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:

Constitutive activation mechanism of a class C GPCR

Abstract

Class C G-protein-coupled receptors (GPCRs) are activated through binding of agonists to the large extracellular domain (ECD) followed by rearrangement of the transmembrane domains (TMDs). GPR156, a class C orphan GPCR, is unique because it lacks an ECD and exhibits constitutive activity. Impaired GPR156–Gi signaling contributes to loss of hearing. Here we present the cryo-electron microscopy structures of human GPR156 in the Go-free and Go-coupled states. We found that an endogenous phospholipid molecule is located within each TMD of the GPR156 dimer. Asymmetric binding of Gα to the phospholipid-bound GPR156 dimer restructures the first and second intracellular loops and the carboxy-terminal part of the elongated transmembrane 7 (TM7) without altering dimer conformation. Our findings reveal that GPR156 is a transducer for phospholipid signaling. Constant binding of abundant phospholipid molecules and the G-protein-induced reshaping of the cytoplasmic face provide a basis for the constitutive activation of GPR156.

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: Overall structure of GPR156.
Fig. 2: Phospholipid-bound structure of GPR156.
Fig. 3: Phospholipid-binding and dimeric structure of GPR156.
Fig. 4: Comparison of the phospholipid-free GPR156 with the Go-free, phospholipid-bound GPR156.
Fig. 5: Rearrangement of the cytoplasmic face of GPR156 induced by Go.
Fig. 6: Interface between Go and GPR156.
Fig. 7: Comparison of the activation mechanism of GPR156 with other class C GPCRs.

Similar content being viewed by others

Data availability

Atomic coordinates and the cryo-EM map have been deposited in the the EM Data Bank (EMD) and Protein Data Bank (PDB), respectively, under the following accession numbers: EMD-35380 and PDB 8IED (GPR156-Go–scFv16), EMD-35377 and PDB 8IEB (GPR156 dimer of GPR156-Go–scFv16), EMD-35378 and PDB 8IEC (Go–scFv16 of GPR156-Go–scFv16), EMD-35390 and PDB 8IEQ (GPR156A/B/C/D), EMD-35382 and PDB 8IEI (GPR156A/B of GPR156A/B/C/D) and EMD-35389 and PDB 8IEP (GPR156C/D of GPR156A/B/C/D). Mass spectroscopy data are deposited on Figshare (https://doi.org/10.6084/m9.figshare.24212226 and https://doi.org/10.6084/m9.figshare.24715704.v1). The trajectories for GPR156-PC and GPR156-PG from the molecular dynamics simulations data are deposited on Zenodo (https://doi.org/10.5281/zenodo.8418994 and https://doi.org/10.5281/zenodo.8419006, respectively). Previously published PDBs used in this study are available under PDB accession codes 7UM5, 7MTR, 7MTS, 6UO8, 7EB2, 7C7S, 7E9H, 7EPA, 7EWL, 7M3J, 7M3F, 6WIV and 7E9G. The AlphaFold2 model is available in ModelArchive (https://www.modelarchive.org) with accession code ma-1015e. Sequence data used in the alignment for Extended Data Fig. 7 are H. sapiens GPR156, GABR1, GABR2, CaSR, mGlu1, mGlu2, mGlu3, mGlu4, mGlu5 and mGlu7 (Uniprot accession codes Q8NFN8, Q9UBS5, O75899, P41180, Q13255, Q14416, Q14832, Q14833, P41594 and Q14831, respectively). Sequence data used in the alignment for Extended Data Fig. 10d are H. sapiens Gi1, Gi2, Gi3, Gs, Gq, G12 and G13 (Uniprot accession codes P63096, P04899, P08754, P63092, P50148, Q03113 and Q14344, respectively). Source data are provided with this paper.

