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:

Structures of the ADGRG2–Gs complex in apo and ligand-bound forms

Abstract

Adhesion G protein-coupled receptors are elusive in terms of their structural information and ligands. Here, we solved the cryogenic-electron microscopy (cryo-EM) structure of apo-ADGRG2, an essential membrane receptor for maintaining male fertility, in complex with a Gs trimer. Whereas the formations of two kinks were determinants of the active state, identification of a potential ligand-binding pocket in ADGRG2 facilitated the screening and identification of dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate and deoxycorticosterone as potential ligands of ADGRG2. The cryo-EM structures of DHEA–ADGRG2–Gs provided interaction details for DHEA within the seven transmembrane domains of ADGRG2. Collectively, our data provide a structural basis for the activation and signaling of ADGRG2, as well as characterization of steroid hormones as ADGRG2 ligands, which might be used as useful tools for further functional studies of the orphan ADGRG2.

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 structure of apo-ADGRG2 in complex with the DNGs.
Fig. 2: Identification of DHEA as agonist and DOC as antagonist of ADGRG2.
Fig. 3: Cryo-EM structure of DHEA-bound ADGRG2.
Fig. 4: Interaction modes of DHEA or DOC with ADGRG2.

Similar content being viewed by others

Data availability

All data generated in this study are included in the main text or supplementary materials. Source data are provided with this paper. The atomic coordinates and the cryo-EM density maps have been deposited in PDB and Electron Microscopy Data Bank (EMDB) databases under accession codes 7XM6 and EMD-30658 for apo-ADGRG2–Gs complex; 7XKD and EMDB-33248 for DHEA–ADGRG2–βT-Gs complex (DHEA upper state), 7XKF and EMDB-33250 for DHEA–ADGRG2–βT-Gs complex (DHEA lower state); 7XKE and EMDB-33249 for DHEA–ADGRG2-FL-Gs complex, respectively. All other data are available upon request to the corresponding authors. Publicly available datasets used in this study are PDB IDs 3SN6 and 7D77.

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Boyden, S. E. et al. Vibratory urticaria associated with a missense variant in ADGRE2. N. Engl. J. Med. 374, 656–663 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Arcos-Burgos, M. et al. A common variant of the latrophilin 3 gene, LPHN3, confers susceptibility to ADHD and predicts effectiveness of stimulant medication. Mol. Psychiatry 15, 1053–1066 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Weston, M. D., Luijendijk, M. W., Humphrey, K. D., Moller, C. & Kimberling, W. J. Mutations in the VLGR1 gene implicate G-protein signaling in the pathogenesis of Usher syndrome type II. Am. J. Hum. Genet. 74, 357–366 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bassilana, F., Nash, M. & Ludwig, M. G. Adhesion G protein-coupled receptors: opportunities for drug discovery. Nat. Rev. Drug Disco. 18, 869–884 (2019).

    Article  CAS  Google Scholar 

  6. Kou, I. et al. Genetic variants in GPR126 are associated with adolescent idiopathic scoliosis. Nat. Genet. 45, 676–679 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Patat, O. et al. Truncating mutations in the adhesion G protein-coupled receptor G2 gene ADGRG2 cause an X-linked congenital bilateral absence of vas deferens. Am. J. Hum. Genet. 99, 437–442 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xiao, P. et al. Tethered peptide activation mechanism of the adhesion GPCRs ADGRG2 and ADGRG4. Nature https://doi.org/10.1038/s41586-022-04590-8 (2022).

  9. Ping, Y. Q. et al. Structural basis for the tethered peptide activation of adhesion GPCRs. Nature https://doi.org/10.1038/s41586-022-04619-y (2022).

  10. Qu, X. et al. Structural basis of tethered agonism of the adhesion GPCRs ADGRD1 and ADGRF1. Nature https://doi.org/10.1038/s41586-022-04580-w (2022).

  11. Barros-Alvarez, X. et al. The tethered peptide activation mechanism of adhesion GPCRs. Nature https://doi.org/10.1038/s41586-022-04575-7 (2022).

