Skip to main content
Top
Published in: Diabetologia 2/2009

Open Access 01-02-2009 | Article

Nutrient-dependent secretion of glucose-dependent insulinotropic polypeptide from primary murine K cells

Authors: H. E. Parker, A. M. Habib, G. J. Rogers, F. M. Gribble, F. Reimann

Published in: Diabetologia | Issue 2/2009

Login to get access

Abstract

Aims/hypothesis

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone with anti-apoptotic effects on the pancreatic beta cell. The aim of this study was to generate transgenic mice with fluorescently labelled GIP-secreting K cells and to use these to investigate pathways by which K cells detect nutrients.

Methods

Transgenic mice were generated in which the GIP promoter drives the expression of the yellow fluorescent protein Venus. Fluorescent cells were purified by flow cytometry and analysed by quantitative RT-PCR. GIP secretion was assayed in primary cultures of small intestine.

Results

Expression of Venus in transgenic mice was restricted to K cells, as assessed by immunofluorescence and measurements of the Gip mRNA and GIP protein contents of purified cells. K cells expressed high levels of mRNA for Kir6.2 (also known as Kcnj11), Sur1 (also known as Abcc8), Sglt1 (also known as Slc5a1), and of the G-protein-coupled lipid receptors Gpr40 (also known as Ffar1), Gpr119 and Gpr120. In primary cultures, GIP release was stimulated by glucose, glutamine and linoleic acid, and potentiated by forskolin plus 3-isobutyl-1-methylxanthine (IBMX), but was unaffected by the artificial sweetener sucralose. Secretion was half-maximal at 0.6 mmol/l glucose and partially mimicked by α-methylglucopyranoside, suggesting the involvement of SGLT1. Tolbutamide triggered secretion under basal conditions, whereas diazoxide suppressed responses in forskolin/IBMX.