References

  1. Gilman, A. G. G proteins: transducers of receptor-generated signals. Annu. Rev. Biochem. 56, 615–649 (1987).

    CAS  PubMed  Google Scholar 

  2. Weis, W. I. & Kobilka, B. K. The molecular basis of G protein-coupled receptor activation. Annu. Rev. Biochem. 87, 897–919 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Chun, L., Zhang, W. & Liu, J. Structure and ligand recognition of class C GPCRs. Acta Pharmacol. Sin. 33, 312–323 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Ellaithy, A., Gonzalez-Maeso, J., Logothetis, D. A. & Levitz, J. Structural and biophysical mechanisms of class C G protein-coupled receptor function. Trends Biochem. Sci. 45, 1049–1064 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Shen, C. et al. Structural basis of GABAB receptor–Gi protein coupling. Nature 594, 594–598 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Seven, A. B. et al. G-protein activation by a metabotropic glutamate receptor. Nature 595, 450–454 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lin, S. et al. Structures of Gi-bound metabotropic glutamate receptors mGlu2 and mGlu4. Nature 594, 583–588 (2021).

    CAS  PubMed  Google Scholar 

  8. Watkins, L. R. & Orlandi, C. In vitro profiling of orphan G protein coupled receptor (GPCR) constitutive activity. Br. J. Pharmacol. 178, 2963–2975 (2021).

    CAS  PubMed  Google Scholar 

  9. Tsutsumi, N. et al. Structural basis for the constitutive activity and immunomodulatory properties of the Epstein–Barr virus-encoded G protein-coupled receptor BILF1. Immunity 54, 1405–1416.e7 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Lin, X. et al. Cryo-EM structures of orphan GPR21 signaling complexes. Nat. Commun. 14, 216 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Xu, L. et al. Cryo-EM structure of constitutively active human Frizzled 7 in complex with heterotrimeric Gs. Cell Res. 31, 1311–1314 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhang, H. et al. Structural basis for chemokine recognition and receptor activation of chemokine receptor CCR5. Nat. Commun. 12, 4151 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Lin, X. et al. Structural basis of ligand recognition and self-activation of orphan GPR52. Nature 579, 152–157 (2020).

    CAS  PubMed  Google Scholar 

  15. Xu, P. et al. Structural insights into the lipid and ligand regulation of serotonin receptors. Nature 592, 469–473 (2021).

    CAS  PubMed  Google Scholar 

  16. Xu, P. et al. Structural identification of lysophosphatidylcholines as activating ligands for orphan receptor GPR119. Nat. Struct. Mol. Biol. 29, 863–870 (2022).

    CAS  PubMed  Google Scholar 

  17. Qu, X. et al. Structural basis of tethered agonism of the adhesion GPCRs ADGRD1 and ADGRF1. Nature 604, 779–785 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ping, Y.-Q. et al. Structural basis for the tethered peptide activation of adhesion GPCRs. Nature 604, 763–770 (2022).

    CAS  PubMed  Google Scholar 

  19. Xiao, P. et al. Tethered peptide activation mechanism of the adhesion GPCRs ADGRG2 and ADGRG4. Nature 604, 771–777 (2022).

    CAS  PubMed  Google Scholar 

  20. Barros-Álvarez, X. et al. The tethered peptide activation mechanism of adhesion GPCRs. Nature 604, 757–762 (2022).

    PubMed  PubMed Central  Google Scholar 

  21. Jeong, E., Kim, Y., Jeong, J. & Cho, Y. Structure of the class C orphan GPCR GPR158 in complex with RGS7-Gβ5. Nat. Commun. 12, 6805 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Smith, E. L. et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci. Transl. Med. 11, eaau7746 (2019).

    PubMed  PubMed Central  Google Scholar 

  23. Patil, D. N. et al. Cryo-EM structure of human GPR158 receptor coupled to the RGS7-Gβ5 signaling complex. Science 375, 86–91 (2021).

    PubMed  PubMed Central  Google Scholar 

  24. Calver, A. R. et al. Molecular cloning and characterisation of a novel GABAB-related G-protein coupled receptor. Mol. Brain. Res. 110, 305–317 (2003).

    CAS  PubMed  Google Scholar 

  25. Kindt, K. S. et al. EMX2-GPR156-Gαi reverses hair cell orientation in mechanosensory epithelia. Nat. Commun. 12, 2861 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Greene, D. et al. Genetic association analysis of 77,539 genomes reveals rare disease etiologies. Nat. Med. 29, 679–688 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ramzan, M. et al. Novel GPR156 variants confirm its role in moderate sensorineural hearing loss. Sci. Rep. 13, 17010 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Kalam, H. et al. Identification of host regulators of Mycobacterium tuberculosis phenotypes uncovers a role for the MMGT1-GPR156 lipid droplet axis in persistence. Cell Host Microbe 31, 978–992 (2023).

