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.

  • Article
  • Published:

Crystal structure of the GLP-1 receptor bound to a peptide agonist

A Corrigendum to this article was published on 12 July 2017

Abstract

Glucagon-like peptide 1 (GLP-1) regulates glucose homeostasis through the control of insulin release from the pancreas. GLP-1 peptide agonists are efficacious drugs for the treatment of diabetes. To gain insight into the molecular mechanism of action of GLP-1 peptides, here we report the crystal structure of the full-length GLP-1 receptor bound to a truncated peptide agonist. The peptide agonist retains an α-helical conformation as it sits deep within the receptor-binding pocket. The arrangement of the transmembrane helices reveals hallmarks of an active conformation similar to that observed in class A receptors. Guided by this structural information, we design peptide agonists with potent in vivo activity in a mouse model of diabetes.

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

Figure 1: The overall structure of GLP-1R in complex with peptide 5.
Figure 2: Molecular details of the agonist peptide binding site in GLP-1R.
Figure 3: Comparison of GLP-1R with GCGR and CRF1R.
Figure 4: Mouse in vivo OGTT.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

References

  1. Todd, J. F. & Bloom, S. R. Incretins and other peptides in the treatment of diabetes. Diabet. Med. 24, 223–232 (2007)

    CAS  PubMed  Google Scholar 

  2. Chatterjee, S., Ghosal, S. & Chatterjee, S. Glucagon-like peptide-1 receptor agonists favorably address all components of metabolic syndrome. World J. Diabetes 7, 441–448 (2016)

    PubMed  PubMed Central  Google Scholar 

  3. Mentlein, R., Gallwitz, B. & Schmidt, W. E. Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7-36)amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur. J. Biochem. 214, 829–835 (1993)

    CAS  PubMed  Google Scholar 

  4. Deacon, C. F., Johnsen, A. H. & Holst, J. J. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J. Clin. Endocrinol. Metab. 80, 952–957 (1995)

    CAS  PubMed  Google Scholar 

  5. Willard, F. S., Bueno, A. B. & Sloop, K. W. Small molecule drug discovery at the glucagon-like peptide-1 receptor. Exp. Diabetes Res. 2012, 709893 (2012)

    PubMed  PubMed Central  Google Scholar 

  6. Mann, R. et al. Peptide binding at the GLP-1 receptor. Biochem. Soc. Trans. 35, 713–716 (2007)

    CAS  PubMed  Google Scholar 

  7. Underwood, C. R. et al. Crystal structure of glucagon-like peptide-1 in complex with the extracellular domain of the glucagon-like peptide-1 receptor. J. Biol. Chem. 285, 723–730 (2010)

    CAS  PubMed  Google Scholar 

  8. Runge, S., Thøgersen, H., Madsen, K., Lau, J. & Rudolph, R. Crystal structure of the ligand-bound glucagon-like peptide-1 receptor extracellular domain. J. Biol. Chem. 283, 11340–11347 (2008)

    CAS  PubMed  Google Scholar 

  9. Siu, F. Y. et al. Structure of the human glucagon class B G-protein-coupled receptor. Nature 499, 444–449 (2013)

    ADS  CAS  PubMed  Google Scholar 

  10. Jazayeri, A. et al. Extra-helical binding site of a glucagon receptor antagonist. Nature 533, 274–277 (2016)

    ADS  CAS  PubMed  Google Scholar 

  11. Haque, T. S. et al. Identification of potent 11mer glucagon-like peptide-1 receptor agonist peptides with novel C-terminal amino acids: homohomophenylalanine analogs. Peptides 31, 950–955 (2010)

    CAS  PubMed  Google Scholar 

  12. Haque, T. S. et al. Exploration of structure-activity relationships at the two C-terminal residues of potent 11mer glucagon-like peptide-1 receptor agonist peptides via parallel synthesis. Peptides 31, 1353–1360 (2010)

    CAS  PubMed  Google Scholar 

  13. Mapelli, C. et al. Eleven amino acid glucagon-like peptide-1 receptor agonists with antidiabetic activity. J. Med. Chem. 52, 7788–7799 (2009)

