Dapagliflozin stimulates glucagon secretion at high glucose: experiments and mathematical simulations of human A-cells

Morten Gram Pedersen, Ingela Ahlstedt, Mickaël F El Hachmane, Sven O Göpel, Morten Gram Pedersen, Ingela Ahlstedt, Mickaël F El Hachmane, Sven O Göpel

Abstract

Glucagon is one of the main regulators of blood glucose levels and dysfunctional stimulus secretion coupling in pancreatic A-cells is believed to be an important factor during development of diabetes. However, regulation of glucagon secretion is poorly understood. Recently it has been shown that Na(+)/glucose co-transporter (SGLT) inhibitors used for the treatment of diabetes increase glucagon levels in man. Here, we show experimentally that the SGLT2 inhibitor dapagliflozin increases glucagon secretion at high glucose levels both in human and mouse islets, but has little effect at low glucose concentrations. Because glucagon secretion is regulated by electrical activity we developed a mathematical model of A-cell electrical activity based on published data from human A-cells. With operating SGLT2, simulated glucose application leads to cell depolarization and inactivation of the voltage-gated ion channels carrying the action potential, and hence to reduce action potential height. According to our model, inhibition of SGLT2 reduces glucose-induced depolarization via electrical mechanisms. We suggest that blocking SGLTs partly relieves glucose suppression of glucagon secretion by allowing full-scale action potentials to develop. Based on our simulations we propose that SGLT2 is a glucose sensor and actively contributes to regulation of glucagon levels in humans which has clinical implications.

Figures

Figure 1. Effect of dapagliflozin (10 nM)…
Figure 1. Effect of dapagliflozin (10 nM) on glucagon secretion from human and mouse islets at different glucose concentrations.
The figure shows estimates from linear mixed-effects statistical modeling with error bars indicating standard errors. For each islet preparation (i.e., each individual or animal) 6–8 groups of size-matched islets were tested for each indicated condition from 3 donors for human data, and from 29 (1 mM glucose), 11 (6 mM glucose) or 18 (11 mM glucose) animals for mouse data.
Figure 2. Model simulation of electrical activity.
Figure 2. Model simulation of electrical activity.
The glucose concentration was raised from 1 mM to 6 mM and 11 mM, followed by simulated dapagliflozin application, as indicated. Simulations were performed by changing the glucose parameter in the SGLT2 submodel, GSGLT2, from 1 mM to 6 mM to 11 mM, and by lowering the KATP conductance from 0.15 to 0.115 nS to model the increase in glucose concentration to 6 or 11 mM, since inhibition of KATP channel activity is maximal already at 6 mM glucose. Dapagliflozin application was assumed to block all SGLT2 transporters, which was modeled by setting the parameter n = 0.
Figure 3. Ion currents during simulated action…
Figure 3. Ion currents during simulated action potentials during low glucose, high glucose, and high glucose in the presence of dapagliflozin.
The upper panels show zooms on action potentials under the various conditions as indicated. The middle and lower panels show the simulated ion currents during the action potentials, as indicated in the legend. Note the different scales on the y-axes of the middle and lower panels.
Figure 4. Schematic representation of the control…
Figure 4. Schematic representation of the control of glucagon secretion by KATP-conductance and SGLT2.