    CAS  PubMed  Google Scholar 

  29. Maeda, S. et al. Development of an antibody fragment that stabilizes GPCR/G-protein complexes. Nat. Commun. 9, 3712 (2018).

    PubMed  PubMed Central  Google Scholar 

  30. Park, J. et al. Structure of human GABAB receptor in an inactive state. Nature 584, 304–309 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Papasergi-Scott, M. M. et al. Structures of metabotropic GABAB receptor. Nature 584, 310–314 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Shaye, H. et al. Structural basis of the activation of a metabotropic GABA receptor. Nature 584, 298–303 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Mao, C. et al. Cryo-EM structures of inactive and active GABAB receptor. Cell Res. 30, 564–573 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Kim, Y., Jeong, E., Jeong, J.-H., Kim, Y. & Cho, Y. Structural basis for activation of the heterodimeric GABAB Receptor. J. Mol. Biol. 432, 5966–5984 (2020).

    CAS  PubMed  Google Scholar 

  35. Koehl, A. et al. Structural insights into the activation of metabotropic glutamate receptors. Nature 566, 79–84 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Gao, Y. et al. Asymmetric activation of the calcium-sensing receptor homodimer. Nature 595, 455–459 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Du, J. et al. Structures of human mGlu2 and mGlu7 homo- and heterodimers. Nature 594, 589–593 (2021).

    CAS  PubMed  Google Scholar 

  38. Jurcik, A. et al. CAVER Analyst 2.0: analysis and visualization of channels and tunnels in protein structures and molecular dynamics trajectories. Bioinformatics 34, 3586–3588 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Suckau, D. et al. A novel MALDI LIFT-TOF/TOF mass spectrometer for proteomics. Anal. Bioanal. Chem. 376, 952–965 (2003).

    CAS  PubMed  Google Scholar 

  40. Carlson, M. L. et al. The peptidisc, a simple method for stabilizing membrane proteins in detergent-free solution. eLife 7, e34085 (2018).

    PubMed  PubMed Central  Google Scholar 

  41. Symons, J. L. et al. Lipidomic atlas of mammalian cell membranes reveals hierarchical variation induced by culture conditions, subcellular membranes, and cell lineages. Soft Matter 17, 288–297 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Pin, J.-P., Galvez, T. & Prézeau, L. Evolution, structure, and activation mechanism of family 3/C G-protein-coupled receptors. Pharmacol. Ther. 98, 325–354 (2003).

    CAS  PubMed  Google Scholar 

  43. Congreve, M., Oswald, C. & Marshall, F. H. Applying structure-based drug design approaches to allosteric modulators of GPCRs. Trends Pharmacol. Sci. 38, 837–847 (2017).

    CAS  PubMed  Google Scholar 

  44. Potterton, L. et al. CCP4i2: the new graphical user interface to the CCP4 program suite. Acta Crystallogr. D. Struct. Biol. 74, 68–84 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Pándy-Szekeres, G. et al. GPCRdb in 2023: state-specific structure models using AlphaFold2 and new ligand resources. Nucleic Acids Res. 51, D395–D402 (2022).

    PubMed Central  Google Scholar 

  47. Flock, T. et al. Universal allosteric mechanism for Gα activation by GPCRs. Nature 524, 173–179 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Nehmé, R. et al. Mini-G proteins: novel tools for studying GPCRs in their active conformation. PLoS One 12, e0175642 (2017).

    PubMed  PubMed Central  Google Scholar 

  49. Liang, J. et al. Structural basis of lysophosphatidylserine receptor GPR174 ligand recognition and activation. Nat. Commun. 14, 1012 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim, K. et al. Structure of a hallucinogen-activated Gq-coupled 5-HT2A serotonin receptor. Cell 182, 1574–1588.e19 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, S. et al. Inactive and active state structures template selective tools for the human 5-HT5A receptor. Nat. Struct. Mol. Biol. 29, 677–687 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    CAS  PubMed  Google Scholar 

  54. Rohou, A. & Grigorieff, N. CTFFIND4: fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    PubMed  PubMed Central  Google Scholar 

  55. Yue, Y. et al. Structural insight into apelin receptor-G protein stoichiometry. Nat. Struct. Mol. Biol. 29, 688–697 (2022).

