Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Metabolic Messengers
  • Published:

Metabolic Messengers: glucagon-like peptide 1

Abstract

Glucagon like peptide-1 (GLP-1), a peptide hormone from the intestinal tract, plays a central role in the coordination of postprandial glucose homeostasis through actions on insulin secretion, food intake and gut motility. GLP-1 forms the basis for a variety of current drugs for the treatment of type 2 diabetes and obesity, as well as new agents currently being developed. Here, we provide a concise overview of the core physiology of GLP-1 secretion and action, and the role of the peptide in human health, disease and therapeutics.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Timeline of GLP-1 discovery and clinical development.
Fig. 2: Food-dependent stimulation of GLP-1 release from small intestine L-cells.
Fig. 3: Major GLP-1 targets.

Similar content being viewed by others

References

  1. Moore, B. On the treatment of diabetus mellitus by acid extract of duodenal mucous membrane. Biochem. J. 1, 28–38 (1906).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Holst, J. J. The physiology of glucagon-like peptide 1. Physiol. Rev. 87, 1409–1439 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Thorens, B. Expression cloning of the pancreatic beta cell receptor for the gluco-incretin hormone glucagon-like peptide 1. Proc. Natl. Acad. Sci. USA 89, 8641–8645 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Nauck, M. A. et al. Normalization of fasting hyperglycaemia by exogenous glucagon-like peptide 1 (7-36 amide) in type 2 (non-insulin-dependent) diabetic patients. Diabetologia 36, 741–744 (1993).

    Article  CAS  PubMed  Google Scholar 

  5. Gribble, F. M. & Reimann, F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat. Rev. Endocrinol. 15, 226–237 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Sjölund, K., Sandén, G., Håkanson, R. & Sundler, F. Endocrine cells in human intestine: an immunocytochemical study. Gastroenterology 85, 1120–1130 (1983).

    Article  PubMed  Google Scholar 

  7. Lewis, J. E. et al. Selective stimulation of colonic L cells improves metabolic outcomes in mice. Diabetologia 63, 1396–1407 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Batterham, R. L. et al. Inhibition of food intake in obese subjects by peptide YY3-36. N. Engl. J. Med. 349, 941–948 (2003).

    Article  CAS  PubMed  Google Scholar 

  9. Gribble, F. M. & Reimann, F. Enteroendocrine cells: chemosensors in the intestinal epithelium. Annu. Rev. Physiol. 78, 277–299 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Brubaker, P. L., Schloos, J. & Drucker, D. J. Regulation of glucagon-like peptide-1 synthesis and secretion in the GLUTag enteroendocrine cell line. Endocrinology 139, 4108–4114 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Edfalk, S., Steneberg, P. & Edlund, H. Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes 57, 2280–2287 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chu, Z. L. et al. 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, 2038–2047 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Hansen, K. B. et al. 2-Oleoyl glycerol is a GPR119 agonist and signals GLP-1 release in humans. J. Clin. Endocrinol. Metab. 96, E1409–E1417 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Mace, O. J., Schindler, M. & Patel, S. The regulation of K- and L-cell activity by GLUT2 and CasR in rat small intestine. J. Physiol. 590, 2917–2936 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Thomas, C. et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab 10, 167–177 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bolognini, D., Tobin, A. B., Milligan, G. & Moss, C. E. The pharmacology and function of receptors for short-chain fatty acids. Mol. Pharmacol. 89, 388–398 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Pais, R., Rievaj, J., Larraufie, P., Gribble, F. & Reimann, F. Angiotensin II type 1 receptor-dependent GLP-1 and PYY secretion in mice and humans. Endocrinology 157, 3821–3831 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pais, R. et al. Role of enteroendocrine L-cells in arginine vasopressin-mediated inhibition of colonic anion secretion. J. Physiol. (Lond.) 594, 4865–4878 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Psichas, A., Glass, L. L., Sharp, S. J., Reimann, F. & Gribble, F. M. Galanin inhibits GLP-1 and GIP secretion via the GAL1 receptor in enteroendocrine L and K cells. Br. J. Pharmacol. 173, 888–898 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Roberge, J. N., Gronau, K. A. & Brubaker, P. L. Gastrin-releasing peptide is a novel mediator of proximal nutrient-induced proglucagon-derived peptide secretion from the distal gut. Endocrinology 137, 2383–2388 (1996).