Conclusions/interpretation

These transgenic mice and primary culture techniques provide novel opportunities to interrogate the mechanisms of GIP secretion. Glucose-triggered GIP secretion was SGLT1-dependent and modulated by KATP channel activity but not determined by sweet taste receptors. Synergistic stimulation by elevated cAMP and glucose suggests that targeting appropriate G-protein-coupled receptors may provide opportunities to modulate GIP release in vivo.
Appendix
Available only for authorised users
Literature
1.
go back to reference Nauck MA, Bartels E, Orskov C, Ebert R, Creutzfeldt W (1993) Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide-1-(7–36) amide infused at near-physiological insulinotropic hormone and glucose concentrations. J Clin Endocrinol Metab 76:912–917PubMedCrossRef Nauck MA, Bartels E, Orskov C, Ebert R, Creutzfeldt W (1993) Additive insulinotropic effects of exogenous synthetic human gastric inhibitory polypeptide and glucagon-like peptide-1-(7–36) amide infused at near-physiological insulinotropic hormone and glucose concentrations. J Clin Endocrinol Metab 76:912–917PubMedCrossRef
2.
4.
go back to reference Getty-Kaushik L, Song DH, Boylan MO, Corkey BE, Wolfe MM (2006) Glucose-dependent insulinotropic polypeptide modulates adipocyte lipolysis and reesterification. Obesity 14:1124–1131PubMedCrossRef Getty-Kaushik L, Song DH, Boylan MO, Corkey BE, Wolfe MM (2006) Glucose-dependent insulinotropic polypeptide modulates adipocyte lipolysis and reesterification. Obesity 14:1124–1131PubMedCrossRef
5.
go back to reference Kim SJ, Nian C, McIntosh CH (2007) Activation of lipoprotein lipase by glucose-dependent insulinotropic polypeptide in adipocytes. A role for a protein kinase B, LKB1, and AMP-activated protein kinase cascade. J Biol Chem 282:8557–8567PubMedCrossRef Kim SJ, Nian C, McIntosh CH (2007) Activation of lipoprotein lipase by glucose-dependent insulinotropic polypeptide in adipocytes. A role for a protein kinase B, LKB1, and AMP-activated protein kinase cascade. J Biol Chem 282:8557–8567PubMedCrossRef
6.
go back to reference Drucker DJ, Nauck MA (2006) The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368:1696–1705PubMedCrossRef Drucker DJ, Nauck MA (2006) The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368:1696–1705PubMedCrossRef
7.
go back to reference Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W (1993) Preserved incretin activity of glucagon-like peptide 1 [7–36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 91:301–307PubMedCrossRef Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W (1993) Preserved incretin activity of glucagon-like peptide 1 [7–36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 91:301–307PubMedCrossRef
8.
go back to reference Xu G, Kaneto H, Laybutt DR et al (2007) Downregulation of GLP-1 and GIP receptor expression by hyperglycaemia—possible contribution to impaired incretin effects in diabetes. Diabetes 56:1551–1558PubMedCrossRef Xu G, Kaneto H, Laybutt DR et al (2007) Downregulation of GLP-1 and GIP receptor expression by hyperglycaemia—possible contribution to impaired incretin effects in diabetes. Diabetes 56:1551–1558PubMedCrossRef
9.
go back to reference Trumper A, Trumper K, Horsch D (2002) Mechanisms of mitogenic and anti-apoptotic signaling by glucose-dependent insulinotropic polypeptide in beta(INS-1)-cells. J Endocrinol 174:233–246PubMedCrossRef Trumper A, Trumper K, Horsch D (2002) Mechanisms of mitogenic and anti-apoptotic signaling by glucose-dependent insulinotropic polypeptide in beta(INS-1)-cells. J Endocrinol 174:233–246PubMedCrossRef
10.
go back to reference Hinke SA, Gelling RW, Pederson RA et al (2002) Dipeptidyl peptidase IV-resistant [D-Ala(2)]glucose-dependent insulinotropic polypeptide (GIP) improves glucose tolerance in normal and obese diabetic rats. Diabetes 51:652–661PubMedCrossRef Hinke SA, Gelling RW, Pederson RA et al (2002) Dipeptidyl peptidase IV-resistant [D-Ala(2)]glucose-dependent insulinotropic polypeptide (GIP) improves glucose tolerance in normal and obese diabetic rats. Diabetes 51:652–661PubMedCrossRef
