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 of ligand recognition and self-activation of orphan GPR52

Abstract

GPR52 is a class-A orphan G-protein-coupled receptor that is highly expressed in the brain and represents a promising therapeutic target for the treatment of Huntington’s disease and several psychiatric disorders1,2. Pathological malfunction of GPR52 signalling occurs primarily through the heterotrimeric Gs protein2, but it is unclear how GPR52 and Gs couple for signal transduction and whether a native ligand or other activating input is required. Here we present the high-resolution structures of human GPR52 in three states: a ligand-free state, a Gs-coupled self-activation state and a potential allosteric ligand-bound state. Together, our structures reveal that extracellular loop 2 occupies the orthosteric binding pocket and operates as a built-in agonist, conferring an intrinsically high level of basal activity to GPR523. A fully active state is achieved when Gs is coupled to GPR52 in the absence of an external agonist. The receptor also features a side pocket for ligand binding. These insights into the structure and function of GPR52 could improve our understanding of other self-activated GPCRs, enable the identification of endogenous and tool ligands, and guide drug discovery efforts that target GPR52.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Crystal structure of GPR52-apo and analysis of ECL2.
Fig. 2: Cryo-EM structure of GPR52 bound to heterotrimeric mini-Gs in the absence of an agonist.
Fig. 3: Structural comparison of apo and mini-Gs-coupled GPR52.
Fig. 4: Crystal structure of GPR52–c17 and a novel ligand-binding pocket.
Fig. 5: Unique ligand-binding mode and structural conformation of GPR52 compared to other class-A GPCRs.

Similar content being viewed by others

Data availability

The coordinates and structure factors for GPR52-Fla-apo, GPR52-Rub-apo, GPR52-Fla–c17 and GPR52–mini-Gs–Nb35 have been deposited in the PDB with accession codes 6LI1, 6LI2, 6LI0 and 6LI3, respectively. The cryo-EM 3D maps of the GPR52–mini-Gs–Nb35 complex have been deposited in the Electron Microscopy Data Bank (EMDB) with accession code EMD-0902. All other data relating to this study are available from the corresponding authors on reasonable request.

References

  1. Yao, Y. et al. A striatal-enriched intronic GPCR modulates huntingtin levels and toxicity. eLife 4, e05449 (2015).

    PubMed Central  Google Scholar 

  2. Komatsu, H. et al. Anatomical transcriptome of G protein-coupled receptors leads to the identification of a novel therapeutic candidate GPR52 for psychiatric disorders. PLoS ONE 9, e90134 (2014).

    ADS  PubMed  PubMed Central  Google Scholar 

  3. Martin, A. L., Steurer, M. A. & Aronstam, R. S. Constitutive activity among orphan class-A G protein coupled receptors. PLoS ONE 10, e0138463 (2015).

    PubMed  PubMed Central  Google Scholar 

  4. Foster, S. R. et al. Discovery of human signaling systems: pairing peptides to G protein-coupled receptors. Cell 179, 895–908 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Krutzke, S. K. et al. Array-based molecular karyotyping in fetal brain malformations: identification of novel candidate genes and chromosomal regions. Birth Defects Res. A Clin. Mol. Teratol. 106, 16–26 (2016).

    CAS  PubMed  Google Scholar 

  6. Setoh, M. et al. Discovery of the first potent and orally available agonist of the orphan G-protein-coupled receptor 52. J. Med. Chem. 57, 5226–5237 (2014).

    CAS  PubMed  Google Scholar 

  7. Nishiyama, K. et al. FTBMT, a novel and selective GPR52 agonist, demonstrates antipsychotic-like and procognitive effects in rodents, revealing a potential therapeutic agent for schizophrenia. J. Pharmacol. Exp. Ther. 363, 253–264 (2017).

    CAS  PubMed  Google Scholar 

  8. Komatsu, H. Novel therapeutic GPCRs for psychiatric disorders. Int. J. Mol. Sci. 16, 14109–14121 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Alavi, M. S., Shamsizadeh, A., Azhdari-Zarmehri, H. & Roohbakhsh, A. Orphan G protein-coupled receptors: the role in CNS disorders. Biomed Pharmacother. 98, 222–232 (2018).