    Article  CAS  PubMed  Google Scholar 

  21. Jang, H. J. et al. Gut-expressed gustducin and taste receptors regulate secretion of glucagon-like peptide-1. Proc. Natl Acad. Sci. USA 104, 15069–15074 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Saltiel, M. Y. et al. Sweet taste receptor activation in the gut is of limited importance for glucose-stimulated GLP-1 and GIP secretion. Nutrients 9, E418 (2017).

    Article  Google Scholar 

  23. Reimann, F. et al. Glucose sensing in L cells: a primary cell study. Cell Metab. 8, 532–539 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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).

    Article  CAS  PubMed  Google Scholar 

  25. Gorboulev, V. et al. Na+-D-glucose cotransporter SGLT1 is pivotal for intestinal glucose absorption and glucose-dependent incretin secretion. Diabetes 61, 187–196 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Reimann, F., Williams, L., da Silva Xavier, G., Rutter, G. A. & Gribble, F. M. Glutamine potently stimulates glucagon-like peptide-1 secretion from GLUTag cells. Diabetologia 47, 1592–1601 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Matsumura, K., Miki, T., Jhomori, T., Gonoi, T. & Seino, S. Possible role of PEPT1 in gastrointestinal hormone secretion. Biochem. Biophys. Res. Commun. 336, 1028–1032 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Diakogiannaki, E. et al. Oligopeptides stimulate glucagon-like peptide-1 secretion in mice through proton-coupled uptake and the calcium-sensing receptor. Diabetologia 56, 2688–2696 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Parker, H. E. et al. Predominant role of active versus facilitative glucose transport for glucagon-like peptide-1 secretion. Diabetologia 55, 2445–2455 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Powell, D. R. et al. LX4211 increases serum glucagon-like peptide 1 and peptide YY levels by reducing sodium/glucose cotransporter 1 (SGLT1)-mediated absorption of intestinal glucose. J. Pharmacol. Exp. Ther. 345, 250–259 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Christiansen, C. B. et al. The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. Am. J. Physiol. Gastrointest. Liver Physiol. 315, G53–G65 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Jørgensen, N. B. et al. Acute and long-term effects of Roux-en-Y gastric bypass on glucose metabolism in subjects with type 2 diabetes and normal glucose tolerance. Am. J. Physiol. Endocrinol. Metab. 303, E122–E131 (2012).

    Article  PubMed  Google Scholar 

  33. Martinussen, C. et al. The effect of acute dual SGLT1/SGLT2 inhibition on incretin release and glucose metabolism after gastric bypass surgery. Am. J. Physiol. Endocrinol. Metab. 318, E956–E964 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. El-Ouaghlidi, A. et al. The dipeptidyl peptidase 4 inhibitor vildagliptin does not accentuate glibenclamide-induced hypoglycemia but reduces glucose-induced glucagon-like peptide 1 and gastric inhibitory polypeptide secretion. J. Clin. Endocrinol. Metab. 92, 4165–4171 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Reimann, F., Tolhurst, G. & Gribble, F. M. G-protein-coupled receptors in intestinal chemosensation. Cell Metab. 15, 421–431 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Goldspink, D. A. et al. Mechanistic insights into the detection of free fatty and bile acids by ileal glucagon-like peptide-1 secreting cells. Mol. Metab. 7, 90–101 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Goldspink, D. A. et al. Labeling and characterization of human GLP-1-secreting L-cells in primary ileal organoid culture. Cell Rep. 31, 107833 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Brighton, C. A. et al. Bile acids trigger GLP-1 release predominantly by accessing basolaterally located G protein-coupled bile acid receptors. Endocrinology 156, 3961–3970 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Christensen, L. W., Kuhre, R. E., Janus, C., Svendsen, B. & Holst, J. J. Vascular, but not luminal, activation of FFAR1 (GPR40) stimulates GLP-1 secretion from isolated perfused rat small intestine. Physiol. Rep. 3, e12551 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Modvig, I. M., Kuhre, R. E. & Holst, J. J. Peptone-mediated glucagon-like peptide-1 secretion depends on intestinal absorption and activation of basolaterally located calcium-sensing receptors. Physiol. Rep. 7, e14056 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Llewellyn-Smith, I. J., Reimann, F., Gribble, F. M. & Trapp, S. Preproglucagon neurons project widely to autonomic control areas in the mouse brain. Neuroscience 180, 111–121 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. Orskov, C., Holst, J. J., Poulsen, S. S. & Kirkegaard, P. Pancreatic and intestinal processing of proglucagon in man. Diabetologia 30, 874–881 (1987).