  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. Bondarev, A. D. et al. Opportunities and challenges for drug discovery in modulating Adhesion G protein-coupled receptor (GPCR) functions. Expert Opin. Drug Disco. 15, 1291–1307 (2020).

    Article  CAS  Google Scholar 

  14. Kuffer, A. et al. The prion protein is an agonistic ligand of the G protein-coupled receptor Adgrg6. Nature 536, 464–468 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Eubelen, M. et al. A molecular mechanism for Wnt ligand-specific signaling. Science https://doi.org/10.1126/science.aat1178 (2018).

  16. Wang, J. et al. RTN4/NoGo-receptor binding to BAI adhesion-GPCRs regulates neuronal development. Cell 184, 5869–5885 e5825 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Liu, D. et al. CD97 promotes spleen dendritic cell homeostasis through the mechanosensing of red blood cells. Science 375, eabi5965 (2022).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. Gupte, J. et al. Signaling property study of adhesion G-protein-coupled receptors. FEBS Lett. 586, 1214–1219 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. An, W. et al. Progesterone activates GPR126 to promote breast cancer development via the Gi pathway. Proc. Natl Acad. Sci. USA 119, e2117004119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang, D. L. et al. Gq activity- and beta-arrestin-1 scaffolding-mediated ADGRG2/CFTR coupling are required for male fertility. eLife https://doi.org/10.7554/eLife.33432 (2018).

  22. Davies, B. et al. Targeted deletion of the epididymal receptor HE6 results in fluid dysregulation and male infertility. Mol. Cell. Biol. 24, 8642–8648 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang, D. et al. Function and therapeutic potential of GPCRs in epididymis. Br. J. Pharmacol. https://doi.org/10.1111/bph.15252 (2020).

  24. Sun, Y. et al. Optimization of a peptide ligand for the adhesion GPCR ADGRG2 provides a potent tool to explore receptor biology. J. Biol. Chem. 296, 100174 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Azimzadeh, P., Talamantez-Lyburn, S. C., Chang, K. T., Inoue, A. & Balenga, N. Spatial regulation of GPR64/ADGRG2 signaling by beta-arrestins and GPCR kinases. Ann. N. Y. Acad. Sci. 1456, 26–43 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Demberg, L. M. et al. Activation of adhesion G protein-coupled receptors: agonist specificity of Stachel sequence-derived peptides. J. Biol. Chem. 292, 4383–4394 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kirchhoff, C., Obermann, H., Behnen, M. & Davies, B. Role of epididymal receptor HE6 in the regulation of sperm microenvironment. Mol. Cell. Endocrinol. 250, 43–48 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Demberg, L. M., Rothemund, S., Schoneberg, T. & Liebscher, I. Identification of the tethered peptide agonist of the adhesion G protein-coupled receptor GPR64/ADGRG2. Biochem. Biophys. Res. Commun. 464, 743–747 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Promel, S. et al. The GPS motif is a molecular switch for bimodal activities of adhesion class G protein-coupled receptors. Cell Rep. 2, 321–331 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rasmussen, S. G. et al. Crystal structure of the beta2 adrenergic receptor-Gs protein complex. Nature 477, 549–555 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yang, F. et al. Structural basis of GPBAR activation and bile acid recognition. Nature https://doi.org/10.1038/s41586-020-2569-1 (2020).

  32. 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, 5211–5216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Visiers, I., Ballesteros, J. A. & Weinstein, H. Three-dimensional representations of G protein-coupled receptor structures and mechanisms. Methods Enzymol. 343, 329–371 (2002).

    Article  PubMed  Google Scholar 

  34. Latorraca, N. R., Venkatakrishnan, A. J. & Dror, R. O. GPCR dynamics: structures in motion. Chem. Rev. 117, 139–155 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Liang, Y. L. et al. Phase-plate cryo-EM structure of a biased agonist-bound human GLP-1 receptor-Gs complex. Nature 555, 121–125 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Hilger, D. et al. Structural insights into differences in G protein activation by family A and family B GPCRs. Science https://doi.org/10.1126/science.aba3373 (2020).

