Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters










Publication year range
1.
Diabetologia ; 49(10): 2368-78, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16924481

ABSTRACT

AIMS/HYPOTHESIS: ATP-sensitive K(+) (K(ATP)) channels couple glucose metabolism to insulin secretion in pancreatic beta cells. In humans, loss-of-function mutations of beta cell K(ATP) subunits (SUR1, encoded by the gene ABCC8, or Kir6.2, encoded by the gene KCNJ11) cause congenital hyperinsulinaemia. Mice with dominant-negative reduction of beta cell K(ATP) (Kir6.2[AAA]) exhibit hyperinsulinism, whereas mice with zero K(ATP) (Kir6.2(-/-)) show transient hyperinsulinaemia as neonates, but are glucose-intolerant as adults. Thus, we propose that partial loss of beta cell K(ATP) in vivo causes insulin hypersecretion, but complete absence may cause insulin secretory failure. MATERIALS AND METHODS: Heterozygous Kir6.2(+/-) and SUR1(+/-) animals were generated by backcrossing from knockout animals. Glucose tolerance in intact animals was determined following i.p. loading. Glucose-stimulated insulin secretion (GSIS), islet K(ATP) conductance and glucose dependence of intracellular Ca(2+) were assessed in isolated islets. RESULTS: In both of the mechanistically distinct models of reduced K(ATP) (Kir6.2(+/-) and SUR1(+/-)), K(ATP) density is reduced by approximately 60%. While both Kir6.2(-/-) and SUR1(-/-) mice are glucose-intolerant and have reduced glucose-stimulated insulin secretion, heterozygous Kir6.2(+/-) and SUR1(+/-) mice show enhanced glucose tolerance and increased GSIS, paralleled by a left-shift in glucose dependence of intracellular Ca(2+) oscillations. CONCLUSIONS/INTERPRETATION: The results confirm that incomplete loss of beta cell K(ATP) in vivo underlies a hyperinsulinaemic phenotype, whereas complete loss of K(ATP) underlies eventual secretory failure.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Hyperinsulinism/genetics , Loss of Heterozygosity , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/genetics , Animals , Blood Glucose/metabolism , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Kinetics , Mice , Mice, Knockout , Potassium Channels/genetics , Receptors, Drug , Sulfonylurea Receptors
2.
Eur J Clin Invest ; 32 Suppl 3: 24-34, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12028372

ABSTRACT

A combination of insulin resistance and pancreatic beta-cell dysfunction underlies most cases of type 2 diabetes. While the interplay of these two impairments is believed to be important in the development and progression of type 2 diabetes, the mechanisms involved are unclear. A number of factors have been suggested as possibly linking insulin resistance and beta-cell dysfunction in the pathogenesis of type 2 diabetes mellitus. Pro-inflammatory cytokines such as tumour necrosis factor-alpha (TNF-alpha) have deleterious effects on both glucose homeostasis and beta-cell function, and can disrupt insulin signalling pathways in both pancreatic beta cells and liver and adipose tissue. The anti-inflammatory activity of the thiazolidinedione anti-diabetic agents is potentially beneficial, given the possible role of pro-inflammatory cytokines in linking insulin resistance with beta-cell dysfunction.


Subject(s)
Adipocytes/metabolism , Cytokines/physiology , Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance/physiology , Islets of Langerhans/physiology , Obesity/metabolism , Cell Cycle/physiology , Diabetes Mellitus, Type 2/etiology , Humans , Insulin/metabolism , Lipid Metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/physiology , Transcription Factors/agonists , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Am J Physiol Endocrinol Metab ; 281(3): E466-71, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500301

ABSTRACT

Amino acids and insulin have anabolic effects in skeletal muscle, but the mechanisms are poorly understood. To test the hypothesis that leucine and insulin stimulate translation initiation in human skeletal muscle by phosphorylating 70-kDa ribosomal protein S6 kinase (p70(S6k)), we infused healthy adults with leucine alone (n = 6), insulin alone (n = 6), or both leucine and insulin (n = 6) for 2 h. p70(S6k) and protein kinase B (PKB) serine(473) phosphorylation were measured in vastus lateralis muscles. Plasma leucine increased from approximately 116 to 343 micromol/l during the leucine-alone and leucine + insulin infusions. Plasma insulin increased to approximately 400 pmol/l during the insulin-alone and leucine + insulin infusions and was unchanged during the leucine-alone infusion. Phosphorylation of p70(S6k) increased 4-fold in response to leucine alone, 8-fold in response to insulin alone, and 18-fold after the leucine + insulin infusion. Insulin-alone and leucine + insulin infusions increased PKB phosphorylation, but leucine alone had no effect. These results show that physiological concentrations of leucine and insulin activate a key mediator of protein synthesis in human skeletal muscle. They suggest that leucine stimulates protein synthesis through a nutrient signaling mechanism independent of insulin, raising the possibility that administration of branched-chain amino acids may improve protein synthesis in insulin-resistant states.