    CAS  PubMed  Google Scholar 

  10. Song, H. et al. Targeting Gpr52 lowers mutant HTT levels and rescues Huntington’s disease-associated phenotypes. Brain 141, 1782–1798 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. García-Nafría, J., Lee, Y., Bai, X., Carpenter, B. & Tate, C. G. Cryo-EM structure of the adenosine A2A receptor coupled to an engineered heterotrimeric G protein. eLife 7, e35946 (2018).

    PubMed  PubMed Central  Google Scholar 

  13. Liang, Y. L. et al. Phase-plate cryo-EM structure of a class B GPCR–G-protein complex. Nature 546, 118–123 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liang, Y. L. et al. Cryo-EM structure of the active, Gs-protein complexed, human CGRP receptor. Nature 561, 492–497 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhao, L. H. et al. Structure and dynamics of the active human parathyroid hormone receptor-1. Science 364, 148–153 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rauser, L., Savage, J. E., Meltzer, H. Y. & Roth, B. L. Inverse agonist actions of typical and atypical antipsychotic drugs at the human 5-hydroxytryptamine2C receptor. J. Pharmacol. Exp. Ther. 299, 83–89 (2001).

    CAS  PubMed  Google Scholar 

  18. Katritch, V. et al. Allosteric sodium in class A GPCR signaling. Trends Biochem. Sci. 39, 233–244 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  Google Scholar 

  20. Ma, Y. et al. Structural basis for apelin control of the human apelin receptor. Structure 25, 858–866 (2017).

    CAS  PubMed  Google Scholar 

  21. Wacker, D. et al. Crystal structure of an LSD-bound human serotonin receptor. Cell 168, 377–389 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ballesteros, J. A. & Weinstein, H. in Receptor Molecular Biology, Methods in Neurosciences vol. 25 (ed. Sealfon, S. C.) 366–428 (Elsevier, 1995).

  23. Wang, S. & Wacker, D. D4 dopamine receptor high-resolution structures enable the discovery of selective agonists. Science 358, 381–386 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Carpenter, B. & Tate, C. G. Engineering a minimal G protein to facilitate crystallisation of G protein-coupled receptors in their active conformation. Protein Eng. Des. Sel. 29, 583–594 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Nakahata, T. et al. Design and synthesis of 1-(1-benzothiophen-7-yl)-1H-pyrazole, a novel series of G protein-coupled receptor 52 (GPR52) agonists. Bioorg. Med. Chem. 26, 1598–1608 (2018).

    CAS  PubMed  Google Scholar 

  26. García-Nafría, J., Nehmé, R., Edwards, P. C. & Tate, C. G. Cryo-EM structure of the serotonin 5-HT1B receptor coupled to heterotrimeric Go. Nature 558, 620–623 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  27. Kang, Y. et al. Cryo-EM structure of human rhodopsin bound to an inhibitory G protein. Nature 558, 553–558 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Koehl, A. et al. Structure of the µ-opioid receptor–Gi protein complex. Nature 558, 547–552 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Maeda, S., Qu, Q., Robertson, M. J., Skiniotis, G. & Kobilka, B. K. Structures of the M1 and M2 muscarinic acetylcholine receptor/G-protein complexes. Science 364, 552–557 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Qi, X. et al. Cryo-EM structure of oxysterol-bound human Smoothened coupled to a heterotrimeric Gi. Nature 571, 279–283 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Draper-Joyce, C. J. et al. Structure of the adenosine-bound human adenosine A1 receptor–Gi complex. Nature 558, 559–563 (2018).

    ADS  CAS  PubMed  Google Scholar 

  32. Kato, H. E. et al. Conformational transitions of a neurotensin receptor 1–Gi1 complex. Nature 572, 80–85 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Ballesteros, J. A. et al. Activation of the β2-adrenergic receptor involves disruption of an ionic lock between the cytoplasmic ends of transmembrane segments 3 and 6. J. Biol. Chem. 276, 29171–29177 (2001).