    CAS  PubMed  Google Scholar 

  56. Bepler, T. et al. Positive-unlabeled convolutional neural networks for particle picking in cryo-electron micrographs. Nat. Methods 16, 1153–1160 (2018).

    Google Scholar 

  57. Sanchez-Garcia, R. et al. DeepEMhancer: a deep learning solution for cryo-EM volume post-processing. Commun. Biol. 4, 874 (2021).

    PubMed  PubMed Central  Google Scholar 

  58. 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 

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

    PubMed  Google Scholar 

  60. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D. Struct. Biol. 74, 531–544 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. 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 

  62. 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 

  63. Moriarty, N. W., Grosse‐Kunstleve, R. W. & Adams, P. D. electronic Ligand Builder and Optimization Workbench (eLBOW): a tool for ligand coordinate and restraint generation. Acta Crystallogr. D. Biol. Crystallogr. 65, 1074–1080 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Afonine, P. V. et al. New tools for the analysis and validation of cryo-EM maps and atomic models. Acta Crystallogr. D. Struct. Biol. 74, 814–840 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Olsen, R. H. J. et al. TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome. Nat. Chem. Biol. 16, 841–849 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Schmidpeter, P. A. M. et al. Anionic lipids unlock the gates of select ion channels in the pacemaker family. Nat. Struct. Mol. Biol. 29, 1092–1100 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Hutchins, P. D., Russell, J. D. & Coon, J. J. LipiDex: an integrated software package for high-confidence lipid identification. Cell Syst. 6, 621–625.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Pluskal, T., Castillo, S., Villar-Briones, A. & Orešič, M. MZmine 2: modular framework for processing, visualizing, and analyzing mass spectrometry-based molecular profile data. BMC Bioinform. 11, 395 (2010).

    Google Scholar 

  69. An, J. N. et al. Effects of periostin deficiency on kidney aging and lipid metabolism. Aging (Albany NY) 13, 22649–22665 (2021).

    CAS  PubMed  Google Scholar 

  70. Breil, C., Vian, M. A., Zemb, T., Kunz, W. & Chemat, F. ‘Bligh and Dyer’ and Folch methods for solid–liquid–liquid extraction of lipids from microorganisms. Comprehension of solvatation mechanisms and towards substitution with alternative solvents. Int. J. Mol. Sci. 18, 708 (2017).

    PubMed  PubMed Central  Google Scholar 

  71. Lee, J. W., Nishiumi, S., Yoshida, M., Fukusaki, E. & Bamba, T. Simultaneous profiling of polar lipids by supercritical fluid chromatography/tandem mass spectrometry with methylation. J. Chromatogr. A 1279, 98–107 (2013).

    CAS  PubMed  Google Scholar 

  72. Lee, J. W. et al. Detailed characterization of alterations in the lipid profiles during autophagic cell death of leukemia cells. RSC Adv. 6, 29512–29518 (2016).

    CAS  Google Scholar 

  73. Shanta, S. R. et al. Binary matrix for MALDI imaging mass spectrometry of phospholipids in both ion modes. Anal. Chem. 83, 1252–1259 (2011).

    CAS  PubMed  Google Scholar 

  74. Fahy, E., Sud, M., Cotter, D. & Subramaniam, S. LIPID MAPS online tools for lipid research. Nucleic Acids Res. 35, W606–W612 (2007).

    PubMed  PubMed Central  Google Scholar 

  75. Sud, M. et al. LMSD: LIPID MAPS structure database. Nucleic Acids Res. 35, D527–D532 (2007).

    CAS  PubMed  Google Scholar 

  76. Noh, S. A. et al. Alterations in lipid profile of the aging kidney identified by MALDI imaging mass spectrometry. J. Proteome Res. 18, 2803–2812 (2019).