Subject(s)
Insulin/pharmacology , Leucine/pharmacology , Muscle, Skeletal/enzymology , Protein Serine-Threonine Kinases , Ribosomal Protein S6 Kinases/metabolism , Adult , Blood Glucose/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Female , Humans , Insulin/blood , Kinetics , Leucine/blood , Male , Phosphorylation , Phosphoserine/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt
4.
Diabetes ; 50(2): 353-60, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272147

ABSTRACT

Recent findings have demonstrated that the branched-chain amino acid leucine can activate the translational regulators, phosphorylated heat- and acid-stable protein regulated by insulin (PHAS-I) and p70 S6 kinase (p70S6k), in an insulin-independent and rapamycin-sensitive manner through mammalian target of rapamycin (mTOR), although the mechanism for this activation is undefined. It has been previously established that leucine-induced insulin secretion by beta-cells involves increased mitochondrial metabolism by oxidative decarboxylation and allosteric activation of glutamate dehydrogenase (GDH). We now show that these same intramitochondrial events that generate signals for leucine-induced insulin exocytosis are required to activate the mTOR mitogenic signaling pathway by beta-cells. Thus, a minimal model consisting of leucine and glutamine as substrates for oxidative decarboxylation and an activator of GDH, respectively, confirmed the requirement for these two metabolic components and mimicked closely the synergistic interactions achieved by a complete complement of amino acids to activate p70s6k in a rapamycin-sensitive manner. Studies using various leucine analogs also confirmed the close association of mitochondrial metabolism and the ability of leucine analogs to activate p70s6k. Furthermore, selective inhibitors of mitochondrial function blocked this activation in a reversible manner, which was not associated with a global reduction in ATP levels. These findings indicate that leucine at physiological concentrations stimulates p70s6k phosphorylation via the mTOR pathway, in part, by serving both as a mitochondrial fuel and an allosteric activator of GDH. Leucine-mediated activation of protein translation through mTOR may contribute to enhanced beta-cell function by stimulating growth-related protein synthesis and proliferation associated with the maintenance of beta-cell mass.


Subject(s)
Islets of Langerhans/physiology , Leucine/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Protein Biosynthesis/physiology , Protein Kinases , Allosteric Regulation , Amino Acids, Cyclic/pharmacology , Cell Line , Decarboxylation , Enzyme Activation , Glutamate Dehydrogenase/metabolism , Glutamic Acid/physiology , Islets of Langerhans/cytology , Isoleucine/pharmacology , Leucine/metabolism , Leucine/pharmacology , Mitochondria/physiology , Models, Biological , Oxidation-Reduction , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Valine/pharmacology
5.
J Biol Chem ; 276(15): 12162-8, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11154699

ABSTRACT

Lipoprotein lipase (LpL) provides tissues with triglyceride-derived fatty acids. Fatty acids affect beta-cell function, and LpL overexpression decreases insulin secretion in cell lines, but whether LpL is regulated in beta-cells is unknown. To test the hypothesis that glucose and insulin regulate LpL activity in beta-cells, we studied pancreatic islets and INS-1 cells. Acute exposure of beta-cells to physiological concentrations of glucose stimulated both total cellular LpL activity and heparin-releasable LpL activity. Glucose had no effect on total LpL protein mass but instead promoted the appearance of LpL protein in a heparin-releasable fraction, suggesting that glucose stimulates the translocation of LpL from intracellular to extracellular sites in beta-cells. The induction of heparin-releasable LpL activity was unaffected by treatment with diazoxide, an inhibitor of insulin exocytosis that does not alter glucose metabolism but was blocked by conditions that inhibit glucose metabolism. In vitro hyperinsulinemia had no effect on LpL activity in the presence of low concentrations of glucose but increased LpL activity in the presence of 20 mm glucose. Using dual-laser confocal microscopy, we detected intracellular LpL in vesicles distinct from those containing insulin. LpL was also detected at the cell surface and was displaced from this site by heparin in dispersed islets and INS-1 cells. These results show that glucose metabolism controls the trafficking of LpL activity in beta-cells independent of insulin secretion. They suggest that hyperglycemia and hyperinsulinemia associated with insulin resistance may contribute to progressive beta-cell dysfunction by increasing LpL-mediated delivery of lipid to islets.


Subject(s)
Glucose/pharmacology , Heparin/metabolism , Insulin Resistance , Insulin/pharmacology , Islets of Langerhans/drug effects , Lipoprotein Lipase/metabolism , Animals , Cell Line , Cell Membrane/enzymology , Islets of Langerhans/enzymology , Islets of Langerhans/physiopathology , Male , Mice , Mice, Inbred C57BL , Protein Transport
6.
J Biol Chem ; 274(39): 27426-32, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488074

ABSTRACT

Lipoprotein lipase (LPL) provides tissues with fatty acids, which have complex effects on glucose utilization and insulin secretion. To determine if LPL has direct effects on glucose metabolism, we studied mice with heterozygous LPL deficiency (LPL+/-). LPL+/- mice had mean fasting glucose values that were up to 39 mg/dl lower than LPL+/+ littermates. Despite having lower glucose levels, LPL+/- mice had fasting insulin levels that were twice those of +/+ mice. Hyperinsulinemic clamp experiments showed no effect of genotype on basal or insulin-stimulated glucose utilization. LPL message was detected in mouse islets, INS-1 cells (a rat insulinoma cell line), and human islets. LPL enzyme activity was detected in the media from both mouse and human islets incubated in vitro. In mice, +/- islets expressed half the enzyme activity of +/+ islets. Islets isolated from +/+ mice secreted less insulin in vitro than +/- and -/- islets, suggesting that LPL suppresses insulin secretion. To test this notion directly, LPL enzyme activity was manipulated in INS-1 cells. INS-1 cells treated with an adeno-associated virus expressing human LPL had more LPL enzyme activity and secreted less insulin than adeno-associated virus-beta-galactosidase-treated cells. INS-1 cells transfected with an antisense LPL oligonucleotide had less LPL enzyme activity and secreted more insulin than cells transfected with a control oligonucleotide. These data suggest that islet LPL is a novel regulator of insulin secretion. They further suggest that genetically determined levels of LPL play a role in establishing glucose levels in mice.


