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:

The tethered peptide activation mechanism of adhesion GPCRs

Abstract

Adhesion G-protein-coupled receptors (aGPCRs) are characterized by the presence of auto-proteolysing extracellular regions that are involved in cell–cell and cell–extracellular matrix interactions1. Self cleavage within the aGPCR auto-proteolysis-inducing (GAIN) domain produces two protomers—N-terminal and C-terminal fragments—that remain non-covalently attached after receptors reach the cell surface1. Upon dissociation of the N-terminal fragment, the C-terminus of the GAIN domain acts as a tethered agonist (TA) peptide to activate the seven-transmembrane domain with a mechanism that has been poorly understood2,3,4,5. Here we provide cryo-electron microscopy snapshots of two distinct members of the aGPCR family, GPR56 (also known as ADGRG1) and latrophilin 3 (LPHN3 (also known as ADGRL3)). Low-resolution maps of the receptors in their N-terminal fragment-bound state indicate that the GAIN domain projects flexibly towards the extracellular space, keeping the encrypted TA peptide away from the seven-transmembrane domain. High-resolution structures of GPR56 and LPHN3 in their active, G-protein-coupled states, reveal that after dissociation of the extracellular region, the decrypted TA peptides engage the seven-transmembrane domain core with a notable conservation of interactions that also involve extracellular loop 2. TA binding stabilizes breaks in the middle of transmembrane helices 6 and 7 that facilitate aGPCR coupling and activation of heterotrimeric G proteins. Collectively, these results enable us to propose a general model for aGPCR activation.

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: Cryo-EM reconstructions for GPR56 and LPHN3.
Fig. 2: Structures of active-state GPR56 and LPHN3 complexes with bound TA peptide.
Fig. 3: TA–peptide interactions.
Fig. 4: G-protein binding by GPR56 and LPHN3.

Similar content being viewed by others

Data availability

The cryo-EM density maps and corresponding coordinates have been deposited in the Electron Microscopy Data Bank and the Protein Data Bank, respectively, under the following accession codes: EMD-25077 and 7SF8 (GPR56 7TM–miniG13) and EMD-25076 and 7SF7 (LPHN3 7TM–miniG13). Source data are provided with this paper.

References

  1. Vizurraga, A., Adhikari, R., Yeung, J., Yu, M. & Tall, G. G. Mechanisms of adhesion G protein–coupled receptor activation. J. Biol. Chem. 295, 14065–14083 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Liebscher, I. et al. A tethered agonist within the ectodomain activates the adhesion G protein-coupled receptors GPR126 and GPR133. Cell Rep. 9, 2018–2026 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Stoveken, H. M. et al. Dihydromunduletone is a small-molecule selective adhesion G protein-coupled receptor antagonist. Mol. Pharmacol. 90, 214–224 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Stoveken, H. M., Hajduczok, A. G., Xu, L. & Tall, G. G. Adhesion G protein-coupled receptors are activated by exposure of a cryptic tethered agonist. Proc. Natl Acad. Sci. USA 112, 6194–6199 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wilde, C. et al. The constitutive activity of the adhesion GPCR GPR114/ADGRG5 is mediated by its tethered agonist. FASEB J. 30, 666–673 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Hamann, J. et al. Adhesion G protein-coupled receptors. Pharmacol Rev. 67, 338–367 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Langenhan, T., Aust, G. & Hamann, J. Sticky signaling-adhesion class G protein-coupled receptors take the stage. Sci. Signal. 6, re3 (2013).

    Article  PubMed  CAS  Google Scholar 

  8. Langenhan, T., Piao, X. & Monk, K. R. Adhesion G protein-coupled receptors in nervous system development and disease. Nat. Rev. Neurosci. 17, 550–561 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Boucard, A. A., Maxeiner, S. & Südhof, T. C. Latrophilins function as heterophilic cell-adhesion molecules by binding to teneurins: regulation by alternative splicing. J. Biol. Chem. 289, 387–402 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Jin, Z., Luo, R. & Piao, X. in G Protein-Coupled Receptors in Health and Disease, Part B Vol. 89 (ed. Tao, Y.-X.) 1–13 (Elsevier, 2009).

