Exp Clin Endocrinol Diabetes 2012; 120(04): 210-216
DOI: 10.1055/s-0031-1298015
Article
© J. A. Barth Verlag in Georg Thieme Verlag KG Stuttgart · New York

Effects of Short and Prolonged Mild Intracellular Nitric Oxide Manipulations on Various Aspects of Insulin Secretion in INS-1E β-Cells

A. Natali
1   Metabolism Unit, Department of Internal Medicine, University of Pisa, Italy and Chronic Diseases Research Centre, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Portugal
,
E. Santini
2   Metabolism Unit, Department of Internal Medicine, University of Pisa, Italy
,
A. Delbarba
3   Center for Study and Research on Obesity, Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milano
,
S. Baldi
2   Metabolism Unit, Department of Internal Medicine, University of Pisa, Italy
,
E. Venturi
2   Metabolism Unit, Department of Internal Medicine, University of Pisa, Italy
,
A. Tulipani
2   Metabolism Unit, Department of Internal Medicine, University of Pisa, Italy
,
E. Nisoli
3   Center for Study and Research on Obesity, Department of Pharmacology, Chemotherapy and Medical Toxicology, University of Milano
,
E. Ferrannini
2   Metabolism Unit, Department of Internal Medicine, University of Pisa, Italy
› Institutsangaben
Weitere Informationen

Publikationsverlauf

received 17. August 2011
first decision 09. November 2011

accepted 02. Dezember 2011

Publikationsdatum:
10. Februar 2012 (online)

Abstract

Objective:

We aimed at evaluating the impact of short and prolonged mild manipulations of intracellular nitric oxide (NO) bioavailability on the main features of insulin secretion and whether NO promotes mitochondrial biogenesis in isolated β-cells.

Materials/Methods:

INS-1E β-cells were exposed to either the intracellular NO donor, hydroxylamine (HA), or the NO synthase inhibitor, L-nitro-arginine-methyl-ester (l-NAME), at concentrations lower than 2.0 mM. Glucose and arginine-induced insulin secretion (GIIS and AIIS) were measured after short (1 h) or prolonged (48 h) exposure to l-NAME 1.0 and 2.0 mM or HA 0.4 and 0.8 mM, lower concentrations were also evaluated for the 1 h effects. Basal insulin secretion (BIS), with either HA or l-NAME added to culture media, and peroxisome proliferators-activated receptor γ coactivator 1α (PGC-1α), nuclear respiratory factor-1 (NRF-1), and mitochondrial DNA transcription factor-A (Tfam) gene expression during chronic HA supplementation were also measured.

Results:

Neither l-NAME nor HA affected insulin release at glucose 3.3 mM or in cell culture (BIS). Both short and prolonged cell exposure to l-NAME potentiated GIIS though with a flat dose-response curve while HA inhibited GIIS only at the highest concentration. AIIS was prevented by short exposure to l-NAME and potentiated by HA, while it did not respond to prolonged incubations. Prolonged cell exposure to HA had no effect on PGC-1α, NRF-1 or Tfam gene expression.

Conclusion:

In INS1E cells an intact NO synthesis is necessary to limit insulin release in response to acute glucose gradients and to fully respond to arginine while intracellular NO enrichment above the physiologic levels further inhibits GIIS and potentiate AIIS only when excessive. Prolonged NO manipulations do not affect AIIS, BIS or mitochondrial biogenesis.