Subject(s)
Hyperinsulinism/genetics , Hyperlipoproteinemia Type I/physiopathology , Hypoglycemia/genetics , Insulin/metabolism , Islets of Langerhans/enzymology , Lipoprotein Lipase/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Cholesterol/blood , Fatty Acids, Nonesterified/blood , Genotype , Glucose Tolerance Test , Heterozygote , Humans , Hyperlipoproteinemia Type I/blood , Hyperlipoproteinemia Type I/genetics , Insulin Secretion , Insulinoma , Islets of Langerhans/metabolism , Lipoprotein Lipase/genetics , Mice , Mice, Knockout , Pancreatic Neoplasms , Rats , Recombinant Proteins/metabolism , Transfection , Triglycerides/blood , Tumor Cells, Cultured
7.
J Biol Chem ; 274(26): 18702-8, 1999 Jun 25.
Article in English | MEDLINE | ID: mdl-10373483

ABSTRACT

Recent studies have identified a beta-cell insulin receptor that functions in the regulation of protein translation and mitogenic signaling similar to that described for insulin-sensitive cells. These findings have raised the novel possibility that beta-cells may exhibit insulin resistance similar to skeletal muscle, liver, and fat. To test this hypothesis, the effects of tumor necrosis factor-alpha (TNFalpha), a cytokine proposed to mediate insulin resistance by interfering with insulin signaling at the level of the insulin receptor and its substrates, was evaluated. TNFalpha inhibited p70(s6k) activation by glucose-stimulated beta-cells of the islets of Langerhans in a dose- and time-dependent manner, with maximal inhibition observed at approximately 20-50 ng/ml, detected after 24 and 48 h of exposure. Exogenous insulin failed to prevent TNFalpha-induced inhibition of p70(s6k), suggesting a defect in the insulin signaling pathway. To further define mechanisms responsible for this inhibition and also to exclude cytokine-induced nitric oxide (NO) as a mediator, the ability of exogenous or endogenous insulin +/- inhibitors of nitric-oxide synthase (NOS) activity, aminoguanidine or N-monomethyl-L-arginine, was evaluated. Unexpectedly, TNFalpha and also interleukin 1 (IL-1)-induced inhibition of p70(s6k) was completely prevented by inhibitors that block NO production. Western blot analysis verified inducible NOS (iNOS) expression after TNFalpha exposure. Furthermore, the ability of IL-1 receptor antagonist protein, IRAP, to block TNFalpha-induced inhibition of p70(s6k) indicated that activation of intra-islet macrophages and the release of IL-1 that induces iNOS expression in beta-cells was responsible for the inhibitory effects of TNFalpha. This mechanism was confirmed by the ability of the peroxisome proliferator-activated receptor-gamma agonist 15-deoxy-Delta12, 14-prostaglandin J2 to attenuate TNFalpha-induced insulin resistance by down-regulating iNOS expression and/or blocking IL-1 release from activated macrophages. Overall, TNFalpha-mediated insulin resistance in beta-cells is characterized by a global inhibition of metabolism mediated by NO differing from that proposed for this proinflammatory cytokine in insulin-sensitive cells.


Subject(s)
Guanidines/pharmacology , Insulin Resistance , Islets of Langerhans/drug effects , Nitric Oxide Synthase/biosynthesis , Nitric Oxide/metabolism , Prostaglandin D2/analogs & derivatives , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazolidinediones , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Chromans/pharmacology , Hypoglycemic Agents/pharmacology , Male , Nitric Oxide Synthase Type II , Phosphorylation , Prostaglandin D2/pharmacology , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/metabolism , Thiazoles/pharmacology , Troglitazone
8.
J Biol Chem ; 273(43): 28178-84, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9774438

