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:

Structural insights into probe-dependent positive allosterism of the GLP-1 receptor

Abstract

Drugs that promote the association of protein complexes are an emerging therapeutic strategy. We report discovery of a G protein-coupled receptor (GPCR) ligand that stabilizes an active state conformation by cooperatively binding both the receptor and orthosteric ligand, thereby acting as a ‘molecular glue’. LSN3160440 is a positive allosteric modulator of the GLP-1R optimized to increase the affinity and efficacy of GLP-1(9-36), a proteolytic product of GLP-1(7-36). The compound enhances insulin secretion in a glucose-, ligand- and GLP-1R-dependent manner. Cryo-electron microscopy determined the structure of the GLP-1R bound to LSN3160440 in complex with GLP-1 and heterotrimeric Gs. The modulator binds high in the helical bundle at an interface between TM1 and TM2, allowing access to the peptide ligand. Pharmacological characterization showed strong probe dependence of LSN3160440 for GLP-1(9-36) versus oxyntomodulin that is driven by a single residue. Our findings expand protein–protein modulation drug discovery to uncompetitive, active state stabilizers for peptide hormone receptors.

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: The pharmacological profile of LSN3160440.
Fig. 2: The overall structure of the GLP-1R–GLP-1–LSN3160440–Gs–Nb35 ternary complex.
Fig. 3: Sequence determinants for LSN3160440 enhancement of GLP-1(9-36) binding.
Fig. 4: Structural determinants of GLP-1R probe dependency for GLP-1 and OXM.

Similar content being viewed by others

Data availability

All relevant data are available from the authors and/or included in the manuscript or Supplementary Information. Atomic coordinates and the cryo-EM density map have been deposited in the Protein Data Bank under accession number 6VCB and Electron Microscopy Data Bank entry ID EMD-21147.

References

  1. Marso, S. P. et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N. Engl. J. Med. 375, 311–322 (2016).

    Article  CAS  Google Scholar 

  2. Marso, S. P. et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 375, 1834–1844 (2016).

    Article  CAS  Google Scholar 

  3. Hernandez, A. F. et al. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 392, 1519–1529 (2018).

    Article  CAS  Google Scholar 

  4. Gerstein, H. C. et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 394, 121–130 (2019).

    Article  CAS  Google Scholar 

  5. Wootten, D. et al. Allosteric modulation of endogenous metabolites as an avenue for drug discovery. Mol. Pharmacol. 82, 281–290 (2012).

    Article  CAS  Google Scholar 

  6. 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).

    Article  CAS  Google Scholar 

  7. Knudsen, L. B. & Pridal, L. Glucagon-like peptide-1-(9-36) amide is a major metabolite of glucagon-like peptide-1-(7-36) amide after in vivo administration to dogs, and it acts as an antagonist on the pancreatic receptor. Eur. J. Pharmacol. 318, 429–435 (1996).

    Article  CAS  Google Scholar 

  8. Chen, X. et al. Substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrazines as inhibitors of c-Met and RON tyrosine kinase receptor and their preparation, pharmaceutical compositions and use in the treatment of cancer. World patent WO2010059771 (2010).

  9. May, L. T., Leach, K., Sexton, P. M. & Christopoulos, A. Allosteric modulation of G protein-coupled receptors. Annu. Rev. Pharmacol. Toxicol. 47, 1–51 (2007).

    Article  CAS  Google Scholar 

  10. Sloop, K. W. et al. Novel small molecule glucagon-like peptide-1 receptor agonist stimulates insulin secretion in rodents and from human islets. Diabetes 59, 3099–3107 (2010).

    Article  CAS  Google Scholar 

  11. Bueno, A. B. et al. Positive allosteric modulation of the glucagon-like peptide-1 receptor by diverse electrophiles. J. Biol. Chem. 291, 10700–10715 (2016).

    Article  CAS  Google Scholar 

  12. Gallwitz, B. et al. Structure/activity characterization of glucagon-like peptide-1. Eur. J. Biochem. 225, 1151–1156 (1994).

    Article  CAS  Google Scholar 

  13. Kruse, A. C. et al. Activation and allosteric modulation of a muscarinic acetylcholine receptor. Nature 504, 101–106 (2013).

    Article  CAS  Google Scholar 

  14. Che, Y., Gilbert, A. M., Shanmugasundaram, V. & Noe, M. C. Inducing protein–protein interactions with molecular glues. Bioorg. Med. Chem. Lett. 28, 2585–2592 (2018).

    Article  CAS  Google Scholar 

  15. Schreiber, S. L. Immunophilin-sensitive protein phosphatase action in cell signaling pathways. Cell 70, 365–368 (1992).

    Article  CAS  Google Scholar 

  16. Liu, J. et al. Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506 complexes. Cell 66, 807–815 (1991).

    Article  CAS  Google Scholar 

  17. Zhang, Y. et al. Cryo-EM structure of the activated GLP-1 receptor in complex with a G protein. Nature 546, 248–253 (2017).

    Article  CAS  Google Scholar 

  18. Venkatesan, K. et al. An empirical framework for binary interactome mapping. Nat. Methods 6, 83–90 (2009).

    Article  CAS  Google Scholar 

  19. Arkin, M. R., Tang, Y. & Wells, J. A. Small-molecule inhibitors of protein–protein interactions: progressing toward the reality. Chem. Biol. 21, 1102–1114 (2014).

    Article  CAS  Google Scholar 

  20. Jochim, A. L. & Arora, P. S. Assessment of helical interfaces in protein–protein interactions. Mol. Biosyst. 5, 924–926 (2009).

    Article  CAS  Google Scholar 

  21. Wells, J. A. & McClendon, C. L. Reaching for high-hanging fruit in drug discovery at protein–protein interfaces. Nature 450, 1001–1009 (2007).

    Article  CAS  Google Scholar 

  22. Beck, B. et al. in Assay Guidance Manual (eds Sittampalam, G. S. et al.) (Eli Lilly & Co., 2004).

  23. Willard, F. S. et al. Small molecule allosteric modulation of the glucagon-like Peptide-1 receptor enhances the insulinotropic effect of oxyntomodulin. Mol. Pharmacol. 82, 1066–1073 (2012).

    Article  CAS  Google Scholar 

  24. Cheng, Y. & Prusoff, W. H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 22, 3099–3108 (1973).

    Article  CAS  Google Scholar 

  25. Sattler, M. et al. A novel small molecule met inhibitor induces apoptosis in cells transformed by the oncogenic TPR-MET tyrosine kinase. Cancer Res. 63, 5462–5469 (2003).

    CAS  PubMed  Google Scholar 

  26. Christopoulos, A. & Kenakin, T. G protein-coupled receptor allosterism and complexing. Pharm. Rev. 54, 323–374 (2002).

    Article  CAS  Google Scholar 

  27. Scheres, S. H. Processing of structurally heterogeneous cryo-EM data in RELION. Methods Enzymol. 579, 125–157 (2016).

    Article  CAS  Google Scholar 

  28. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  Google Scholar 

  29. Zhang, K. Gctf: Real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  CAS  Google Scholar 

  30. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, https://doi.org/10.7554/eLife.42166 (2018).

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

    Article  CAS  Google Scholar 

  32. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D. 60, 2126–2132 (2004).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  34. Williams, C. J. et al. MolProbity: more and better reference data for improved all-atom structure validation. Protein Sci. 27, 293–315 (2018).

    Article  CAS  Google Scholar 

  35. Lomize, M. A., Pogozheva, I. D., Joo, H., Mosberg, H. I. & Lomize, A. L. OPM database and PPM web server: resources for positioning of proteins in membranes. Nucleic Acids Res. 40, D370–D376 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to B. Anderson, A. Castano, B. Czeskis, C. Corkins, T. Gopalappa, D. Jett, C.R. Logan, Y. Qian, M. Russell, H. Wang, J. Wyss and R. Zink for advice and technical support. We also appreciate long-standing support for the project by C. Montrose-Rafizadeh, G. Zhu, J. Moyers and R. Gimeno. The cryo-EM data were collected at NanoImaging Service.

Author information

Authors and Affiliations

Authors

Contributions

A.B.B., F.J.A., G.R.C., A.J. and I.R. designed and synthesized the small molecule ligands related to and including LSN3160440. F.S.W., J.D.H., D.B.W., A.D.S., C.S. and K.W.S. designed and/or performed the pharmacological studies. J.D.H., D.B.W., A.D.S., B.C. and C.S. performed the mutagenesis studies. J.F. performed the insulin secretion studies. B.S. and D.F. prepared cryo-EM grids. B.S. performed cryo-EM data processing, model building and refinement. D.F. and B.S. expressed and purified the ligand–receptor complex. M.V. and Q.C. designed, performed and analyzed the molecular dynamics simulations. A.B.B., F.S.W., B.S., J.D.H., D.B.W., A.D.S., M.V., Q.C., C.S., J.F., D.F., B.K.K. and K.W.S. performed data analysis. A.B.B., F.S.W., J.D.H., M.V., B.S., B.K.K. and K.W.S. wrote the manuscript. A.B.B., T.S.K., B.K.K. and K.W.S. supervised the project.

Corresponding authors

Correspondence to Brian K. Kobilka or Kyle W. Sloop.

Ethics declarations

Competing interests

A.B.B., F.S.W., J.D.H., D.B.W., A.D.S., M.V., Q.C., C.S., B.C., J.F., F.J.A., G.R.C., A.J., I.R. and K.W.S. are employees of Eli Lilly and Company and may own company stock. B.S., D.F., T.S.K. and B.K.K. are employees of or consultants for ConfometRx. T.S.K. and B.K.K. cofounded ConfometRx.

Additional information

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

Extended data

Extended Data Fig. 1 LSN3160440 increases the affinity of GLP-1(9-36) for the GLP-1R.

a, The potency of unlabeled GLP-1(9-36) to displace [125I]GLP-1(7-36) binding to the GLP-1R was measured at different concentrations of LSN3160440. b, Data were replotted to generate a modified Schild plot and LSN3160440 KB (5.7 µM) and α (407) parameters were calculated using the allosteric Schild equation. Data are a representative example of eight independent experiments for which summary statistics are mean (SD): pKB 5.14 (0.2) and log α 2.64 (0.4).

Extended Data Fig. 2 LSN3160440 enhances insulin secretion in a glucose-, ligand-, and GLP-1R-dependent manner.

a-b, Insulin secretion in islets isolated from wild-type (WT, grey bars) and Glp-1r knockout (KO, white bars) mice at high or low glucose. Insulin levels were quantified in media from cultures of WT and Glp-1r KO mouse (C57/Bl6 background) islets treated with GLP-1(7-36), GIP(1-42), GLP-1(9-36), LSN3160440, or GLP-1(9-36) plus LSN3160440 in the presence of 11.2 mM glucose (a) or 2.8 mM glucose (b). For each treatment, mean (SEM) insulin concentrations were determined in six individual wells containing three islets per well. * p = 0.0002 and ** p < 0.0001 using one-way ANOVA followed by Dunnett’s comparison versus the vehicle response. Data are representative of three (a) or two (b) independent experiments. c, Time course of plasma insulin concentrations in fasted, anesthetized Wistar rats treated with either vehicle, GLP-1(7-36) (3 nmol/kg), LSN3160440 (5 mg/kg), GLP-1(9-36) (50 nmol/kg), or LSN3160440 (5 mg/kg) plus 5, 20, or 50 nmol/kg of GLP-1(9-36), dosed immediately before intravenous administration of a glucose bolus (0.5 g/kg). Data are presented as the mean (SEM) from five animals per treatment group and are representative of three independent experiments.

Extended Data Fig. 3 Cryo-EM data processing and validation.

a, Cryo-EM data processing flow chart. b, Gold standard Fourier shell correlation (FSC) curves of two individual half maps indicating an average resolution of 3.3 Å at 0.143 FSC threshold. c, Cross-validation of model to cryo-EM density map. The model was refined against one half map, and FSC curves were calculated between this model and the final cryo-EM map (full dataset, blue) of the outcome of model refinement with a half map versus the same map (orange), and of the outcome of model refinement with a half map versus the other half map (green). d, Density map colored by local resolution. e, Local resolution near the binding site for LSN3160440. LSN3160440 density is indicated by white dashed circle.

Extended Data Fig. 4 Model of the GLP-1R/GLP-1/LSN3160440/Gs/Nb35 complex in the cryo-EM density map.

Cryo-EM density map and the model are shown for all seven transmembrane (TM) helices and helix 8 of GLP-1R, GLP-1(7-37), and LSN3160440 (zoomed in). All the residues in GLP-1(7-37) are resolved.

Extended Data Fig. 5 Allosteric pharmacology of LSN3160440 binding site mutants in the GLP-1R.

Potentiation of the cAMP accumulation produced by GLP-1(9-36) in combination with various fixed concentrations of positive allosteric modulator using mutant GLP-1R transiently transfected into HEK293 cells. LSN3160440: a, L142A, b, Y145A, c, K202A, and d, L142A, Y145A, K202A. GLP-1R with BETP: g, L142A, h, Y145A, i, K202A, and j, L142A, Y145A, K202A. Each graph is a single experiment. Replicate data was generated in a concentration response format with altered compound testing concentrations but gave analogous data. N values for replication: a = 2, b = 2, c = 2, d = 3, g = 2, h = 2, i = 2, j = 3. The magnitude of allosteric modulator affinity and efficacy cooperativity could not be quantified using the operational model of allosterism due to the efficacy of GLP-1(9-36) alone, being below the noise level in these experiments. Therefore, we used an empirical approach to quantify allosterism. Concentration response curves were individually fit to the 4-parameter logistic model to obtain potency and efficacy for GLP-1(9-36). The maximal efficacy obtained at any single concentration of modulator is plotted for e, LSN3160440 and k, BETP. EC50 values at different concentrations of modulators were calculated and are plotted versus modulator concentrations for f, LSN3160440, and l, BETP.

Extended Data Fig. 6 Fluorescent competition binding assay on WT and mutant GLP-1R.

a, GLP-1R (WT), b, GLP-1R triple mutant (L142A, K202A, Y145A), c, GLP-1R (K202A), d, GLP-1R (Y145A), e, GLP-1R (Y145F). Competitive binding of GLP-1(9-36) in the presence and absence of 10 µM positive allosteric modulator (PAM) LSN3160440 was measured using 4 nM labeled exendin-4. Data are presented as the mean (SEM) of three independent experiments.

Extended Data Fig. 7 MD simulation experiments.

Interaction of ligand with protein based on initial 50 ns simulation. Interaction of Lys202 with N3 of benzimidazole is bridged with water 45% of simulation time. Stacking interaction with Tyr145 is present 98% of simulation time. Benzimidazole oscillates 1 Å in RMSD from its original position from the cryo-EM structure. a, 2D ligand protein interaction diagram over the initial 50 ns simulation. b, 3D representation of the MMGBSA energetically Representative MD frame at 47 ns of simulation. Waters bridging interactions with ligand are shown as ball-and-stick model. Piperidine is also interacting with waters which at times bridge interaction with POPC (shown as maroon sticks) phosphates.

Supplementary information

Supplementary Information

Supplementary Tables 1–6 and Note 1.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bueno, A.B., Sun, B., Willard, F.S. et al. Structural insights into probe-dependent positive allosterism of the GLP-1 receptor. Nat Chem Biol 16, 1105–1110 (2020). https://doi.org/10.1038/s41589-020-0589-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0589-7

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research