  11. Salzman, G. S. et al. Structural basis for regulation of GPR56/ADGRG1 by its alternatively spliced extracellular domains. Neuron 91, 1292–1304 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Purcell, R. H. & Hall, R. A. Adhesion G protein-coupled receptors as drug targets. Annu. Rev. Pharmacol. Toxicol. 58, 429–449 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Aust, G., Zhu, D., Van Meir, E. G. & Xu, L. Adhesion GPCRs in tumorigenesis. Handb. Exp. Pharmacol. 234, 369–396 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Scholz, N. Cancer cell mechanics: adhesion G protein-coupled receptors in action? Front. Oncol. 8, 59 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Piao, X. et al. G protein-coupled receptor-dependent development of human frontal cortex. Science 303, 2033–2036 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  16. Iguchi, T. et al. Orphan G protein-coupled receptor GPR56 regulates neural progenitor cell migration via a Gα12/13 and Rho pathway. J. Biol. Chem. 283, 14469–14478 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Luo, R. et al. G protein-coupled receptor 56 and collagen III, a receptor-ligand pair, regulates cortical development and lamination. Proc. Natl Acad. Sci. USA 108, 12925–12930 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sando, R., Jiang, X. & Südhof, T. C. Latrophilin GPCRs direct synapse specificity by coincident binding of FLRTs and teneurins. Science 363, eaav7969 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Li, J. et al. Structural basis for teneurin function in circuit-wiring: a toxin motif at the synapse. Cell 173, 735–748.e715 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mathiasen, S. et al. G12/13 is activated by acute tethered agonist exposure in the adhesion GPCR ADGRL3. Nat. Chem. Biol. 16, 1343–1350 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cruz-Ortega, J. S. & Boucard, A. A. Actin cytoskeleton remodeling defines a distinct cellular function for adhesion G protein-coupled receptors ADGRL/latrophilins 1, 2 and 3. Biol. Open 8, bio039826 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wallis, D. et al. Initial characterization of mice null for Lphn3, a gene implicated in ADHD and addiction. Brain Res. 1463, 85–92 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Araç, D. et al. A novel evolutionarily conserved domain of cell-adhesion GPCRs mediates autoproteolysis. EMBO J. 31, 1364–1378 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Lu, Y. C. et al. Structural basis of latrophilin–FLRT–UNC5 interaction in cell adhesion. Structure 23, 1678–1691 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jackson, V. A. et al. Structural basis of latrophilin–FLRT interaction. Structure 23, 774–781 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jackson, V. A. et al. Super-complexes of adhesion GPCRs and neural guidance receptors. Nat. Commun. 7, 11184 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ping, Y.-Q. et al. Structures of the glucocorticoid-bound adhesion receptor GPR97–Go complex. Nature 589, 620–626 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Wootten, D., Simms, J., Miller, L. J., Christopoulos, A. & Sexton, P. M. Polar transmembrane interactions drive formation of ligand-specific and signal pathway-biased family B G protein-coupled receptor conformations. Proc. Natl Acad. Sci. USA 110, 5211 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ballesteros, J. A. & Weinstein, H. in Methods in Neurosciences Vol. 25 (ed. Sealfon, S. C.) 366–428 (Academic, 1995).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hilger, D. et al. Structural insights into differences in G protein activation by family A and family B GPCRs. Science 369, eaba3373 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Stoveken, H. M., Larsen, S. D., Smrcka, A. V. & Tall, G. G. Gedunin- and khivorin-derivatives are small-molecule partial agonists for adhesion G protein-coupled receptors GPR56/ADGRG1 and GPR114/ADGRG5. Mol. Pharmacol. 93, 477–488 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  37. Macfarlane, S. R., Seatter, M. J., Kanke, T., Hunter, G. D. & Plevin, R. Proteinase-activated receptors. Pharmacol. Rev. 53, 245–282 (2001).

    CAS  PubMed  Google Scholar 

  38. Vu, T. K., Hung, D. T., Wheaton, V. I. & Coughlin, S. R. Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation. Cell 64, 1057–1068 (1991).

    Article  CAS  PubMed  Google Scholar 

  39. Kim, K. et al. Structure of a hallucinogen-activated Gq-coupled 5-HT2A serotonin receptor. Cell 182, 1574–1588.e1519 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bayburt, T. H., Grinkova, Y. V. & Sligar, S. G. Self-assembly of discoidal phospholipid bilayer nanoparticles with membrane scaffold proteins. Nano Lett. 2, 853–856 (2002).