    CAS  PubMed  Google Scholar 

  34. Greasley, P. J., Fanelli, F., Rossier, O., Abuin, L. & Cotecchia, S. Mutagenesis and modelling of the α1b-adrenergic receptor highlight the role of the helix 3/helix 6 interface in receptor activation. Mol. Pharmacol. 61, 1025–1032 (2002).

    CAS  PubMed  Google Scholar 

  35. Shapiro, D. A., Kristiansen, K., Weiner, D. M., Kroeze, W. K. & Roth, B. L. Evidence for a model of agonist-induced activation of 5-hydroxytryptamine 2A serotonin receptors that involves the disruption of a strong ionic interaction between helices 3 and 6. J. Biol. Chem. 277, 11441–11449 (2002).

    CAS  PubMed  Google Scholar 

  36. Romo, T. D., Grossfield, A. & Pitman, M. C. Concerted interconversion between ionic lock substates of the β2 adrenergic receptor revealed by microsecond timescale molecular dynamics. Biophys. J. 98, 76–84 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang, S. et al. Structure of the D2 dopamine receptor bound to the atypical antipsychotic drug risperidone. Nature 555, 269–273 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Munk, C. et al. An online resource for GPCR structure determination and analysis. Nat. Methods 16, 151–162 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Audet, M. & Stevens, R. C. Emerging structural biology of lipid G protein-coupled receptors. Protein Sci. 28, 292–304 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Kakarala, K. K. & Jamil, K. Sequence-structure based phylogeny of GPCR class A rhodopsin receptors. Mol. Phylogenet. Evol. 74, 66–96 (2014).

    CAS  PubMed  Google Scholar 

  41. Chun, E. et al. Fusion partner toolchest for the stabilization and crystallization of G protein-coupled receptors. Structure 20, 967–976 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Roth, C. B., Hanson, M. A. & Stevens, R. C. Stabilization of the human β2-adrenergic receptor TM4–TM3–TM5 helix interface by mutagenesis of Glu1223.41, a critical residue in GPCR structure. J. Mol. Biol. 376, 1305–1319 (2008).

    CAS  PubMed  Google Scholar 

  43. Lv, X. et al. In vitro expression and analysis of the 826 human G protein-coupled receptors. Protein Cell 7, 325–337 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Koehler Leman, J., Mueller, B. K. & Gray, J. J. Expanding the toolkit for membrane protein modeling in Rosetta. Bioinformatics 33, 754–756 (2017).

    PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    PubMed  PubMed Central  Google Scholar 

  55. Cardone, G., Heymann, J. B. & Steven, A. C. One number does not fit all: mapping local variations in resolution in cryo-EM reconstructions. J. Struct. Biol. 184, 226–236 (2013).

    PubMed  Google Scholar 

  56. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  58. Zhang, S. et al. Molecular matchmaking between the popular weight-loss herb Hoodia gordonii and GPR119, a potential drug target for metabolic disorder. Proc. Natl Acad. Sci. USA 111, 14571–14576 (2014).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  59. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Yuan, Y., Pei, J. & Lai, L. LigBuilder 2: a practical de novo drug design approach. J. Chem. Inf. Model. 51, 1083–1091 (2011).

    CAS  PubMed  Google Scholar 

  61. Robert, X. & Gouet, P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res. 42, W320–W324 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. The PyMOL Molecular Graphics system v.1.8.x (Schrödinger, 2015).

Download references

Acknowledgements

This work was supported by the National Key Research and Development Program of China, grant 2018YFA0507000 (to F.X. and S.Z.), National Natural Science Foundation of China (31525007 to M.L., 81861128023 to F.X. and 31971178 to S.Z.), Shanghai Municipal Education Commission−Gaofeng Clinical Medicine Grant Support (20181711 to J.W.) and Shanghai Outstanding Academic Leader funding (19XD1422800 to F.X.). The diffraction data were collected at BL41XU at SPring-8 with JASRI proposal 2019A2704. We thank J. Liu, N. Chen and L. Xue of the BV facility at the iHuman Institute, ShanghaiTech University for support with protein expression; the staff of the Electron Microscopy System and Mass Spectrometry System at Shanghai Institute of Precision Medicine for technical support and assistance in data collection; M. Cao for help with the collection and analysis of cryo-EM data; Q. Sun and the Bio-EM facility at ShanghaiTech University for technical support; V. Katritch and P. Popov for suggestions on construct mutations; M. Hanson for help with processing of X-ray data; and R. C. Stevens for encouraging this work.