11.
go back to reference O’Harte FP, Mooney MH, Kelly CM, Flatt PR (2000) Improved glycaemic control in obese diabetic ob/ob mice using N-terminally modified gastric inhibitory polypeptide. J Endocrinol 165:639–648PubMedCrossRef O’Harte FP, Mooney MH, Kelly CM, Flatt PR (2000) Improved glycaemic control in obese diabetic ob/ob mice using N-terminally modified gastric inhibitory polypeptide. J Endocrinol 165:639–648PubMedCrossRef
12.
13.
go back to reference Miyawaki K, Yamada Y, Yano H et al (1999) Glucose intolerance caused by a defect in the entero-insular axis: a study in gastric inhibitory polypeptide receptor knockout mice. Proc Natl Acad Sci U S A 96:14843–14847PubMedCrossRef Miyawaki K, Yamada Y, Yano H et al (1999) Glucose intolerance caused by a defect in the entero-insular axis: a study in gastric inhibitory polypeptide receptor knockout mice. Proc Natl Acad Sci U S A 96:14843–14847PubMedCrossRef
14.
go back to reference Miyawaki K, Yamada Y, Ban N et al (2002) Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nat Med 8:738–742PubMedCrossRef Miyawaki K, Yamada Y, Ban N et al (2002) Inhibition of gastric inhibitory polypeptide signaling prevents obesity. Nat Med 8:738–742PubMedCrossRef
15.
go back to reference Althage MC, Ford EL, Wang S, Tso P, Polonsky KS, Wice BM (2008) Targeted ablation of GIP-producing cells in transgenic mice reduces obesity and insulin resistance induced by a high fat diet. J Biol Chem 283:18365–18376PubMedCrossRef Althage MC, Ford EL, Wang S, Tso P, Polonsky KS, Wice BM (2008) Targeted ablation of GIP-producing cells in transgenic mice reduces obesity and insulin resistance induced by a high fat diet. J Biol Chem 283:18365–18376PubMedCrossRef
16.
go back to reference Irwin N, McClean PL, O’Harte FP, Gault VA, Harriott P, Flatt PR (2007) Early administration of the glucose-dependent insulinotropic polypeptide receptor antagonist (Pro(3))GIP prevents the development of diabetes and related metabolic abnormalities associated with genetically inherited obesity in ob/ob mice. Diabetologia 50:1532–1540PubMedCrossRef Irwin N, McClean PL, O’Harte FP, Gault VA, Harriott P, Flatt PR (2007) Early administration of the glucose-dependent insulinotropic polypeptide receptor antagonist (Pro(3))GIP prevents the development of diabetes and related metabolic abnormalities associated with genetically inherited obesity in ob/ob mice. Diabetologia 50:1532–1540PubMedCrossRef
17.
go back to reference McClean PL, Irwin N, Cassidy RS, Holst JJ, Gault VA, Flatt PR (2007) GIP receptor antagonism reverses obesity, insulin resistance, and associated metabolic disturbances induced in mice by prolonged consumption of high-fat diet. Am J Physiol Endocrinol Metab 293:E1746–E1755PubMedCrossRef McClean PL, Irwin N, Cassidy RS, Holst JJ, Gault VA, Flatt PR (2007) GIP receptor antagonism reverses obesity, insulin resistance, and associated metabolic disturbances induced in mice by prolonged consumption of high-fat diet. Am J Physiol Endocrinol Metab 293:E1746–E1755PubMedCrossRef
18.
go back to reference Ebert R, Creutzfeldt W (1982) Influence of gastric inhibitory polypeptide antiserum on glucose-induced insulin secretion in rats. Endocrinology 111:1601–1606PubMedCrossRef Ebert R, Creutzfeldt W (1982) Influence of gastric inhibitory polypeptide antiserum on glucose-induced insulin secretion in rats. Endocrinology 111:1601–1606PubMedCrossRef
19.
go back to reference Tseng CC, Kieffer TJ, Jarboe LA, Usdin TB, Wolfe MM (1996) Postprandial stimulation of insulin release by glucose-dependent insulinotropic polypeptide (GIP). Effect of a specific glucose-dependent insulinotropic polypeptide receptor antagonist in the rat. J Clin Invest 98:2440–2445PubMedCrossRef Tseng CC, Kieffer TJ, Jarboe LA, Usdin TB, Wolfe MM (1996) Postprandial stimulation of insulin release by glucose-dependent insulinotropic polypeptide (GIP). Effect of a specific glucose-dependent insulinotropic polypeptide receptor antagonist in the rat. J Clin Invest 98:2440–2445PubMedCrossRef
20.
go back to reference McClean PL, Gault VA, Irwin N, McCluskey JT, Flatt PR (2008) Daily administration of the GIP-R antagonist (Pro3)GIP in streptozotocin-induced diabetes suggests that insulin-dependent mechanisms are critical to anti-obesity-diabetes actions of (Pro3)GIP. Diabetes Obes Metab 10:336–342PubMedCrossRef McClean PL, Gault VA, Irwin N, McCluskey JT, Flatt PR (2008) Daily administration of the GIP-R antagonist (Pro3)GIP in streptozotocin-induced diabetes suggests that insulin-dependent mechanisms are critical to anti-obesity-diabetes actions of (Pro3)GIP. Diabetes Obes Metab 10:336–342PubMedCrossRef
21.
go back to reference Elliot RM, Morgan LM, Tredger JA, Deacon S, Wright J, Marks V (1993) Glucagon-like peptide-1 (7–36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J Endocrinol 138:159–166CrossRef Elliot RM, Morgan LM, Tredger JA, Deacon S, Wright J, Marks V (1993) Glucagon-like peptide-1 (7–36)amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J Endocrinol 138:159–166CrossRef
22.
go back to reference Meier JJ, Nauck MA (2004) Glucose-dependent insulinotropic polypeptide/gastric inhibitory polypeptide. Best Pract Res Clin Endocrinol Metab 18:587–606PubMedCrossRef Meier JJ, Nauck MA (2004) Glucose-dependent insulinotropic polypeptide/gastric inhibitory polypeptide. Best Pract Res Clin Endocrinol Metab 18:587–606PubMedCrossRef
23.
go back to reference Buffa R, Polak JM, Pearse AGE, Solcia E, Grimelius L, Capella C (1975) Identification of the intestinal cell storing gastric inhibitory peptide. Histochemistry 43:249–255PubMedCrossRef Buffa R, Polak JM, Pearse AGE, Solcia E, Grimelius L, Capella C (1975) Identification of the intestinal cell storing gastric inhibitory peptide. Histochemistry 43:249–255PubMedCrossRef
24.
go back to reference Buchan AM, Polak JM, Capella C, Solcia E, Pearse AG (1978) Electronimmunocytochemical evidence for the K cell localization of gastric inhibitory polypeptide (GIP) in man. Histochemistry 56:37–44PubMedCrossRef Buchan AM, Polak JM, Capella C, Solcia E, Pearse AG (1978) Electronimmunocytochemical evidence for the K cell localization of gastric inhibitory polypeptide (GIP) in man. Histochemistry 56:37–44PubMedCrossRef
25.
go back to reference Sykes S, Morgan LM, English J, Marks V (1980) Evidence for preferential stimulation of gastric inhibitory polypeptide secretion in the rat by actively transported carbohydrates and their analogues. J Endocrinol 85:201–207PubMedCrossRef Sykes S, Morgan LM, English J, Marks V (1980) Evidence for preferential stimulation of gastric inhibitory polypeptide secretion in the rat by actively transported carbohydrates and their analogues. J Endocrinol 85:201–207PubMedCrossRef
26.
go back to reference Flatt PR, Kwasowski P, Bailey CJ (1989) Stimulation of gastric inhibitory polypeptide release in ob/ob mice by oral administration of sugars and their analogues. J Nutr 119:1300–1303PubMed Flatt PR, Kwasowski P, Bailey CJ (1989) Stimulation of gastric inhibitory polypeptide release in ob/ob mice by oral administration of sugars and their analogues. J Nutr 119:1300–1303PubMed
27.
go back to reference Fushiki T, Kojima A, Imoto T, Inoue K, Sugimoto E (1992) An extract of Gymnema sylvestre leaves and purified gymnemic acid inhibits glucose-stimulated gastric inhibitory peptide secretion in rats. J Nutr 122:2367–2373PubMed Fushiki T, Kojima A, Imoto T, Inoue K, Sugimoto E (1992) An extract of Gymnema sylvestre leaves and purified gymnemic acid inhibits glucose-stimulated gastric inhibitory peptide secretion in rats. J Nutr 122:2367–2373PubMed
28.
go back to reference Rindi G, Grant SG, Yiangou Y et al (1990) Development of neuroendocrine tumors in the gastrointestinal tract of transgenic mice. Heterogeneity of hormone expression. Am J Pathol 136:1349–1363PubMed Rindi G, Grant SG, Yiangou Y et al (1990) Development of neuroendocrine tumors in the gastrointestinal tract of transgenic mice. Heterogeneity of hormone expression. Am J Pathol 136:1349–1363PubMed
