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 basis for the cooperative allosteric activation of the free fatty acid receptor GPR40

Abstract

Clinical studies indicate that partial agonists of the G-protein-coupled, free fatty acid receptor 1 GPR40 enhance glucose-dependent insulin secretion and represent a potential mechanism for the treatment of type 2 diabetes mellitus. Full allosteric agonists (AgoPAMs) of GPR40 bind to a site distinct from partial agonists and can provide additional efficacy. We report the 3.2-Å crystal structure of human GPR40 (hGPR40) in complex with both the partial agonist MK-8666 and an AgoPAM, which exposes a novel lipid-facing AgoPAM-binding pocket outside the transmembrane helical bundle. Comparison with an additional 2.2-Å structure of the hGPR40–MK-8666 binary complex reveals an induced-fit conformational coupling between the partial agonist and AgoPAM binding sites, involving rearrangements of the transmembrane helices 4 and 5 (TM4 and TM5) and transition of the intracellular loop 2 (ICL2) into a short helix. These conformational changes likely prime GPR40 to a more active-like state and explain the binding cooperativity between these ligands.

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: AgoPAM AP8 interacts with positive cooperativity with the partial agonist MK-8666 in functional assays.
Figure 2: Structure of hGPR40 in complex with the partial agonist MK-8666 and the AgoPAM AP8.
Figure 3: Binding interactions of AgoPAM AP8.
Figure 4: AgoPAM-binding site rationalizes potency disconnect in dog and human species.
Figure 5: Structural comparisons of the ternary GPR40–MK-8666–AP8 structure versus the binary GPR40–MK-8666 structure.
Figure 6: Structural alignment of GPR40 complexes with active-state GPCRs.
Figure 7: A putative activation mechanism of partial agonists and AgoPAMs.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Choi, Y.J., Shin, D. & Lee, J.Y. G-protein coupled receptor 40 agonists as novel therapeutics for type 2 diabetes. Arch. Pharm. Res. 37, 435–439 (2014).

    Article  CAS  Google Scholar 

  2. Kotarsky, K., Nilsson, N.E., Flodgren, E., Owman, C. & Olde, B. A human cell surface receptor activated by free fatty acids and thiazolidinedione drugs. Biochem. Biophys. Res. Commun. 301, 406–410 (2003).

    Article  CAS  Google Scholar 

  3. Flodgren, E. et al. GPR40 is expressed in glucagon producing cells and affects glucagon secretion. Biochem. Biophys. Res. Commun. 354, 240–245 (2007).

    Article  CAS  Google Scholar 

  4. Shapiro, H., Shachar, S., Sekler, I., Hershfinkel, M. & Walker, M.D. Role of GPR40 in fatty acid action on the beta cell line INS-1E. Biochem. Biophys. Res. Commun. 335, 97–104 (2005).

    Article  CAS  Google Scholar 

  5. Lin, D.C. et al. Identification and pharmacological characterization of multiple allosteric binding sites on the free fatty acid 1 receptor. Mol. Pharmacol. 82, 843–859 (2012).

    Article  CAS  Google Scholar 

  6. Srivastava, A. et al. High-resolution structure of the human GPR40 receptor bound to allosteric agonist TAK-875. Nature 513, 124–127 (2014).

    Article  CAS  Google Scholar 

  7. Hirozane, Y., Motoyaji, T., Maru, T., Okada, K. & Tarui, N. Generating thermostabilized agonist-bound GPR40/FFAR1 using virus-like particles and a label-free binding assay. Mol. Membr. Biol. 31, 168–175 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Zhang, D. et al. Two disparate ligand-binding sites in the human P2Y1 receptor. Nature 520, 317–321 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Hauge, M. et al. GPR40 (FFAR1)—combined Gs and Gq signaling in vitro is associated with robust incretin secretagogue action ex vivo and in vivo. Mol. Metab. 4, 3–14 (2014).

    Article  Google Scholar 

  12. Lee, A.G. Lipid-protein interactions in biological membranes: a structural perspective. Biochim. Biophys. Acta 1612, 1–40 (2003).

    Article  CAS  Google Scholar 

  13. Vinothkumar, K.R. Structure of rhomboid protease in a lipid environment. J. Mol. Biol. 407, 232–247 (2011).

    Article  CAS  Google Scholar 

  14. Sum, C.S. et al. Identification of residues important for agonist recognition and activation in GPR40. J. Biol. Chem. 282, 29248–29255 (2007).