ABSTRACT

Amino acids have been identified as important signaling molecules involved in pancreatic beta-cell proliferation, although the cellular mechanism responsible for this effect is not well defined. We previously reported that amino acids are required for glucose or exogenous insulin to stimulate phosphorylation of PHAS-I (phosphorylated heat- and acid-stable protein regulated by insulin), a recently discovered regulator of translation initiation during cell mitogenesis. Here we demonstrate that essential amino acids, in particular branched-chain amino acids (leucine, valine, and isoleucine), are largely responsible for mediating this effect. The transamination product of leucine, alpha-ketoisocaproic acid, also stimulates PHAS-I phosphorylation although the transamination products of isoleucine and valine are ineffective. Since amino acids are secretagogues for insulin secretion by beta-cells, we investigated whether endogenous insulin secreted by beta-cells is involved. Interestingly, branched-chain amino acids stimulate phosphorylation of PHAS-I independent of endogenous insulin secretion since genistein (10 microM) and herbimycin A (1 microM), two tyrosine kinase inhibitors in the insulin signaling pathway, exert no effect on amino acid-induced phosphorylation of PHAS-I. Furthermore, branched-chain amino acids retain their ability to induce phosphorylation of PHAS-I under conditions that block insulin secretion from beta-cells. In exploring the signaling pathway responsible for these effects, we find that rapamycin (25 nM) inhibits the ability of branched-chain amino acids to stimulate the phosphorylation of PHAS-I and p70(s6) kinase, suggesting that the mammalian target of rapamycin signaling pathway is involved. The branched-chain amino acid, leucine, also exerts similar effects on PHAS-I phosphorylation in isolated pancreatic islets. In addition, we find that amino acids are necessary for insulin-like growth factor (IGF-I) to stimulate the phosphorylation of PHAS-I indicating that a requirement for amino acids may be essential for other beta-cell growth factors in addition to insulin and IGF-I to activate this signaling pathway. We propose that amino acids, in particular branched-chain amino acids, may promote beta-cell proliferation either by stimulating phosphorylation of PHAS-I and p70(s6k) via the mammalian target of rapamycin pathway and/or by facilitating the proliferative effect mediated by growth factors such as insulin and IGF-I.


Subject(s)
Amino Acids, Branched-Chain/pharmacology , Carrier Proteins , Islets of Langerhans/drug effects , Phosphoproteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Animals , Cell Division , Drug Synergism , Gene Expression Regulation , Growth Substances/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Male , Phosphorylation/drug effects , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction , Sirolimus/pharmacology
9.
Diabetes ; 47(4): 583-91, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9568691

ABSTRACT

Interleukin-1beta (IL-1beta) has been implicated as an effector molecule of beta-cell destruction in autoimmune diabetes. IL-1beta inhibits insulin secretion from pancreatic beta-cells by stimulating the expression of inducible nitric oxide synthase (iNOS) that generates the free radical nitric oxide. IL-1beta also induces the coexpression of the inducible isoform of cyclooxygenase (COX-2) that results in the overproduction of proinflammatory prostaglandins. The current studies were designed to characterize the involvement of protease(s) in the signaling pathway of IL-1beta-induced iNOS and COX-2 expression by rat islets and transformed rat pancreatic beta-cells. Because of the limitations of cell numbers of purified primary beta-cells obtained from rat islets, biochemical and molecular studies were performed using the rat insulinoma beta-cell line RINm5F. A serine protease inhibitor, Nalpha-P-tosyl-L-lysine chloromethyl ketone (TLCK), and a proteasome complex (26S) inhibitor, MG 132, inhibited IL-1beta-induced nitrite formation, an oxidation product of nitric oxide produced by iNOS, in a concentration-dependent manner, with complete inhibition observed at 100 micromol/l and 10 micromol/l, respectively. Both TLCK and MG 132 also inhibited iNOS gene expression at the level of mRNA and protein. In an analogous manner, TLCK (100 micromol/l) and MG 132 (10 micromol/l) inhibited IL-1beta-induced COX-2 enzyme activity (PGE2 formation) and COX-2 gene expression at the level of mRNA and protein. In human islets, the proteasome inhibitor MG 132 also inhibited the formation of the products of iNOS and COX-2 enzyme activity, nitrite, and PGE2, respectively. These findings suggest that the inhibitory action of TLCK and MG 132 on iNOS and COX-2 expression precedes transcription. The transcription factor NFkappaB is essential for activation of a number of cytokine-inducible enzymes and was evaluated as a possible site of protease action necessary for IL-1beta-induced coexpression of iNOS and COX-2. TLCK and MG 132 inhibited both IL-1beta-induced activation of NFkappaB and degradation of IkappaBalpha by islets and RINm5F cells. These results implicate protease activation as an early signaling event in IL-1beta-induced inhibition of beta-cell function. This study also suggests that IL-1beta-induced iNOS and COX-2 coexpression by pancreatic beta-cells share a common signaling pathway in utilizing the proteasome complex (26S) and the transcription factor NFkappaB, and it identifies sites of intervention to prevent the overproduction of their inflammatory products.


Subject(s)
Dinoprostone/biosynthesis , Interleukin-1/metabolism , Islets of Langerhans/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Animals , Cell Line, Transformed , Cyclooxygenase 2 , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , In Vitro Techniques , Islets of Langerhans/enzymology , Isoenzymes/biosynthesis , Leupeptins/pharmacology , Male , Membrane Proteins , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phosphorylation , Prostaglandin-Endoperoxide Synthases/biosynthesis , RNA, Messenger/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Tosyllysine Chloromethyl Ketone/pharmacology , Tumor Cells, Cultured
10.
J Biol Chem ; 273(8): 4485-91, 1998 Feb 20.
Article in English | MEDLINE | ID: mdl-9468502