    CAS  PubMed  Google Scholar 

  77. Liebisch, G. et al. Quantitative measurement of different ceramide species from crude cellular extracts by electrospray ionization tandem mass spectrometry (ESI–MS/MS). J. Lipid Res. 40, 1539–1546 (1999).

    CAS  PubMed  Google Scholar 

  78. Hsu, F.-F. & Turk, J. Structural determination of sphingomyelin by tandem mass spectrometry with electrospray ionization. J. Am. Soc. Mass Spectr. 11, 437–449 (2000).

    CAS  Google Scholar 

  79. Pi, J., Wu, X. & Feng, Y. Fragmentation patterns of five types of phospholipids by ultra-high-performance liquid chromatography electrospray ionization quadrupole time-of-flight tandem mass spectrometry. Anal. Methods 8, 1319–1332 (2016).

    CAS  Google Scholar 

  80. Sugawara, T., Aida, K., Duan, J. & Hirata, T. Analysis of glucosylceramides from various sources by liquid chromatography–ion trap mass spectrometry. J. Oleo Sci. 59, 387–394 (2010).

    CAS  PubMed  Google Scholar 

  81. Gu, M., Kerwin, J. L., Watts, J. D. & Aebersold, R. Ceramide profiling of complex lipid mixtures by electrospray ionization mass spectrometry. Anal. Biochem. 244, 347–356 (1997).

    CAS  PubMed  Google Scholar 

  82. Abraham, M. J. et al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 1, 19–25 (2015).

    Google Scholar 

  83. Huang, J. et al. CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14, 71–73 (2017).

    CAS  PubMed  Google Scholar 

  84. Abagyan, R., Totrov, M. & Kuznetsov, D. ICM—a new method for protein modeling and design: applications to docking and structure prediction from the distorted native conformation. J. Comput. Chem. 15, 488–506 (1994).

    CAS  Google Scholar 

  85. Jo, S., Kim, T., Iyer, V. G. & Im, W. CHARMM‐GUI: a web‐based graphical user interface for CHARMM. J. Comput. Chem. 29, 1859–1865 (2008).

    CAS  PubMed  Google Scholar 

  86. 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).

    CAS  PubMed  Google Scholar 

  87. Wu, E. L. et al. CHARMM‐GUI Membrane Builder toward realistic biological membrane simulations. J. Comput. Chem. 35, 1997–2004 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Hess, B., Bekker, H., Berendsen, H. J. C. & Fraaije, J. G. E. M. LINCS: a linear constraint solver for molecular simulations. J. Comput. Chem. 18, 1463–1472 (1997).

    CAS  Google Scholar 

  89. Bussi, G., Donadio, D. & Parrinello, M. Canonical sampling through velocity rescaling. J. Chem. Phys. 126, 014101 (2007).

    PubMed  Google Scholar 

  90. McGibbon, R. T. et al. MDTraj: a modern open library for the analysis of molecular dynamics trajectories. Biophys. J. 109, 1528–1532 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Michaud‐Agrawal, N., Denning, E. J., Woolf, T. B. & Beckstein, O. MDAnalysis: a toolkit for the analysis of molecular dynamics simulations. J. Comput. Chem. 32, 2319–2327 (2011).

    PubMed  PubMed Central  Google Scholar 

  92. Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods 17, 261–272 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Koh for technical help and Y. Kim, C. Lee, J. Lee, K. Kim, S. H. Ryu (POSTECH), Y. Yu (Kookmin U.), M. Jin (GIST) and J. Kim (SNU) for helpful comments. This work was supported by grants from the National Research Foundation of Korea (NRF) funded by the Korean government (MEST, No. 2021R1A2C301335711 and 2019M3E5D6066058 to Y.C.), the Bio & Medical Technology Development Program (NRF-2019M3E5D3073567 to K.P.K.), the Ministry of Science and ICT (grant number 2022R1A2C1005885 to J.H.), the BK21 program (Ministry of Education to Y.C.), Wellcome Trust Investigator Award (221795/Z/20/Z to X.Q., D.W., C.V.R.) and internal funding from the University of Southern California Dornsife College (to V.K). Cryo-EM data were acquired at the Core Research Facility, Pusan National University. Computing resources were provided by the Center for Advanced Research Computing (CARC) at the University of Southern California (https://carc.usc.edu).