29.
go back to reference Mortensen K, Christensen LL, Holst JJ, Ørskov C (2003) GLP-1 and GIP are colocalized in a subset of endocrine cells in the small intestine. Regul Pept 114:189–196PubMedCrossRef Mortensen K, Christensen LL, Holst JJ, Ørskov C (2003) GLP-1 and GIP are colocalized in a subset of endocrine cells in the small intestine. Regul Pept 114:189–196PubMedCrossRef
30.
go back to reference Gribble FM, Williams L, Simpson AK, Reimann F (2003) A novel glucose-sensing mechanism contributing to glucagon-like peptide-1 secretion from the GLUTag cell line. Diabetes 52:1147–1154PubMedCrossRef Gribble FM, Williams L, Simpson AK, Reimann F (2003) A novel glucose-sensing mechanism contributing to glucagon-like peptide-1 secretion from the GLUTag cell line. Diabetes 52:1147–1154PubMedCrossRef
31.
go back to reference Reimann F, Gribble FM (2002) Glucose-sensing in glucagon-like peptide-1-secreting cells. Diabetes 51:2757–2763PubMedCrossRef Reimann F, Gribble FM (2002) Glucose-sensing in glucagon-like peptide-1-secreting cells. Diabetes 51:2757–2763PubMedCrossRef
32.
go back to reference Li L, Wice BM (2005) Bombesin and nutrients independently and additively regulate hormone release from GIP/Ins cells. Am J Physiol Endocrinol Metab 288:E208–E215PubMedCrossRef Li L, Wice BM (2005) Bombesin and nutrients independently and additively regulate hormone release from GIP/Ins cells. Am J Physiol Endocrinol Metab 288:E208–E215PubMedCrossRef
33.
go back to reference Wang SY, Chi MM, Li L, Moley KH, Wice BM (2003) Studies with GIP/Ins cells indicate secretion by gut K cells is KATP channel independent. Am J Physiol Endocrinol Metab 284:E988–E1000PubMed Wang SY, Chi MM, Li L, Moley KH, Wice BM (2003) Studies with GIP/Ins cells indicate secretion by gut K cells is KATP channel independent. Am J Physiol Endocrinol Metab 284:E988–E1000PubMed
34.
go back to reference Jang HJ, Kokrashvili Z, Theodorakis MJ et al (2007) Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A 104:15069–15074PubMedCrossRef Jang HJ, Kokrashvili Z, Theodorakis MJ et al (2007) Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc Natl Acad Sci U S A 104:15069–15074PubMedCrossRef
35.
go back to reference Margolskee RF, Dyer J, Kokrashvili Z et al (2007) T1R3 and gustducin in gut sense sugars to regulate expression of Na+ -glucose cotransporter 1. Proc Natl Acad Sci U S A 104:15075–15080PubMedCrossRef Margolskee RF, Dyer J, Kokrashvili Z et al (2007) T1R3 and gustducin in gut sense sugars to regulate expression of Na+ -glucose cotransporter 1. Proc Natl Acad Sci U S A 104:15075–15080PubMedCrossRef
36.
go back to reference Zhang Y, Muyrers JP, Testa G, Stewart AF (2000) DNA cloning by homologous recombination in Escherichia coli. Nat Biotechnol 18:1314–1317PubMedCrossRef Zhang Y, Muyrers JP, Testa G, Stewart AF (2000) DNA cloning by homologous recombination in Escherichia coli. Nat Biotechnol 18:1314–1317PubMedCrossRef
37.
go back to reference Mace OJ, Affleck J, Patel N, Kellett GL (2007) Sweet taste receptors in rat small intestine stimulate glucose absorption through apical GLUT2. J Physiol 582:379–392PubMedCrossRef Mace OJ, Affleck J, Patel N, Kellett GL (2007) Sweet taste receptors in rat small intestine stimulate glucose absorption through apical GLUT2. J Physiol 582:379–392PubMedCrossRef
38.
go back to reference Polak JM, Pearse AGE, Grimelius L, Marks V (1975) Gastrointestinal apudosis in obese hyperglycaemic mice. Virchows Arch B Cell Path 19:135–150 Polak JM, Pearse AGE, Grimelius L, Marks V (1975) Gastrointestinal apudosis in obese hyperglycaemic mice. Virchows Arch B Cell Path 19:135–150
39.
go back to reference Habib AM, Reimann F, Parker HE, Gribble FM (2007) Expression of glucose-sensing machinery in intestinal GLP-1 secreting cells. Diabetologia 50(Suppl 1):S287 (Abstract) Habib AM, Reimann F, Parker HE, Gribble FM (2007) Expression of glucose-sensing machinery in intestinal GLP-1 secreting cells. Diabetologia 50(Suppl 1):S287 (Abstract)
