Skip to main content
Erschienen in: Diabetology International 2/2016

27.02.2016 | Review Article

Is zinc an intra-islet regulator of glucagon secretion?

verfasst von: Hisamitsu Ishihara, Claes B. Wollheim

Erschienen in: Diabetology International | Ausgabe 2/2016

Einloggen, um Zugang zu erhalten

Abstract

More than a decade ago, zinc was suggested to have a role as an intra-islet regulator of glucagon secretion. Several lines of experimental evidence have since provided support for this hypothesis, though contradictory observations have also been reported. Meanwhile, Slc30A/ZnT8, a zinc transporter expressed in insulin and glucagon secretory granules, was identified. Furthermore, genome wide association analyses revealed it to be a candidate causative gene for type 2 diabetes mellitus. Recent progress in gene manipulation in animals yielded considerable information on the role of zinc in islet cells. In this mini-review, data pertaining the roles played by zinc in islet hormone secretion are summarized and discussed.
Literatur
1.
Zurück zum Zitat Gromada J, Franklin I, Wollheim CB. α-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocrine Rev. 2007;28:84–116.CrossRef Gromada J, Franklin I, Wollheim CB. α-cells of the endocrine pancreas: 35 years of research but the enigma remains. Endocrine Rev. 2007;28:84–116.CrossRef
2.
Zurück zum Zitat Gylfe E, Gilon P. Glucose regulation of glucagon secretion. Diab Res Clin Prac. 2014;103:1–10.CrossRef Gylfe E, Gilon P. Glucose regulation of glucagon secretion. Diab Res Clin Prac. 2014;103:1–10.CrossRef
3.
Zurück zum Zitat Ishihara H, Maechler P, Gjinovci A, Herrera PL, Wollheim CB. Islet β-cell secretion determines glucagon release from neighbouring α-cells. Nature Cell Biol. 2003;5:330–5.CrossRefPubMed Ishihara H, Maechler P, Gjinovci A, Herrera PL, Wollheim CB. Islet β-cell secretion determines glucagon release from neighbouring α-cells. Nature Cell Biol. 2003;5:330–5.CrossRefPubMed
4.
Zurück zum Zitat Marliss EB, Wollheim CB, Blondel B, Orci L, Lambert AE, Stauffacher W, Like AA, Renold AE. Insulin and glucagon release from monolayer cell cultures of pancreas from newborn rats. Eur J Clin Invest. 1973;3:16–26.CrossRefPubMed Marliss EB, Wollheim CB, Blondel B, Orci L, Lambert AE, Stauffacher W, Like AA, Renold AE. Insulin and glucagon release from monolayer cell cultures of pancreas from newborn rats. Eur J Clin Invest. 1973;3:16–26.CrossRefPubMed
5.
Zurück zum Zitat Leclercq-Meyer V, Marchand J, Malaisse WJ. An arginine-like effect of the “fumalate + glutamate + pyruvate” mixture on glucagon release. Life Sci. 1977;20:1193–8.CrossRefPubMed Leclercq-Meyer V, Marchand J, Malaisse WJ. An arginine-like effect of the “fumalate + glutamate + pyruvate” mixture on glucagon release. Life Sci. 1977;20:1193–8.CrossRefPubMed
6.
Zurück zum Zitat Takahashi R, Ishihara H, Tamura A, Yamaguchi S, Yamada T, Takei D, Katagiri H, Endou H, Oka Y. Cell type specific activation of metabolism reveals that β-cell secretion suppresses glucagon release from α-cells in rat pancreatic islets. Am J Physiol. Endocrinol Metab. 2006;290:E308–16.CrossRefPubMed Takahashi R, Ishihara H, Tamura A, Yamaguchi S, Yamada T, Takei D, Katagiri H, Endou H, Oka Y. Cell type specific activation of metabolism reveals that β-cell secretion suppresses glucagon release from α-cells in rat pancreatic islets. Am J Physiol. Endocrinol Metab. 2006;290:E308–16.CrossRefPubMed
7.
Zurück zum Zitat Ishihara H, Wang H, Drewes LR, Wollheim CB. Overexpression of monocarboxylate transporter and lactate dehydrogenase alters insulin secretory responses to pyruvate and lactate in β cells. J. Clin. Invest. 1999;104:1621–9.CrossRefPubMedPubMedCentral Ishihara H, Wang H, Drewes LR, Wollheim CB. Overexpression of monocarboxylate transporter and lactate dehydrogenase alters insulin secretory responses to pyruvate and lactate in β cells. J. Clin. Invest. 1999;104:1621–9.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Pullen TJ, Sylow L, Sun G, Halestrap AP, Richter EA, Rutter GA. Overexpression of monocarboxylate transporter-1 (Slc16A1) in mouse pancreatic β-cells leads to relative hyperinsulinism during exercise. Diabetes. 2012;61:1719–25.CrossRefPubMedPubMedCentral Pullen TJ, Sylow L, Sun G, Halestrap AP, Richter EA, Rutter GA. Overexpression of monocarboxylate transporter-1 (Slc16A1) in mouse pancreatic β-cells leads to relative hyperinsulinism during exercise. Diabetes. 2012;61:1719–25.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Ravier MA, Rutter GA. Glucose or insulin, but not zinc ions, inhibit glucagon secretion from mouse pancreatic and α-cells. Diabetes. 2005;54:1789–97.CrossRefPubMed Ravier MA, Rutter GA. Glucose or insulin, but not zinc ions, inhibit glucagon secretion from mouse pancreatic and α-cells. Diabetes. 2005;54:1789–97.CrossRefPubMed
10.
Zurück zum Zitat Quoix N, Cheng-Xue R, Mattart L, Zeinoun Z, Guiot Y, Beauvois MC, Henquin JC, Gilon P. Glucose and pharmacological modulators of ATP-sensitive K channels control [Ca2+]c by different mechanisms in isolated mouse β-cells. Diabetes. 2009;58:412–21.CrossRefPubMedPubMedCentral Quoix N, Cheng-Xue R, Mattart L, Zeinoun Z, Guiot Y, Beauvois MC, Henquin JC, Gilon P. Glucose and pharmacological modulators of ATP-sensitive K channels control [Ca2+]c by different mechanisms in isolated mouse β-cells. Diabetes. 2009;58:412–21.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Ramracheya R, Ward C, Shigeto M, Walker JN, Amisten S, Zhang Q, Johnson PR, Rorsman P, Braun M. Membrane potential-dependent inactivation of voltage-gated ion channels in & α-cells inhibits glucagon secretion from human islets. Diabetes. 2010;59:2198–208.CrossRefPubMedPubMedCentral Ramracheya R, Ward C, Shigeto M, Walker JN, Amisten S, Zhang Q, Johnson PR, Rorsman P, Braun M. Membrane potential-dependent inactivation of voltage-gated ion channels in & α-cells inhibits glucagon secretion from human islets. Diabetes. 2010;59:2198–208.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Franklin I, Gromada J, Gjinovci A, Theander S, Wollheim CB. β-cell secretory products activate and β-cell ATP-dependent potassium channels to inhibit glucagon release. Diabetes. 2005;54:1808–15.CrossRefPubMed Franklin I, Gromada J, Gjinovci A, Theander S, Wollheim CB. β-cell secretory products activate and β-cell ATP-dependent potassium channels to inhibit glucagon release. Diabetes. 2005;54:1808–15.CrossRefPubMed
13.
Zurück zum Zitat Le Marchand SJ, Piston DW. Glucose suppression of glucagon secretion: metabolic and calcium responses from & #x03B1;-cells in intact mouse pancreatic islets. J Biol Chem. 2010;285:14389–98.CrossRefPubMedPubMedCentral Le Marchand SJ, Piston DW. Glucose suppression of glucagon secretion: metabolic and calcium responses from & #x03B1;-cells in intact mouse pancreatic islets. J Biol Chem. 2010;285:14389–98.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Chimienti F. Zinc, pancreatic islet cell function and diabetes: new insights into an old story. Nutri Resear Rev. 2013;26:1–11.CrossRef Chimienti F. Zinc, pancreatic islet cell function and diabetes: new insights into an old story. Nutri Resear Rev. 2013;26:1–11.CrossRef
15.
Zurück zum Zitat Fadista J, Vikman P, Laakso EO, Mollet IG, Esguerra JL, et al. Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc Natl Acad Sci USA. 2014;111:13924–9.CrossRefPubMedPubMedCentral Fadista J, Vikman P, Laakso EO, Mollet IG, Esguerra JL, et al. Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc Natl Acad Sci USA. 2014;111:13924–9.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Flannick J, Thorleifsson G, Beer NL, Jacobs SB, Grarup N, et al. Loss-of-function mutations in SLC30A8 protect against type 2 diabetes. Nat Genet. 2014;46:357–63.CrossRefPubMedPubMedCentral Flannick J, Thorleifsson G, Beer NL, Jacobs SB, Grarup N, et al. Loss-of-function mutations in SLC30A8 protect against type 2 diabetes. Nat Genet. 2014;46:357–63.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Gyulkhandanyan AV, Lu H, Lee SC, Bhattachajee A, Wijesekara N, Manning Fox JE, MacDonald PE, Chimienti F Dai FF, Wheeler MB. Investigation of transport mechanisms and regulation of intracellular Zn2+ in pancreatic α-cells. Regulation of glucagon secretion by zinc: lessons from the β cell-specific Znt8 knockout mouse model. J Biol Chem. 2008;283:10184–97.CrossRefPubMed Gyulkhandanyan AV, Lu H, Lee SC, Bhattachajee A, Wijesekara N, Manning Fox JE, MacDonald PE, Chimienti F Dai FF, Wheeler MB. Investigation of transport mechanisms and regulation of intracellular Zn2+ in pancreatic α-cells. Regulation of glucagon secretion by zinc: lessons from the β cell-specific Znt8 knockout mouse model. J Biol Chem. 2008;283:10184–97.CrossRefPubMed
18.
Zurück zum Zitat Zhou H, Zhang T, Harmon JS, Bryan J, Robertson RP. Zinc, not insulin, regulates the rat α-cell response to hypoglycemia in vivo. Diabetes. 2007;56:1107–12.CrossRefPubMed Zhou H, Zhang T, Harmon JS, Bryan J, Robertson RP. Zinc, not insulin, regulates the rat α-cell response to hypoglycemia in vivo. Diabetes. 2007;56:1107–12.CrossRefPubMed
19.
Zurück zum Zitat Bancila V, Cens T, Monnier D, Chanson F, Faure C, Durant Y, Bloc A. Two SUR-1 specific histidine residues mandatory for zinc-induced activation of the rat KATP channel. J Biol Chem. 2005;280:8793–9.CrossRefPubMed Bancila V, Cens T, Monnier D, Chanson F, Faure C, Durant Y, Bloc A. Two SUR-1 specific histidine residues mandatory for zinc-induced activation of the rat KATP channel. J Biol Chem. 2005;280:8793–9.CrossRefPubMed
20.
Zurück zum Zitat Drobinskaya I, Neumaier F, Perverzev A, Hescheler J, Schneider T. Diethyldithiocarbamate-mediated zinc ion chelation reveals role of Cav2.3 channels in glucagon secretion. Biochem Biophys Acta. 2015;1853:953–64.CrossRefPubMed Drobinskaya I, Neumaier F, Perverzev A, Hescheler J, Schneider T. Diethyldithiocarbamate-mediated zinc ion chelation reveals role of Cav2.3 channels in glucagon secretion. Biochem Biophys Acta. 2015;1853:953–64.CrossRefPubMed
21.
Zurück zum Zitat Hardy AB, Serino AS, Wijesekara N, Chimienti F, Wheeler MB. Regulation of glucagon secretion by zinc: lessons from the β cell-specific Znt8 knockout mouse model. Diab Obes Metab. 2011;13(Suppl. 1):112–7.CrossRef Hardy AB, Serino AS, Wijesekara N, Chimienti F, Wheeler MB. Regulation of glucagon secretion by zinc: lessons from the β cell-specific Znt8 knockout mouse model. Diab Obes Metab. 2011;13(Suppl. 1):112–7.CrossRef
22.
Zurück zum Zitat Kawamori D, Kurpad AJ, Hu J, Liew CW, Shih JL, Ford EL, Herrera PL, Polonsky KS, McGuinness OP, Kulkarni RN. Insulin signaling in alpha cells modulates glucagon secretion in vivo. Cell Metab. 2009;9:350–61.CrossRefPubMedPubMedCentral Kawamori D, Kurpad AJ, Hu J, Liew CW, Shih JL, Ford EL, Herrera PL, Polonsky KS, McGuinness OP, Kulkarni RN. Insulin signaling in alpha cells modulates glucagon secretion in vivo. Cell Metab. 2009;9:350–61.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Pound LD, Sarkar SA, Benninger RK, Wang Y, Suwanichkul A, et al. Deletion of the mouse Slc30a8 gene encoding zinc transporter-8 results in impaired insulin secretion. Biochem J. 2009;421:371–6.CrossRefPubMedPubMedCentral Pound LD, Sarkar SA, Benninger RK, Wang Y, Suwanichkul A, et al. Deletion of the mouse Slc30a8 gene encoding zinc transporter-8 results in impaired insulin secretion. Biochem J. 2009;421:371–6.