ABSTRACT

Although glucose regulates the biosynthesis of a variety of beta cell proteins at the level of translation, the mechanism responsible for this effect is unknown. We demonstrate that incubation of pancreatic islets with elevated glucose levels results in rapid and concentration-dependent phosphorylation of PHAS-I, an inhibitor of mRNA cap-binding protein, eukaryotic initiation factor (eIF)-4E. Our initial approach was to determine if this effect is mediated by the metabolism of glucose and activation of islet cell protein kinases, or whether insulin secreted from the beta cell stimulates phosphorylation of PHAS-I via an insulin-receptor mechanism as described for insulin-sensitive cells. In support of the latter mechanism, inhibitors of islet cell protein kinases A and C exert no effect on glucose-stimulated phosphorylation of PHAS-I, whereas the phosphatidylinositol 3-kinase inhibitor, wortmannin, the immunosuppressant, rapamycin, and theophylline, a phosphodiesterase inhibitor, promote marked dephosphorylation of PHAS-I. In addition, exogenous insulin and endogenous insulin secreted by the beta cell line, betaTC6-F7, increase phosphorylation of PHAS-I, suggesting that beta cells of the islet, in part, mediate this effect. Studies with beta cell lines and islets indicate that amino acids are required for glucose or exogenous insulin to stimulate the phosphorylation of PHAS-I, and amino acids alone dose-dependently stimulate the phosphorylation of PHAS-I, which is further enhanced by insulin. Furthermore, rapamycin inhibits by approximately 62% the increase in total protein synthesis stimulated by high glucose concentrations. These results indicate that glucose stimulates PHAS-I phosphorylation via insulin interacting with its own receptor on the beta cell which may serve as an important mechanism for autoregulation of protein synthesis by translation.


Subject(s)
Carrier Proteins , Glucose/metabolism , Insulin/pharmacology , Islets of Langerhans/drug effects , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Animals , Cells, Cultured , Eukaryotic Initiation Factor-4E , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Male , Peptide Initiation Factors/metabolism , Phosphorylation , Protein Binding , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction
11.
J Biol Chem ; 273(6): 3308-13, 1998 Feb 06.
Article in English | MEDLINE | ID: mdl-9452447

ABSTRACT

Diabetic patients with hyperglycemia (high blood glucose) have frequent and persistent bacterial infections linked to significantly diminished bactericidal activity and macrophage function. Interleukin-1 (IL-1), released primarily from activated macrophages, is a key mediator of effective host defense against microorganisms. We observe that hyperglycemic levels of D-glucose (8-20 mM) inhibit the release of IL-1 by lipopolysaccharide-stimulated RAW 264.7 murine macrophage cells. An inhibitor of glucose transport and metabolism, 2-deoxyglucose, prevents this inhibition of IL-1 release. High levels (8-20 mM) of fructose and mannose (but not galactose or L-glucose) also inhibit the release of IL-1 activity, suggesting that metabolism is required for IL-1 inhibition. Immunoprecipitation and activity measurements demonstrate that high glucose levels block the release of IL-1 but do not inhibit IL-1 production. High glucose levels (20 mM) increase protein kinase C (PKC) activity, and inhibitors of PKC block the inhibitory effects of glucose. Phorbol 12-myristate 13-acetate, an agonist of PKC, mimics glucose-induced inhibition of IL-1 release. These results demonstrate that high glucose levels inhibit IL-1 release (but not production) by RAW 264. 7 murine macrophages, and this inhibition is mediated by PKC activation. These studies suggest that persistent infections in hyperglycemic patients may be due to an inhibition of IL-1 release from macrophages.


Subject(s)
Blood Glucose/metabolism , Hyperglycemia/blood , Interleukin-1/antagonists & inhibitors , Macrophages/metabolism , Protein Kinase C/metabolism , Animals , Binding, Competitive , Cell Line , Deoxyglucose/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 2 , Interleukin-1/metabolism , Macrophages/drug effects , Macrophages/enzymology , Mice , Monosaccharide Transport Proteins/metabolism , Nitric Oxide/metabolism , Protein Kinase C/antagonists & inhibitors
12.
Biochim Biophys Acta ; 1361(3): 313-20, 1997 Oct 24.
Article in English | MEDLINE | ID: mdl-9375806

ABSTRACT

The cytokine interleukin-1beta (IL-1beta) has been shown to inhibit insulin secretion and destroy pancreatic islets by a mechanism that involves the expression of inducible nitric oxide synthase (iNOS), and the production of nitric oxide (NO). Insulin containing beta-cells, selectively destroyed during the development of autoimmune diabetes, appear to be the islet cellular source of iNOS following treatment with IL-1beta. In this study we have evaluated the presence of type I IL-1 signaling receptors on purified pancreatic beta-cells. We show that the interleukin-1 receptor antagonist protein (IRAP) prevents IL-1beta-induced nitrite formation and IL-1beta-induced inhibition of insulin secretion by isolated islets and primary beta-cells purified by fluorescence-activated cell sorting (FACS). The protective effects of IRAP correlate with an inhibition of IL-1beta-induced iNOS expression by islets and FACS purified beta-cells. To provide direct evidence to support beta-cell expression of IL-1 type I signaling receptors, we show that antiserum specific for the type I IL-1 receptor neutralizes IL-1beta-induced nitrite formation by RINm5F cells, and that RINm5F cells express the type I IL-1 receptor at the protein level. Using reverse transcriptase-polymerase chain reaction (RT-PCR), the expression of type I IL-1 signaling receptors by FACS purified beta-cells and not alpha-cells is demonstrated. These results provide direct support for the expression of type I IL-1 receptors by primary pancreatic beta-cells, the cell type selectively destroyed during the development of autoimmune diabetes.