    CAS  PubMed  Google Scholar 

  14. Serrano-Vega, M. J., Magnani, F., Shibata, Y. & Tate, C. G. Conformational thermostabilization of the beta1-adrenergic receptor in a detergent-resistant form. Proc. Natl Acad. Sci. USA 105, 877–882 (2008)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lebon, G. et al. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature 474, 521–525 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  16. White, J. F. et al. Structure of the agonist-bound neurotensin receptor. Nature 490, 508–513 (2012)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yang, L. et al. Conformational states of the full-length glucagon receptor. Nat. Commun. 6, 7859 (2015)

    ADS  CAS  PubMed  Google Scholar 

  18. Hollenstein, K. et al. Structure of class B GPCR corticotropin-releasing factor receptor 1. Nature 499, 438–443 (2013)

    ADS  CAS  PubMed  Google Scholar 

  19. Wootten, D., Simms, J., Miller, L. J., Christopoulos, A. & Sexton, P. M. Polar transmembrane interactions drive formation of ligand-specific and signal pathway-biased family B G protein-coupled receptor conformations. Proc. Natl Acad. Sci. USA 110, 5211–5216 (2013)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tom Burnley, B ., Afonine, P. V ., Adams, P. D. & Gros, P. Modelling dynamics in protein crystal structures by ensemble refinement. eLife 2012, 1–29 (2012)

    Google Scholar 

  21. Yang, D. et al. Structural determinants of binding the seven-transmembrane domain of the glucagon-like peptide-1 receptor (GLP-1R). J. Biol. Chem. 291, 12991–13004 (2016)

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Dods, R. L. & Donnelly, D. The peptide agonist-binding site of the glucagon-like peptide-1 (GLP-1) receptor based on site-directed mutagenesis and knowledge-based modelling. Biosci. Rep. 36, e00285 (2015)

    PubMed  Google Scholar 

  23. Wootten, D. et al. The extracellular surface of the GLP-1 receptor is a molecular trigger for biased agonism. Cell 165, 1632–1643 (2016)

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Coopman, K. et al. Residues within the transmembrane domain of the glucagon-like peptide-1 receptor involved in ligand binding and receptor activation: modelling the ligand-bound receptor. Mol. Endocrinol. 25, 1804–1818 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Xiao, Q., Jeng, W. & Wheeler, M. B. Characterization of glucagon-like peptide-1 receptor-binding determinants. J. Mol. Endocrinol. 25, 321–335 (2000)

    CAS  PubMed  Google Scholar 

  26. Koole, C. et al. Second extracellular loop of human glucagon-like peptide-1 receptor (GLP-1R) has a critical role in GLP-1 peptide binding and receptor activation. J. Biol. Chem. 287, 3642–3658 (2012)

    CAS  PubMed  Google Scholar 

  27. Wood, V. et al. The genome sequence of Schizosaccharomyces pombe. Nature 415, 871–880 (2002)

    ADS  CAS  PubMed  Google Scholar 

  28. Moon, M. J. et al. Ligand binding pocket formed by evolutionarily conserved residues in the glucagon-like peptide-1 (GLP-1) receptor core domain. J. Biol. Chem. 290, 5696–5706 (2015)

    CAS  PubMed  PubMed Central  Google Scholar 

  29. López de Maturana, R., Treece-Birch, J., Abidi, F., Findlay, J. B. C. & Donnelly, D. Met-204 and Tyr-205 are together important for binding GLP-1 receptor agonists but not their N-terminally truncated analogues. Protein Pept. Lett. 11, 15–22 (2004)

    PubMed  Google Scholar 

  30. Koth, C. M. et al. Molecular basis for negative regulation of the glucagon receptor. Proc. Natl Acad. Sci. USA 109, 14393–14398 (2012)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mason, J. S. et al. High end GPCR design: crafted ligand design and druggability analysis using protein structure, lipophilic hotspots and explicit water networks. In Silico Pharmacol. 1, 23 (2013)

    PubMed Central  Google Scholar 

  32. Goodford, P. J. A computational procedure for determining energetically favorable binding sites on biologically important macromolecules. J. Med. Chem. 28, 849–857 (1985)

    CAS  PubMed  Google Scholar 

  33. Sciabola, S. et al. High-throughput virtual screening of proteins using GRID molecular interaction fields. J. Chem. Inf. Model. 50, 155–169 (2010)

    CAS  PubMed  Google Scholar 

  34. Rasmussen, S. G. F. et al. Crystal structure of the β2 adrenergic receptor-Gs protein complex. Nature 477, 549–555 (2011)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Deupi, X. & Standfuss, J. Structural insights into agonist-induced activation of G-protein-coupled receptors. Curr. Opin. Struct. Biol. 21, 541–551 (2011)

    CAS  PubMed  Google Scholar 

  36. Tehan, B. G., Bortolato, A., Blaney, F. E., Weir, M. P. & Mason, J. S. Unifying family A GPCR theories of activation. Pharmacol. Ther. 143, 51–60 (2014)

    CAS  PubMed  Google Scholar 

  37. Wheeler, M. B. et al. Functional expression of the rat glucagon-like peptide-I receptor, evidence for coupling to both adenylyl cyclase and phospholipase-C. Endocrinology 133, 57–62 (1993)

    CAS  PubMed  Google Scholar 

  38. Vilsbøll, T., Agersø, H., Krarup, T. & Holst, J. J. Similar elimination rates of glucagon-like peptide-1 in obese type 2 diabetic patients and healthy subjects. J. Clin. Endocrinol. Metab. 88, 220–224 (2003)

    PubMed  Google Scholar 

  39. Kabsch, W. Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Winn, M. D. et al. Overview of the CCP4 suite and current developments. Acta Crystallogr. D Biol. Crystallogr. 67, 235–242 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  41. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007)

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Murshudov, G. N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D Biol. Crystallogr. 67, 355–367 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Nicholls, R. A., Long, F. & Murshudov, G. N. Low-resolution refinement tools in REFMAC5. Acta Crystallogr. D Biol. Crystallogr. 68, 404–417 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Adams, P. D . et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010)

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21 (2010)

    CAS  PubMed  Google Scholar 

  47. Diederichs. K. & Karplus, P. A. Better models by discarding data? Acta Crystallogr D Biol Crystallogr. 69, 1215–1222 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank various colleagues past and present who have helped with the project. In particular we would like to acknowledge the contribution of K. Hollenstein, M. Koglin and C. Larner. We thank G. Brown for his help with coordinating peptide synthesis and radio-labelling and C. Scully for his assistance with the GRID analysis. We are grateful to R. Owen, J. Waterman and D. Axford at I24, Diamond Light Source, Oxford, UK for technical support.

Author information

Authors and Affiliations

Authors

Contributions

J.K., N.J.R. and A.J. carried out the conformational thermostabilization of constructs and determined the stability of the StaR in a panel of reagents/additives. A.H.B. and I.T. carried out the in vitro pharmacology. A.J.H.B. managed the in vivo studies. M.C. and S.P.A. designed the novel peptides, aided by A.B. and J.S.M. who designed the homology models and carried out in silico analyses of peptide binding. M.R. and J.C.E. designed the crystallization construct, and with C.F.V. performed and optimized protein expression and purification. M.R. and C.F.V. performed and optimized protein crystallization. M.R. and A.S.D. harvested crystals, collected and processed X-ray diffraction data, and solved and refined the structure. Project management was carried out by A.J., R.M.C., F.H.M. and M.W. The manuscript was prepared by M.R., A.J., A.S.D., A.J.H.B., M.C., R.M.C. and F.H.M. All authors contributed to the final editing and approval of the manuscript.

Corresponding author

Correspondence to Fiona H. Marshall.

Ethics declarations

Competing interests

All authors are employees of Heptares Therapeutics Ltd and are shareholders in Sosei Group Corporation.

Additional information

Reviewer Information Nature thanks T. Schwartz, C. Siebold and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 Peptide 5 and in vitro pharmacology of wild-type GLP-1R.

a, Two-dimensional chemical plot of peptide 5 used in this study. b, Pharmacological characterization of 3H-peptide 1 at the wild-type GLP-1R. Affinity of peptide 1 for wild-type GLP-1R construct was measured using homologous competition experiments against three different concentration of 3H-peptide 1. cpm, counts per minute. Data are representative of four independent experiments and the Kd values are calculated as the arithmetic mean and s.e.m. c, Heterologous competition binding of exendin-4 and exendin-3 at the wild-type GLP-1R determined using 3H-peptide 1. Affinity constants (Ki) were calculated from IC50 values, using the Cheng–Prusoff equation and results are given as the arithmetic mean ± s.e.m.

Extended Data Figure 2 Stability, pharmacological characterization and functional activity of GLP-1R StaR.

a, Thermal stability (Tm) comparison of GLP-1R wild-type and StaR (containing the following point mutations: T207E, Q211A, D215R, L232F, G295A, T298A, C329A, P358A, G361A, H363V and V405A). Thermal stability was measured following solubilization in n-dodecyl-β-d-maltopyranoside supplemented with cholesteryl hemisuccinate (see Methods). Data representative of two independent experiments with Tm values calculated as the arithmetic mean and the standard deviation of the mean. b, Pharmacological characterization of GLP-1R wild-type and StaR. Affinity of peptide 1 for wild-type and StaR constructs was measured using homologous competition experiments against three different concentrations of 3H-peptide 1. Data representative of three independent experiments and the Kd values calculated as the arithmetic mean and the standard deviation of the mean. The difference in the means is not statistically significant as analysed by two-tailed t-test. c, d, cAMP response of GLP-1R wild-type and StaR in the presence of the indicated peptide agonists. e, Reported pEC50 values for each peptide agonist. Data presented is the arithmetic mean of three independent experiments. Error bars represent s.e.m. P values are calculated by multi-parametric two-way ANOVA.

Extended Data Figure 3 The GLP-1R StaR B-factors and electron density.

a, b, B-factor putty representation of the GLP-1R–peptide-5 structure (rainbow spectrum, blue to red = lowest to highest B-factors). c, d, representative 2Fo − Fc electron density contoured at 1.0σ across the orthosteric peptide-binding pocket of GLP-1R.

Extended Data Figure 4 Walkthrough of peptide 5 interactions with GLP-1R StaR.

ak, Views moving from the N to C terminus of peptide 5. Four overlayed models generated by PHENIX ensemble refinement are shown in each panel to demonstrate the confidence that can be assigned to interactions with the receptor described in this study.

Extended Data Figure 5 Structural superposition of the GLP-1R peptide 5 crystal structure with the crystal structure of the isolated GLP-1R extracellular domain in complex with the GLP-1 peptide.

a, Tilted view from membrane of the GLP-1R represented as cartoon (cyan) with the peptide 5 agonist in stick representation and carbon, nitrogen and oxygen atoms coloured yellow, blue and red respectively; the ECD solved in isolation from the TMD of GLP-1R in cartoon representation is coloured orange, with the GLP-1 peptide coloured magenta. The superposition was achieved using equivalent residues from peptide 5 and the GLP-1 peptide. b, Rotation of the superposed structures in a to view from extracellular space, the relative difference in orientation of the ECD is denoted.

Extended Data Figure 6 Evaluation of the lipophilic hotspots on GLP-1R and interactions with peptide 5.

a, GRID hotspot analysis of the binding mode of peptide 5 showing the overlap of the Cap1, X2 and X3 groups of peptide 5 with lipophilic regions of the GLP-1R. GLP-1R is represented as cartoon (cyan) with the ECD coloured brown. The peptide 5 agonist is shown in stick representation with carbon, nitrogen and oxygen atoms coloured yellow, blue and red respectively. The c1 = GRID map is represented as mesh (orange) and contoured at −2.5 kcal mol−1. b, View as in a rotated by 180°.

Extended Data Figure 7 In vitro pharmacological and pharmacokinetic profiles of selected peptides.

ad, Insulinotropic activities of GLP-1 and selected peptides on isolated rat pancreatic islets. Results are presented as mean ± s.e.m. (n = 6 each group) and analysed using a one-way analysis of variance and Dunnett’s post hoc test. Significant differences from basal responses are indicated with an asterisk (*P < 0.05). e, Pharmacokinetics of peptide 2 (1 mg kg−1), peptide 5 (1 mg kg−1) and peptide 8 (0.5 mg kg−1) following intravenous administration in male Sprague Dawley rats. Results presented as mean ± s.e.m. (n = 3). f, Pharmacokinetics of peptide 8 following subcutaneous administration in male CD1 mice. Results presented as mean ± s.e.m. (n = 3).

Extended Data Table 1 Truncated GLP-1 peptide analogues and associated biological data
Extended Data Table 2 Data collection and refinement statistics for GLP-1R StaR complexed with peptide 5
Extended Data Table 3 Pharmacokinetic evaluation of peptide 2, 5 and 8 detailed in this study

Supplementary information

Supplementary information

This file contains Supplementary Text and Data, 2 Supplementary Tables and Supplementary references. (PDF 413 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jazayeri, A., Rappas, M., Brown, A. et al. Crystal structure of the GLP-1 receptor bound to a peptide agonist. Nature 546, 254–258 (2017). https://doi.org/10.1038/nature22800

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature22800

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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