 
  • References

  • 1 Antoine MH, Ouedraogo R, Sergooris J et al. Hydroxylamine, a nitric oxide donor, inhibits insulin release and activates K+ATP channels. Eur J Pharmacol 1996; 313: 229-235
  • 2 Beffy P, Lajoix AD, Masiello P et al. A constitutive nitric oxide synthase modulates insulin secretion in the INS-1 cell line. Mol Cell Endocrinol 2001; 183: 41-48
  • 3 Cnop M, Welsh N, Jonas JC et al. Mechanisms of pancreatic beta-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 2005; 54 (Suppl. 02) S97-S107
  • 4 Dandona P, Aljada A, Chaudhuri A et al. Endothelial dysfunction, inflammation and diabetes. Rev Endocr Metab Disord 2004; 5: 189-197
  • 5 Ding Y, Rana RS. Nitric oxide does not initiate but potentiates glucose-induced insulin secretion in pancreatic beta-cells. Biochem Biophys Res Commun 1998; 251: 699-703
  • 6 Drews G, Krippeit-Drews P. No synthase activity does not influence electrical activity of mouse pancreatic B-cells. Biochem Biophys Res Commun 1995; 210: 914-920
  • 7 Gross R, Roye M, Manteghetti M et al. Mechanisms involved in the effect of nitric oxide synthase inhibition on L-arginine-induced insulin secretion. Br J Pharmacol 1997; 120: 495-501
  • 8 Gross R, Roye M, Manteghetti M et al. Alterations of insulin response to different beta cell secretagogues and pancreatic vascular resistance induced by N omega-nitro-L-arginine methyl ester. Br J Pharmacol 1995; 116: 1965-1972
  • 9 Guillausseau PJ, Meas T, Virally M et al. Abnormalities in insulin secretion in type 2 diabetes mellitus. Diabetes Metab 2008; 34 (Suppl. 02) S43-S48
  • 10 Henningsson R, Salehi A, Lundquist I. Role of nitric oxide synthase isoforms in glucose-stimulated insulin release. Am J Physiol Cell Physiol 2002; 283: C296-C304
  • 11 Jitrapakdee S, Wutthisathapornchai A, Wallace JC et al. Regulation of insulin secretion: role of mitochondrial signalling. Diabetologia 2010; 53: 1019-1032
  • 12 Jones PM, Persaud SJ, Bjaaland T et al. Nitric oxide is not involved in the initiation of insulin secretion from rat islets of Langerhans. Diabetologia 1992; 35: 1020-1027
  • 13 Kaneko Y, Ishikawa T, Amano S et al. Dual effect of nitric oxide on cytosolic Ca2+ concentration and insulin secretion in rat pancreatic beta-cells. Am J Physiol Cell Physiol 2003; 284: C1215-C1222
  • 14 Krippeit-Drews P, Welker S, Drews G. Effects of the nitric oxide synthase inhibitor N omega nitro-L-arginine methyl ester on electrical activity and ion channels of mouse pancreatic B cells. Biochem Biophys Res Commun 1996; 224: 199-205
  • 15 Laffranchi R, Gogvadze V, Richter C et al. Nitric oxide (nitrogen monoxide, NO) stimulates insulin secretion by inducing calcium release from mitochondria. Biochem Biophys Res Commun 1995; 217: 584-591
  • 16 Lajoix AD, Badiou S, Peraldi-Roux S et al. Protein inhibitor of neuronal nitric oxide synthase (PIN) is a new regulator of glucose-induced insulin secretion. Diabetes 2006; 55: 3279-3288
  • 17 Lajoix AD, Pugniere M, Roquet F et al. Changes in the dimeric state of neuronal nitric oxide synthase affect the kinetics of secretagogue-induced insulin response. Diabetes 2004; 53: 1467-1474
  • 18 Lajoix AD, Reggio H, Chardes T et al. A neuronal isoform of nitric oxide synthase expressed in pancreatic beta-cells controls insulin secretion. Diabetes 2001; 50: 1311-1323
  • 19 Laychock SG, Modica ME, Cavanaugh CT. L-arginine stimulates cyclic guanosine 3′,5′-monophosphate formation in rat islets of Langerhans and RINm5F insulinoma cells: evidence for L-arginine:nitric oxide synthase. Endocrinology 1991; 129: 3043-3052
  • 20 Matsuura N, Ishikawa T, Abe S et al. Nitric oxide-cyclic GMP system potentiates glucose-induced rise in cytosolic Ca2+ concentration in rat pancreatic beta-cells. Life Sci 1999; 65: 1515-1522
  • 21 Meredith M, Rabaglia ME, Corbett JA et al. Dual functional effects of interleukin-1beta on purine nucleotides and insulin secretion in rat islets and INS-1 cells. Diabetes 1996; 45: 1783-1791
  • 22 Mosen H, Salehi A, Henningsson R et al. Nitric oxide inhibits, and carbon monoxide activates, islet acid alpha-glucoside hydrolase activities in parallel with glucose-stimulated insulin secretion. J Endocrinol 2006; 190: 681-693
  • 23 Natali A, Ferrannini E. Hypertension, insulin resistance, and the metabolic syndrome. Endocrinol Metab Clin North Am 2004; 33: 417-429
  • 24 Natali A, Toschi E, Baldeweg S et al. Clustering of insulin resistance with vascular dysfunction and low-grade inflammation in type 2 diabetes. Diabetes 2006; 55: 1133-1140
  • 25 Nisoli E, Clementi E, Paolucci C et al. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science 2003; 299: 896-899
  • 26 Nisoli E, Falcone S, Tonello C et al. Mitochondrial biogenesis by NO yields functionally active mitochondria in mammals. Proc Natl Acad Sci USA 2004; 101: 16507-16512
  • 27 Nunemaker CS, Buerk DG, Zhang M et al. Glucose-induced release of nitric oxide from mouse pancreatic islets as detected with nitric oxide-selective glass microelectrodes. Am J Physiol Endocrinol Metab 2007; 292: E907-E912
  • 28 Panagiotidis G, Alm P, Lundquist I. Inhibition of islet nitric oxide synthase increases arginine-induced insulin release. Eur J Pharmacol 1992; 229: 277-278
  • 29 Salehi A, Carlberg M, Henningson R et al. Islet constitutive nitric oxide synthase: biochemical determination and regulatory function. Am J Physiol 1996; 270: C1634-C1641
  • 30 Scarpulla RC. Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. Biochim Biophys Acta 2010;
  • 31 Schmidt HH, Warner TD, Ishii K et al. Insulin secretion from pancreatic B cells caused by L-arginine-derived nitrogen oxides. Science 1992; 255: 721-723
  • 32 Smukler SR, Tang L, Wheeler MB et al. Exogenous nitric oxide and endogenous glucose-stimulated beta-cell nitric oxide augment insulin release. Diabetes 2002; 51: 3450-3460
  • 33 Spinas GA, Laffranchi R, Francoys I et al. The early phase of glucose-stimulated insulin secretion requires nitric oxide. Diabetologia 1998; 41: 292-299
  • 34 Sunouchi T, Suzuki K, Nakayama K et al. Dual effect of nitric oxide on ATP-sensitive K+ channels in rat pancreatic beta cells. Pflugers Arch 2008; 456: 573-579
  • 35 Tsuura Y, Ishida H, Shinomura T et al. Endogenous nitric oxide inhibits glucose-induced insulin secretion by suppression of phosphofructokinase activity in pancreatic islets. Biochem Biophys Res Commun 1998; 252: 34-38
  • 36 Vincent SR. Nitric oxide and arginine-evoked insulin secretion. Science 1992; 258: 1376-1378