    Article  CAS  PubMed  Google Scholar 

  43. Rouillé, Y. et al. Proteolytic processing mechanisms in the biosynthesis of neuroendocrine peptides: the subtilisin-like proprotein convertases. Front. Neuroendocrinol. 16, 322–361 (1995).

    Article  PubMed  Google Scholar 

  44. Larraufie, P. et al. Important role of the GLP-1 axis for glucose homeostasis after bariatric surgery. Cell Rep. 26, 1399–1408.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Song, Y. et al. Gut-proglucagon-derived peptides are essential for regulating glucose homeostasis in mice. Cell Metab. 30, 976–986.e3 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ban, K. et al. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 117, 2340–2350 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Richards, P. et al. Identification and characterization of GLP-1 receptor-expressing cells using a new transgenic mouse model. Diabetes 63, 1224–1233 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Pyke, C. et al. GLP-1 receptor localization in monkey and human tissue: novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 155, 1280–1290 (2014).

    Article  PubMed  Google Scholar 

  49. He, S. et al. Gut intraepithelial T cells calibrate metabolism and accelerate cardiovascular disease. Nature 566, 115–119 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Baggio, L. L. et al. GLP-1 receptor expression within the human heart. Endocrinology 159, 1570–1584 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dhir, G. & Cusi, K. Glucagon like peptide-1 receptor agonists for the management of obesity and non-alcoholic fatty liver disease: a novel therapeutic option. J. Investig. Med. 66, 7–10 (2018).

    Article  PubMed  Google Scholar 

  52. Gromada, J., Holst, J. J. & Rorsman, P. Cellular regulation of islet hormone secretion by the incretin hormone glucagon-like peptide 1. Pflugers Arch. 435, 583–594 (1998).

    Article  CAS  PubMed  Google Scholar 

  53. Creutzfeldt, W. The [pre-] history of the incretin concept. Regul. Pept. 128, 87–91 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Nauck, M. A., Bartels, E., Orskov, C., Ebert, R. & Creutzfeldt, W. 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, 912–917 (1993).

    CAS  PubMed  Google Scholar 

  55. Gasbjerg, L. S. et al. Separate and combined glucometabolic effects of endogenous glucose-dependent insulinotropic polypeptide and glucagon-like peptide 1 in healthy individuals. Diabetes 68, 906–917 (2019).

    Article  CAS  PubMed  Google Scholar 

  56. Jørgensen, N. B. et al. Exaggerated glucagon-like peptide 1 response is important for improved β-cell function and glucose tolerance after Roux-en-Y gastric bypass in patients with type 2 diabetes. Diabetes 62, 3044–3052 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Hansen, L., Deacon, C. F., Orskov, C. & Holst, J. J. Glucagon-like peptide-1-(7-36)amide is transformed to glucagon-like peptide-1-(9-36)amide by dipeptidyl peptidase IV in the capillaries supplying the L cells of the porcine intestine. Endocrinology 140, 5356–5363 (1999).

    Article  CAS  PubMed  Google Scholar 

  58. Vahl, T. P. et al. Glucagon-like peptide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein mediate the effects of endogenous GLP-1 on glucose tolerance in rats. Endocrinology 148, 4965–4973 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Woerle, H. J., Carneiro, L., Derani, A., Göke, B. & Schirra, J. The role of endogenous incretin secretion as amplifier of glucose-stimulated insulin secretion in healthy subjects and patients with type 2 diabetes. Diabetes 61, 2349–2358 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chambers, A. P. et al. The role of pancreatic preproglucagon in glucose homeostasis in mice. Cell Metab. 25, 927–934.e3 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Svendsen, B. et al. Insulin secretion depends on intra-islet glucagon signaling. Cell Rep. 25, 1127–1134.e2 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. 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).

    Article  CAS  PubMed  Google Scholar 

  63. Gasbjerg, L. S. et al. Evaluation of the incretin effect in humans using GIP and GLP-1 receptor antagonists. Peptides 125, 170183 (2020).