References

    1. Baron A. D., Schaeffer L., Shragg P. & Kolterman O. G. Role of Hyperglucagonemia in Maintenance of Increased Rates of Hepatic Glucose Output in Type II Diabetics. Diabetes 36, 274–283 (1987).
    1. D’Alessio D. The role of dysregulated glucagon secretion in type 2 diabetes. Diabetes, Obesity and Metabolism 13, 126–132 (2011).
    1. Segel S. A., Paramore D. S. & Cryer P. E. Hypoglycemia-Associated Autonomic Failure in Advanced Type 2 Diabetes. Diabetes 51, 724–733 (2002).
    1. Cryer P. E. Hypoglycaemia: The limiting factor in the glycaemic management of type I and type II diabetes. Diabetologia 45, 937–948 (2002).
    1. Bolli G., Tsalikian E., Haymond M., Cryer P. E. & Gerich J. E. Defective glucose couterregulation after subcutaneous insulin in nonsinslin-depedent diabeted mellitus. Paradoxical suppression of glucose utilization and lack of compensatory increase in glucose production, roles of insulin resistance, abnormal neuroendocrine responses, and islet paracrine interactions. J. Clin. Invest. 73, 1532–1541 (1984).
    1. Meneilly G. S., Cheung E. & Tuokko H. Counterregulatory Hormone Responses to Hypoglycemia in the Elderly Patient with Diabetes. Diabetes 43, 403–410 (1994).
    1. Shamoon H. et al.. Increased epinephrine and skeletal muscle responses to hypoglycemia in non-insulin-dependent diabetes mellitus. J. Clin. Invest. 93, 2562–2571 (1994).
    1. Jamison R. A. et al.. Hyperglucagonemia precedes a decline in insulin secretion and causes hyperglycemia in chronically glucose-infused rats. American Journal of Physiology - Endocrinology and Metabolism 301, E1174–E1183 (2011).
    1. Lee Y. et al.. Metabolic manifestations of insulin deficiency do not occur without glucagon action. Proceedings of the National Academy of Sciences 109, 14972–14976 (2012).
    1. Buchanan K. D. & Mawhinney W. A. A. Insulin Control of Glucagon Release From Insulin-deficient Rat Islets. Diabetes 22, 801–803 (1973).
    1. Greenbaum C. J., Havel P. J., Taborsky J. & Klaff L. J. Intra-islet insulin permits glucose to directly suppress pancreatic A-cell function. J. Clin. Pharmacol. 88, 767–773 (1991).
    1. Muller W. A., Faloona G. R. & Unger R. H. The effect of experimental insulin deficiency on glucagon secretion. J. Clin. Invest. 50, 1992–1999 (1971).
    1. Maruyama H., Hisatomi A., Orci L., Grodsky G. & Unger R. Insulin within islets is a physiologic glucagon release inhibitor. J. Clin. Invest. 74, 2296–2299 (1984).
    1. Ishihara H., Maechler P., Gjinovci A., Herrera P.-L. & Wollheim C. B. Islet [beta]-cell secretion determines glucagon release from neighbouring [alpha]-cells. Nat Cell Biol 5, 330–335 (2003).
    1. Cheng-Xue R. et al.. Tolbutamide Controls Glucagon Release From Mouse Islets Differently Than Glucose: Involvement of KATP Channels From Both α-Cells and δ-Cells. Diabetes 62, 1612–1622 (2013).
    1. Vieira E., Salehi A. & Gylfe E. Glucose inhibits glucagon secretion by a direct effect on mouse pancreatic alpha cells. Diabetologia 50, 370–379 (2007).
    1. de Heer J., Rasmussen C., Coy D. H. & Holst J. J. Glucagon-like peptide-1, but not glucose-dependent insulinotropic peptide, inhibits glucagon secretion via somatostatin (receptor subtype 2) in the perfused rat pancreas. Diabetologia 51, 2263–2270 (2008).
    1. Liu Y.-J., Vieira E. & Gylfe E. A store-operated mechanism determines the activity of the electrically excitable glucagon-secreting pancreatic [alpha]-cell. Cell Calcium 35, 357–365 (2004).
    1. Göpel S. O. et al.. Regulation of glucagon release in mouse alpha-cells by KATP channels and inactivation of TTX-sensitive Na+ channels. J Physiol (Lond) 528, 509–520 (2000).
    1. MacDonald P. et al.. A K ATP channel-dependent pathway within alpha cells regulates glucagon release from both rodent and human islets of Langerhans. Plos Biol 5, 1236–1247 (2007).
    1. Diderichsen P. & Göpel S. O. Modelling the electrical activity of pancreatic A-cells based on experimental data from intact mouse islest. J. Biol. Physics 32, 209–229 (2006).
    1. Ramracheya R. et al.. Membrane Potential-Dependent Inactivation of Voltage-Gated Ion Channels in α-Cells Inhibits Glucagon Secretion From Human Islets. Diabetes 59, 2198–2208 (2010).
    1. Merovci A. et al.. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. The Journal of Clinical Investigation 124, 509–514 (2014).
    1. Ferrannini E. et al.. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J. Clin. Inv. 124, 499–508 (2014).
    1. Bonner C. et al.. Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion. Nat Med 21, 512–517 (2015).
    1. Kasichayanula S. et al.. Pharmacokinetics and pharmacodynamics of dapagliflozin, a novel selective inhibitor of sodium–glucose co-transporter type 2, in Japanese subjects without and with type 2 diabetes mellitus. Diabetes, Obesity and Metabolism 13, 357–365 (2011).
    1. Parent L., Supplisson S., Loo D. & Wright E. M. Electrogenic properties of the cloned Na+/glucose cotransporter: I. Voltage-clamp studies. J. Membr. Biol. 125, 49–62 (1992).
    1. Hirayama B. A. et al.. Kinetic and specificity differences between rat, human, and rabbit Na+-gllucose cotransporters (SGLT-1). Am. J. Physiol. 270, G919–926 (1996).
    1. Mackenzie B., Loo D. D. F., Panayotova-Heiermann M. & Wright E. M. Biophysical characteristics of the pig kidney Na+/glucose cotransporter SGLT2 reveal a common mechanism for SGLT1 and SGLT2. J. Biol. Chem. 271, 32678–32683 (1996).
    1. Parker H. E. et al.. Predominant role of active versus facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia 55, 2445–2455 (2012).
    1. Gribble F. M., Williams L., Simpson A. K. & Reimann F. A Novel Glucose-Sensing Mechanism Contributing to Glucagon-Like Peptide-1 Secretion From the GLUTag Cell Line. Diabetes 52, 1147–1154 (2003).
    1. Zhang Q. et al.. Role KATP channels in glucose-regulated glucagon secretion and impaired counterregulation in Type 2 diabetes. Cell Metabolism 18, 871–882 (2013).
    1. Watts M. & Sherman A. Modeling the Pancreatic α-Cell: Dual Mechanisms of Glucose Suppression of Glucagon Secretion. Biophysical Journal 106, 741–751 (2014).
    1. Montefusco F. & Pedersen M. G. Mathematical modelling of local calcium and regulated exocytosis during inhibition and stimulation of glucagon secretion from pancreatic alpha-cells. The Journal of Physiology 593, 4519–4530 (2015).
    1. Göpel S. et al.. Capacitance measurements of exocytosis in mouse pancreatic {alpha}-, {beta}- and {delta}-cells within intact islets of Langerhans. J Physiol (Lond) 556, 711–726 (2004).
    1. Hummel C. S. et al.. Glucose transport by human renal Na+/d-glucose cotransporters SGLT1 and SGLT2. American Journal of Physiology - Cell Physiology 300, C14–C21 (2011).
    1. Heimberg H., De Vos A., Pipeleers D., Thorens B. & Schuit F. Differences in Glucose Transporter Gene Expression between Rat Pancreatic α- and β-Cells Are Correlated to Differences in Glucose Transport but Not in Glucose Utilization. Journal of Biological Chemistry 270, 8971–8975 (1995).
    1. Sweet I. R. & Matschinsky F. M. Are there kinetic advantages of GLUT2 in pancreatic glucose sensing? Diabetologia 40, 112–119 (1997).
    1. Barg S., Galvanovskis J., Göpel S. O., Rorsman P. & Eliasson L. Tight coupling between electrical activity and exocytosis in mouse glucagon-secreting alpha-cells. Diabetes 49, 1500–1510 (2000).
    1. Göpel S., Kanno T., Barg S., Galvanovskis J. & Rorsman P. Voltage-gated and resting membrane currents recorded from B-cells in intact mouse pancreatic islets. The Journal of Physiology 521, 717–728 (1999).
    1. Arkhammar P., Nilsson T., Rorsman P. & Berggren P. O. Inhibition of ATP-regulated K+ channels precedes depolarization-induced increase in cytoplasmic free Ca2+ concentration in pancreatic beta-cells. Journal of Biological Chemistry 262, 5448–5454 (1987).
    1. Bohannon N. V., Lorenzi M., Grodsky G. M. & Karam J. H. Stimulatory Effects of Tolbutamide Infusion on Plasma Glucagon in Insulin-Dependent Diabetic Subjects. Journal of Clinical Endocrinology & Metabolism 54, 459–462 (1982).
    1. Silvestre R. A., Miralles P., Moreno P., Villanueva M. L. & Marco J. Somatostatin, insulin and glucagon secretion by the perfused pancreas from the cysteamine-treated rat. Biochemical and Biophysical Research Communications 134, 1291–1297 (1986).
    1. Loreti L., Sugase T. & Foa P. P. Diurnal variations of serum insulin, total glucagon, cortisol, glucose and free fatty acids in normal and diabetic subjects before and after treatment with chlorpropamide. Hormone Res. 5, 278–292 (1974).
    1. Leung Y. M. et al.. Insulin Regulates Islet α-Cell Function by Reducing KATP Channel Sensitivity to Adenosine 5′-Triphosphate Inhibition. Endocrinology 147, 2155–2162 (2006).
    1. Bailey C. J. et al.. Efficacy and safety of dapagliflozin monotherapy in people with Type 2 diabetes: a randomized double-blind placebo-controlled 102-week trial. Diabetic Medicine 32, 531–541 (2015).
    1. Halimi S. & Verges B. Adverse effects and safety of SGLT-2 inhibitors. Diabetes & Metabolism 40, S28–S34 (2014).
    1. Anderson S. L. Dapagliflozin efficacy and safety: a perspective review. Ther. Adv. Drug Saf. 5, 242–254 (2014).
    1. R-Core-Team. R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. URL (2013).
    1. Pinheiro J., Bates D., Debroy S., Deepayan S. & Team, R. D. C. nlme: Linear and Nonlinear Mixed Effects Rodels. R package version 3.1-111 (2013).

Source: PubMed

3
구독하다