Subject(s)
Islets of Langerhans/metabolism , Receptors, Interleukin-1/metabolism , Animals , Cell Line , Flow Cytometry , Insulin/metabolism , Insulin Antagonists/pharmacology , Insulin Secretion , Interleukin 1 Receptor Antagonist Protein , Male , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/pharmacology
13.
Mol Pharmacol ; 52(3): 398-405, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9281601

ABSTRACT

Aspirin and aspirin-like drugs are the most commonly indicated agents for the treatment of inflammation. Mechanisms of action for these drugs, however, are not clearly understood. In this study, we examined the effects of aspirin on production of nitric oxide (NO), a proinflammatory mediator, and show that aspirin inhibits NO production by transformed pancreatic beta cells (RINm5F) and rat islets in a concentration-dependent manner with an IC50 value of approximately 3 mM. Therapeutic concentrations of aspirin (1-5 mM) that block NO production affected neither nuclear factor-kappaB activation nor inducible NO synthase (iNOS) mRNA transcription but potently inhibited iNOS protein expression by both RINm5F cells and rat islets. The effects of aspirin on islet function were examined by measuring glucose-stimulated insulin secretion in the presence of various concentrations of aspirin. Aspirin (1-5 mM) did not affect insulin secretion at basal or glucose-stimulated conditions, whereas higher concentrations of aspirin (10-20 mM) significantly increased basal insulin secretion. Aspirin at high concentrations of 10 and 20 mM inhibited de novo protein synthesis as demonstrated by inhibition of [35S]methionine incorporation into total islet protein and by inhibition of rabbit reticulocyte expression by Brome mosaic virus mRNA, suggesting that inhibition of iNOS expression at these high concentrations of aspirin may be due to the impairment of the translational machinery. These findings indicate that inhibition of iNOS expression and NO production may explain, in part, the beneficial effects of aspirin as an anti-inflammatory agent at therapeutic concentrations, whereas inhibition of de novo protein synthesis may possibly explain clinical and side effects of aspirin in the inflamed tissues and organs such as stomach and kidney that may accumulate high concentrations of aspirin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Insulinoma/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Nitric Oxide/biosynthesis , Pancreatic Neoplasms/metabolism , Protein Biosynthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Aspirin/toxicity , Cell Line, Transformed/drug effects , Cells, Cultured , Cytosol/metabolism , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Interleukin-1/antagonists & inhibitors , Interleukin-1/pharmacology , Islets of Langerhans/physiology , Kinetics , Male , NF-kappa B/drug effects , NF-kappa B/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitrites/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , omega-N-Methylarginine/pharmacology
15.
J Biol Chem ; 271(37): 22672-8, 1996 Sep 13.
Article in English | MEDLINE | ID: mdl-8798439

ABSTRACT

The bioactivity of interleukin-1 (IL-1), a major proinflammatory cytokine, can be modulated by a variety of factors including inhibitors of IL-1 production and release and receptor blockade by IL-1 receptor antagonist and by binding to nonsignaling soluble receptors. This study demonstrates that the free radical nitric oxide (NO) is also a regulator of IL-1 bioactivity. Lipopolysaccharide-activated murine macrophage RAW264.7 cells, and lipopolysaccharide plus interferon-gamma-activated murine peritoneal macrophages release IL-1 bioactivity, which is increased 10-fold over control levels by 24 h. NG-Monomethyl -arginine (NMMA), a nitric oxide synthase (NOS) inhibitor, almost completely inhibits the release of IL-1 bioactivity from activated macrophages in a time- and concentration-dependent manner with an IC50 of 50 microM. IL-1 activity was determined by thymocyte proliferation bioassay and by a new spectrophotometric bioassay based on IL-1-specific induction of NOS and NO production by an insulinoma cell line, RINm5F. Neither NO nor NOS inhibitors present in the macrophage supernatant interfere with the bioassays. Aminoguanidine and iodonium diphenyl, mechanistically unrelated NOS inhibitors, also prevent the release of IL-1 activity from RAW 264.7 cells. The addition of the NO donor S-nitroso-acetylpenicillamine reconstituted the release of IL-1 bioactivity inhibited by NMMA in a concentration-dependent manner. NO appears to increase the amount of IL-1 protein released by activated macrophages as determined by enzyme-linked immunosorbent assay, but not by mechanisms involving cell death nor modification of IL-1 precursor processing. A cGMP donor, 8-bromo-cGMP, dose-dependently reverses NMMA inhibition of bioactive IL-1 release, suggesting that NO regulates IL-1 release by a cGMP-dependent mechanism. These observations suggest that NO stimulation of the activity of IL-1, a key mediator of the immune response, may be a potentially important mechanism for control of IL-1 activity in vivo.


Subject(s)
Interleukin-1/metabolism , Macrophages/metabolism , Nitric Oxide/metabolism , Animals , Cell Line , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine , omega-N-Methylarginine/administration & dosage , omega-N-Methylarginine/pharmacology
16.
Am J Physiol ; 270(6 Pt 1): C1581-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8764139

ABSTRACT

Insulin-dependent diabetes mellitus (IDDM) is an autoimmune disease that is characterized by selective destruction of insulin-secreting beta-cells. Cytokines have been implicated as effector molecules that participate in both islet inflammation and beta-cell destruction during the development of IDDM. In this study, the effects of cytokines on the expression of inducible nitric oxide synthase (iNOS) and inducible cyclooxygenase (COX-2) by human islets were examined. In combination, the cytokines, human recombinant interleukin-1 beta (IL-1 beta), human recombinant tumor necrosis factor-alpha (TNF-alpha), and human recombinant interferon-gamma (IFN-gamma), induce the time-dependent formation of nitrite and prostaglandin E2 (PGE2) by human islets. The nitric oxide synthase inhibitor NG-monomethyl-L-arginine (L-NMMA) completely inhibits cytokine-induced nitrite formation and attenuates PGE2 production by human islets. L-NMMA does not inhibit cytokine-induced expression of COX-2 by human islets, suggesting that nitric oxide may directly activate cyclooxygenase, an effect that has been previously demonstrated for isolated rat islets. This combination of cytokines (IL-1 beta, TNF-alpha, and IFN-gamma) also induces the expression of iNOS mRNA by human islets as demonstrated by both reverse transcriptase-polymerase chain reaction and Northern blot analysis. We further show that the tyrosine kinase inhibitors genistein and herbimycin A prevent IL-1 beta plus IFN-gamma-induced expression of COX-2 and iNOS and the production of PGE2 and nitric oxide by human islets. These results demonstrate that cytokines induce the expression of iNOS and COX-2 by human islets and that cytokine-induced expression of both COX-2 and iNOS by human islets appears to require the activation of a tyrosine kinase(s).


Subject(s)
Cyclooxygenase Inhibitors/metabolism , Cytokines/pharmacology , Islets of Langerhans/metabolism , Isoenzymes/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Arginine/analogs & derivatives , Arginine/pharmacology , Base Sequence , Benzoquinones , Enzyme Induction , Enzyme Inhibitors/pharmacology , Genistein , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Isoflavones/pharmacology , Lactams, Macrocyclic , Molecular Probes , Molecular Sequence Data , Quinones/pharmacology , Recombinant Proteins/pharmacology , Rifabutin/analogs & derivatives , Tumor Necrosis Factor-alpha/pharmacology , omega-N-Methylarginine
17.
Anal Biochem ; 236(1): 14-9, 1996 Apr 05.
Article in English | MEDLINE | ID: mdl-8619479

ABSTRACT

Cytokines inhibit glucose-induced insulin secretion from pancreatic beta-cells by stimulating the expression of nitric oxide synthase and the increased production of nitric oxide (NO). We have found that the rat insulinoma cell line, RINm5F, responds specifically and linearly to murine and human interleukin-1beta (IL-1beta) and IL-1alpha in the range of 0.1 to 1 unit/ml to produce nitric oxide. Other cytokines, including IL-2, IL-4, IL-6, IL-9, IL-11, IL-15, tumor necrosis factor-alpha, interferon-gamma, and lipopolysaccharide fail to stimulate nitric oxide formation by RINm5F cells either alone or in combination. In addition, these cytokines do not significantly potentiate or attenuate the IL-1 response. This unprecedented specificity to IL-1 has been further developed as a sensitive and specific assay for IL-1 bioactivity. Quantitation by this new bioassay of human IL-1beta and IL-1 released from activated murine peritoneal macrophages showed a close correlation with the quantitation of IL-1 by enzyme immunoassay (ELISA). This new bioassay, which is specific, nonradioactive and inexpensive, represents a significant improvement over current bioassays for IL-1.


Subject(s)
Interleukin-1/analysis , Nitric Oxide/biosynthesis , Animals , Biological Assay , Culture Media/chemistry , Humans , Immunoenzyme Techniques , Macrophage Activation , Mice , Nitrites/analysis , Rats , Recombinant Proteins , Spectrophotometry , Tumor Cells, Cultured
18.
Endocrinology ; 137(3): 825-30, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8603591

ABSTRACT

Recent evidence indicates that nitric oxide (NO) produced after expression of inducible NO synthase (iNOS) mediates cytokine-induced inhibition of insulin secretion by pancreatic islets. The current studies were designed to characterize the involvement of immediate-early response genes, c-fos and c-jun, in interleukin 1 (IL-1)-induced expression of iNOS. iNOS messenger RNA (mRNA) expression by both rat islets and RINm5F cells was time dependent, with maximal expression observed after an approximately 3- to 6-h exposure to IL-1. IL-1 also stimulated rapid and transient expression of c-fos and c-jun by both rat islets and RINm5F cells, with maximal mRNA accumulation detected 30-60 min after IL-1 treatment. IL-1-induced protein synthesis of Fos and Jun was observed as early as 30 min, peaked between 3-5 h, and decreased by 8 h after IL-1 treatment. Temporal correlation of Fos and Jun expression and iNOS gene induction suggested that Fos and Jun might regulate iNOS gene transcription by rodent pancreatic beta-cells. The present study, however, indicates that IL-1 induced expression of Fos and Jun does not seem to participate in the regulation of iNOS and mRNA expression, because: 1) cycloheximide (1 microM) completely inhibited Fos expression but had no inhibitory effect on iNOS mRNA levels; and 2) tyrosine kinase inhibitors genistein and herbimycin A completely inhibited IL-1 induced iNOS expression but did not block c-fos and c-jun expression. These results indicate that two separate signaling pathways may exist for induction of c-fos and c- jun and iNOS genes and that de novo synthesis of Fos and Jun does not participate in the regulation of iNOS gene expression.


Subject(s)
Interleukin-1/pharmacology , Islets of Langerhans/metabolism , Nitric Oxide Synthase/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Animals , Cells, Cultured , Gene Expression Regulation, Enzymologic , Male , Rats , Rats, Sprague-Dawley , Transcriptional Activation
19.
Biochim Biophys Acta ; 1300(1): 63-72, 1996 Mar 29.
Article in English | MEDLINE | ID: mdl-8608164

ABSTRACT

Inflammatory cytokines may participate in the destruction of pancreatic islets during the pathogenesis of insulin-dependent diabetes mellitus, and the cytokine interleukin-1 (IL-1) strongly inhibits insulin secretion from rat pancreatic islets by a process which involves induction of expression of the inducible isoform of nitric oxide synthase and the overproduction of nitric oxide. The signaling events between IL-1 receptor occupancy and induction of nitric oxide synthase in rat islets involve activation of the transcriptional activator NFkappa B. Because sphingomyelin hydrolysis has been implicated as a signaling process both in NFkappa B activation and in IL-1 action in some cells, we have examined the potential involvement of sphingomyelin hydrolysis in the induction of islet nitric oxide overproduction by IL-1. Rat islet sphingomyelin pools were radiolabeled with [3H]choline, and sphingomyelin was then isolated by normal phase HPLC. Electrospray ionization-mass spectrometric analysis revealed islet sphingomyelin consists of at least 4 distinct molecular species, and the most abundant of them contained sphingosine as the long chain base and a residue of palmitic acid as the fatty acid substituent. Molecular species containing residues of stearic acid and arachidic acid were also observed. Neither interleukin-1 nor tumor necrosis factor-alpha was found to induce hydrolysis of islet sphingomyelin species, and neither an exogenous, cell-permeant ceramide species (N-acetyl-D-sphingosine) nor exogenous sphingomyelinase mimicked or potentiated the effect of IL-1 to increase rat islet nitric oxide generation, as reflected by nitrite production. Similar findings were obtained with RINm5F insulinoma cells and with mouse pancreatic islets. These findings provide the first information on the molecular species of sphingomyelin in pancreatic islets and suggest that sphingomyelin hydrolysis is not involved in the signaling pathway whereby IL-1 induces the overproduction of nitric oxide by pancreatic islets.


Subject(s)
Interleukin-1/pharmacology , Islets of Langerhans/drug effects , Nitric Oxide/metabolism , Sphingomyelins/metabolism , Animals , Cells, Cultured , Ceramides/pharmacology , Chromatography, High Pressure Liquid , Hydrolysis , In Vitro Techniques , Islets of Langerhans/cytology , Male , Mass Spectrometry , Mice , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
20.
J Neuroimmunol ; 64(2): 123-33, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632054

ABSTRACT

This study assessed the role of de novo nitric oxide (NO) production in the pathogenesis of experimental allergic encephalomyelitis (EAE) by using aminoguanidine (AG), an inhibitor of nitric oxide synthase (NOS), which preferentially inhibits the cytokine- and endotoxin-inducible isoform of NOS versus the constitutive isoforms consisting of endothelial and neuronal NOS. The maximum clinical severity of EAE and the duration of illness were significantly reduced or totally inhibited by twice daily subcutaneous injection of 100 mg/kg body weight AG. Histochemical staining for NADPH diaphorase, which detects enzymatic activity of NOS, revealed positive reactivity in untreated EAE rats both in parenchymal blood vessel walls and in anterior horn cell neurons, while normal rats and rats with EAE treated with AG showed predominantly the neuronal positivity. Moreover, this NADPH staining pattern was further supported by the immunohistochemical findings that endothelial NOS (eNOS) expression was increased in blood vessels in the inflamed lesions of untreated EAE rats and that inducible NOS (iNOS) was detected in some inflammatory cells, while treatment with AG could significantly reduce both iNOS and eNOS production. These results suggest that: (i) both iNOS and eNOS are upregulated in inflamed areas of the rat central nervous system in EAE; (ii) increased NO production plays a role in the development of clinical signs in EAE; and (iii) selective inhibitors of iNOS and/or eNOS may have therapeutic potential for the treatment of certain autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Biomarkers/analysis , CD4-CD8 Ratio/drug effects , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Enzyme Inhibitors/therapeutic use , Female , Guanidines/therapeutic use , Immunohistochemistry , NADPH Dehydrogenase/metabolism , Nitric Oxide Synthase/metabolism , Rats , Rats, Inbred Lew , Spinal Cord/chemistry , Spinal Cord/pathology , Spinal Cord/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...