    Article  CAS  Google Scholar 

  15. Tikhonova, I.G. & Poerio, E. Free fatty acid receptors: structural models and elucidation of ligand binding interactions. BMC Struct. Biol. 15, 16 (2015).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  17. Kang, Y. et al. Crystal structure of rhodopsin bound to arrestin by femtosecond X-ray laser. Nature 523, 561–567 (2015).

    Article  CAS  Google Scholar 

  18. Carpenter, B., Nehmé, R., Warne, T., Leslie, A.G. & Tate, C.G. Structure of the adenosine A(2A) receptor bound to an engineered G protein. Nature 536, 104–107 (2016).

    Article  CAS  Google Scholar 

  19. Huang, W. et al. Structural insights into μ-opioid receptor activation. Nature 524, 315–321 (2015).

    Article  CAS  Google Scholar 

  20. Katritch, V., Cherezov, V. & Stevens, R.C. Structure-function of the G protein-coupled receptor superfamily. Annu. Rev. Pharmacol. Toxicol. 53, 531–556 (2013).

    Article  CAS  Google Scholar 

  21. Wacker, D. et al. Structural features for functional selectivity at serotonin receptors. Science 340, 615–619 (2013).

    Article  CAS  Google Scholar 

  22. Wang, C. et al. Structural basis for molecular recognition at serotonin receptors. Science 340, 610–614 (2013).

    Article  CAS  Google Scholar 

  23. Venkatakrishnan, A.J. et al. Molecular signatures of G-protein-coupled receptors. Nature 494, 185–194 (2013).

    Article  CAS  Google Scholar 

  24. Moro, O., Lameh, J., Högger, P. & Sadée, W. Hydrophobic amino acid in the i2 loop plays a key role in receptor-G protein coupling. J. Biol. Chem. 268, 22273–22276 (1993).

    CAS  PubMed  Google Scholar 

  25. Burstein, E.S., Spalding, T.A. & Brann, M.R. The second intracellular loop of the m5 muscarinic receptor is the switch which enables G-protein coupling. J. Biol. Chem. 273, 24322–24327 (1998).

    Article  CAS  Google Scholar 

  26. Warne, T. et al. Structure of a beta1-adrenergic G-protein-coupled receptor. Nature 454, 486–491 (2008).

    Article  CAS  Google Scholar 

  27. Jaakola, V.P. et al. The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science 322, 1211–1217 (2008).

    Article  CAS  Google Scholar 

  28. Perez-Aguilar, J.M., Shan, J., LeVine, M.V., Khelashvili, G. & Weinstein, H. A functional selectivity mechanism at the serotonin-2A GPCR involves ligand-dependent conformations of intracellular loop 2. J. Am. Chem. Soc. 136, 16044–16054 (2014).

    Article  CAS  Google Scholar 

  29. Kobilka, B. The structural basis of G-protein-coupled receptor signaling (Nobel Lecture). Angew. Chem. Int. Edn Engl. 52, 6380–6388 (2013).

    Article  CAS  Google Scholar 

  30. Cooke, R. & Congreve, M. Allosteric binding: structures reveal new ways to tame G protein-coupled receptors. Future Med. Chem. 8, 2007–2007 (2016).

    Article  Google Scholar 

  31. Molecular Operating Environment (MOE) 2013.08 (Chemical Computing Group ULC, 2017).

  32. Kenakin, T. Allosteric agonist modulators. J. Recept. Signal Transduct. Res. 27, 247–259 (2007).

    Article  CAS  Google Scholar 

  33. Ehlert, F.J. Estimation of the affinities of allosteric ligands using radioligand binding and pharmacological null methods. Mol. Pharmacol. 33, 187–194 (1988).

    CAS  PubMed  Google Scholar 

  34. Caffrey, M. & Cherezov, V. Crystallizing membrane proteins using lipidic mesophases. Nat. Protoc. 4, 706–731 (2009).

    Article  CAS  Google Scholar 

  35. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  37. Vonrhein, C. et al. Data processing and analysis with the autoPROC toolbox. Acta Crystallogr. D Biol. Crystallogr. 67, 293–302 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  42. BUSTER v. 2.11.6 (Global Phasing Ltd., 2016).

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

    Article  CAS  Google Scholar 

  44. The PyMOL Molecular Graphics System v. 1.8 (Schrödinger, LLC).

  45. Jones, G., Willett, P., Glen, R.C., Leach, A.R. & Taylor, R. Development and validation of a genetic algorithm for flexible docking. J. Mol. Biol. 267, 727–748 (1997).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This manuscript is dedicated to the late Frank K. Brown for his groundbreaking contributions to drug discovery and his vision and tenacity in making this research possible. We also thank K. Hollenstein and H. Krishnamurthy for helpful discussions and technical insights. This research used resources at the Industrial Macromolecular Crystallography Association Collaborative Access Team (IMCA-CAT) beamline 17-ID, supported by the companies of the Industrial Macromolecular Crystallography Association through a contract with Hauptman-Woodward Medical Research Institute. We thank K. Battaile and A. M. Mulichak for data collection assistance and L. J. Keefe for direction of IMCA-CAT operations. This research was performed at the Advanced Photon Source, a US Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under Contract No. DE-AC02-06CH11357.

Author information

Authors and Affiliations

Authors

Contributions

S.M.S., S. Sharma, A.B.W., J.M.J., and S.J.A. conceived and managed the project with support from F.K.B., M.W., A.D.H., K.J.L. and J.D.H. N.B., S.J.A., D.L.H., J.H., T.H., M.K., H.W., A.B.W., P.S. and S.T. designed, optimized, purified and characterized receptor constructs for structural studies. J.L. and S.B.P. crystallized the receptor in LCP. J.L. supervised data collection and J.L., C.V. and G.B. processed synchrotron data. J.L. solved and refined the structures. J.L., J.M.J., J.W., A.B.W., J.D.S., S.M.S. and S. Sharma interpreted the structure and designed experiments. J.W., J.H. and D.L.H. prepared cells for binding studies. J.W., J.D.S. and A.B.W. designed the binding experiments. J.W. carried out radioligand-binding assays with cells. B.T.F., A.B.W., N.B.H. and S. Souza conceived and performed cell-based functional assays. J.L., N.B., J.W., A.B.W., J.M.J., S. Sharma, and S.M.S. analyzed the data and compiled the figures for the manuscript. J.M.J. and B.S.S. designed and performed docking and molecular modeling. H.R.C., S.L.C., Y.G., H.J., M.W.M., B.P., and C.W.P. selected and synthesized compounds for pharmacological profiling, receptor purification, and crystallization studies. J.L., N.B. and J.W. wrote the manuscript with contributions from J.M.J., A.B.W., S. Sharma and S.M.S.

Corresponding authors

Correspondence to Jun Lu or Stephen M Soisson.

Ethics declarations

Competing interests

All authors, with the exception of G.B. and C.V., are current or past employees of Merck Research Laboratories.

Integrated supplementary information

Supplementary Figure 1 Activation of GPR40 by select synthetic ligands.

(a) Chemical structures of the synthetic agonists used in this study. Partial agonists [TAK875, MK-8666 & P4] and AgoPAMs [AP4 & AP8] chemical structures. (b) Dose-response curves for the five agonists shown in (a) were generated monitoring IP accumulation in HEK293 cells expressing human GPR40. Data are expressed as a percentage of the control response of an in-house partial agonist, and fitted to a standard 4-parameter non-linear regression model. EC50s were determined for each test compound using a custom in-house developed software package. Each agonist EC50 and Emax was calculated at least six times (mean parameters ± SD in Supplementary Table 1) with representative curves (n=1) illustrated here.

Supplementary Figure 2 Saturation binding of [3H]P4 or [3H]AP8 to hGPR40 variants.

Panels show [3H]P4 or [3H]AP8 total (black circle), non-specific (open black circle) and specific (red circle) binding to the WT receptor (a & b) or the triple mutant (c & d) with a T4L insertion. Each study was repeated twice (N=2), with symbols in the representative graphs shown denoting the mean values of a single experiment ± SD (n=2). The mean parameters of these and other individual experiments are shown in Supplementary Table 3.

Supplementary Figure 3 GPR40 purification, LCP crystallization drop image and the electron density map for the AgoPAM AP8

(a) Schematic of GPR40 purification. Purification begins from isolated membranes from insect cells. (b) Final SEC trace shows that only select fractions of >95% purity were selected for the final pool. This pool corresponds to the monomeric form of GPR40. Purity is observed in the SDS-Page gel shown below the chromatogram. (c) Crystals obtained in lipidic cubic phase viewed under (Left) regular light and (Right) circularly polarized light. (d) The stereo image of the omit maps for AP8 in the ternary complex (3.5σ).

Supplementary Figure 4 Snake-plot diagram of the GPR40 construct and summary of AgoPAM AP8 interactions.

(a) Snake-plot diagram of the GPR40 construct(s) in crystallization. Thermostabilizing mutations are shown in red, Cysteines forming disulfide bond are shown in yellow, the affinity tags are shown in light blue, most conserved residues for each TM are colored in dark gray, residues interacting with MK-8666 are colored in green and residues interacting with AP8 are in gold. Note Tyr913.37 is colored half green and half gold since it is the only amino acid making direct interactions with both MK-8666 and AP8. (b) AgoPAM AP8 interactions with GPR40 using Molecular Operating Environment (MOE)46. Schematic representation of the interactions between AP8 and the AgoPAM binding site following the MOE standard structure preparation and 3D (rule-based) protonation protocols.

Supplementary Figure 5 Cell based [3H]AP8 and [3H]P4 saturation binding to hGPR40 WT receptor and binding-pocket mutants.

[3H]AP8 (a-b) or [3H]P4 (c-d) binding to WT (black circle), Y44F (red circle), Y114F (green circle), S123A (blue circle), G95F (yellow circle), A99L (purple circle) and Al02W (cyan circle). Each study was repeated twice (N=2), with symbols in the representative graphs shown denoting the mean values of a single experiment ± SD (n=2). KD values are displayed in Table 2. Data is transformed with a log plot of x-axis to illustrate effects on KD (b & d).

Supplementary Figure 6 Identification & application of [3H]AP4 for interrogation of the AgoPAM binding site in dog GPR40.

(a-f) [3H]AP4 binds with high affinity to dog GPR40. Saturation binding of [3H]P4 (a & d, 1.5 & 2.4 nM respectively), [3H]AP8 (b & e, 1.6 & >125 nM respectively) or [3H]AP4 (c & f, 1.3 & 12.8 nM respectively) to human (a-c) or dog (d-f) GPR40. Total (black circle), non-specific (open black circle) and specific (red circle) binding are denoted as described. Each experiment was repeated twice with representative graphs (n=2) shown. Notably, while [3H]AP8 is a suitable tool to profile AgoPAM binding to human GPR40, weaker observed affinity for dog GPR40 dictated replacement with the higher affinity tool, [3H]AP4. (g-j) AgoPAM affinity can be modulated in human and dog GPR40 by mutagenesis of position 102. Human GPR40 [3H]AP8 binding affinity is reduced by replacing Ala102 with larger side chains (g) while [3H]P4 KD (h) is not significantly changed [human GPR40 WT (black circle), A102V (red circle), A102I (green circle) and A102W (blue circle)]. Dog GPR40 [3H]AP4 binding affinity is modulated bi-directionally by varying the bulk of the side chain at Val102 (i) while [3H]P4 KD (j) is not significantly changed [dog GPR40 WT (black circle), V102A (red circle), V102I (green circle) and V102W (blue circle)]. Each study was repeated twice (N=2), with symbols in the representative graphs shown denoting the mean values of a single experiment ± SD (n=2). KD values are displayed in Supplementary Table 6. Data is transformed with a log plot of x-axis to illustrate effects on KD.

Supplementary Figure 7 Examination of binding and cooperativity in the hGPR40 quadruple mutant with a T4L insertion.

(a-b) Saturation binding of [3H]P4 or [3H]AP8 to Quad hGPR40. [3H]P4 (a) or [3H]AP8 (b) total (black circle), non-specific (open black circle) and specific (red circle) binding to the quadruple mutant with a T4L insertion. (c-d) MK-8666 binds with high affinity to displace [3H]P4 from hGPR40 quadruple mutant-T4L (red circle), while augmenting the binding of [3H]AP8 to the same receptor (red circle). In a reciprocal manner, AP8 displaces [3H]AP8 from the hGPR40 quadruple mutant-T4L (black circle), while augmenting the binding of [3H]P4 to the same receptors (black circle). Each study was repeated twice (N=2), with symbols in the representative graphs shown denoting the mean values of a single experiment ± SD (n=2).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–8. (PDF 2191 kb)

Conformational changes between GPR40 binary complex and ternary complex upon the binding of the AgoPAM AP8.

A video showing the conformation changes in hGPR40 from the binary complex with MK-8666 to the ternary complex with both MK-8666 and AP8, made using a morphing technique. (MPG 8328 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, J., Byrne, N., Wang, J. et al. Structural basis for the cooperative allosteric activation of the free fatty acid receptor GPR40. Nat Struct Mol Biol 24, 570–577 (2017). https://doi.org/10.1038/nsmb.3417

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3417

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