  37. 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 

  38. Singh, J. P. et al. Activation of GPR56, a novel adhesion GPCR, is necessary for nuclear androgen receptor signaling in prostate cells. PLoS ONE 15, e0226056 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bokoch, M. P. et al. Ligand-specific regulation of the extracellular surface of a G-protein-coupled receptor. Nature 463, 108–112 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang, L. et al. Cryo-EM structure of the AVP-vasopressin receptor 2-Gs signaling complex. Cell Res. 31, 932–934 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, F. et al. Molecular basis of ligand recognition and activation of human V2 vasopressin receptor. Cell Res. 31, 929–931 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Mindell, J. A. & Grigorieff, N. Accurate determination of local defocus and specimen tilt in electron microscopy. J. Struct. Biol. 142, 334–347 (2003).

    Article  PubMed  Google Scholar 

  44. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife https://doi.org/10.7554/eLife.42166 (2018).

  46. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nat. Methods 11, 63–65 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

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

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Huang, J. & MacKerell, A. D. Jr. CHARMM36 all-atom additive protein force field: validation based on comparison to NMR data. J. Comput. Chem. 34, 2135–2145 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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).

    Article  CAS  PubMed  Google Scholar 

  53. Li, T. et al. Homocysteine directly interacts and activates the angiotensin II type I receptor to aggravate vascular injury. Nat. Commun. 9, 11 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Lee, M. H. et al. The conformational signature of beta-arrestin2 predicts its trafficking and signalling functions. Nature 531, 665–668 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yang, F. et al. Structure, function and pharmacology of human itch receptor complexes. Nature 600, 164–169 (2021).

    Article  CAS  PubMed  Google Scholar 

  56. Cheng, J. et al. Autonomous sensing of the insulin peptide by an olfactory G protein-coupled receptor modulates glucose metabolism. Cell Metab. 34, 240–255 e210 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key R&D Program of China (grant nos. 2018YFC1003600 to X.Y., 2019YFA0904200 to J.-P.S. and P.X.), the National Natural Science Foundation of China (grant nos. 92057121 to X.Y., 31971195 to P.X., 82090024 and 91949202 to F.Y. and 32000850 to Z.-M.L), the National Science Fund for Distinguished Young Scholars (grant no. 81825022 to J.-P.S.), the Major Fundamental Research Program of Natural Science Foundation of Shandong Province, China (grant nos. ZR2021ZD18 to X.Y. and ZR2020ZD39 to J.-P.S.), Shandong Provincial Natural Science Fund for Excellent Young Scholars (grant no. ZR2021YQ18 to P.X.), the Key Research and Development Program of Shandong Province (grant nos. 2021ZLGX02 to J.-P.S. and GG201709260059 to P.X.), the Key Research Project of the Natural Science Foundation of Beijing, China (Z20J00129 to J.-P.S.), the Rolling program of ChangJiang Scholars and Innovative Research Team in University (grant no. IRT_17R68 to X.Y.), the Fundamental Research Funds for the Central Universities (grant no. 2021JCG020 to J.-P.S. and P.X.). We thank the Cryo-Electron Microscopy Center of Southern University of Science and Technology for supporting our project and we are grateful to X. Ma, Y. Gao and L. Fu for their technical help during cryo-EM data collection. We thank the HPC Cloud Platform of Shandong University and HPC-Service Station in the Cryo-EM center at the Southern University of Science and Technology for data processing support. We also thank Y. Yu at Translational Medicine Core Facility of Advanced Medical Research Institute, Shandong University for her assistance with EnVision multimode plate reader (PerkinElmer).

Author information

Authors and Affiliations

Authors

Contributions

J.-P.S. and X.Y. conceived, designed and supervised the overall project. Z.L. and J.-P.S. designed and supervised the cryo-EM sample preparation, data collection and structural refinement. J.-P.S., X.Y., F.Y., Z.L., P.X., Z.Y., S.G. and H.L. participated in data analysis and interpretation. S.G., P.X. and D.Y. generated the ADGRG2 insect cell expression construct, established the ADGRG2–Gs complex formation strategy and prepared protein samples for cryo-EM. S.G. and P.X. generated viruses and infected Sf9 cells for large quantitative protein production. H.L. performed steroid ligands screening assay for ADGRG2. S.G. and Y.-Q.P. prepared negative staining. R.-Q.B. evaluated the sample by negative-stain EM. R.-Q.B., C.-Y.X. and B.-S.Y. prepared the cryo-EM grids, collected the cryo-EM data and performed cryo-EM map calculations under the supervision of Z.L. R.-Q.B. and Z.L. performed the model building and refinement with assistance from Z.-L.Z., D.Y. and Q.-T.H. C.Z. performed molecular dynamics stimulations. J.-P.S., X.Y., P.X., S.-C.G., Z.Y. and H.L. designed all the mutations for determination of ligand-binding sites. H.L., S.-C.G., C.-H.L., D.J. and D.-L.Z. generated all constructs and mutants for the cell-based assays. H.L., S.-C.G., R.-J.Z., S.H., C.-S.M. and Y.-T.X. performed ELISAs and cAMP accumulation assays. Z.Y. performed FlAsH–BRET assays. C.-X.Z., X.-N.Z., H.-C.L., X.-H.Z. and H.Z. performed the western blot assay. S.G., H.L., P.X., Z.Y., J.-Y.L. and W.-M.X. prepared the figures. J.-P.S., X.Y. and Z.L. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Fan Yi, Zhongmin Liu, Jin-Peng Sun or Xiao Yu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Bryan Roth, Jean-Ju Chung and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Reconstitution of ADGRG2-Gs complex.

a, SDS-PAGE of samples of in vitro reconstituted apo-ADGRG2-AA-120-CT in complex with wild-type Gs heterotrimer, dominant negative Gs (DNGs) heterotrimer, or DNGs heterotrimer stabilized with Nb35 after Flag-tagged antibody pulling down. Full SDS-PAGE scans were shown. Experiments were repeated three times with similar results. b, ADGRG2-120-AA-CT (120-CT) showed higher expression yield in Sf9-insect cell expression system compared with wild-type ADGRG2. Full SDS-PAGE scans were shown. Experiments were repeated three times with similar results. c, Representative concentration dependent curve of the VPM-p15-induced cAMP accumulation in HEK293 cells overexpressing similar levels (as shown in Extended Data Fig. 1d) of wild-type ADGRG2 or ADGRG2-120-AA-CT using Glosensor assay. Values are the mean ± SEM of three independent experiments performed in triplicate (n = 3). d, ELISA experiments showing similar expression levels of wild-type ADGRG2 and ADGRG2-120-AA-CT on HEK293 cell surface when indicated amounts of plasmids were transiently transfected. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). Comparison between ADGRG2-WT and ADGRG2-AA-CT was determined by two-sided one-way ANOVA with Tukey’s test. ns, no statistical significance (P = 0.7149). e-f, Representative elution profile and SDS-PAGE of the size-exclusion chromatography peak of apo-ADGRG2-AA-120-CT-DNGs (apo-ADGRG2-FL-DNGs) (e) or DHEA-ADGRG2-βT-120-CT-DNGs (DHEA-ADGRG2-βT-DNGs) (f) complex. Full SDS-PAGE scans were shown. Experiments were repeated three times with similar results. g, Two-dimensional class averages of negative staining sample showing distinct N-terminal structural features of the apo-ADGRG2-FL-DNGs complex. Scale bar: 10 nm. h-j, Representative cryo-EM micrograph (scale bar: 20 nm) and 2D class averages (scale bar: 50 nm) of the apo-ADGRG2-FL-DNGs (h), DHEA-ADGRG2-βT-DNGs (i), and DHEA-ADGRG2-AA-120-CT-mini-Gs (DHEA-ADGRG2-FL-mini-Gs) (j) complexes. The 2D class average for apo-ADGRG2-FL-DNGs complex was shown in Fig. 1b. Representative Cryo-EM micrographs from 9,997 movies (h) or 5,222 movies (i) or 4,780 movies (j) and representative two-dimensional class averages determined using approximately 3.46 million (i) or 0.66 million (j) particles from 2D classification were shown. k-m, Schematic flow chart of the image processing steps for the apo-ADGRG2-FL-DNGs (k), DHEA-ADGRG2-βT-DNGs (l), and DHEA-ADGRG2-FL-mini-Gs (m) complexes.

Source data

Extended Data Fig. 2 Local resolution of cryo-EM map and biochemical validation of the apo-ADGRG2-FL-DNGs, DHEA-ADGRG2-βT-DNGs, and DHEA-ADGRG2-FL-mini-Gs complex structures.

a-c, Local resolution map of the final 3D reconstruction of the apo-ADGRG2-FL-DNGs (a), DHEA-ADGRG2-βT-DNGs (b), and DHEA-ADGRG2-FL-mini-Gs (c) complexes estimated using RESMAP. d-f, Gold-standard Fourier shell correlation (FSC) and cross-validation of model to cryo-EM density maps corresponding to apo-ADGRG2- DNGs (d), DHEA-ADGRG2-βT-DNGs (e), or DHEA-ADGRG2- FL-mini-Gs (f) complexes, respectively. g-h, Cryo-EM density maps and the models of apo-ADGRG2-FL -DNGs (g) and DHEA-ADGRG2-FL- mini-Gs (h) complexes are shown for all transmembrane helices, helix 8, DHEA (h), and α-helix 5 of Gαs. Notably, the EM density between the I755 and P762 could not be defined. The density map of DHEA-ADGRG2-βT-DNGs complexes is shown in Supplementary Fig. 2.

Extended Data Fig. 3 Detailed descriptions of ECL2 region of ADGRG2.

a, Cryo-EM density maps of ECL2 region in apo-ADGRG2-FL DNGs, DHEA-ADGRG2-FL- mini-Gs and DHEA-ADGRG2-βT-DNGs complex structures, respectively. b, Structural presentation of the ECL2 of ADGRG2 in apo-ADGRG2- FLDNGs, DHEA-ADGRG2-FL- mini-Gs and DHEA-ADGRG2-βT-DNGs complex structures, respectively.

Extended Data Fig. 4 Key structural elements in TM6 and TM7 of ADGRG2.

a-b, Structural comparison of DNGs-coupled apo-ADGRG2 (slate) with Gs-coupled β2AR (PDB: 3SN6, gray) at the “PIF motif” region (a). Note that ADGRG2 does not contain similar residues of the triad I3.40aP5.50aF6.44a motif in Class A GPCR subfamily, and the equivalent residues at the I3.40aP5.50aF6.44a motif of ADGRG2 were separated from each other without tight packing (b). c, Cryo-EM densities of the conserved “upper quaternary core (UQC)” of ADGRG2 consisting of F7013.44, F7865.43, M7043.47 and W8386.53. d, Cryo-EM densities of the key residues stabilizing the TM6-TM7 kinks in apo-ADGRG2-DNGs complex. e, Sequence alignment of residues forming the TM6-TM7 kinks in ADGRG subfamily members and selected receptors from other adhesion GPCR subfamilies. The conserved residues are highlighted with colors. f, ELISA experiments showing similar expression levels of wild-type (WT) ADGRG2 and its mutants on HEK293 cell surface when indicated amounts of plasmids were transiently transfected. Related to Extended Data Fig. 4g. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). Comparison between wild-type ADGRG2 and its mutants was determined by two-sided one-way ANOVA with Tukey’s test. n.s., no statistical significance. (P = 0.5819, 0.5777, 0.4031 from left to right) g, Representative dose response curves of VPM-p15 induced cAMP accumulation in HEK293 cells over-expressing wild-type (WT) ADGRG2 or its mutants. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). h, Structural comparison of the TM6 (upper) and TM7 (lower) helices of Apo-ADGRG2-DNGs (slate) with those of ADGRF1 (PDB: 7WU3, pink), ADGRD1 (PDB: 7EPT, yellow-orange), ADGRG5 (PDB: 7EQ1, sky-blue), ADGRG1 (PDB: 7SF8, pale-cyan), ADGRL3 (PDB: 7SF7, gray), ADGRG4 (PDB: 7WUJ, pale-green) and ADGRG2-β (PDB: 7WUQ, yellow) (from left to right).

Source data

Extended Data Fig. 5 Identification of DHEA as the agonist of ADGRG2.

a, Vertical cross section of the putative ligand binding pocket in apo-ADGRG2 (left panel). The net-shaped deep layer of the pocket is outlined, and the surrounding residues are indicated (right panel). b, EM densities of the putative ligand binding pocket residues of DNGs-coupled apo ADGRG2. Data are correlated to a. c, Comparison between pocket residues in structures of DNGs-coupled apo ADGRG2 and Go-coupled ADGRG3-Cortisol (PDB: 7D77). Apo ADGRG2 was depicted in light blue and ADGRG3 was depicted in yellow. d-l, Representative dose response curves of the steroid ligands induced cAMP accumulation in HEK293 cells over-expressing wild-type mADGRG2 or hADGRG2 using Glosensor assay. Note that the 0.1% DMSO was used as the control. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). m, ELISA experiments showing similar expression levels of mADGRG2 and human GPR126 on HEK293 cell surface when indicated amounts of plasmids were transiently transfected. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). ns, no statistical significance. Comparison between ADGRG2 and GPR126 was determined by two-sided one-way ANOVA with Tukey’s test. (P = 0.6177) n-o, Representative dose response curves of VPM-p15, DHEA or DHEAS induced cAMP accumulation (n) or PrPc induced cAMP accumulation (o) in HEK293 cells over-expressing mADGRG2 or GPR126. The transiently transfected with indicated amount of mADGRG2 and GPR126 plasmid were shown in m. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). p, Bar graph presentation of the efficacies of VPM-p15, DHEA or DHEAS toward mADGRG2 and PrPc toward GPR126. The efficacies generated from data shown in n and o are normalized and presented as the response percentage for VPM-p15. The similar expression levels of mADGRG2 and GPR126 (ADGRG6) were validated by ELISA (as shown in m). Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). ***P < 0.001; **P < 0.01, DHEA or DHEAS-induced ADGRG2 activation or PrPc-induced GPR126 activation compared with VPM-p15-inducedADGRG2 activation using two-sided one-way ANOVA with Tukey’s test (P = 0.0004, 0.001, 0.0003 from left to right).

Source data

Extended Data Fig. 6 Screening of steroid hormone for ADGRG2-Gq coupling and the ligand induced-conformational changes within ADGRG2 extracellular domains.

a, Heatmap represents the Gq signaling activation induced by steroid ligands (100 μM) or VPM-p15 (100 μM) in mADGRG2- (left panel) or hADGRG2- (right panel) -overexpressing HEK293 cells. The 0.1% DMSO was used as the control, VPM-p15 was applied as a positive control. b, Concentration dependent Gq activation measured by G trimer dissociation BRET assay in HEK293 cells overexpressing ATIR in response to AII stimulation. Representative curves from three independent experiments were shown (n = 3). c, Concentration dependent Gq activation measured by G trimer dissociation BRET assay in HEK293 cells overexpressing mADGRG2-FL in response to DHEA or VPM-p15 stimulation. Representative curves from three independent experiments were shown (n = 3). d, Schematic representation of the FlAsH-BRET assay design. NanoLuc (Nluc) was inserted at the N-terminus of ADGRG2-β, and the FlAsH motif (CCPGCC) was incorporated in the designated positions at the extracellular loops (ECLs) of the receptor. e, Detailed description of the FlAsH motif incorporation sites at the ECLs of ADGRG2 according to 3-dimensional structure shown in b. f, ELISA experiments showing similar expression levels of the wild-type ADGRG2 and six FlAsH-BRET sensors. Data are normalized to the expression level of the wild-type ADGRG2. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). n.s., no statistical significance; Comparison between wild-type ADGRG2 and six FlAsH-BRET sensors was determined by two-sided one-way ANOVA with Tukey’s test (P = 0.9776, 0.821, 0.5814, 0.3245, 0.2459, 0.9261 from left to right). g-h, Representative dose response curve of six ADGRG2 FlAsH-BRET sensors in response to DHEA (e) or DOC (f) stimulation. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). i-j, The maximal response of ADGRG2 FlAsH-BRET sensors upon DHEA (g) or DOC (h) stimulation. Data are derived from the dose-response curves in e and f. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). **P < 0.01; ***P < 0.001; ND, not detected; Comparison between FlAsH-BRET sensors stimulated with DHEA or DOC and those stimulated with control vehicle. were determined by two-sided one-way ANOVA with Tukey’s test (g: P = 0.0004, <0.0001; h: P = 0.0002, 0.0015, 0.0013, 0.0006 from left to right).

Source data

Extended Data Fig. 7 Interaction modes of DHEA with ADGRG2.

a, Schematic diagrams showing the sequence features of the ADGRG2 constructs used for the reconstitution of DHEA-ADGRG2-DNGs complex. The N-terminal Stachel sequence truncated ADGRG2-β subunit construct (ADGRG2-βT) was used. The C-tail was replaced with that of GPR120. b, Representative concentration dependent curve of the DHEA-induced cAMP accumulation in HEK293 cells overexpressing ADGRG2-βT or control (pcDNA3.1) using Glosensor assay. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). c-d, Orthogonal views of and structural models for the DHEA-ADGRG2-βT-120CT-DNGs-Nb35 (c) and DHEA-ADGRG2-AA-120CT-miniGs-Nb35 (d) complexes. The DHEA densities were highlighted with red dashed squares. e, Fitting of the DHEA (Right panel) or CHS (Left panel) into the EM density of the ligand binding pocket in the DHEA-ADGRG2-βT-DNGs complex structure. The DHEA can be fit into the EM density while CHS cannot. f, Representative concentration dependent curve of the DHEA-induced cAMP accumulation in ADGRG2-βT-overexpressed HEK293 cells in the absence or presence of 100 μM CHS. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). g, Diagram of DHEA interactions in the ligand binding pocket of ADGRG2. Hydrogen bonds are depicted as red dashed lines. h, Assignment of DHEA (yellow) in the ligand binding pocket of ADGRG2 (green). DHEA could be fit into the density in two conformational states. In the ‘upper’ state, DHEA forms a hydrogen bond (red dashed line) with a water molecule via its 3-hydroxyl group. i, Superposition of the structures of DHEA-ADGRG2-FL and DHEA-ADGRG2-βT complexes indicate that DHEA adopt a similar pose in the orthosteric ligand pocket of ADGRG2. For clarity, only the vertical cross section of ADGRG2-FL was shown. DHEA in DHEA-ADGRG2-FL is pink while in DHEA-ADGRG2-βT is yellow. j, Clear EM densities enabled unambiguous assignment of DHEA, a water molecule, and surrounding residues in the ligand binding pocket of ADGRG2-FL. The hydrogen bond formed between DHEA and water molecule is shown in red dashed line.

Source data

Extended Data Fig. 8 Effects of mutations of putative binding pocket residues of ADGRG2 on DHEA binding.

a-c, Representative dose response curves of DHEA induced cAMP accumulation in HEK293 cells over-expressing wild-type ADGRG2 or its mutants using Glosensor assay. The dashed lines depicting data of wild-type ADGRG2 in (a) are represented again in (b) and (c) for comparison. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). The similar expression levels of wild-type receptors and mutants were validated by ELISA (as shown in Supplementary Fig. 5a-b). d and h, The efficacy changes of DHEA-induced cAMP accumulation forwild-type and mutant ADGRG2 (d) and the maximal response of ADGRG2 FlAsH-BRET sensor S3 (annotated as WT) and the sensor-based alanine mutants to DHEA stimulation (h). Data are correlated to a-c and e-g. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). **P < 0.01; *P < 0.05; n.s., no statistical significance; ND, not detected; Comparison between wild-type ADGRG2 (d) or FlAsH-BRET sensor S3 (h) and their mutants were determined by two-sided one-way ANOVA with Tukey’s test (d: P = 0.0139, 0.0101, 0.0088, 0.0416, 0.4114, 0.8433, 0.2144, 0.0061; h: P = 0.0195, 0.1243, 0.0073, 0.0846, 0.0327 from left to right). e-g, Representative concentration dependent curve of ADGRG2 FlAsH-BRET sensor S3 (annotated as WT) and the sensor-based alanine mutants in response to DHEA stimulation. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). The similar expression levels of wild-type receptors and mutants were validated by ELISA (as shown in Supplementary Fig. 5c). i, Representative dose response curves of VPM-p15 induced cAMP accumulation in HEK293 cells over-expressing wild-type ADGRG2 or its mutants using Glosensor assay. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). j, The effect of mutations on the efficacy of VPM-p15-induced cAMP accumulation. The bar graph was generated from data shown in i. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). **P < 0.01; ***P < 0.01; ND, not detected; Comparison between wild-type ADGRG2 and its mutants was determined by two-sided one-way ANOVA with Tukey’s test (P = 0.0009, 0.0012 from top to bottom).

Source data

Extended Data Fig. 9 Comparison of ligand binding pocket between DHEA-ADGRG2-FL and Cortisol-ADGRG3.

a, Comparison of ligand binding pocket between DHEA-ADGRG2-FL and Cortisol-ADGRG3 (PDB: 7D77). Compared with Cortisol, the DHEA has a 30˚ tilting toward TM5-TM6. The angles for DHEA-ADGRG2-FL TM6-TM7 kinks were labeled by two-way arrows in red. b, Structural representation of three conserved large hydrophobic residues forming direct contacts with ligands shared by both DHEA-ADGRG2-FL and Cortisol-ADGRG3. c, Sequence alignment of conserved pocket residues involved in ligand recognition in DHEA-coupled ADGRG2-FL and Cortisol-bound ADGRG3.

Extended Data Fig. 10 Effects of mutations of putative binding pocket residues of ADGRG2 on DOC binding.

a, RMSD analysis of DOC-ADGRG2 during 200-ns molecular dynamics simulation trajectories by GROMACS. RMSDs of DOC (lower panel) or DOC binding pocket residues (upper panel) are shown. The initial modeled complex state after equilibration (0 ns) was used for calculation. b, Binding mode of DOC in ADGRG2 according to computational simulation. c, Diagram of DOC interactions in the ligand binding pocket of ADGRG2. Hydrogen bonds are depicted as red dashed lines. d-f, Representative dose response curve of ADGRG2 FlAsH-BRET sensor S3 (annotated as WT) and the sensor-based alanine mutants in response to DOC stimulation. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). The expression levels of ADGRG2 mutants were normalized to equivalent levels of their wild-type (WT) receptors by adjusting the amount of plasmid transfected according to ELISA assay data shown in Supplementary Fig. 5c. g, The maximal response of ADGRG2 FlAsH-BRET sensor S3 (annotated as WT) and the sensor-based alanine mutants to DOC stimulation. Data are derived from the dose-response curves in Extended Data Fig. 10d-f and are normalized to the maximal response of the sensor S3. Values are mean ± SEM from three independent experiments performed in triplicates (n = 3). *P < 0.05; n.s., no significant difference; ND, not detected; FlAsH-BRET sensor S3 based alanine mutants were compared to the sensor S3. All data were analyzed by two-sided one-way ANOVA with Tukey’s test (P = 0.0673, 0.0220, 0.2403, 0.3698, 0.1591, 0.2406, 0.7043, 0.1264, 0.2, 0.8616 from left to right).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–5 and Tables 1–11.

Reporting Summary.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor 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

Lin, H., Xiao, P., Bu, RQ. et al. Structures of the ADGRG2–Gs complex in apo and ligand-bound forms. Nat Chem Biol 18, 1196–1203 (2022). https://doi.org/10.1038/s41589-022-01084-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01084-6

This article is cited by

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