40.
go back to reference Reimann F, Williams L, da Silva Xavier G, Rutter GA, Gribble FM (2004) Glutamine potently stimulates glucagon-like peptide-1 secretion from GLUTag cells. Diabetologia 47:1592–1601PubMedCrossRef Reimann F, Williams L, da Silva Xavier G, Rutter GA, Gribble FM (2004) Glutamine potently stimulates glucagon-like peptide-1 secretion from GLUTag cells. Diabetologia 47:1592–1601PubMedCrossRef
41.
go back to reference Hirasawa A, Tsumaya K, Awaji T et al (2005) Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med 11:90–94PubMedCrossRef Hirasawa A, Tsumaya K, Awaji T et al (2005) Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med 11:90–94PubMedCrossRef
42.
go back to reference Chu ZL, Carroll C, Alfonso J et al (2008) A role for intestinal endocrine cell-expressed g protein-coupled receptor 119 in glycemic control by enhancing glucagon-like peptide-1 and glucose-dependent insulinotropic peptide release. Endocrinology 149:2038–2047PubMedCrossRef Chu ZL, Carroll C, Alfonso J et al (2008) A role for intestinal endocrine cell-expressed g protein-coupled receptor 119 in glycemic control by enhancing glucagon-like peptide-1 and glucose-dependent insulinotropic peptide release. Endocrinology 149:2038–2047PubMedCrossRef
43.
go back to reference Overton HA, Babbs AJ, Doel SM et al (2006) Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents. Cell Metab 3:167–175PubMedCrossRef Overton HA, Babbs AJ, Doel SM et al (2006) Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents. Cell Metab 3:167–175PubMedCrossRef
44.
go back to reference Díez-Sampedro A, Lostao MP, Wright EM, Hirayama BA (2000) Glycoside binding and translocation in Na(+)-dependent glucose cotransporters: comparison of SGLT1 and SGLT3. J Membr Biol 176:111–117PubMedCrossRef Díez-Sampedro A, Lostao MP, Wright EM, Hirayama BA (2000) Glycoside binding and translocation in Na(+)-dependent glucose cotransporters: comparison of SGLT1 and SGLT3. J Membr Biol 176:111–117PubMedCrossRef
45.
go back to reference Fujita Y, Speck M, Asadi A, Webb T, Kieffer TM (2008) Incretin release from gut is enhanced by sugar but not by sweeteners in vivo. 68th Scientific Session of the American Diabetes Association in San Francisco 2008: 1451-P (Abstract). Available from http://professional.diabetes.org/Abstracts Fujita Y, Speck M, Asadi A, Webb T, Kieffer TM (2008) Incretin release from gut is enhanced by sugar but not by sweeteners in vivo. 68th Scientific Session of the American Diabetes Association in San Francisco 2008: 1451-P (Abstract). Available from http://​professional.​diabetes.​org/​Abstracts
46.
go back to reference Nielsen LB, Ploug KB, Swift P et al (2007) Co-localisation of the Kir6.2/SUR1 channel complex with glucagon-like peptide-1 and glucose-dependent insulinotrophic polypeptide expression in human ileal cells and implications for glycaemic control in new onset type 1 diabetes. Eur J Endocrinol 156:663–671PubMedCrossRef Nielsen LB, Ploug KB, Swift P et al (2007) Co-localisation of the Kir6.2/SUR1 channel complex with glucagon-like peptide-1 and glucose-dependent insulinotrophic polypeptide expression in human ileal cells and implications for glycaemic control in new onset type 1 diabetes. Eur J Endocrinol 156:663–671PubMedCrossRef
47.
go back to reference Uldry M, Thorens B (2004) The SLC2 family of facilitated hexose and polyol transporters. Pflugers Arch 447:480–489PubMedCrossRef Uldry M, Thorens B (2004) The SLC2 family of facilitated hexose and polyol transporters. Pflugers Arch 447:480–489PubMedCrossRef
Metadata
Title
Nutrient-dependent secretion of glucose-dependent insulinotropic polypeptide from primary murine K cells
Authors
H. E. Parker
A. M. Habib
G. J. Rogers
F. M. Gribble
F. Reimann
Publication date
01-02-2009
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 2/2009
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-008-1202-x

Other articles of this Issue 2/2009

Diabetologia 2/2009 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine