Skip to main content
Top

20-04-2015 | Dapagliflozin | ReviewPaper | Article

Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion

Authors: Caroline Bonner, Julie Kerr-Conte, Valéry Gmyr, Gurvan Queniat, Ericka Moerman, Julien Thévenet, Cédric Beaucamps, Nathalie Delalleau, Iuliana Popescu, Willy J Malaisse, Abdullah Sener, Benoit Deprez, Amar Abderrahmani, Bart Staels, François Pattou

Abstract

Type 2 diabetes (T2D) is characterized by chronic hyperglycemia resulting from a deficiency in insulin signaling, because of insulin resistance and/or defects in insulin secretion; it is also associated with increases in glucagon and endogenous glucose production (EGP)1. Gliflozins, including dapagliflozin, are a new class of approved oral antidiabetic agents that specifically inhibit sodium-glucose co-transporter 2 (SGLT2) function in the kidney2, 3, 4, 5, thus preventing renal glucose reabsorption and increasing glycosuria in diabetic individuals while reducing hyperglycemia. However, gliflozin treatment in subjects with T2D increases both plasma glucagon and EGP6, 7 by unknown mechanisms. In spite of the rise in EGP, T2D patients treated with gliflozin have lower blood glucose levels than those receiving placebo, possibly because of increased glycosuria6, 7; however, the resulting increase in plasma glucagon levels represents a possible concerning side effect, especially in a patient population already affected by hyperglucagonemia. Here we demonstrate that SGLT2 is expressed in glucagon-secreting alpha cells of the pancreatic islets. We further found that expression of SLC5A2 (which encodes SGLT2) was lower and glucagon (GCG) gene expression was higher in islets from T2D individuals and in normal islets exposed to chronic hyperglycemia than in islets from non-diabetics. Moreover, hepatocyte nuclear factor 4-α (HNF4A) is specifically expressed in human alpha cells, in which it controls SLC5A2 expression, and its expression is downregulated by hyperglycemia. In addition, inhibition of either SLC5A2 via siRNA-induced gene silencing or SGLT2 via dapagliflozin treatment in human islets triggered glucagon secretion through KATP channel activation. Finally, we found that dapagliflozin treatment further promotes glucagon secretion and hepatic gluconeogenesis in healthy mice, thereby limiting the decrease of plasma glucose induced by fasting. Collectively, these results identify a heretofore unknown role of SGLT2 and designate dapagliflozin an alpha cell secretagogue.

Nat Med 2015;21:512–517. doi: 10.1038/nm.3828

Supplementary figures appear at the end of the article.

Literature
  1. Kahn, S.E., Cooper, M.E. & Del Prato, S. Pathophysiology and treatment of type 2 diabetes: perspectives on the past, present, and future. Lancet 383, 1068–1083 (2014).
  2. Demaris, K.M. & White, J.R. Dapagliflozin, an SGLT2 inhibitor for the treatment of type 2 diabetes. Drugs Today (Barc) 49, 289–301 (2013).
  3. Jabbour, S.A. & Goldstein, B.J. Sodium glucose co-transporter 2 inhibitors: blocking renal tubular reabsorption of glucose to improve glycaemic control in patients with diabetes. Int. J. Clin. Pract. 62, 1279–1284 (2008).
  4. Komoroski, B. et al. Dapagliflozin, a novel, selective SGLT2 inhibitor, improved glycemic control over 2 weeks in patients with type 2 diabetes mellitus. Clin. Pharmacol. Ther. 85, 513–519 (2009).
  5. Nair, S. & Wilding, J.P. Sodium glucose cotransporter 2 inhibitors as a new treatment for diabetes mellitus. J. Clin. Endocrinol. Metab. 95, 34–42 (2010).
  6. Merovci, A. et al. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. J. Clin. Invest. 124, 509–514 (2014).
  7. Ferrannini, E. et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J. Clin. Invest. 124, 499–508 (2014).
  8. Biddinger, S.B. & Kahn, C.R. From mice to men: insights into the insulin resistance syndromes. Annu. Rev. Physiol. 68, 123–158 (2006).
  9. DeFronzo, R.A., Simonson, D. & Ferrannini, E. Hepatic and peripheral insulin resistance: a common feature of type 2 (non-insulin-dependent) and type 1 (insulin-dependent) diabetes mellitus. Diabetologia 23, 313–319 (1982).
  10. Campbell, P.J., Mandarino, L.J. & Gerich, J.E. Quantification of the relative impairment in actions of insulin on hepatic glucose production and peripheral glucose uptake in non-insulin-dependent diabetes mellitus. Metabolism 37, 15–21 (1988).
  11. Consoli, A. Role of liver in pathophysiology of NIDDM. Diabetes Care 15, 430–441 (1992).
  12. Dinneen, S., Alzaid, A., Turk, D. & Rizza, R. Failure of glucagon suppression contributes to postprandial hyperglycaemia in IDDM. Diabetologia 38, 337–343 (1995).
  13. 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).
  14. Unger, R.H., Aguilar-Parada, E., Muller, W.A. & Eisentraut, A.M. Studies of pancreatic alpha cell function in normal and diabetic subjects. J. Clin. Invest. 49, 837–848 (1970).
  15. Alford, F.P. et al. Glucagon control of fasting glucose in man. Lancet 2, 974–977 (1974).
  16. Gargani, S. et al. Adaptive changes of human islets to an obesogenic environment in the mouse. Diabetologia 56, 350–358 (2013).
  17. Yi, P., Park, J.S. & Melton, D.A. Betatrophin: a hormone that controls pancreatic beta cell proliferation. Cell 153, 747–758 (2013).
  18. Stoffel, M. & Duncan, S.A. The maturity-onset diabetes of the young (MODY1) transcription factor HNF4alpha regulates expression of genes required for glucose transport and metabolism. Proc. Natl. Acad. Sci. USA 94, 13209–13214 (1997).
  19. Rhee, J. et al. Regulation of hepatic fasting response by PPARgamma coactivator-1α (PGC-1): requirement for hepatocyte nuclear factor 4α in gluconeogenesis. Proc. Natl. Acad. Sci. USA 100, 4012–4017 (2003).
  20. Love-Gregory, L.D. et al. A common polymorphism in the upstream promoter region of the hepatocyte nuclear factor-4α gene on chromosome 20q is associated with type 2 diabetes and appears to contribute to the evidence for linkage in an Ashkenazi Jewish population. Diabetes 53, 1134–1140 (2004).
  21. Weedon, M.N. et al. Common variants of the hepatocyte nuclear factor-4α P2 promoter are associated with type 2 diabetes in the U.K. population. Diabetes 53, 3002–3006 (2004).
  22. Han, S. et al. Dapagliflozin, a selective SGLT2 inhibitor, improves glucose homeostasis in normal and diabetic rats. Diabetes 57, 1723–1729 (2008).
  23. Leibiger, B. et al. Glucagon regulates its own synthesis by autocrine signaling. Proc. Natl. Acad. Sci. USA 109, 20925–20930 (2012).
  24. Gylfe, E. & Gilon, P. Glucose regulation of glucagon secretion. Diabetes Res. Clin. Pract. 103, 1–10 (2014).
  25. Zhang, Q. et al. Role of KATP channels in glucose-regulated glucagon secretion and impaired counterregulation in type 2 diabetes. Cell Metab. 18, 871–882 (2013).
  26. Mueckler, M. & Thorens, B. The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 34, 121–138 (2013).
  27. Fujimori, Y. et al. Remogliflozin etabonate, in a novel category of selective low-affinity sodium glucose cotransporter (SGLT2) inhibitors, exhibits antidiabetic efficacy in rodent models. J. Pharmacol. Exp. Ther. 327, 268–276 (2008).
  28. Jurczak, M.J. et al. SGLT2 deletion improves glucose homeostasis and preserves pancreatic beta-cell function. Diabetes 60, 890–898 (2011).
