. In polycystic ovary syndrome women who have normal receptor autophosphorylation, it remains likely that signaling mechanisms downstream of the receptor are abnormal, since these women are also insulin resistant. This distinctive post-insulin-binding defect appears to be genetic, since it is present in cells removed from the in vivo environment for generations.
Collapse
Affiliation(s)
- A Dunaif
- Department of Medicine, Penn State University College of Medicine, Hershey 17033
| |
Collapse
|
97
|
Liu F, Roth RA. Identification of serines-1035/1037 in the kinase domain of the insulin receptor as protein kinase C alpha mediated phosphorylation sites. FEBS Lett 1994; 352:389-92. [PMID: 7926007 DOI: 10.1016/0014-5793(94)00996-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A new site of serine phosphorylation (Ser-1035/1037) has been identified in the kinase domain of the insulin receptor. Mutant receptors missing these two serines were expressed in Chinese hamster ovary cells overexpressing protein kinase C alpha. These mutant receptors lacked a phorbol ester-stimulated phosphoserine containing tryptic peptide as demonstrated by both high percentage polyacrylamide/urea gel electrophoresis and two-dimensional tlc. Moreover, a synthetic peptide with the sequence of this tryptic peptide was phosphorylated by isolated protein kinase C alpha and co-migrated with the phosphopeptide from in vivo labeled receptor. These results indicate that serine-1035 and/or 1037 in the kinase domain of the insulin receptor are phosphorylated in response to activation of protein kinase C alpha.
Collapse
Affiliation(s)
- F Liu
- Department of Molecular Pharmacology, Stanford University School of Medicine, CA 94305
| | | |
Collapse
|
98
|
Lewis RE, Volle DJ, Sanderson SD. Phorbol ester stimulates phosphorylation on serine 1327 of the human insulin receptor. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(18)47188-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
99
|
Baier G, Baier-Bitterlich G, Meller N, Coggeshall KM, Giampa L, Telford D, Isakov N, Altman A. Expression and biochemical characterization of human protein kinase C-theta. EUROPEAN JOURNAL OF BIOCHEMISTRY 1994; 225:195-203. [PMID: 7925438 DOI: 10.1111/j.1432-1033.1994.00195.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In this study, the recently identified human protein kinase C-theta (PKC-theta) isoform has been biochemically characterized in detail. An antiserum raised against the unique V3 domain of PKC-theta identified an 80-kDa protein in all human T-cell lines tested, in erythroleukemia K562 cells and in histiocytic lymphoma U-937 cells, but not in a B-lymphoma line (Raji) or in several melanoma, carcinoma, schwanoma or astrocytoma lines, confirming, at the protein level, its predominant expression in hematopoietic cell lines, in particular T cells. Immunoreactive PKC-theta was detected almost exclusively in the cytosolic compartment of unstimulated Jurkat T cells. Stimulation with phorbol ester, however, caused rapid translocation to the membrane. In order to compare the properties of PKC-theta with a representative member of the Ca(2+)-dependent PKC enzymes, full-length cDNAs encoding PKC-theta or PKC-alpha were transiently expressed in COS-1 cells, and recombinant enzymes were partially purified via a six-histidine peptide tag. The catalytic activity of these PKC enzymes was assayed against distinct substrates in the absence and presence of known PKC cofactors. Significant differences were found with respect to activation requirements and substrate preferences between PKC-theta and PKC-alpha. Both enzymes were stimulated by phospholipid and phorbol ester, and were active towards a PKC-derived substrate peptide corresponding to the pseudosubstrate site of PKC. In contrast to PKC-alpha, however, full activation of PKC-theta did not require Ca2+, and its basal activity towards histone H1 was not stimulated by lipid cofactors. Additionally, a myelin-basic-protein-(MBP)-derived peptide, which was readily phosphorylated by PKC-alpha, was a poor substrate for PKC-theta. Similar to PKC-alpha, transient PKC-theta overexpression in murine EL4 thymoma cells caused an approximately 2.5-fold increase in the phorbol-12-myristate-13-acetate-induced transcriptional activation of an interleukin-2 promoter-reporter gene construct. The unique expression and functional properties of PKC-theta suggest that it may play a specialized role in T-cell signaling pathways.
Collapse
Affiliation(s)
- G Baier
- La Jolla Institute for Allergy and Immunology
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Zachayus JL, Cherqui G, Plas C. Protein kinase C and insulin receptor beta-subunit serine phosphorylation in cultured foetal rat hepatocytes. Mol Cell Endocrinol 1994; 105:11-20. [PMID: 7529733 DOI: 10.1016/0303-7207(94)90030-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In digitonin-permeabilized cultured foetal hepatocytes, insulin receptor beta-subunit was highly phosphorylated on serine residues in the presence of [gamma-32P]ATP and Ca2+, a process enhanced after short exposure to insulin with no detectable insulin receptor autophosphorylation. By contrast with this situation, experiments performed with isolated foetal insulin receptors revealed an insulin stimulation of both serine phosphorylation and tyrosine autophosphorylation. In permeabilized cells, insulin receptor beta-subunit phosphorylation was increased after a 2-min exposure to phorbol 12-myristate 13-acetate (PMA) prior to applying the permeabilization/phosphorylation step, while it was inhibited by chronic treatment with PMA leading to protein kinase C (PKC) down modulation. The PKC specific inhibitor, GF109203X, strikingly reduced basal and insulin-enhanced phosphorylation of insulin receptor beta-subunit in permeabilized cells, but failed to exert any effect with isolated receptors. Labelling of glycogen from [U-14C]glucose determined 1 h after a 10-min transitory exposure to insulin and/or modulators of PKC activity showed that PMA prevented insulin glycogenic response, whereas GF109203X was ineffective. Thus, although not directly responsible for insulin receptor serine phosphorylation in cultured foetal hepatocytes, PKC physiologically regulates this process which may inhibit insulin receptor tyrosine kinase activity. This regulation is independent of the antagonistic effect of PMA-activated PKC on insulin glycogenic response.
Collapse
Affiliation(s)
- J L Zachayus
- Laboratoire de Biologie, U.F.R. Odontologie, Université Paris 7, Institut Biomédical des Cordeliers, France
| | | | | |
Collapse
|