Author information

Authors and Affiliations

Authors

Contributions

X.L. performed cloning, protein purification, crystallization, X-ray data collection, mutagenesis, cAMP functional assays and structural analysis; M. Li, N.W. and X.L. purified the GPR52 and mini-Gs–Nb35 proteins; M. Li prepared cryo-EM samples, collected data and determined structures; Y.W. performed structural analysis, molecular docking and structural similarity network analysis; J.W. performed model building and refinement; Z.L. processed the diffraction data with HKL2000 and solved the three crystal structures; S.G. designed ECL2 peptides and assisted with the data processing from cell-based functional assays; G.-W.H. was responsible for structure quality control; S.L. assisted with cryo-EM data collection and analysis; Y.Y. assisted with X-ray data collection; X.W. assisted with cell-based functional assays; X.X. supervised functional assays; Y.C. supervised structural analysis; S.Z. supervised structural analysis, molecular docking and structural similarity network analysis; and F.X. conceived the project and designed and supervised all experiments. All authors contributed to data interpretation and preparation of the manuscript. X.L., F.X. and M. Lei wrote the manuscript and F.X., M. Lei and J.W. orchestrated the project.

Corresponding authors

Correspondence to Jian Wu, Ming Lei or Fei Xu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Bryan Roth 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 Fig. 1 Engineering and crystallization of GPR52.

a, Schematic of the GPR52 constructs that were used for crystallization (residues 17—340). Thermostabilizing mutations (red) are A130W, A264L, W278Q, C314P, S318A, N321D and V323T. Cysteine residues (yellow), TEV cleavage site (pink) and disulfide bonds (orange dashed lines) are shown. b, Left, analytical size-exclusion chromatography of GPR52. Experiments were repeated three times with similar results. Right, superposition of GPR52-Fla-apo, GPR52-Rub-apo and GPR52–c17 structures shows the overall conserved helical arrangement. c, Crystal images of the GPR52–c17 complex (left), GPR52-Rub-apo (middle) and GPR52-Fla-apo (right). Experiments were repeated three times with similar results. d, ECL2 comparison of two apo structures (GPR52-Fla-apo, orange; GPR52-Rub-apo, cyan). The crystal packing of GPR52-Fla-apo (middle) and GPR52-Rub-apo (right) is also shown. Helix 8 of GPR52-Rub-apo is highlighted in red.

Extended Data Fig. 2 Effects of GPR52 mutations on the potency of c17.

a, Basal activity of GPR52 mutants. Response-level values were compared with wild-type GPR52 by two-way ANOVA without repeated measures, followed by Dunnett’s post hoc test (****P < 0.0001). Data are mean ± s.e.m. (n = 3) b, Mapping of mutated residues (green) onto GPR52 crystal structures. c17 is shown in orange. c, Summary of functional potency (pEC50) values of c17 on the GPR52 mutants. Data are mean ± s.e.m. (n = 3). d, Relative surface expression levels of mutant constructs were monitored by a FACS staining assay (Methods) and normalized to the expression levels of wild-type GPR52. Data are mean ± s.e.m. (n = 3). NA, not available.

Extended Data Fig. 3 Comparison of GPR52 with other class-A GPCRs.

a, Comparison of the ECL2 ALM-occupied orthosteric binding pocket of GPR52 with agonist-bound pockets of A1R (PDB 6D9H), A2AR (PDB 6GDG) and 5-HT1BR (PDB 6G79). b, PIF motif comparison of mini-Gs-coupled GPR52 (green) and A2AR (yellow; PDB 6GDG). c, Side view (left), cytoplasmic view (middle) and extracellular view (right) of A2AR in the mini-Gs-coupled state (green, PDB 6GDG) compared with the adenosine analogue (NECA; PDB 2YDV)-bound state (pink). d, Side view (left), cytoplasmic view (middle) and extracellular view (right) of 5-HT1BR in the mini-Go-coupled state (in complex with donitriptan) (green; PDB 6G79) compared with the ergotamine-bound state (pink; PDB 4IAR). e, DRY motif of GPR52-apo, 5-HT1BR–ergotamine (PDB 4IAR) and A2AR–NECA (PDB 2YDV).

Extended Data Fig. 4 Cryo-EM analysis of the GPR52–mini-Gs–Nb35 complex.

a, b, Size-exclusion chromatography profile (a) and corresponding SDS–PAGE gel (b) of the purified GPR52–mini-Gs–Nb35 complex. Experiments were repeated three times with similar results. c, Representative reference-free 2D cryo-EM average of the GPR52–mini-Gs–Nb35 complex. d, GPR52 with point mutations A130W3.41 and C314P7.50 maintained around 50% of the activity relative to the wild-type protein, according to the cAMP response level. Data are mean ± s.e.m. (n = 3). e, Representative cryo-EM micrograph of the GPR52–mini-Gs–Nb35 complex. f, Reference-free 2D averages of the GPR52–mini-Gs–Nb35 complex. g, Final 3D density map coloured according to the local resolution. h, Gold-standard FSC curves, showing the overall nominal resolution at 3.3 Å. i, Angular distribution of the particles used for the final reconstruction of the GPR52–mini-Gs–Nb35 complex.

Extended Data Fig. 5 Flow chart for the cryo-EM data processing and structure determination of the GPR52–mini-Gs–Nb35 complex.

See Methods for details. The final reconstruction has an average resolution of 3.3 Å. All the images in this figure were created in UCSF Chimera.

Extended Data Fig. 6 Cryo-EM map quality and ECL2 comparison.

a, Atomic model of GPR52 transmembrane helices, ECL2 and ICL2 in the cryo-EM density map. The molecular model is shown in stick representation and the cryo-EM map as mesh. b, Stereo views of the electron density maps of ECL2. Left, the 2Fo − Fc map of ECL2 from the GPR52-Rub-apo crystal structure. Right, the electron density map of ECL2 from the GPR52–mini-Gs complex structure.

Extended Data Fig. 7 Comparison of the GPR52–mini-Gs interface with that of other GPCR–G-protein complexes.

a, b, Front view (a) and back view (b) of the GPR52–mini-Gs interface. GPR52 (centre) and mini-Gs (right) are in surface representation and coloured according to the electrostatic potential (blue, positive; red, negative). c, The GPR52–mini-Gs interface. GPR52 and mini-Gs are in cartoon representation and coloured in green and grey, respectively. A magnified view of the interface is shown on the right in surface representation. dg, Magnified views of the interface between other receptors and G proteins in surface representation. h, Buried surface area of the interfaces between receptors and G proteins, calculated by PyMOL.

Extended Data Fig. 8 Docking position of GPR52 agonists, structural similarity network and sequence alignment of GPR52.

a, Docking position of GPR52 agonists with different scaffolds: WO-459 (left); 7m (centre) and FTBMT (right). The PR52 residues that are involved in ligand binding are shown as green sticks and c17 is shown as grey sticks, for reference. b, Structural similarity network of class-A GPCRs with reported inactive structures. c, Sequence alignment of GPR52 and GPR21 (yellow, less than 5.0 Å to ligand; green indicates key residues for structural features).

Extended Data Table 1 Data collection and structure refinement statistics
Extended Data Table 2 Cryo-EM data collection and refinement statistics

Supplementary information

Supplementary Information

This file contains Supplementary Figure 1 and Supplementary Tables 1 to 4.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, X., Li, M., Wang, N. et al. Structural basis of ligand recognition and self-activation of orphan GPR52. Nature 579, 152–157 (2020). https://doi.org/10.1038/s41586-020-2019-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2019-0

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: 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