Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Structural basis of sphingosine-1-phosphate receptor 1 activation and biased agonism

An Author Correction to this article was published on 10 January 2022

This article has been updated

Abstract

Sphingosine-1-phosphate receptor 1 (S1PR1) is a master regulator of lymphocyte egress from the lymph node and an established drug target for multiple sclerosis (MS). Mechanistically, therapeutic S1PR1 modulators activate the receptor yet induce sustained internalization through a potent association with β-arrestin. However, a structural basis of biased agonism remains elusive. Here, we report the cryo-electron microscopy (cryo-EM) structures of Gi-bound S1PR1 in complex with S1P, fingolimod-phosphate (FTY720-P) and siponimod (BAF312). In combination with functional assays and molecular dynamics (MD) studies, we reveal that the β-arrestin-biased ligands direct a distinct activation path in S1PR1 through the extensive interplay between the PIF and the NPxxY motifs. Specifically, the intermediate flipping of W2696.48 and the retained interaction between F2656.44 and N3077.49 are the key features of the β-arrestin bias. We further identify ligand–receptor interactions accounting for the S1PR subtype specificity of BAF312. These structural insights provide a rational basis for designing novel signaling-biased S1PR modulators.

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 the S1PR1–Gi complex.
Fig. 2: Ligand-binding pocket of S1PR1.
Fig. 3: Comparison between inactive and active S1PR1 structures.
Fig. 4: MD simulation study of ligand-bound S1PR1.
Fig. 5: S1PR subtype specificity of BAF312.
Fig. 6: A model of S1PR1 activation and Gi- versus β-arrestin-biased mechanisms triggered by the flip of W2696.48.

Similar content being viewed by others

Data availability

All data produced or analyzed in this study are included in the main text or the Supplementary Materials. A reporting summary for this article is available as a Supplementary Information file. The cryo-EM density maps and atomic coordinates have been deposited in the Electron Microscopy Data Bank (EMDB) and PDB under accession numbers EMD-32461 and 7WF7 for the S1P-bound S1PR1–Gi complex, EMD-31225 and 7EO2 for the FTY720-P-bound S1PR1–Gi complex and EMD-31226 and 7EO4 for the BAF312-bound S1PR1–Gi complex. All other data relating to this study are available from the corresponding author on reasonable request. All other published PDB codes cited in this paper are 3V2Y and 6VMS. Source data are provided with this paper.

Change history

References

  1. Cartier, A. & Hla, T. Sphingosine 1-phosphate: lipid signaling in pathology and therapy. Science 366, eaar5551 (2019).

  2. Spiegel, S. Sphingosine-1-phosphate: from insipid lipid to a key regulator. J. Biol. Chem. 295, 3371–3384 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bryan, A. M. & Del Poeta, M. Sphingosine-1-phosphate receptors and innate immunity. Cell Microbiol. 20, e12836 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Brinkmann, V. et al. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat. Rev. Drug Discov. 9, 883–897 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Chun, J., Kihara, Y., Jonnalagadda, D. & Blaho, V. A. Fingolimod: lessons learned and new opportunities for treating multiple sclerosis and other disorders. Annu. Rev. Pharmacol. Toxicol. 59, 149–170 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kawahara, A. et al. The sphingolipid transporter Spns2 functions in migration of zebrafish myocardial precursors. Science 323, 524–527 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Vu, T. M. et al. Mfsd2b is essential for the sphingosine-1-phosphate export in erythrocytes and platelets. Nature 550, 524–528 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Oo, M. L. et al. Immunosuppressive and anti-angiogenic sphingosine 1-phosphate receptor-1 agonists induce ubiquitinylation and proteasomal degradation of the receptor. J. Biol. Chem. 282, 9082–9089 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Sykes, D. A. et al. Investigating the molecular mechanisms through which FTY720-P causes persistent S1P1 receptor internalization. Br. J. Pharmacol. 171, 4797–4807 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Murakami, A. et al. Sphingosine 1-phosphate (S1P) regulates vascular contraction via S1P3 receptor: investigation based on a new S1P3 receptor antagonist. Mol. Pharmacol. 77, 704–713 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Scott, L. J. Siponimod: a review in secondary progressive multiple sclerosis. CNS Drugs 34, 1191–1200 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. Olsen, R. H. J. et al. TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome. Nat. Chem. Biol. 16, 841–84 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Duan, J. et al. Cryo-EM structure of an activated VIP1 receptor–G protein complex revealed by a NanoBiT tethering strategy. Nat. Commun. 11, 4121 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Punjani, A. & Fleet, D. J. 3D variability analysis: resolving continuous flexibility and discrete heterogeneity from single particle cryo-EM. J. Struct. Biol. 213, 107702 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Picard, L. P., Schonegge, A. M. & Bouvier, M. Structural insight into G protein-coupled receptor signaling efficacy and bias between Gs and β-arrestin. ACS Pharmacol. Transl. Sci. 2, 148–154 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Okude, J. et al. Identification of a conformational equilibrium that determines the efficacy and functional selectivity of the μ-opioid receptor. Angew. Chem. Int. Ed. Engl. 54, 15771–15776 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wingler, L. M. et al. Angiotensin analogs with divergent bias stabilize distinct receptor conformations. Cell 176, 468–478 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Suomivuori, C. M. et al. Molecular mechanism of biased signaling in a prototypical G protein-coupled receptor. Science 367, 881–887 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Violin, J. D., Crombie, A. L., Soergel, D. G. & Lark, M. W. Biased ligands at G-protein-coupled receptors: promise and progress. Trends Pharmacol. Sci. 35, 308–316 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Wingler, L. M., McMahon, C., Staus, D. P., Lefkowitz, R. J. & Kruse, A. C. Distinctive activation mechanism for angiotensin receptor revealed by a synthetic nanobody. Cell 176, 479–490 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wingler, L. M. et al. Angiotensin and biased analogs induce structurally distinct active conformations within a GPCR. Science 367, 888–892 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lagane, B. et al. Mutation of the DRY motif reveals different structural requirements for the CC chemokine receptor 5-mediated signaling and receptor endocytosis. Mol. Pharmacol. 67, 1966–1976 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Barak, L. S., Oakley, R. H., Laporte, S. A. & Caron, M. G. Constitutive arrestin-mediated desensitization of a human vasopressin receptor mutant associated with nephrogenic diabetes insipidus. Proc. Natl Acad. Sci. USA 98, 93–98 (2001).

    Article  CAS  PubMed  Google Scholar 

  26. Nakajima, K. & Wess, J. Design and functional characterization of a novel, arrestin-biased designer G protein-coupled receptor. Mol. Pharmacol. 82, 575–582 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bouley, R. et al. Functional role of the NPXXY motif in internalization of the type 2 vasopressin receptor in LLC-PK1 cells. Am. J. Physiol. Cell Physiol. 285, C750–C762 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Hunyady, L., Bor, M., Baukal, A. J., Balla, T. & Catt, K. J. A conserved NPLFY sequence contributes to agonist binding and signal transduction but is not an internalization signal for the type 1 angiotensin II receptor. J. Biol. Chem. 270, 16602–16609 (1995).

    Article  CAS  PubMed  Google Scholar 

  29. Prioleau, C., Visiers, I., Ebersole, B. J., Weinstein, H. & Sealfon, S. C. Conserved helix 7 tyrosine acts as a multistate conformational switch in the 5HT2C receptor. Identification of a novel “locked-on” phenotype and double revertant mutations. J. Biol. Chem. 277, 36577–36584 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Yang, D. et al. G protein-coupled receptors: structure- and function-based drug discovery. Signal Transduct. Target. Ther. 6, 7 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhao, C. et al. Structural insights into sphingosine-1-phosphate recognition and ligand selectivity of S1PR3–Gi signaling complexes. Cell Res https://doi.org/10.1038/s41422-021-00567-w (2021)

  32. Yuan, Y. et al. Structures of signaling complexes of lipid receptors S1PR1 and S1PR5 reveal mechanisms of activation and drug recognition. Cell Res https://doi.org/10.1038/s41422-021-00566-x (2021).

  33. Xia, R. et al. Cryo-EM structure of the human histamine H1 receptor/Gq complex. Nat. Commun. 12, 2086 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. 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 

  36. Wagner, T. et al. SPHIRE-crYOLO is a fast and accurate fully automated particle picker for cryo-EM. Commun. Biol. 2, 218 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

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

  38. 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 

  39. Zivanov, J., Nakane, T. & Scheres, S. H. W. A Bayesian approach to beam-induced motion correction in cryo-EM single-particle analysis. IUCrJ 6, 5–17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Yin, J. et al. Structure of a D2 dopamine receptor–G-protein complex in a lipid membrane. Nature 584, 125–129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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 

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

    Article  PubMed  Google Scholar 

  44. Wang, R. Y. et al. Automated structure refinement of macromolecular assemblies from cryo-EM maps using Rosetta. eLife 5, e17219 (2016).

  45. 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 

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

    Article  CAS  PubMed  Google Scholar 

  47. Inoue, A. et al. Illuminating G-protein-coupling selectivity of GPCRs. Cell 177, 1933–1947 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  49. Fiser, A., Do, R. K. G. & Sali, A. Modeling of loops in protein structures. Protein Sci. 9, 1753–1773 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Best, R. B. et al. Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone φ, ψ and side-chain χ1 and χ2 dihedral angles. J. Chem. Theory Comput. 8, 3257–3273 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Vanommeslaeghe, K. et al. CHARMM general force field: a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J. Comput. Chem. 31, 671–690 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, H., Pluhackova, K., Jiang, Z. & Bockmann, R. A. Binding characteristics of sphingosine-1-phosphate to ApoM hints to assisted release mechanism via the ApoM calyx-opening. Sci. Rep. 6, 30655 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Berendsen, H. J. C., Vanderspoel, D. & Vandrunen, R. Gromacs—a message-passing parallel molecular-dynamics implementation. Comput. Phys. Commun. 91, 43–56 (1995).

    Article  CAS  Google Scholar 

  54. Hoover, W. G. Canonical dynamics—equilibrium phase-space distributions. Phys. Rev. A Gen. Phys. 31, 1695–1697 (1985).

    Article  CAS  PubMed  Google Scholar 

  55. Parrinello, M. & Rahman, A. Crystal structure and pair potentials: a molecular-dynamics study. Phys. Rev. Lett. 45, 4 (1980).

    Article  Google Scholar 

  56. Darden, T., York, D. & Pedersen, L. Particle mesh Ewald: an Nlog(N) method for Ewald sums in large systems. J. Chem. Phys. 98, 3 (1993).

    Article  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Harris, C. R. et al. Array programming with NumPy. Nature 585, 357–362 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hunter, J. D. Matplotlib: a 2D graphics environment. Comput. Sci. Eng. 9, 90–95 (2007).

    Article  Google Scholar 

Download references

Acknowledgements

We thank the Cryo-EM facility of Harbin Institute of Technology for sample screening and data collection. We thank P. Lu at Westlake University for instruction on ligand docking. This work was supported by the Startup Funds of HIT Center for Life Sciences and the National Natural Science Foundation of China (32070048 to Y.H.). We thank H.E. Xu of Shanghai Institute of Materia Medica, Chinese Academy of Sciences, for the original LgBiT and pFastbac-2×MBP plasmids. We thank K. Sato, S. Nakano and A. Inoue at Tohoku University for their assistance in plasmid preparation and cell-based GPCR assays. This work was supported by KAKENHI 21H04791 (A.I.), 21H051130 (A.I.) and JPJSBP120213501 (A.I.) from the Japan Society for the Promotion of Science (JSPS); Grant-in-Aid for JSPS Fellows 19J11256 (K.K.); the PRIME JP19gm5910013 (A.I.), the LEAP JP20gm0010004 (A.I.) and the BINDS JP20am0101095 (A.I.) from the Japan Agency for Medical Research and Development (AMED); JST Moonshot Research and Development Program JPMJMS2023 (A.I.) from the Japan Science and Technology Agency (JST); Daiichi Sankyo Foundation of Life Science (A.I.); Takeda Science Foundation (A.I.); Ono Medical Research Foundation (A.I.) and The Uehara Memorial Foundation (A.I.).

Author information

Authors and Affiliations

Authors

Contributions

Y.H. initiated the project. Y.H. and A.I. designed the experiments, analyzed data and supervised the whole project. Z.X. made the expression constructs, purified the proteins, prepared and screened the grids, made the mutations and performed BRET2 assays. T.I. performed the MD simulation and analyzed data. K.K. and R.K. performed pilot studies for the NanoBiT GPCR signaling assays and edited the manuscript. Y.Q. and R.X. made the mutations and prepared the plasmids. M.-X.S. and X.-H.C. set up the BRET2 experiments. A.Z. and C.G. collected data with Y.H., and Y.H. solved the structures. Z.H. analyzed the data and edited the manuscript. Y.H., A.I. and T.I. wrote the manuscript. All authors contributed to data interpretation and preparation of the manuscript.

Corresponding authors

Correspondence to Asuka Inoue or Yuanzheng He.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Chemical Biology thanks Jesús Giraldo and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Sample preparation of S1PR1/Gi complexes.

a, Snake-shape diagram of the S1PR1-LgBiT construct used in the complex assembling. The diagram is adopted and modified from the GPCRdb (https://www.gpcrdb.org/protein/s1pr1_human/). The pink color highlights the position where the F1333.41W mutation is introduced. b, The BRET2 Gi dissociation assay to examine the F133W3.41 activity to S1P. Data are presented as mean values ± SEM, n = 3-4. c, The BRET2 Gi dissociation assay to examine the LgBiT-fused construct activity to S1P. Data are presented as mean values ± SEM, n = 3-4. d, Size exclusion column profile of S1PR1/Gi complex. e, Bias signaling activity of S1P, FTY720-P and BAF312. Gi dissociation, β-arrestin recruitment and β-arrestin endosomal translocation responses in the wild-type S1PR1 were tested by the NanoBiT-based assays. For the individual experiments performed in parallel, pharmacological parameters were normalized to S1P and presented as a logarithm of relative intrinsic activity (RAi; EC50/Emax value relative to that of S1P; Methods). Data are presented as mean values ± SEM, n = 6-7 (dots). Adjusted P values were calculated by the two-way ANOVA with the Dunnett’s post-hoc test.

Source data

Extended Data Fig. 2 Representative data process of S1PR1/Gi complexes.

A flow-chart of the cryo-EM data process of the S1PR1/Gi complex.

Extended Data Fig. 3 Local resolution of S1PR1/Gi complexes.

a, Local resolution analysis of the S1PR1/Gi complexes. b, FSC curve of the S1PR1/Gi complexes refined by the Non-uniform refinement in cryoSPARC, the resolution is assessed by model to map, FSC = 0.5.

Extended Data Fig. 4 Representative density map of S1PR1/Gi complexes and 3D variability analysis.

a-c, The cryo-EM density map of the representative regions of the S1PR1/Gi complexes. The density map is drawn by pymol at the contour level of 3.0. d, Representative image of the 3D variability analysis of the S1PR1 cryo-EM maps. The yellow arrow marks the position of ligand and the red arrow marks TM1.

Extended Data Fig. 5 Ligand binding pocket of S1PR1.

a, Cross section of a hydrophobicity analysis in the S1P-bound S1PR1 structure. b, An electrostatic potential surface analysis of the S1P-binding pocket. c, Mutant study to assess ligands response by the NanoBiT-Gi dissociation assay. Dashed lines in the mutant panels represent the wild-type (WT) S1PR1 response. Data are presented as mean values ± SEM, n = 3-5. Note that in many data points, error bars are smaller than the size of symbols and thus are not visible. For details of the ∆pEC50, Emax and error values, see Supplementary Table 2.

Extended Data Fig. 6 Mutant studies assessing ligand recognition.

a, Surface expression of the S1PR1 mutants analyzed by the flow cytometry. b, c, Pharmacological parameters for the Gi-coupling activity analyzed by the NanoBiT-Gi dissociation assay. For the individual experiments performed in parallel, data were normalized to the wild-type (WT) S1PR1 and presented as surface expression level (a), Emax (b) and ∆pEC50 (c). Data are presented as mean values ± SEM, n = 3-5 (dots). NA, parameter not available owing to a lack of ligand response. Mutants with lowered surface expression levels (E1213.29A, W2696.48A and E2947.36×35A) are shown as grey bars.

Source data

Extended Data Fig. 7 MD simulation study and mutant study assessing the ligand binding modes.

a, Key residues distance distributions with S1P (cyan), FTY720-P (green), BAF312 (purple), and ML056 (pink). Left panel, K34N-term and oxygens in the negatively-charged head groups (phosphate groups in S1P, FTY720-P, and ML056; carboxyl group in BAF312); middle panel, R1203.28 and the negatively-charged head groups; right panel, R1203.28 and N1012.60 or E1213.29. b, Representative snapshots of the key polar residues (K34N-term, N1012.60, R1203.28, and E1213.29) with the ligands during the simulations. Black dashed lines represent hydrogen bonds. c, The related position of ligands to W2696.48. FTY: FTY720-P, green color. BAF: BAF312, blue color. S1P, cyan color. d, The hydrophobic interaction between the lower part of the ligand binding pocket and the distal end of the benzyloxy oxime moiety of BAF312. e, Functional study for the F1253.33A mutation assessed by the BRET2 Gi dissociation assay. Data are presented as mean values ± SEM, n = 3-4. f, The outward displacement (arrow) of F2736.52 upon ligand binding.

Source data

Extended Data Fig. 8 MD simulation study assessing ligand-induced S1PR1 activation.

a, Distance distribution matrix of the key residues with S1P (cyan), FTY720-P (green), BAF312 (purple), and ML056 (pink). b, Interaction traces of three replicas (blue, red, and orange) with each ligand. Distance of the nearest non-hydrogen atom pairs is drawn with thick 2-ns moving average traces and thin unsmoothed traces (2-ps interval). c, Plots of averaged distances in Interactions 1-3 from the three replicas (the same color schemes as in b). Bars and error bars represent mean and SEM, respectively. Dotted lines indicate distances in the ML056-bound S1PR1 structure (PDB ID: 3V2Y). d, Fractions of contacts in Interactions 1-3 from the three replicas. Contact thresholds were set as the distances in the ML056-bound S1PR1. Statistical analyses were performed using the ordinary one-way ANOVA followed by the Dunnett’s post hoc test (c, d).

Source data

Extended Data Fig. 9 Key residues and ligand structures of the biased agonism.

a, Flow cytometry-based surface expression analysis for the mutants studied in the β-arrestin assays. The vertical dashed line separates mutants that are included in Fig. 4c (left) and Extended Data Fig. 8b (right). Note that the expression data of the W2696.48A mutant is identical to the one shown in Extended Data Fig. 5c. Data are presented as mean values ± SEM, n = 3-5 (dots). b, Mutant study to assess S1P response of the indicated mutants by the NanoBiT-Gi dissociation assay and the β-arrestin recruitment assay. Dashed lines and the long dashed dotted lines in the mutant panels represent the WT and the mock responses, respectively. Data are presented as mean values ± SEM, n = 3-4. Note that, in many data points, error bars are smaller than the size of symbols and thus are not visible. c, Bias profiles of etrasimod and S1P receptor agonist 1. Gi dissociation, β-arrestin recruitment and β-arrestin endosomal translocation responses in the wild-type S1PR1 were tested by the NanoBiT-based assays. For the individual experiments performed in parallel, data were normalized to S1P and presented as a logarithm of relative intrinsic activity (RAi; EC50/Emax value relative to that of S1P; Methods). Data are presented as mean values ± SEM, n = 3-4.

Source data

Extended Data Fig. 10 Comparison with published S1PR structures.

A comparison of S1P-, FTY720-P- and BAF312-bound S1PR1 with published S1P-bound S1PR3, FTY720-P-bound S1PR3 and BAF312-bound S1PR1, respectively. The dashed arrow marks the acyl-chain tail of the S1P molecule pointing the different directions.

Supplementary information

Supplementary Information

Supplementary Tables 1–3 and Fig. 1.

Reporting Summary

Supplementary Video 1

Supplementary Video 1. Molecule simulation of S1P-bound S1PR1.

Supplementary Video 2

Supplementary Video 2. Molecule simulation of FTY720-P-bound S1PR1.

Supplementary Video 3

Supplementary Video 3. 3DVa of S1P-, FTY720-P- and BAF312-bound S1PR1.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

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. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, Z., Ikuta, T., Kawakami, K. et al. Structural basis of sphingosine-1-phosphate receptor 1 activation and biased agonism. Nat Chem Biol 18, 281–288 (2022). https://doi.org/10.1038/s41589-021-00930-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00930-3

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research