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:

Insights into distinct signaling profiles of the µOR activated by diverse agonists

This article has been updated

Abstract

Drugs targeting the μ-opioid receptor (μOR) are the most effective analgesics available but are also associated with fatal respiratory depression through a pathway that remains unclear. Here we investigated the mechanistic basis of action of lofentanil (LFT) and mitragynine pseudoindoxyl (MP), two μOR agonists with different safety profiles. LFT, one of the most lethal opioids, and MP, a kratom plant derivative with reduced respiratory depression in animal studies, exhibited markedly different efficacy profiles for G protein subtype activation and β-arrestin recruitment. Cryo-EM structures of μOR-Gi1 complex with MP (2.5 Å) and LFT (3.2 Å) revealed that the two ligands engage distinct subpockets, and molecular dynamics simulations showed additional differences in the binding site that promote distinct active-state conformations on the intracellular side of the receptor where G proteins and β-arrestins bind. These observations highlight how drugs engaging different parts of the μOR orthosteric pocket can lead to distinct signaling outcomes.

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: Distinct ligands for μOR.
Fig. 2: Structures of μOR-Gi complex activated by MP and LFT.
Fig. 3: Semiconserved ligand interaction network for MP and LFT.
Fig. 4: Simulations of the μOR reveal distinct binding pocket conformations favored by MP, DAMGO and LFT.
Fig. 5: In simulations with the G protein removed, the μOR adopts two active intracellular conformations, with MP and LFT favoring different conformations.

Similar content being viewed by others

Data availability

The atomic coordinates for MP–μOR-Gi1 and LFT–μOR-Gi1-scFv complexes have been deposited in the Protein Data Bank with the accession codes 7T2G and 7T2H, respectively. The EM maps for MP–μOR-Gi1 and LFT–μOR-Gi1-scFv complexes have been deposited in EMDB with the accession codes EMD-25612 and EMD-25613, respectively. The composite non-model-based density-modified map for MP–µOR-Gi1 is deposited to the Electron Microscopy Data Bank (EMDB) as the main map and used for model building. The locally refined individual maps are deposited as additional maps.

Change history

  • 01 December 2022

    In the version of this article initially published, due to a processing error, additional traces appeared in Figure 4b, which has since been corrected in the HTML and PDF versions of the article.

References

  1. Masuho, I. et al. Distinct profiles of functional discrimination among G proteins determine the actions of G protein-coupled receptors. Sci. Signal. 8, ra123 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Lamberts, J. T., Jutkiewicz, E. M., Mortensen, R. M. & Traynor, J. R. mu-Opioid receptor coupling to Gαo plays an important role in opioid antinociception. Neuropsychopharmacology 36, 2041–2053 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Stanley, T. H. The history and development of the fentanyl series. J. Pain Symptom Manag. 7, S3–S7 (1992).

    Article  CAS  Google Scholar 

  4. Bot, G., Blake, A. D., Li, S. & Reisine, T. Fentanyl and its analogs desensitize the cloned mu opioid receptor. J. Pharmacol. Exp. Ther. 285, 1207–1218 (1998).

    CAS  PubMed  Google Scholar 

  5. Schmid, C. L. et al. Bias factor and therapeutic window correlate to predict safer opioid analgesics. Cell 171, 1165–1175 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chakraborty, S. et al. A novel mitragynine analog with low-efficacy mu opioid receptor agonism displays antinociception with attenuated adverse effects. J. Med. Chem. 64, 13873–13892 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Meert, T. F., Lu, H. R., van Craenndonck, H. & Janssen, P. A. Comparison between epidural fentanyl, sufentanil, carfentanil, lofentanil and alfentanil in the rat: analgesia and other in vivo effects. Eur. J. Anaesthesiol. 5, 313–321 (1988).

    CAS  PubMed  Google Scholar 

  8. Prozialeck, W. C., Jivan, J. K. & Andurkar, S. V. Pharmacology of kratom: an emerging botanical agent with stimulant, analgesic and opioid-like effects. J. Am. Osteopath. Assoc. 112, 792–799 (2012).

    PubMed  Google Scholar 

  9. Kruegel, A. C. et al. 7-Hydroxymitragynine is an active metabolite of mitragynine and a key mediator of its analgesic effects. ACS Cent. Sci. 5, 992–1001 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kamble, S. H. et al. Metabolism of a kratom alkaloid metabolite in human plasma increases its opioid potency and efficacy. ACS Pharmacol. Transl. Sci. 3, 1063–1068 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chakraborty, S. et al. Oxidative Metabolism as a Modulator of Kratom’s Biological Actions. J. Med. Chem. 64, 16553–16572. https://doi.org/10.1021/acs.jmedchem.1c01111(2021).

  12. Varadi, A. et al. Mitragynine/corynantheidine pseudoindoxyls as opioid analgesics with mu agonism and delta antagonism, which do not recruit β-arrestin-2. J. Med. Chem. 59, 8381–8397 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Conibear, A. E. & Kelly, E. A biased view of μ-opioid receptors? Mol. Pharmacol. 96, 542–549 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bohn, L. M., Gainetdinov, R. R., Lin, F. T., Lefkowitz, R. J. & Caron, M. G. μ-Opioid receptor desensitization by β-arrestin-2 determines morphine tolerance but not dependence. Nature 408, 720–723 (2000).

    Article  CAS  PubMed  Google Scholar 

  15. Groer, C. E. et al. An opioid agonist that does not induce μ-opioid receptor–arrestin interactions or receptor internalization. Mol. Pharmacol. 71, 549–557 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. DeWire, S. M. et al. A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. J. Pharmacol. Exp. Ther. 344, 708–717 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Manglik, A. et al. Structure-based discovery of opioid analgesics with reduced side effects. Nature 537, 185–190 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gillis, A. et al. Low intrinsic efficacy for G protein activation can explain the improved side effect profiles of new opioid agonists. Sci. Signal. https://doi.org/10.1126/scisignal.aaz3140 (2020).

  19. Bachmutsky, I., Wei, X. P., Durand, A. & Yackle, K. β-arrestin 2 germline knockout does not attenuate opioid respiratory depression. eLife 10, e62552 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kliewer, A. et al. Phosphorylation-deficient G-protein-biased μ-opioid receptors improve analgesia and diminish tolerance but worsen opioid side effects. Nat. Commun. 10, 1–11 (2019).

    Article  Google Scholar 

  21. He, L. et al. Pharmacological and genetic manipulations at the µ-opioid receptor reveal arrestin-3 engagement limits analgesic tolerance and does not exacerbate respiratory depression in mice. Neuropsychopharmacology https://doi.org/10.1038/s41386-021-01054-x (2021).

  22. Raffa, R. B., Martinez, R. P. & Connelly, C. D. G-protein antisense oligodeoxyribonucleotides and μ-opioid supraspinal antinociception. Eur. J. Pharmacol. 258, R5–R7 (1994).

    Article  CAS  PubMed  Google Scholar 

  23. Leck, K. J. et al. Deletion of guanine nucleotide binding protein αz subunit in mice induces a gene dose dependent tolerance to morphine. Neuropharmacology 46, 836–846 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhou, Y. et al. Predicted mode of binding to and allosteric modulation of the μ-opioid receptor by kratom’s alkaloids with reported antinociception in vivo. Biochemistry 60, 1420–1429 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Robertson, M. J., van Zundert, G. C., Borrelli, K. & Skiniotis, G. GemSpot: a pipeline for robust modeling of ligands into cryo-EM maps. Structure 28, 707–716 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ballesteros, J. A. & Weinstein, H. Integrated methods for the construction of three-dimensional models and computational probing of structure-function relations in G protein-coupled receptors. in Methods in Neurosciences, Vol. 25 (Elsevier, 1995).

  29. Huang, W. et al. Structural insights into µ-opioid receptor activation. Nature 524, 315–321 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Manglik, A. et al. Crystal structure of the μ-opioid receptor bound to a morphinan antagonist. Nature 485, 321–326 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Friesner, R. A. et al. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein−ligand complexes. J. Med. Chem. 49, 6177–6196 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Mahinthichaichan, P., Vo, Q. N., Ellis, C. R. & Shen, J. Kinetics and mechanism of fentanyl dissociation from the μ-opioid receptor. JACS Au 1, 2208–2215 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wingler, L. M. & Lefkowitz, R. J. Conformational basis of G protein-coupled receptor signaling versatility. Trends Cell Biol. 30, 736–747 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. Grim, T. W., Acevedo-Canabal, A. & Bohn, L. M. Toward directing opioid receptor signaling to refine opioid therapeutics. Biol. Psychiatry 87, 15–21 (2020).

    Article  CAS  PubMed  Google Scholar 

  36. Faouzi, A., Varga, B. R. & Majumdar, S. Biased opioid ligands. Molecules https://doi.org/10.3390/molecules25184257 (2020).

  37. 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 

  38. 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).

  39. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jacobson, M. P., Friesner, R. A., Xiang, Z. & Honig, B. On the role of the crystal environment in determining protein side-chain conformations. J. Mol. Biol. 320, 597–608 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Vanommeslaeghe, K. et al. CHARMM General Force Field (CGenFF): 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 

  47. Ghanouni, P. et al. The effect of pH on β2 adrenoceptor function: evidence for protonation-dependent activation. J. Biol. Chem. 275, 3121–3127 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Mahalingam, M., Martínez-Mayorga, K., Brown, M. F. & Vogel, R. Two protonation switches control rhodopsin activation in membranes. Proc. Natl Acad. Sci. USA 105, 17795–17800 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Betz, R. Dabble https://doi.org/10.5281/zenodo.836914 (2017).

  50. J, H. et al. CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14, 71–73 (2016).

    Google Scholar 

  51. Case, D. A. et al. Amber 2020 (University of California, 2020).

  52. Lee, T. S. et al. GPU-accelerated molecular dynamics and free energy methods in Amber18: performance enhancements and new features. J. Chem. Inf. Model. 58, 2043–2050 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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  CAS  Google Scholar 

  54. Roe, D. R. & Cheatham, T. E. PTRAJ and CPPTRAJ: software for processing and analysis of molecular dynamics trajectory data. J. Chem. Theory Comput. 9, 3084–3095 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996).

    Article  CAS  PubMed  Google Scholar 

  56. Venkatakrishnan, A. J. et al. Uncovering patterns of atomic interactions in static and dynamic structures of proteins. Preprint at bioRxiv https://doi.org/10.1101/840694 (2019).

  57. Lyman, E. & Zuckerman, D. M. Ensemble-based convergence analysis of biomolecular trajectories. Biophys. J. 91, 164–172 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kenakin, T., Watson, C., Muniz-Medina, V., Christopoulos, A. & Novick, S. A simple method for quantifying functional selectivity and agonist bias. ACS Chem. Neurosci. 3, 193–203 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Black, J. W. & Leff, P. Operational models of pharmacological agonism. Proc. R. Soc. Lond. B Biol. Sci. 220, 141–162 (1983).

    Article  CAS  PubMed  Google Scholar 

  60. Uprety, R. et al. Controlling opioid receptor functional selectivity by targeting distinct subpockets of the orthosteric site. eLife https://doi.org/10.7554/eLife.56519 (2021).

Download references

Acknowledgements

This work was supported by the Swiss National Science Foundation Early Postdoctoral Mobility grant no. P2ELP3_187989 (D.A.); the European Molecular Biology Organization Long-Term Fellowship grant no. ALTF 544-2019 (D.A.); a Stanford Graduate Fellowship (J.M.P.); the Human Frontier Science Program Long-Term Fellowship grant no. LT000916/2018-L (C.-M.S.); the National Institutes of Health grants no. R01GM127359 (R.O.D.), no. DA045884 (S.M.) and no. R37DA036246 (B.K.K. and G.S.); and the Mathers Foundation (G.S. and B.K.K.). B.K.K. is a Chan Zuckerberg Biohub Investigator. An award of computer time was provided by the INCITE program. This research used resources of the Oak Ridge Leadership Computing Facility, which is a US Department of Energy Office of Science User Facility supported under contract no. DE-AC05-00OR22725. A.I. was funded by grant nos. PRIME 19gm5910013, LEAP 19gm0010004 and BINDS JP20am0101095 from the Japan Agency for Medical Research and Development (AMED); grant nos. KAKENHI 21H04791 and 21H05113 from the Japan Society for the Promotion of Science (JSPS); and JST Moonshot Research and Development Program grant no. JPMJMS2023 from Japan Science and Technology Agency (JST). We thank F. M. N. Kadji, K. Sato, Y. Sugamura and A. Inoue at Tohoku University for plasmid construction and the cell-based GPCR assays; and Y. Laloudakis, S. Hollingsworth and N. Latorraca for helpful discussions.

Author information

Authors and Affiliations

Authors

Contributions

W.H. and H.W. prepared protein samples for structural studies and conducted biochemical assays. Q.Q. and A.B.S. collected and processed the cryo-EM data and generated the maps. Q.Q., M.J.R. and A.B.S. built and refined models. S.C. synthesized MP. T.C. and J.F.D. performed signaling profile assays under the supervision of B.L.R. A.I. performed NanoBiT experiments. D.A. and J.M.P. performed and analyzed molecular dynamics simulations, with input from C.-M.S., under the supervision of R.O.D. Q.Q., W.H., D.A., J.M.P., A.B.S., S.M., R.O.D., B.K.K. and G.S. interpreted the data and wrote the manuscript with inputs from all authors. B.K.K. and G.S. supervised the project.

Corresponding authors

Correspondence to Susruta Majumdar, Ron O. Dror, Brian K. Kobilka or Georgios Skiniotis.

Ethics declarations

Competing interests

G.S. is a cofounder of and consultant for Deep Apple Therapeutics. B.K.K. is a cofounder of and consultant for ConfometRx. S.M. is a cofounder of Sparian biosciences. S.M. has filed a provisional patent (US20220024923A1) on MP and related molecules.

Peer review

Peer review information

Nature Chemical Biology thanks the anonymous reviewers 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 Activity characterization of diverse ligands on μOR.

a, Efficacy of compounds LFT, MP and Morphine at human μOR of Gi1, GoA, GoB and Gz activation, and recruitment of β-arrestin-2 using the BRET assay are shown as a percentage of receptor activation relative to the full agonist, DAMGO.MP had significantly lower G-protein as well as arrestin among ligands tested (****p < 0.0001). Statistical significance was determined using one-way ANOVA followed by Dunnett’s multiple comparison test. Data represent mean + /- SEM from five independent experiments. b, Dose-dependent activation of Gi1 signaling in NanoBiT Gi1-dissociation assay and activation of β-arr1 and β-arr2 signaling in NanoBiT Arrestin-recruitment assay on wild type human μOR. Data are the means (+/- SEM) from four independent experiments and summarized at bottom panel. c, Efficacy of LFT, MP, and DAMGO at mouse μOR for Gi1, Gi3, GoA, GoB and Gz activation measured by GTPase-GloTM assay (n = 3 independent replicates, means + /-SEM are represented).

Extended Data Fig. 2 Cryo-EM process for LFT-μOR-Gi1-scFv and MP-μOR-Gi1 complexes.

Representative raw micrographs (a) and 2D classification averages (b) for LFT-μOR-Gi1-scFv and MP-μOR-Gi1, respectively. c, Workflow of cryo-EM data processing of LFT (left) and MP (right). d, Angular distribution of projections for cryo-EM maps and gold-standard FSC curves of half-maps (0.143 cutoff). e, Local resolution of LFT-μOR-Gi1-scFv cryo-EM map. f, Local resolution of composite cryo-EM maps of MP-μOR-Gi1 after local refinement and non-model based density modification (Phenix Resolve Cryo-EM).

Extended Data Fig. 3 Conserved μOR-Gi1 conformation activated by diverse agonists.

a, Alignment of MP (orange), LFT (red) and DAMGO (blue) bound μOR-Gi1 complexes onto BU72-bound μOR (grey), with nanobody and scFv removed for clarity. b, Close view of the ligand binding orthosteric pocket show nearly identical poses for residues involved in different agonists interaction, except that Q214 orients its side-chain towards TM3 upon MP engagement, which results in the loss of Q214-Y326 interaction observed among LFT, DAMGO and BU72. Orientation of GPCR activation feature motifs P5.50-I3.40-F6.44 (c), DR3.50Y and NP7.50xxY7.53 (d), and residues lined on the major interface between μOR intracellular site composed of TM2-3, TM5-7 and ICL1-3 (e) and C-terminal α5 helix of Gα subunit (f) are quite similar, suggesting a canonical conformation for Gi1 heterotrimer coupled μOR activated by DAMGO, LFT and MP.

Extended Data Fig. 4 Interaction network comparison among diverse μOR ligands.

a, Structurally distinct agonists such as morphinan BU72 (grey), enkephalin-like DAMGO (blue), synthetic LFT (magenta), novel alkaloid MP (green) and morphinan antagonist βFNA (black) all occupy the central pocket (cp) of wide orthosteric binding site. b, Viewed from extracellular side, functional moieties of the BU72, DAMGO and LFT penetrate into a sub-pocket between TM2 and TM3 (sp1), while MP indole ring explores a new arena composed of TM1, TM2 and TM7 (sp2). c, Schematic interaction diagrams highlight a conserved salt-bridge/hydrogen-bond interaction between μOR D147 (red sphere) and a tertiary amine (NH+) on BU72, DAMGO, LFT and MP, in addition to the major hydrophobic interaction network, calculated by Maestro (Schrödinger Release 2018-4: Maestro, Schrödinger, LLC, New York, NY, 2018).

Extended Data Fig. 5 Binding poses of fentanyl analogues revealed by docking and simulation.

a, Chemical structures of fentanyl and its derivatives, carfentanil and lofentanil. The 4-carbomethoxy moiety added to the piperidinyl group of fentanyl makes carfentanil over 100 times more potent at the μOR, while further addition of the 3-methyl group slightly enhances lofentanil in efficacy compared to carfentanil. b-e, Potential binding poses of carfentanil and fentanyl. The binding pose of LFT in the LFT-μOR cryo-EM structure (magenta sticks) is overlaid with (b) the docked pose of carfentanil (teal sticks), (c) the fentanyl pose modelled using the cryo-EM pose of LFT (pink sticks), (d) the first docked pose of fentanyl (orange stick), and (e) the second docked pose of fentanyl (yellow sticks). The first docked pose is in an orientation similar to that of LFT. The second docked pose is in an orientation opposite to that of LFT and has more favorable GlideScore and Emodel than the first. f, Simulations with fentanyl initiated in the poses of panels c, d, and e are shown in the first, second, and third columns, respectively (see Methods). For each simulation frame, the RMSD of fentanyl relative to the initial pose in that simulation is computed after alignment on the receptor. The first docked pose shifts to the pose shown in panel c in eight out of ten simulations.

Extended Data Fig. 6 Frequencies of key interactions in the binding pocket with different ligands bound.

a, Frequency of key inter-residue hydrogen bonds in simulations with various ligands bound (see Methods). b, Frequency of key protein-ligand interactions in simulations with various ligands bound (see Methods). Pi-pi interactions are aromatic stacking interactions between Y7.43 and the ligand. Lower and upper box boundaries indicate the 25th and 75th percentiles, respectively. The light gray line inside the box denotes the median. Lower and upper error lines represent 10th and 90th percentiles, respectively. Empty circles represent data points falling outside the 10th to 90th percentiles. Individual data points are frequencies in each independent simulation. Each boxplot is computed over six independent simulations with the same ligand bound (see Methods).

Extended Data Fig. 7 Intracellular TM7 rotation.

a, Intracellular TM7 rotation in Fig. 5d and Extended Data Fig. 11 was calculated as described in Methods. i and i + 1 denote consecutive residues. The view is from the extracellular side. The reference structure is represented in black, and the simulation frame is represented in orange. The computed angle is denoted by θ. b, The counterclockwise rotation in the intracellular portion of TM7 (when viewed from the extracellular side; see Fig. 5d) during the transition from the canonical active state to the alternative state leads to an inward shift as measured by a decrease in the P7.50–L2.46 distance. Relative to DAMGO, MP favors the canonical active state, whereas LFT favors the alternative state. Each distribution is computed over 6 independent simulations with the same ligand bound (see Methods).

Extended Data Fig. 8 Interaction between Q2.60 and Y7.43 increases the probability that the intracellular coupling site adopts the alternative state conformation.

a, The distribution of intracellular TM7 rotation when the Q2.60–Y7.43 interaction is formed (left panel) and when it is not (right panel). b, The distribution of P7.50–L2.46 distance when the Q2.60–Y7.43 interaction is formed (left panel) and when it is not (right panel). Distributions are computed across all 18 simulations (6 independent simulations with each of the 3 ligands bound; see Methods).

Extended Data Fig. 9 The transition from the canonical active conformation to the alternative conformation involves changes in an interaction network in the core of the receptor.

The transition from the canonical active conformation to the alternative conformation involves a shift in the hydrogen bonding network in the sodium binding pocket, as shown through representative frames from our μOR simulations (viewed from the extracellular side; see Methods). In the canonical active conformation, N861.50 forms a hydrogen bond with S3297.46, and D1142.50 forms a hydrogen bond with N3327.49. In the alternative conformation, these interactions are broken and replaced by D1142.50–S3297.46 and D1142.50–N1503.35 hydrogen bonds. The μOR cryo-EM structure (grey) is represented by the MP-µOR-Gi1 structure reported in this manuscript.

Extended Data Fig. 10 Comparison of the intracellular conformations observed for the μOR and the AT1R.

a, Simulations indicate that in both the μOR and the AT1R, the transition from the canonical active conformation to the alternative conformation involves a counterclockwise twist at TM7 (viewed from the extracellular side; bottom panels), leading to relaxation of the kink in the NPxxY region and the inward movement of P7.50 (viewed from the extracellular side; top panels). Both at the μOR and the AT1R, P7.50 is translated inward in the alternative conformation with respect to the canonical active conformation and the inactive conformation (top panels). b, The µOR alternative conformation differs from the AT1R alternative conformation in that the intracellular end of TM7 shows an inward displacement in μOR compared to the AT1R alternative conformation. The interaction between R3.50 and D8.47 does not allow the downward Y7.53 rotamer observed at the AT1R. The canonical active and alternative conformations shown here for the μOR are representative frames from our μOR simulations (see Methods). The canonical active and alternative conformations shown here for the AT1R are representative simulation frames (PDB files included in the Supplementary Material of Suomivuori, et al34. The inactive μOR and AT1R structures are the inactive μOR and AT1R crystal structures, respectively (PDB IDs: 4DKL and 4YAY).

Supplementary information

Supplementary Information

Supplementary Figs. 1–7 and Tables 1 and 2.

Reporting Summary

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qu, Q., Huang, W., Aydin, D. et al. Insights into distinct signaling profiles of the µOR activated by diverse agonists. Nat Chem Biol 19, 423–430 (2023). https://doi.org/10.1038/s41589-022-01208-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01208-y

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