    Article  ADS  CAS  Google Scholar 

  41. Denisov, I. G., Grinkova, Y. V., Lazarides, A. A. & Sligar, S. G. Directed self-assembly of monodisperse phospholipid bilayer nanodiscs with controlled size. JACS 126, 3477–3487 (2004).

    Article  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  44. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  45. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  48. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. E. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protoc. 10, 845–858 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kreutz, B. et al. A new approach to producing functional Gα subunits yields the activated and deactivated structures of Gα12/13 proteins. Biochemistry 45, 167–174 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr. D 75, 861–877 (2019).

    Article  CAS  Google Scholar 

  53. 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 

  54. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    Article  CAS  PubMed  Google Scholar 

  55. 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 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Lee, J. et al. CHARMM-GUI input generator for NAMD, Gromacs, Amber, Openmm, and CHARMM/OpenMM simulations using the CHARMM36 additive force field. Biophys. J. 110, 641a (2016).

    Article  ADS  Google Scholar 

  57. Jorgensen, W. L., Chandrasekhar, J., Madura, J. D., Impey, R. W. & Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 79, 926–935 (1983).

    Article  ADS  CAS  Google Scholar 

  58. Phillips, J. C. et al. Scalable molecular dynamics with NAMD. J. Comput. Chem. 26, 1781–1802 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ryckaert, J.-P., Ciccotti, G. & Berendsen, H. J. C. Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes. J. Comput. Phys. 23, 327–341 (1977).

    Article  ADS  CAS  Google Scholar 

  60. Miyamoto, S. & Kollman, P. A. Settle: an analytical version of the SHAKE and RATTLE algorithm for rigid water models. J. Comput. Chem. 13, 952–962 (1992).

    Article  CAS  Google Scholar 

  61. Dror, R. O. et al. Structural basis for nucleotide exchange in heterotrimeric G proteins. Science 348, 1361–1365 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Montabana at the Stanford cEMc facility and N. Elad at the Weizmann Institute of Science microscopy unit for support with data collection, Demet Arac and Katherine Leon for early discussions on LPHN. This work was supported by the Mathers Foundation (MF-1804-00129) (G.S.), the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (no. 949364) and the Minerva Foundation (M.S.-B.) and R01s GM120110 and NS103946 (G.G.T.). E.H.Y. is the recipient of the IASH Fellowships for Israeli Postdoctoral Fellows. A.V. is supported by NHLBI F31-HL152563.

Author information

Authors and Affiliations

Authors

Contributions

X.B.-A. and G.G.T. designed GPR56 and G-protein constructs. D.M., M.S.-B. and G.G.T. designed LPHN3 constructs. X.B.-A. expressed and purified active GPR56 and LPHN3 receptors, G proteins and complexes, prepared cryo-EM grids, oversaw data collection, processed cryo-EM data for complexes, modelled the structures, analysed the structural data and prepared the manuscript. R.M.N. performed mutagenesis experiments, cloned G-protein constructs, purified G proteins, assisted in the purification of complexes, analysed data and prepared the manuscript. A.V. and H.S. conducted G-protein activation experiments. D.M. expressed and purified NTF-bound LPHN3 receptors, prepared cryo-EM grids, collected and processed cryo-EM data and assisted in figure preparation. F.H. purified and processed cryo-EM data for FL-GPR56. O.P. prepared cryo-EM grids, performed data collection. M.M.P.-S. performed G-protein activity assays and assisted in figure preparation. M.J.R. performed and analysed molecular dynamic simulations. E.H.Y. Purified LPHN3 and reconstituted in nanodiscs. A.B.S. assisted in cryo-EM data processing. M.C.P. assisted in protein purifications and mutagenesis. J.G.M. assisted in biochemical assays and cloning. A.V., F.E.K. and G.G.T. purified G proteins and prepared aGPCR membranes. X.B.-A., R.M.N., M.S.-B., G.G.T. and G.S. wrote the manuscript. G.S. supervised the project.

Corresponding authors

Correspondence to Moran Shalev-Benami, Gregory G. Tall or Georgios Skiniotis.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Antony Boucard, Aashish Manglik and the other, anonymous, reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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 NTF-bound LPHN3 purification and cryo-EM processing.

a, Design for the NTF-bound LPHN3 construct used for structural studies. Human LPHN3 (V1, residues 495–1138) was cloned into pFastBac containing a hemagglutinin signal peptide tag (HA) and a cleavable N-terminal FLAG-tag. The construct included a C-terminal cleavable GFP followed by a His6 tag. b, Size exclusion chromatography (SEC) profile of LPHN3. Samples corresponding to the main monomeric (grey) fractions were combined and used for cryo-EM studies. c, Coomassie stained SDS-PAGE of the pooled protein sample visualized by cryo-EM showing the presence of the N-terminal ECR and the 7TM domain. d, Anti-FLAG Western-blot analysis of the LPHN3 sample purified in detergent. e, Cryo-EM reference-free 2D class averages of LPHN3 purified in detergent and f, processing flow chart of the NTF-bound LPHN3 sample, including particle selection, 2D and 3D classifications. g, Size exclusion chromatography (SEC) of LPHN3 embedded in lipid nanodiscs composed of MSP1D1 and brain polar lipids (BPL). Yellow bars indicate fluorescence of lipids (Ex: 295 nm, Em: 330 nm). Arrowed bars indicate signal overload. Grey shaded area shows fractions that were used for the cryo-EM analysis. h, SDS-PAGE stained with InstantBlue, showing purified MSP1D1, purified LPHN3 in DDM, reconstituted LPHN3 in lipid nanodiscs before SEC, and selected SEC fractions of the nanodiscs. Bold fractions were used for cryo-EM. i, Representative reference free 2D class averages of LPHN3 embedded in lipid nanodiscs.

Extended Data Fig. 2 Full-length GPR56 purification, cryo-EM processing and low resolution 3D maps for NTF bound GPR56 and LPHN3.

a, Design for the full-length cleavage deficient (CD, H381S) GPR56 construct used for structural studies. Human GPR56 (V2-FL) was cloned into pFastBac containing a hemagglutinin signal peptide tag (HA) and a cleavable N-terminal FLAG-tag. The construct included a C-terminal cleavable His6 tag. b, Size exclusion chromatography (SEC) profile of full-length GPR56. Fractions corresponding to the monomeric peak (grey) were collected and used for structural studies. c, Coomassie stained SDS-PAGE of the FL-GPR56 sample used for cryo-EM analysis. d, Cryo-EM data processing workflow of the FL-GPR56 sample. e, f. Low resolution 3D maps of NTF-bound receptor conformations with docked structures of the ECR of e, GPR56 and f, LPHN3. The GAIN domains and TA peptides are colored in blue and cyan for GPR56 and in magenta and light pink for LPHN3. Domains are labeled. Docked available ECR crystal structures corresponding to PDB 5KVM and 4DLQ for GPR56 and LPHN1, respectively. Scale bars are provided in the left bottom corner.

Extended Data Fig. 3 Construct design and purification of active-state GPR56 and LPHN3 in complex with miniG13 protein.

a, Design for the tethered agonist complex constructs used in the study. Receptors are presented on top, where sequences corresponding to the TA and 7TM regions of GPR56 and LPHN3 were inserted after a hemagglutinin signal peptide (HA) and a methionine residue. Expression vector for the miniG13 heterotrimer presented at the bottom. b, The MiniGα13/i15 sequence. Residues at the N-terminus corresponding to Gαi2 sequence are in grey. The linker replacing the alpha helical domain is in yellow. Residues corresponding to the stabilizing mutations G57DS1H1.03, E58NS1H1.04, S248DS4.07, E251DS4H3.03, I271DH3.08, I355AH5.04, V358IH5.07 are underlined and presented in bold. c, Size-exclusion chromatography (SEC) profiles of purified miniG13-coupled GPR56 (left) and -LPHN3 (right) with insets showing Coomassie-stained SDS-PAGE of the SEC complex peaks.

Extended Data Fig. 4 Single-particle cryo-EM processing workflow and reconstructions of the GPR56/miniG13 complex.

a, Workflow of cryo-EM data processing for the active-state tethered agonist bound 7TM-GPR56/miniG13 complex. b, Angular distribution heat map of particle projections for 7TM-GPR56/miniG13 reconstruction. c, Gold standard Fourier shell correlation (FSC) curve for receptor and miniG13 reconstructions. Dashed line represents the overall nominal resolution of each reconstruction at 0.143 FSC calculated by CryoSPARC. d, Overall composite cryo-EM map for the 7TM-GPR56/miniG13 complex with chain assignments for its components. e, Cryo-EM density for TMs 1–7, the α5 helix of miniGα13/i and the bound tethered agonist for the 7TM-GPR56/miniG13 complex.

Extended Data Fig. 5 Single-particle cryo-EM processing workflow and reconstructions of the LPHN3/miniG13 complex.

a, Workflow of cryo-EM data processing for the active-state tethered agonist bound 7TM-LPHN3/miniG13 complex. b, Angular distribution heat map of particles for 7TM-LPHN3/miniG13 reconstruction. c, Gold standard Fourier shell correlation (FSC) curve for receptor and miniG13 reconstructions. Dashed line represents the overall nominal resolution of each reconstruction at 0.143 FSC calculated by CryoSPARC. d, Overall composite cryo-EM map for the 7TM-LPHN3/miniG13 complex with chain assignments for its components. e, Cryo-EM density for TMs 1–7, the α5 helix of miniGα13/i and the bound tethered agonist for the 7TM-LPHN3/miniG13 complex.

Extended Data Fig. 6 Kinetic G13 GTPγS binding assays for GPR56 and LPHN3 mutants and cell surface abundances of selected mutants.

Kinetic measurements of receptor-stimulated G protein 13 [35S]- GTPγS binding in membranes normalized to the activities of wild type (WT) GPR56 or LPHN3. 7TM/CTF-only truncated receptors with a, b, point mutations at the TA residues. c, d, TA-interacting point mutants. e, f, G protein interaction site point mutants. g, h, 7TM core-stabilizing point mutants. Note: GPR56 Q644A and LPHN3 E948A were found at low abundance, thus potentially explaining their reduced activities. i, Equivalent amounts of WT, W6176.53A, F6377.42A, and F4542.64A full-length GPR56 holoreceptors were activated by ice-cold urea treatment to dissociate NTFs from CTFs prior to measurement of G13 initial GTPγS binding rates at 20 °C. The urea-dependent changes in approximated initial linear rates demonstrate that wild type GPR56 was activated by urea significantly more than each mutant, indicating that the mutations impart reduced functional activity and that the mutant receptors are not completely dysfunctional or mis-folded. Data represent the average of each kinetic reaction measured as technical triplicates with error bars representing +/− S.D. Unpaired, two-tailed student’s t tests were used to determine significance between initial rates. * = p < 0.05, **** = p < 0.0001. j, Relative aGPCR cell surface levels for selected mutants and WT receptors were measured by intact cell biotinylation, streptavidin pulldown and anti-His tag immunoblotting

Source data

Extended Data Fig. 7 Western Blot quantification of relative abundances of mutant receptors evaluated in G13 GTPγS assays.

a, Relative abundances of CTF-only truncated GPR56 receptors in membrane homogenates determined by immunoblotting for anti-His tag. b, Relative abundances of CTF-only truncated LPHN3 receptors in membrane homogenates determined by immunoblotting for anti-His tag. c, Relative abundances of holoreceptor GPR56 NTFs and CTFs before and after treatment of membrane homogenates with ice-cold 6M urea. CTF was immunoblotted for via a GPR56-specific CTF antibody, and NTF was immunoblotted for via a GPR56-specific NTF antibody. *Multiple glycosylated NTF bands. Data represent the mean band intensity of western blots performed in triplicate with error bars representing +/- S.D. Unpaired, two-tailed student’s t tests were used to determine significance between wild type and mutant receptors with reduced abundances. * = p < 0.05

Source data

Extended Data Fig. 8 Additional structural elements involved in the active conformation of TA-bound GPR56 and LPHN3 7TM domain, and MD simulations for TA-bound LPHN3.

a, b, Density corresponding to the TA peptide and ECL2 in GPR56 (a) and LPHN3 (b), indicating that the TA and loop are penetrating the 7TM cavity. c, d, Residues surrounding the toggle switch residue (W6.53) in GPR56 (c) and LPHN3 (d). Electrostatic interactions are shown as dotted grey lines. e, Superposition of GPR56 and LPHN3, showing similarities in 7TM domain conformation. f, G13 GTPγS binding activity for mutants LPHN3 (magenta) and GPR56 (blue) that interact with W6.53. Data represent mean of biologically independent reactions performed in triplicate with error bars representing +/− S.D. RM one-way ANOVA was used to determine significance between mutants and WT. g-h, Molecular dynamics simulations for the LPHN3 tethered agonist and its binding to the 7TM domain. g, Four snapshots of the LPHN3 TA peptide MD simulations in solution spaced 200 ns apart. h, Average secondary structure percentages of LPHN3 peptide from MD simulations. ‘𝛼’ refers to the very broad range of -160 ≤ 𝜑; ≤ -20; -120 ≤ 𝜓 ≤ 50; ‘β’, beta-sheet; ‘PP2’, polyproline 2. i, j, Cryo-EM structure of LPHN3 colored by the difference in RMSF values between wild-type MD simulations and simulations with the tethered agonist (TA) region (dark gray) removed. Positive values indicate an increase in flexibility when the TA is deleted. I and j correspond to two different color scales

Source data

Extended Data Fig. 9 Structural comparisons of receptor and bound G13 protein for GPR56 and LPHN3, and experimental data for G13 N-terminal construct design.

a-b, Overall structural comparison of 7TM domains of G protein coupled GPR56 (blue) with active state Family B1 receptors: GLP1R (PDB ID: 5VAI; receptor in brown, GLP1 peptide in tan), GCGR (PDB ID: 6WPW; receptor in dark green, glucagon derivative ZP3780 in light green) and calcitonin receptor (PDB ID: 5UZ7; orange). a, side view showing similarities in 7TM domain topology and b, top view with superposition of B1 agonists with GPR56 TA in the orthosteric site. c, Superposition of glucocorticoid ligand-bound GPR97 (PDB ID: 7D77, light grey) with GPR56 (blue) and LPHN3 (magenta). Arrows are indicating differences in TM1, TM6 and TM7 between the ligand and to TA-bound structures. d, Top view of superimposed GPR56 and LPHN3 complexes showing positioning of mini-G13 N-terminal helix (αN) with respect to the receptor TMs. e, Superposition of GPR56 bound mini-Gα13 (gold) vs. 5HT1A (PDB ID: 6G79) bound mini-Gαo (green). f, GTPγS binding assay for recombinant G13 proteins in which the authentic N-terminus of Gα13 was replaced with 15 or 29 residues of the Gαi2 αN to improve expression, stability and ability to interact with receptor. Stimulation of G13/i29 nucleotide exchange by both receptors GPR56 (blue) and LPHN3 (magenta) was reduced substantially when compared to wild type G13 or G13/i15. Receptor constructs used in this assay are the TA-decrypted GPR56 and LPHN3. Data displayed as mean of reactions (n = 18 for all except GPR56 + G13/i29, n = 17, and LPHN3 + G13/i29, n = 16) with error bars representing +/− S.E.M. Statistical significance between experimental condition and corresponding control group was calculated using Mann-Whitney analysis, n.s. = not significant, **** = p < 0.0001. g, G protein binding through α5 helix of mini-Gα13 (gold) by GPR56 (blue) and mini-Gαo (red) by GPR97 (PDB ID: 7D77, in white) showing substantially greater opening of TM5-6 in the GPR56 TA-bound structure.

Source data

Extended Data Table 1 Cryo-EM data collection, model refinement and validation statistics
Extended Data Table 2 Tethered agonist interaction distances in active-state aGPCRs and activity of GPR56 BFPP related mutants

Supplementary information

Supplementary Fig. 1

Raw Western blot data for the assessment of relative abundances of CTF-only truncated receptors used in functional in vitro assays.

Reporting Summary

Peer Review File

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Barros-Álvarez, X., Nwokonko, R.M., Vizurraga, A. et al. The tethered peptide activation mechanism of adhesion GPCRs. Nature 604, 757–762 (2022). https://doi.org/10.1038/s41586-022-04575-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04575-7

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