    Article  CAS  PubMed  Google Scholar 

  64. Wu, T., Rayner, C. K., Young, R. L. & Horowitz, M. Gut motility and enteroendocrine secretion. Curr. Opin. Pharmacol. 13, 928–934 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Meier, J. J. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 8, 728–742 (2012).

    Article  CAS  PubMed  Google Scholar 

  66. Krieger, J. P. et al. Knockdown of GLP-1 receptors in vagal afferents affects normal food intake and glycemia. Diabetes 65, 34–43 (2016).

    Article  CAS  PubMed  Google Scholar 

  67. Nauck, M. A., Quast, D. R., Wefers, J. & Meier, J. J. GLP-1 receptor agonists in the treatment of type 2 diabetes: state-of-the-art. Mol. Metab. https://doi.org/10.1016/j.molmet.2020.101102 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Knudsen, L. B. & Lau, J. The discovery and development of liraglutide and semaglutide. Front. Endocrinol. (Lausanne) 10, 155 (2019).

    Article  PubMed  Google Scholar 

  69. Jepsen, S. L. et al. Paracrine crosstalk between intestinal L- and D-cells controls secretion of glucagon-like peptide-1 in mice. Am. J. Physiol. Endocrinol. Metab. 317, E1081–E1093 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Svane, M. S. et al. Peptide YY and glucagon-like peptide-1 contribute to decreased food intake after Roux-en-Y gastric bypass surgery. Int. J. Obes. (Lond) 40, 1699–1706 (2016).

    Article  CAS  PubMed  Google Scholar 

  71. Williams, E. K. et al. Sensory neurons that detect stretch and nutrients in the digestive system. Cell 166, 209–221 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Lu, V. B. et al. Adenosine triphosphate is co-secreted with glucagon-like peptide-1 to modulate intestinal enterocytes and afferent neurons. Nat. Commun. 10, 1029 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Trapp, S. & Cork, S. C. PPG neurons of the lower brain stem and their role in brain GLP-1 receptor activation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 309, R795–R804 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kreisler, A. D. & Rinaman, L. Hindbrain glucagon-like peptide-1 neurons track intake volume and contribute to injection stress-induced hypophagia in meal-entrained rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 310, R906–R916 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Holt, M. K. et al. Preproglucagon neurons in the nucleus of the solitary tract are the main source of brain GLP-1, mediate stress-induced hypophagia, and limit unusually large intakes of food. Diabetes 68, 21–33 (2019).

    Article  CAS  PubMed  Google Scholar 

  76. Cheng, W. et al. Leptin receptor-expressing nucleus tractus solitarius neurons suppress food intake independently of GLP1 in mice. JCI Insight 5, 134359 (2020).

    Article  PubMed  Google Scholar 

  77. Sun, F. et al. Impact of GLP-1 receptor agonists on blood pressure, heart rate and hypertension among patients with type 2 diabetes: a systematic review and network meta-analysis. Diabetes Res. Clin. Pract. 110, 26–37 (2015).

    Article  CAS  PubMed  Google Scholar 

  78. Giblett, J. P., Clarke, S. J., Dutka, D. P. & Hoole, S. P. Glucagon-like peptide-1: a promising agent for cardioprotection during myocardial ischemia. JACC Basic Transl. Sci. 1, 267–276 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Holt, M. K. et al. PPG neurons in the nucleus of the solitary tract modulate heart rate but do not mediate GLP-1 receptor agonist-induced tachycardia in mice. Mol. Metab. 39, 101024 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Müller, T. D. et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 30, 72–130 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Cheng, H. & Leblond, C. P. Origin, differentiation and renewal of the four main epithelial cell types in the mouse small intestine. III. Entero-endocrine cells. Am. J. Anat. 141, 503–519 (1974).

    CAS  PubMed  Google Scholar 

  82. Mumphrey, M. B., Patterson, L. M., Zheng, H. & Berthoud, H. R. Roux-en-Y gastric bypass surgery increases number but not density of CCK-, GLP-1-, 5-HT-, and neurotensin-expressing enteroendocrine cells in rats. Neurogastroenterol. Motil. 25, e70–e79 (2013).

    Article  CAS  PubMed  Google Scholar 

  83. Nauck, M., Stöckmann, F., Ebert, R. & Creutzfeldt, W. Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 29, 46–52 (1986).