  29. Kojima, N., Williams, J.M., Takahashi, T., Miyata, N. & Roman, R.J. Effects of a new SGLT2 inhibitor, luseogliflozin, on diabetic nephropathy in T2DN rats. J. Pharmacol. Exp. Ther. 345, 464–472 (2013).
  30. Powell, D.R. et al. Improved glycemic control in mice lacking Sglt1 and Sglt2. Am. J. Physiol. Endocrinol. Metab. 304, E117–E130 (2013).
  31. Tahara, A. et al. Antidiabetic effects of SGLT2-selective inhibitor ipragliflozin in streptozotocin-nicotinamide-induced mildly diabetic mice. J. Pharmacol. Sci. 120, 36–44 (2012).
  32. Suzuki, M. et al. Tofogliflozin, a potent and highly specific sodium/glucose cotransporter 2 inhibitor, improves glycemic control in diabetic rats and mice. J. Pharmacol. Exp. Ther. 341, 692–701 (2012).
  33. Liang, Y. et al. Effect of canagliflozin on renal threshold for glucose, glycemia, and body weight in normal and diabetic animal models. PLoS ONE 7, e30555 (2012).
  34. Luippold, G., Klein, T., Mark, M. & Grempler, R. Empagliflozin, a novel potent and selective SGLT-2 inhibitor, improves glycaemic control alone and in combination with insulin in streptozotocin-induced diabetic rats, a model of type 1 diabetes mellitus. Diabetes Obes. Metab. 14, 601–607 (2012).
  35. Yamaguchi, K. et al. In vitro-in vivo correlation of the inhibition potency of sodium-glucose cotransporter inhibitors in rat: a pharmacokinetic and pharmacodynamic modeling approach. J. Pharmacol. Exp. Ther. 345, 52–61 (2013).
  36. Devenny, J.J. et al. Weight loss induced by chronic dapagliflozin treatment is attenuated by compensatory hyperphagia in diet-induced obese (DIO) rats. Obesity (Silver Spring) 20, 1645–1652 (2012).
  37. Nagata, T. et al. Selective SGLT2 inhibition by tofogliflozin reduces renal glucose reabsorption under hyperglycemic but not under hypo- or euglycemic conditions in rats. Am. J. Physiol. Endocrinol. Metab. 304, E414–E423 (2013).
  38. Cryer, P.E., Davis, S.N. & Shamoon, H. Hypoglycemia in diabetes. Diabetes Care 26, 1902–1912 (2003).
  39. DeFronzo, R.A., Ferrannini, E. & Simonson, D.C. Fasting hyperglycemia in non-insulin-dependent diabetes mellitus: contributions of excessive hepatic glucose production and impaired tissue glucose uptake. Metabolism 38, 387–395 (1989).
  40. Jurysta, C. et al. Glucose transport by acinar cells in rat parotid glands. Cell. Physiol. Biochem. 29, 325–330 (2012).
  41. Magen, D., Sprecher, E., Zelikovic, I. & Skorecki, K. A novel missense mutation in SLC5A2 encoding SGLT2 underlies autosomal-recessive renal glucosuria and aminoaciduria. Kidney Int. 67, 34–41 (2005).
  42. Kerr-Conte, J. et al. Upgrading pretransplant human islet culture technology requires human serum combined with media renewal. Transplantation 89, 1154–1160 (2010).
  43. Dorrell, C. et al. Transcriptomes of the major human pancreatic cell types. Diabetologia 54, 2832–2844 (2011).
  44. Lukowiak, B. et al. Identification and purification of functional human beta-cells by a new specific zinc-fluorescent probe. J. Histochem. Cytochem. 49, 519–528 (2001).
  45. Schulthess, F.T. et al. CXCL10 impairs beta cell function and viability in diabetes through TLR4 signaling. Cell Metab. 9, 125–139 (2009).
  46. Lefebvre, B. et al. Efficient gene delivery and silencing of mouse and human pancreatic islets. BMC Biotechnol. 10, 28 (2010).
  47. Obermeier, M. et al. In vitro characterization and pharmacokinetics of dapagliflozin (BMS-512148), a potent sodium-glucose cotransporter type II inhibitor, in animals and humans. Drug Metab. Dispos. 38, 405–414 (2010).
  48. Cheng-Xue, R. et al. Tolbutamide controls glucagon release from mouse islets differently than glucose: involvement of K(ATP) channels from both alpha-cells and delta-cells. Diabetes 62, 1612–1622 (2013).
  49. Abderrahmani, A. et al. Human high-density lipoprotein particles prevent activation of the JNK pathway induced by human oxidised low-density lipoprotein particles in pancreatic beta cells. Diabetologia 50, 1304–1314 (2007).
  50. de Jonge, H.J. et al. Evidence based selection of housekeeping genes. PLoS ONE 2, e898 (2007).

©2015 Nature America, Inc. All rights reserved.



Be confident that your patient care is up to date

Medicine Matters is being incorporated into Springer Medicine, our new medical education platform. 

Alongside the news coverage and expert commentary you have come to expect from Medicine Matters diabetes, Springer Medicine's complimentary membership also provides access to articles from renowned journals and a broad range of Continuing Medical Education programs. Create your free account »