Author information

Authors and Affiliations

Authors

Contributions

J.S. carried out protein expression, purification and structure determination with the help of J.P. J.S. carried out data collection with the help of J.H. J.P. and J.S. performed biochemical experiments with the help of K.K. and J.-Y.L. K.P.K. and J.J. performed mass spectroscopy and phospholipid characterization analysis. X.Q., D.W. and C.V.R. performed comparative lipidomics analysis. J.H.L. and V.K. performed molecular dynamics simulations. J.S., J.P. and Y.C. designed the research; Y.C. wrote the manuscript with the help of J.S., J.P. and K.P.K.

Corresponding authors

Correspondence to Kwang Pyo Kim or Yunje Cho.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Bryan Roth and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Katarzyna Ciazynska, in collaboration with the Nature Structural & Molecular Biology team.

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 Purification of the GPR156-Go complex.

a–c, Size-exclusion chromatography profile (a), SDS-PAGE gel (b), and negative-staining microscopy analysis (c) of the purified GPR156-Go complex. Purification experiments of the GPR156-Go complex in (b) were performed independently at least three times. Negative-staining microscopy analysis (c) was performed once. d, Representative cryo-EM micrograph of the GPR156-Go complex. The cryo-EM data collection of GPR156-Go was performed once. e, 2D class averages of the Go-free GPR156. f, 2D class averages of the GPR156-Go complex. g, Comparison of constitutive activities of GPR156 and GABAB in G-protein activation, measured by BRET2 assay. The activity of GABAB was observed in the absence or presence of 100 µM GABA. Data were mean ± SEM from three independent experiments, performed in technical triplicate. Statistical differences in ΔBRET were analyzed by two-way ANOVA with Dunnett’s post hoc test. h, cAMP inhibition assay for GPR156 C-terminal truncation. Mock transfected with an empty vector was used as a negative control. cAMP production was normalized to a percent of WT activity. i, Surface expression levels of WT and GPR156 mutants in the cAMP assay, measured by ELISA. Surface expression levels of GPR156 mutants were normalized to a percent of WT surface expression level. Data in h and i were mean ± SEM from at least three independent experiments, performed in technical triplicate. Statistical differences were analyzed by one-way ANOVA with Dunnett’s post hoc test, compared to WT (NS, not significant; ***P < 0.001; ****P < 0.0001). j, k, A model of GPR156 predicted from AlphaFold2 was used for initial docking and model building. The N-terminal (1 to 39) and the C-terminal (336 to 814) regions are disordered and omitted in the figure. The model is colored according to the predicted Local Distance Difference Test (pLDDT) score (j). The Predicted-Alignment-Error (PAE) plot of the GPR156 model (k).

Source data

Extended Data Fig. 2 Flow chart of cryo-EM and data processing.

Cryo-EM processing chart of the G-protein coupled and G-protein free GPR156.

Extended Data Fig. 3 Analysis of the quality of the cryo-EM map.

a, b, Angular distributions, Fourier shell correlation curves, and globally refined cryo-EM density maps of GPR156-Go (a) and GPR156 alone (b). c–f, Angular distributions, Fourier shell correlation curves and locally refined cryo-EM density maps marked local resolution of GPR156 dimer (c), G-protein-scFv16 (d), GPR156A/B (e), and GPR156C/D (f). g, h, Fourier shell correlation curves of the model versus the map generated through PHENIX.Mtriage64 of globally and locally refined GPR156-Go (g) and GPR156 alone (h). i, j, Global fitting of the structures of GPR156-Go into the composite map (i) and GPR156 alone into the globally refined map (j). k, Cryo-EM densities and fitted atomic models. GF-GPR156, Go, GPR156A, GPR156B, and GC-GPR156 are shown in yellow, pink, salmon, cyan, and green, respectively.