    Article  CAS  PubMed  Google Scholar 

  84. Vollmer, K. et al. Hyperglycemia acutely lowers the postprandial excursions of glucagon-like peptide-1 and gastric inhibitory polypeptide in humans. J. Clin. Endocrinol. Metab. 94, 1379–1385 (2009).

    Article  CAS  PubMed  Google Scholar 

  85. Wang, J. et al. Mutant neurogenin-3 in congenital malabsorptive diarrhea. N. Engl. J. Med. 355, 270–280 (2006).

    Article  CAS  PubMed  Google Scholar 

  86. Jackson, R. S. et al. Small-intestinal dysfunction accompanies the complex endocrinopathy of human proprotein convertase 1 deficiency. J. Clin. Invest. 112, 1550–1560 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kay, R. G. et al. Peptidomic analysis of endogenous plasma peptides from patients with pancreatic neuroendocrine tumours. Rapid Commun. Mass Spectrom. 32, 1414–1424 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Tan, M., Lamendola, C., Luong, R., McLaughlin, T. & Craig, C. Safety, efficacy and pharmacokinetics of repeat subcutaneous dosing of avexitide (exendin 9-39) for treatment of post-bariatric hypoglycaemia. Diabetes Obes. Metab. 22, 1406–1416 (2020).

    Article  CAS  PubMed  Google Scholar 

  89. Deacon, C. F. Peptide degradation and the role of DPP-4 inhibitors in the treatment of type 2 diabetes. Peptides 100, 150–157 (2018).

    Article  CAS  PubMed  Google Scholar 

  90. Thethi, T. K., Pratley, R. & Meier, J. J. Efficacy, safety and cardiovascular outcomes of once-daily oral semaglutide in patients with type 2 diabetes: The PIONEER programme. Diabetes Obes. Metab. 22, 1263–1277 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhang, X. et al. Differential GLP-1R binding and activation by peptide and non-peptide agonists. Mol. Cell 80, 485–500.e7 (2020).

    Article  CAS  PubMed  Google Scholar 

  92. Lucey, M. et al. Disconnect between signalling potency and in vivo efficacy of pharmacokinetically optimised biased glucagon-like peptide-1 receptor agonists. Mol. Metab. 37, 100991 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Tschöp, M. H. et al. Unimolecular polypharmacy for treatment of diabetes and obesity. Cell Metab. 24, 51–62 (2016).

    Article  PubMed  Google Scholar 

  94. Frias, J. P. et al. Efficacy and safety of LY3298176, a novel dual GIP and GLP-1 receptor agonist, in patients with type 2 diabetes: a randomised, placebo-controlled and active comparator-controlled phase 2 trial. Lancet 392, 2180–2193 (2018).

    Article  CAS  PubMed  Google Scholar 

  95. Parker, V. E. R. et al. Efficacy, safety, and mechanistic insights of cotadutide, a dual receptor glucagon-like peptide-1 and glucagon agonist. J. Clin. Endocrinol. Metab. 105, dgz047 (2020).

    Article  PubMed  Google Scholar 

  96. Ämmälä, C. et al. Targeted delivery of antisense oligonucleotides to pancreatic β-cells. Sci. Adv. 4, eaat3386 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Reiner, T. et al. Accurate measurement of pancreatic islet beta-cell mass using a second-generation fluorescent exendin-4 analog. Proc. Natl. Acad. Sci. USA 108, 12815–12820 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Research in the laboratories of F.M.G. and F.R. is primarily supported by Wellcome (106262/Z/14/Z and 106263/Z/14/Z) and MRC-UK (MRC_MC_UU_12012/3) funding.

Author information

Authors and Affiliations

Authors

Contributions

F.M.G. and F.R. co-wrote the article.

Corresponding authors

Correspondence to Fiona M. Gribble or Frank Reimann.

Ethics declarations

Competing interests

F.M.G. is a consultant for Kallyope, and the laboratories of F.M.G. and F.R. receive additional research funding from AstraZeneca, Eli Lilly and LGC.

Additional information

Peer review information Primary Handling Editor: Christoph Schmitt. Nature Metabolism thanks Brian Finan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gribble, F.M., Reimann, F. Metabolic Messengers: glucagon-like peptide 1. Nat Metab 3, 142–148 (2021). https://doi.org/10.1038/s42255-020-00327-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-00327-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing