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.

  • Letter
  • Published:

High-resolution structure of the human GPR40 receptor bound to allosteric agonist TAK-875

Abstract

Human GPR40 receptor (hGPR40), also known as free fatty-acid receptor 1 (FFAR1), is a G-protein-coupled receptor that binds long-chain free fatty acids to enhance glucose-dependent insulin secretion1. Novel treatments for type-2 diabetes mellitus2 are therefore possible by targeting hGPR40 with partial or full agonists. TAK-875, or fasiglifam, is an orally available, potent and selective partial agonist3 of hGPR40 receptor, which reached phase III clinical trials for the potential treatment of type-2 diabetes mellitus4. Data from clinical studies indicate that TAK-875, which is an ago-allosteric modulator of hGPR40 (ref. 3), demonstrates improved glycaemic control and low hypoglycaemic risk in diabetic patients5. Here we report the crystal structure of hGPR40 receptor bound to TAK-875 at 2.3 Å resolution. The co-complex structure reveals a unique binding mode of TAK-875 and suggests that entry to the non-canonical binding pocket most probably occurs via the lipid bilayer. The atomic details of the extensive charge network in the ligand binding pocket reveal additional interactions not identified in previous studies and contribute to a clear understanding of TAK-875 binding to the receptor. The hGPR40–TAK-875 structure also provides insights into the plausible binding of multiple ligands to the receptor, which has been observed in radioligand binding6 and Ca2+ influx assay studies3. Comparison of the transmembrane helix architecture with other G-protein-coupled receptors suggests that the crystallized TAK-875-bound hGPR40 complex is in an inactive-like state.

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: Structure of hGPR40 in complex with the partial agonist TAK-875.
Figure 2: Binding interactions of TAK-875.
Figure 3: Surface representation of hGPR40 showing multiple putative binding sites.
Figure 4: Structure alignment of active and inactive GPCR structures.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Data deposits

Coordinates and structure factors have been deposited in Protein Data Bank under accession number 4PHU.

References

  1. Itoh, Y. et al. Free fatty acids regulate insulin secretion from pancreatic β cells through GPR40. Nature 422, 173–176 (2003)

    Article  CAS  ADS  PubMed  Google Scholar 

  2. McGarry, J. D. & Dobbins, R. L. Fatty acids, lipotoxicity and insulin secretion. Diabetologia 42, 128–138 (1999)

    Article  CAS  PubMed  Google Scholar 

  3. Yabuki, C. et al. A novel antidiabetic drug, fasiglifam/TAK-875, acts as an ago-allosteric modulator of FFAR1. PLoS ONE 8, :e76280. (2013)

  4. Negoro, N. et al. Discovery of TAK-875: a potent, selective, and orally bioavailable GPR40 agonist. ACS Med. Chem. Lett. 1, 290–294 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burant, C. F. et al. TAK-875 versus placebo or glimepiride in type 2 diabetes mellitus: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet 379, 1403–1411 (2012)

    Article  CAS  PubMed  Google Scholar 

  6. Lin, D. C.-H. 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  PubMed  PubMed Central  Google Scholar 

  7. 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  PubMed  Google Scholar 

  8. Kotarsky, K. et al. A human cell surface receptor activated by free fatty acids and thiazolidinedione drugs. Biochem. Biophys. Res. Commun. 301, 406–410 (2003)

    Article  CAS  PubMed  Google Scholar 

  9. Shapiro, H. et al. Role of GPR40 in fatty acid action on the β cell line INS-1E. Biochem. Biophys. Res. Commun. 335, 97–104 (2005)

    Article  CAS  PubMed  Google Scholar 

  10. Hirozane, Y. et al. Generating thermostabilized agonist bound GPR40/FFAR1 using virus-like particles and label-free binding assay. Mol. Mem. Biol (in the press)

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

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hanson, M. A. et al. Crystal structure of a lipid G protein-coupled receptor. Science 335, 851–855 (2012)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  14. Granier, S. et al. Structure of the δ-opioid receptor bound to naltrindole. Nature 485, 400–404 (2012)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  15. Zhang, C. et al. High-resolution crystal structure of human protease-activated receptor 1. Nature 492, 387–392 (2012)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  16. Wu, B. et al. Structures of the CXCR4 Chemokine GPCR with small-molecule and cyclic peptide antagonists. Science 330, 1066–1071 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  17. Wu, H. et al. Structure of the human κ-opioid receptor in complex with JDTic. Nature 485, 327–332 (2012)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  18. Hildebrand, P. W. et al. A ligand channel through the G protein coupled receptor opsin. PLoS ONE 4, :e4382. (2009)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  19. Mercier, R. W. et al. hCB2 ligand-interaction landscape: cysteine residues critical to biarylpyrazole antagonist binding motif and receptor modulation. Chem. Biol. 17, 1132–1142 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hurst, D. P. et al. A lipid pathway for ligand binding is necessary for a cannabinoid G protein-coupled receptor. J. Biol. Chem. 285, 17954–17964 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Negoro, N. et al. Optimization of (2,3-dihydro-1-benzofuran-3-yl)acetic acids: discovery of a non-free fatty acid-like, highly bioavailable G protein-coupled receptor 40/free fatty acid receptor 1 agonist as a glucose-dependent insulinotropic agent. J. Med. Chem. 55, 3960–3974 (2012)

    Article  CAS  PubMed  Google Scholar 

  22. 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  PubMed  Google Scholar 

  23. Sum, C. S. et al. Two arginine-glutamate ionic locks near the extracellular surface of FFAR1 gate receptor activation. J. Biol. Chem. 284, 3529–3536 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  25. De Lean, A. et al. A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled β-adrenergic receptor. J. Biol. Chem. 255, 7108–7117 (1980)

    CAS  PubMed  Google Scholar 

  26. Katritch, V. et al. Structure-function of the G protein-coupled receptor superfamily. Annu. Rev. Pharmacol. Toxicol. 53, 531–556 (2013)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  28. Rasmussen, S. G. F. et al. Structure of a nanobody-stabilized active state of the β2 adrenoceptor. Nature 469, 175–180 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  29. Standfuss, J. et al. Crystal structure of a thermally stable rhodopsin mutant. J. Mol. Biol. 372, 1179–1188 (2007)

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Choe, H. W. et al. Transmembrane signaling by GPCRs: insight from rhodopsin and opsin structures. Neuropharmacology 60, 52–57 (2011)

    Article  CAS  PubMed  Google Scholar 

  31. Rosenbaum, D. M. et al. GPCR engineering yields high-resolution structural insights into β2-adrenergic receptor function. Science 318, 1266–1273 (2007)

    Article  CAS  ADS  PubMed  Google Scholar 

  32. Caffrey, M. & Cherezov, V. Crystallizing membrane proteins using lipidic mesophases. Nature Protocols 4, 706–731 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cherezov, V. et al. Rastering strategy for screening and centring of microcrystal samples of human membrane proteins with a sub-10 µm size X-ray synchrotron beam. J. R. Soc. Interface 6, S587–S597 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fischetti, R. F. et al. Mini-beam collimator enables microcrystallography experiments on standard beamlines. J. Synchrotron Radiat. 16, 217–225 (2009)

    Article  CAS  PubMed  PubMed Central  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  PubMed  Google Scholar 

  36. Evans, P. R. Scaling and assessment of data quality. Acta Crystallogr. D 62, 72–82 (2006)

    Article  PubMed  Google Scholar 

  37. McNicholas, S. et al. Presenting your structures: the CCP4mg molecular-graphics software. Acta Crystallogr. D 67, 386–394 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Vagin, A. & Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr. D 66, 22–25 (2010)

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  40. Vagin, A. A. et al. REFMAC5 dictionary: organization of prior chemical knowledge and guidelines for its use. Acta Crystallogr. D 60, 2184–2195 (2004)

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. P. Wilson, M. Hixon and K. Goodwill for review and feedback on the manuscript; T. Ho and S. Okubo for molecular cloning support; T. Sjoberg for membrane preparation; and C. Dillard for cell culture support. We also thank the staff of the Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, which operates Advanced Light Source beamline 5.0.3, and the staff of GM/CA, Argonne National Laboratory, which operates Advanced Photon Source beamline 23ID-D. The Berkeley Center for Structural Biology is supported in part by the National Institutes of Health and National Institute of General Medical Sciences. The GM/CA has been funded in whole or in part with Federal funds from the National Cancer Institute (Y1-CO-1020) and the National Institute of General Medical Sciences (Y1-GM-1104). The Advanced Light Source and Advanced Photon Source are supported by the Director, Office of Science, Office of Basic Energy Sciences of the US Department of Energy under contract numbers DE-AC02-05CH11231 and DE-AC02-06CH11357, respectively.

Author information

Authors and Affiliations

Authors

Contributions

A.S. guided construct and expression optimization, developed purification procedures and purified receptor protein, characterized crystallization constructs, performed crystallization and structure analysis, and was responsible for project strategy. J.Y. performed crystallization and crystal harvesting, structure determination and structure analysis. Y.H. performed mutant screening and characterized mutants and crystallization constructs. G.S. and W.L. performed data collection, data processing and structure analysis. G.K. performed molecular biology, construct optimization and expression screening. F.G. performed construct characterization, protein purification and crystallization. K.A. guided construct optimization and performed molecular biology. A.J. and A.I. performed homology modelling to aid molecular refinement. A.J. discussed ligand-binding site in the structure. J.N. performed protein purification and crystallization; K.O. supported experimental design and data discussion for mutant screening. F.G., A.S., J.Y., G.K., G.S. and W.L. prepared the figures. All the authors contributed to manuscript writing.

Corresponding author

Correspondence to Ankita Srivastava.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Crystallization construct and crystallization drop image.

a, Snake-plot diagram of the hGPR40 expression construct. The affinity tags and the linker at the insertion of T4L are shown in blue. Thermostabilizing mutations are shown in red. b, Cartoon representation of the hGPR40–TAK-875 structure with the mutations highlighted in red. c, Crystals obtained in lipidic cubic phase could be observed using circularly polarized light.

Extended Data Figure 2 LC–MS-based TAK-875 binding to hGPR40 crystallization constructs.

Panels above show TAK-875-specific binding to the WT receptor, the quadruple mutant with T4L insertion and the T4L-inserted quadruple mutant with GS linker. The last construct corresponds to the crystal structure. The table summarizes the binding affinity of WT and modified constructs, which remains in the range 12–16 nM. , Specific binding; , total binding; □, Sf9 control membranes; n = 3; s.e.m., standard error of the mean.

Extended Data Figure 3 Comparison of ligand-binding-pocket access.

hGPR40 (white), δ-opioid (orange) and S1P1 (purple) are compared for access to the ligand binding pocket. The extracellular view of the accessible surface (a, d, g) reveals that unlike δ-opioid, which has the canonical solvent-accessible binding pocket, S1P1 and hGPR40 ligand access is obscured by extracellular loops. From a lateral view (b, e, h) in the plane of the membrane, TAK-875 entry to hGPR40 is distinct from the other receptors. S1P1 ligand accessibility is also seen from a lateral view (c, f, i) in the plane of the membrane; however, the position is 120° rotated in the plane of the membrane. The accessible views observed for hGPR40 (b), δ-opioid (d) and S1P1 (i) are illustrated as ribbons with ligands as sticks (k, j, l respectively).

Extended Data Figure 4 Comparison of ECL2 among peptide-binding receptors and the related hGPR40.

a, ECL2 between transmembrane 4 and transmembrane 5 of hGPR40 (grey) shares the short hairpin loop region and disulphide tether to transmembrane 3 seen previously only in the peptide-binding receptor family members NTSR1 (green), PAR1 (cyan), δ-opioid (purple) and CXCR4 (orange). Auxiliary loop of hGPR40 is shown in pink colour in ECL2 region. b, Structure-based alignment of transmembrane 4, transmembrane 5 and the hairpin loop reveal very low sequence identity despite the conserved structural features. A conserved Trp in transmembrane 4, a conserved Pro in transmembrane 5, and the Cys involved in the disulphide bond to transmembrane 3 are the only conserved residues. hGPR40 residues coloured in pink correspond to the auxiliary loop of ECL2 shown in a.

Extended Data Figure 5 The ECL2 loop.

Top (a) and lateral (b, c) views of the ECL2 loop to highlight polar interactions (left) and B-factors (right) of ECL2 residues. The interactions are shown as black dotted lines (left) and TAK-875 is green. ECL2 is shown in cyan with residue side chains in the left, and as a ribbon cartoon on the right, with the ribbon’s thickness and colour scaled according to the B-factors (ranging from 34 Å2 shown in blue to 88 Å2 shown in red within the ECL2 loop). The numerous polar interactions within the ECL2 loop and with water molecules and residues outside of the ECL2 loop, in addition to the disulphide-bond to transmembrane 3, stabilize this region to a large extent. This is reflected in the B-factors, which show relatively low mobility for most of the ECL2 loop. (Inside, intracellular side; outside, extracellular side.)

Extended Data Figure 6 TAK-875 interactions with hGPR40 based on the computational chemistry program MOE.

The figure above illustrates the colour code and bonding interaction based on the crystal structure. The carboxylic acid is buried within a very hydrophobic region where it makes ionic/polar interactions with Arg 1835.39, Arg 2587.35, Tyr 913.37 and Tyr 2406.51 while the other end of the molecule is exposed outside the receptor.

Extended Data Figure 7 Evaluation of LC–MS-based TAK-875 binding to WT and binding-pocket residue mutants of WT hGPR40.

Panels above show TAK-875-specific binding to the WT receptor and the WT receptor containing single point mutations of binding-pocket key residues. Table summarizes the measured binding affinity of WT and single point mutants. , Specific binding; , total binding; □, VLP control membranes; n = 3; s.e.m., standard error of the mean.

Extended Data Figure 8 Evaluation of LC–MS-based TAK-875 binding to WT and thermostabilized receptor binding-pocket residue mutants of hGPR40.

Panels above show TAK-875 specific binding to the WT receptor and thermostabilized receptor containing single point mutations of binding-pocket key residues. Table summarizes the measured binding affinity of WT and single point mutants. , Specific binding; , total binding; □, VLP control membranes; n = 2; s.e.m., standard error of the mean.

Extended Data Table 1 Binding affinity summary of WT hGPR40, crystallization constructs and TAK-875 binding-pocket mutations
Extended Data Table 2 X-ray data collection and refinement statistics

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Srivastava, A., Yano, J., Hirozane, Y. et al. High-resolution structure of the human GPR40 receptor bound to allosteric agonist TAK-875. Nature 513, 124–127 (2014). https://doi.org/10.1038/nature13494

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13494

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