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Nicolson TJ, Bellomo EA, Wijesekara N, Loder MK, Baldwin JM, et al. Insulin storage and glucose homeostasis in mice null for the granule zinc transporter ZnT8 and studies of the type 2 diabetes-associated variants. Diabetes. 2009;58:2070–83.CrossRefPubMedPubMedCentral Nicolson TJ, Bellomo EA, Wijesekara N, Loder MK, Baldwin JM, et al. Insulin storage and glucose homeostasis in mice null for the granule zinc transporter ZnT8 and studies of the type 2 diabetes-associated variants. Diabetes. 2009;58:2070–83.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Lemaire K, Ravier MA, Schraenen A, Creemers JW, Van de Plas R, et al. Insulin crystallization depends on zinc transporter ZnT8 expression, but is not required for normal glucose homeostasis in mice. Proc Natl Acad Sci USA. 2009;106:14872–7.CrossRefPubMedPubMedCentral Lemaire K, Ravier MA, Schraenen A, Creemers JW, Van de Plas R, et al. Insulin crystallization depends on zinc transporter ZnT8 expression, but is not required for normal glucose homeostasis in mice. Proc Natl Acad Sci USA. 2009;106:14872–7.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Wijesekara N, Dai FF, Hardy AB, Giglou PR, Bhattacharjee A, Koshkin V, Chimienti F, Gaisano HY, Rutter GA, Wheeler MB. Beta cell-specific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion. Diabetologia. 2010;53:1656–68.CrossRefPubMed Wijesekara N, Dai FF, Hardy AB, Giglou PR, Bhattacharjee A, Koshkin V, Chimienti F, Gaisano HY, Rutter GA, Wheeler MB. Beta cell-specific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion. Diabetologia. 2010;53:1656–68.CrossRefPubMed
27.
Zurück zum Zitat Tamaki M, Fujitani Y, Hara A, Uchida T, Tamura Y, et al. The diabetes-susceptible gene SLC30A8/ZnT8 regulates hepatic insulin clearance. J Clin Invest. 2013;123:4513–24.CrossRefPubMedPubMedCentral Tamaki M, Fujitani Y, Hara A, Uchida T, Tamura Y, et al. The diabetes-susceptible gene SLC30A8/ZnT8 regulates hepatic insulin clearance. J Clin Invest. 2013;123:4513–24.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Mitchell RK, Hu M, Chabosseau PL, Cane MC, Meur G, Bellomo EA, Carzaniga R, Collinson LM, Li WH, Hodson DJ, Rutter GA. Molecular Genetic regulation of Slc30a8/ZnT8 reveals a positive association with glucose tolerance. Mol Endocrinol. 2016;30:77–91.CrossRefPubMed Mitchell RK, Hu M, Chabosseau PL, Cane MC, Meur G, Bellomo EA, Carzaniga R, Collinson LM, Li WH, Hodson DJ, Rutter GA. Molecular Genetic regulation of Slc30a8/ZnT8 reveals a positive association with glucose tolerance. Mol Endocrinol. 2016;30:77–91.CrossRefPubMed
29.
Zurück zum Zitat Solomou A, Meur G, Hodson DJ, Tomas A, Li SM, Philippe E, Herrera PL, Magnan C, Rutter GA. The zinc transporter Slc30a8/ZnT8 is required in a subpopulation of pancreatic α-cells for hypoglycemia-induced glucagon secretion. J Biol Chem. 2015;290:21432–42.CrossRefPubMedPubMedCentral Solomou A, Meur G, Hodson DJ, Tomas A, Li SM, Philippe E, Herrera PL, Magnan C, Rutter GA. The zinc transporter Slc30a8/ZnT8 is required in a subpopulation of pancreatic α-cells for hypoglycemia-induced glucagon secretion. J Biol Chem. 2015;290:21432–42.CrossRefPubMedPubMedCentral
Metadaten
Titel
Is zinc an intra-islet regulator of glucagon secretion?
verfasst von
Hisamitsu Ishihara
Claes B. Wollheim
Publikationsdatum
27.02.2016
Verlag
Springer Japan
Erschienen in
Diabetology International / Ausgabe 2/2016
Print ISSN: 2190-1678
Elektronische ISSN: 2190-1686
DOI
https://doi.org/10.1007/s13340-016-0259-x

Weitere Artikel der Ausgabe 2/2016

Diabetology International 2/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.