Extended Data Fig. 4 Key features of GPR156.

a, Cryo-EM map of the GPR156 tetramer. b, Interface between the head-to-head dimer of GPR156. c, Interaction between TM7 of GPR156C and ICL2 of GPR156B. H-bonds (Q314-D155 and E321-V158) and hydrophobic interactions (F318-V158 and I325-I159) are highlighted. d, e, Aligned structures of the two GPR156 dimers in two views; front view (d), bottom view. Structures of the ICL2s are encircled (e). f–h, Structural comparison of GPR156B and GABAB-Gi (PDB:7EB2 ref. 5) with respect to ECL2 (f), TM7 (g), and ICLs (h). i, Cryo-EM map of the GPR156-Go complex. j, k, Aligned structures of an GPR156 alone dimer with the GPR156-Go complex in two views; front view (j), bottom view (k). l, A density on top of the ECL2 in two different views. m, Close-up view of the interactions between Y1463.55 (nGC) and H248ICL3 (GC).

Extended Data Fig. 5 Comparison of the dimeric arrangement of GPR156 with other class C GPCRs in inactive and active states.

a–i, The TMDs of Class C GPCRs were aligned with GC-GPR156 (black line) and shown in the extracellular (top) view. GPR156-Go (a), inactive GABAB (red; PDB: 7C7S ref. 33) (b), active GABAB (orange; 7EB2 ref. 5) (c), active mGlu4 (beige; 7E9H ref. 7) (d), inactive mGlu2 (yellow; 7EPA ref. 37) (e), active mGlu2 (green; 7MTS ref. 6) (f), apo GPR158 (pink; 7EWL ref. 21) (g), inactive CaSR (blue; 7M3J ref. 36) (h), active CaSR (purple; 7M3F ref. 36) (i). The gray filled line represents nGC-GPR156.

Extended Data Fig. 6 Characterization of phospholipid in GPR156.

a, Comparative lipidomics analysis of endogenous lipids bound to purified recombinant GPR156. PC and PE are enrichment in GPR156 fraction relative to total cellular lysate. Bars show mean ± standard deviation from three independent experiments (dots). b–c, GPR156 activation as measured by GTPase-Glo assay for GPR156-Gi peptidisc containing PE (b) or GPR156 in LMNG (c). A peptidisc containing GPRC5D-Gi and PE was incubated with PG as a control. Lower levels of residual GTP indicate higher level of G-protein activity. Data in (b) and (c) were mean ± SEM from three independent experiments, performed in technical duplicate. Statistical differences were analyzed by one-way ANOVA with Dunnett’s post hoc test. d‒f, Comparison of molecular dynamics simulation of GPR156 Go-coupled complexes with PG versus PC. d, Root-mean-square-fluctuation (r.m.s.f.) calculated of each subunit in the complexes; shading refers to 95 % confidence interval (n = 5). e–f, Distribution of the closest distances between the sidechain of R2796.57 and the phospholipids against the closest distances between the backbone of C216ECL2 and the phospholipids on the nGC protomer (e) and on the GC protomer (f). The shading refers to density estimated with a multivariate gaussian kernel; the marginal distribution (by count) is shown on the sidebar. The black points in the background is the datapoints collected every 0.5 ns. All distances were calculated using only the heavy atoms. The horizontal and vertical dotted line refers to 3.5 Å. g, Mutational effect on the F215ECL2 adjacent to the phospholipid head in G-protein activation, measured by BRET2 assay. Data were mean ± SEM from four independent experiments, performed in technical triplicate. Statistical differences were analyzed by two-way ANOVA with Dunnett’s post hoc test, compared to WT (NS, not significant; *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001). h‒i, Comparison of PC- and PG-bound GPR156 with representative snapshots from MD simulations of GPR156. Close-up views of a phospholipid-binding site in the simulations of PC-bound (h) and PG-bound GPR156 (i). nGC protomer and GC protomer are shown on top and bottom, respectively. Three representative snapshots (970, 985, and 1000 ns) of a simulation trajectory are displayed.

Source data

Extended Data Fig. 7 Sequence alignment of GPR156 with other human class C GPCRs.

The conserved residues are marked in yellow. The alignment was output from GPCRdb (gpcrdb.org) and edited by using snapgene (snapgene.com).

Extended Data Fig. 8 Phospholipids in GPR156.

a, In one protomer, the phenyl ring is flipped by 85° toward the dimer interface, creating space for the lateral movement of the phospholipid. We refer this conformer to as an open form. The W229 indole ring can be repositioned upon the conformational change of the F275 ring. b, In another protomer, F275 is packed against the fatty-acyl chain of the phospholipid to form a closed conformation. c, Density at the top half of the dimer interface, in which a CLR molecule is modelled. d, e, Densities near V223 (d) and W284 (e) in GPR156. f, Comparison of the phospholipid-binding in GPR156 with that of GABAB2. g, Comparison of the phospholipid-binding in GPR156 with the PAM-binding in mGlu2 and CaSR. h–j, cAMP inhibition assay for GPR156 mutated at the phospholipid binding site (h), dimer interface (i, j). Data were mean ± SEM from at least three independent experiments, performed in technical triplicate. Statistical differences were analyzed by one-way ANOVA with Dunnett’s post hoc test, compared to WT (NS, not significant; **P < 0.01; ***P < 0.001; ****P < 0.0001).

Source data

Extended Data Fig. 9 Structural transition of the cytoplasmic face of GPR156 in the Go protein-coupled state.

a–c, Comparison of the Go-free, nGC-, and GC-protomers in three different views; front (a), extracellular (b), and cytoplasmic view (c). d–e, Aligned ICL1s (d) and ICL2s (e) of the GPR156B with GC-protomers. The major structural differences in the C-terminal loop and ICLs are indicated by red arrows. f, cAMP inhibition assay for GPR156 mutated at ICL2. Data were mean ± SEM from three independent experiments. Statistical differences were analyzed by one-way ANOVA with Dunnett’s post hoc test, compared to WT. g–l, Comparison of the TMD and ICLs of GPR156 with those of other class C GPCRs–Gi; GABAB-Gi1 (PDB: 7EB2 ref. 5), mGlu2-Gi1 (7MTS ref. 6), mGlu4-Gi3 (7E9H ref. 7). Comparison of the TMD of GPR156 GC-protomer with those of other class C GPCRs bound to Gi in three different views; front (g), extracellular (h), and cytoplasmic view (i). Comparison of ICL1 (j), ICL2 (k), and ICL3 and the C-terminal loop (l). m, Mutational effect on CTL of GPR156 in G-protein activation, measured by BRET2 assay. Data were mean ± SEM from three independent experiments, performed in technical triplicate. Statistical differences were analyzed by two-way ANOVA with Dunnett’s post hoc test, compared to WT (NS, not significant; **P < 0.01; ***P < 0.001; ****P < 0.0001).

Source data

Extended Data Fig. 10 Go binding to GPR156.

a–c, Comparison of Go binding between GPR156 and other class C GPCRs: GABAB-Gi1 (PDB: 7EB2 ref. 5) (a), mGlu2-Gi1 (7MTS ref. 6) (b), mGlu4-Gi3 (7E9H ref. 7) (c). The red arrows indicate the structural differences in the receptors and the G proteins. d. Sequence alignment of the residues in the α5 helix in different human Gα proteins. Residues interacting with GPR156 are marked with a light green circle. Absolutely conserved and highly conserved (≥50%) residues are marked with orange and yellow colors, respectively.

Supplementary information

Supplementary Information

Supplementary Notes 1–7, Videos 1–3, and Tables 1–4.

Reporting Summary

Peer Review File

Supplementary Video 1

Dynamics of TM4 and ICL2 of the GPR156B protomer revealed by 3D variability analysis.

Supplementary Video 2

Flexibility of CTL in the GPR156 GC protomer.

Supplementary Video 3

Rigid body movement of the GC protomer with respect to the G protein.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig./Table 1

Unprocessed gel

Source Data Extended Data Fig./Table 1

Statistical source data.

Source Data Extended Data Fig./Table 6

Statistical source data.

Source Data Extended Data Fig./Table 8

Statistical source data.

Source Data Extended Data Fig./Table 9

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shin, J., Park, J., Jeong, J. et al. Constitutive activation mechanism of a class C GPCR. Nat Struct Mol Biol 31, 678–687 (2024). https://doi.org/10.1038/s41594-024-01224-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-024-01224-7

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