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:

Recognition of methamphetamine and other amines by trace amine receptor TAAR1

Abstract

Trace amine-associated receptor 1 (TAAR1), the founding member of a nine-member family of trace amine receptors, is responsible for recognizing a range of biogenic amines in the brain, including the endogenous β-phenylethylamine (β-PEA)1 as well as methamphetamine2, an abused substance that has posed a severe threat to human health and society3. Given its unique physiological role in the brain, TAAR1 is also an emerging target for a range of neurological disorders including schizophrenia, depression and drug addiction2,4,5. Here we report structures of human TAAR1–G-protein complexes bound to methamphetamine and β-PEA as well as complexes bound to RO5256390, a TAAR1-selective agonist, and SEP-363856, a clinical-stage dual agonist for TAAR1 and serotonin receptor 5-HT1AR (refs. 6,7). Together with systematic mutagenesis and functional studies, the structures reveal the molecular basis of methamphetamine recognition and underlying mechanisms of ligand selectivity and polypharmacology between TAAR1 and other monoamine receptors. We identify a lid-like extracellular loop 2 helix/loop structure and a hydrogen-bonding network in the ligand-binding pockets, which may contribute to the ligand recognition in TAAR1. These findings shed light on the ligand recognition mode and activation mechanism for TAAR1 and should guide the development of next-generation therapeutics for drug addiction and various neurological disorders.

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: Functional assessment of four TAAR1 agonists in activating TAAR1 and overall structure of METH–TAAR1–Gs complex.
Fig. 2: METH-binding pocket of TAAR1.
Fig. 3: The recognition of β-PEA by TAAR1.
Fig. 4: Structural basis of SEP-363856 polypharmacology and selectivity.
Fig. 5: Conserved and divergent features of agonist recognition at TAAR1.

Similar content being viewed by others

Data availability

Density maps and structure coordinates have been deposited in the Electron Microscopy Data Bank (EMDB) and the Protein Data Bank (PDB) with accession codes EMD-37347 and 8W87 for METH–TAAR1–Gs complex; EMD-37348 and 8W88 for SEP-363856-bound TAAR1–Gs complex; and EMD-37349 and 8W89 for β-PEA-bound TAAR1–Gs complex; EMD-37350 and 8W8A for RO5256390–TAAR1–Gs complex; and EMD-37351 and 8W8B for SEP-363856 bounded 5-HT1AR–Gi protein complex. Source data are provided with this paper.

References

  1. Gainetdinov, R. R., Hoener, M. C. & Berry, M. D. Trace amines and their receptors. Pharmacol. Rev. 70, 549–620 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Liu, J., Wu, R. & Li, J.-X. TAAR1 and psychostimulant addiction. Cell. Mol. Neurobiol. 40, 229–238 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Most overdose deaths involve illicitly manufactured fentanyls. CDC www.cdc.gov/drugoverdose/featured-topics/overdose-deaths-data.html (2020).

  4. Berry, M. D., Gainetdinov, R. R., Hoener, M. C. & Shahid, M. Pharmacology of human trace amine-associated receptors: therapeutic opportunities and challenges. Pharmacol. Ther. 180, 161–180 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Dodd, S. et al. Trace amine-associated receptor 1 (TAAR1): a new drug target for psychiatry? Neurosci. Biobehavi. Rev. 120, 537–541 (2021).

    Article  CAS  Google Scholar 

  6. Heffernan, M. L. R. et al. Ulotaront: A TAAR1 agonist for the treatment of schizophrenia. ACS Med. Chem. Lett. 13, 92–98 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Nair, P. C. et al. Binding of SEP-363856 within TAAR1 and the 5HT1A receptor: implications for the design of novel antipsychotic drugs. Mol. Psychiatry 27, 88–94 (2022).

    Article  CAS  PubMed  Google Scholar 

  8. Schep, L. J., Slaughter, R. J. & Beasley, D. M. The clinical toxicology of metamfetamine. Clin. Toxicol. 48, 675–694 (2010).

    Article  CAS  Google Scholar 

  9. Jayanthi, S., Daiwile, A. P. & Cadet, J. L. Neurotoxicity of methamphetamine: main effects and mechanisms. Exp. Neurol. 344, 113795 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chan, B. et al. Pharmacotherapy for methamphetamine/amphetamine use disorder—a systematic review and meta-analysis. Addiction 114, 2122–2136 (2019).

    Article  PubMed  Google Scholar 

  11. Bisagno, V. & Cadet, J. L. in Handbook of Neurotoxicity (ed. Kostrzewa, R.M.) 563–585 (Springer, 2022).

  12. Grandy, D. K. in Neuropathology of Drug Addictions and Substance Misuse (ed. Preedy, V. R.) 108–116 (Academic Press, 2016).

  13. Miller, G. M. The emerging role of trace amine-associated receptor 1 in the functional regulation of monoamine transporters and dopaminergic activity. J. Neurochem. 116, 164–176 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Berry, M. D. Mammalian central nervous system trace amines. Pharmacologic amphetamines, physiologic neuromodulators. J. Neurochem. 90, 257–271 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Tonelli, M. & Cichero, E. Trace amine associated receptor 1 (TAAR1) modulators: a patent review (2010–present). Expert Opin.Ther. Pat. 30, 137–145 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Xu, Z. & Li, Q. TAAR agonists. Cell. Mol. Neurobiol. 40, 257–272 (2020).

    Article  CAS  PubMed  Google Scholar 

  17. Achat-Mendes, C., Lynch, L. J., Sullivan, K. A., Vallender, E. J. & Miller, G. M. Augmentation of methamphetamine-induced behaviors in transgenic mice lacking the trace amine-associated receptor 1. Pharmacol. Biochem. Behav. 101, 201–207 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Cotter, R. et al. The trace amine-associated receptor 1 modulates methamphetamine’s neurochemical and behavioral effects. Front. Neurosci. 9, 39 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Underhill, S. M. et al. Amphetamines signal through intracellular TAAR1 receptors coupled to Gα13 and GαS in discrete subcellular domains. Mol. Psychiatry 26, 1208–1223 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Eyun, S.-i, Moriyama, H., Hoffmann, F. G. & Moriyama, E. N. Molecular evolution and functional divergence of trace amine–associated receptors. PLoS ONE 11, e0151023 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Accorroni, A. & Zucchi, R. in Trace Amines and Neurological Disorders (eds Farooqui, T. & Farooqui, A. A.) 151–164 (Academic Press, 2016).

  22. Liberles, S. D. Trace amine-associated receptors: ligands, neural circuits, and behaviors. Curr. Opin. Neurobiol. 34, 1–7 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Alnefeesi, Y. et al. Trace amine-associated receptor 1 (TAAR1): potential application in mood disorders: a systematic review. Neurosci. Biobehav. Rev. 131, 192–210 (2021).

    Article  CAS  PubMed  Google Scholar 

  24. Lothar, L. et al. Trace amine-associated receptor 1 modulates dopaminergic activity. J. Pharmacol. Exp. Ther. 324, 948 (2008).

    Article  Google Scholar 

  25. Revel, F. G. et al. A new perspective for schizophrenia: TAAR1 agonists reveal antipsychotic- and antidepressant-like activity, improve cognition and control body weight. Mol. Psychiatry 18, 543–556 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Wu, R. & Li, J.-X. Potential of ligands for trace amine-associated receptor 1 (TAAR1) in the management of substance use disorders. CNS Drugs 35, 1239–1248 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Galley, G., Stalder, H., Goergler, A., Hoener, M. C. & Norcross, R. D. Optimisation of imidazole compounds as selective TAAR1 agonists: discovery of RO5073012. Bioorg. Med. Chem. Lett. 22, 5244–5248 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Galley, G. et al. Discovery and characterization of 2-aminooxazolines as highly potent, selective, and orally active TAAR1 agonists. ACS Med. Chem. Lett. 7, 192–197 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Raony, Í., Domith, I., Lourenco, M. V., Paes-de-Carvalho, R. & Pandolfo, P. Trace amine-associated receptor 1 modulates motor hyperactivity, cognition, and anxiety-like behavior in an animal model of ADHD. Prog. Neuropsychopharmacol. Biol. Psychiatry 117, 110555 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. Pei, Y. et al. Activation of the trace amine-associated receptor 1 prevents relapse to cocaine seeking. Neuropsychopharmacology 39, 2299–2308 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Barak, L. S. et al. Pharmacological characterization of membrane-expressed human trace amine-associated receptor 1 (TAAR1) by a bioluminescence resonance energy transfer cAMP biosensor. Mol. Pharmacol. 74, 585–594 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Saarinen, M. et al. TAAR1 dependent and independent actions of the potential antipsychotic and dual TAAR1/5-HT(1A) receptor agonist SEP-383856. Neuropsychopharmacology 47, 2319–2329 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Reese, E. A., Bunzow, J. R., Arttamangkul, S., Sonders, M. S. & Grandy, D. K. Trace amine-associated receptor 1 displays species-dependent stereoselectivity for isomers of methamphetamine, amphetamine, and para-hydroxyamphetamine. J. Pharmacol. Exp. Ther. 321, 178–186 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Chun, E. et al. Fusion partner toolchest for the stabilization and crystallization of G protein-coupled receptors. Structure 20, 967–976 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, P. et al. The structural basis of the dominant negative phenotype of the Gαi1β1γ2 G203A/A326S heterotrimer. Acta Pharmacol. Sin. 37, 1259–1272 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  39. Isberg, V. et al. Generic GPCR residue numbers - aligning topology maps while minding the gaps. Trends Pharmacol. Sci. 36, 22–31 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Zhuang, Y. et al. Structural insights into the human D1 and D2 dopamine receptor signaling complexes. Cell 184, 931–942.e918 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tan, E. S. et al. The molecular basis of species-specific ligand activation of trace amine-associated receptor 1 (TAAR(1)). ACS Chem. Biol. 4, 209–220 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cichero, E., Espinoza, S., Gainetdinov, R. R., Brasili, L. & Fossa, P. Insights into the structure and pharmacology of the human trace amine-associated receptor 1 (hTAAR1): homology modelling and docking studies. Chem. Biol. Drug Des. 81, 509–516 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Liao, S., Pino, M. J. Jr., Deleon, C., Lindner-Jackson, M. & Wu, C. Interaction analyses of hTAAR1 and mTAAR1 with antagonist EPPTB. Life Sci. 300, 120553 (2022).

    Article  CAS  PubMed  Google Scholar 

  44. Reese, E. A. et al. Exploring the determinants of trace amine-associated receptor 1’s functional selectivity for the stereoisomers of amphetamine and methamphetamine. J. Med. Chem. 57, 378–390 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Huang, S. et al. GPCRs steer G(i) and G(s) selectivity via TM5-TM6 switches as revealed by structures of serotonin receptors. Mol. Cell 82, 2681–2695.e2686 (2022).

    Article  CAS  PubMed  Google Scholar 

  46. Su, M. et al. Structures of β1-adrenergic receptor in complex with Gs and ligands of different efficacies. Nat. Commun. 13, 4095 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kaplan, A. L. et al. Bespoke library docking for 5-HT2A receptor agonists with antidepressant activity. Nature 610, 582–591 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tan, Y. et al. Structural insights into the ligand binding and Gi coupling of serotonin receptor 5-HT5A. Cell Discov. 8, 50 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Borowsky, B. et al. Trace amines: Identification of a family of mammalian G protein-coupled receptors. Proc. Natl Acad. Sci. USA 98, 8966–8971 (2001).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zhang, Y. et al. Single-particle cryo-EM structural studies of the β2AR–Gs complex bound with a full agonist formoterol. Cell Discov. 6, 45 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Imai, S. et al. Structural equilibrium underlying ligand-dependent activation of β2-adrenoreceptor. Nat. Chem. Biol. 16, 430–439 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Staus, D. P. et al. Allosteric nanobodies reveal the dynamic range and diverse mechanisms of G-protein-coupled receptor activation. Nature 535, 448–452 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. Lam, V. M. et al. Behavioral effects of a potential novel TAAR1 antagonist. Front. Pharmacol. 9, 953 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Bradaia, A. et al. The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system. Proc. Natl Acad. Sci. USA 106, 20081–20086 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  55. Guo, L. et al. Structural basis of amine odorant perception by a mammal olfactory receptor. Nature 618, 193–200 (2023).

    Article  ADS  CAS  PubMed  Google Scholar 

  56. Carpenter, B., Nehmé, R., Warne, T., Leslie, A. G. & Tate, C. G. Structure of the adenosine A(2A) receptor bound to an engineered G protein. Nature 536, 104–107 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  57. García-Nafría, J., Lee, Y., Bai, X., Carpenter, B. & Tate, C. G. Cryo-EM structure of the adenosine A(2A) receptor coupled to an engineered heterotrimeric G protein. eLife 7, e35946 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  59. 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 

  60. 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 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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 

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

    Article  ADS  PubMed  Google Scholar 

  64. Croll, T. I. ISOLDE: a physically realistic environment for model building into low-resolution electron-density maps. Acta Crystallogr. D 74, 519–530 (2018).

    Article  ADS  CAS  Google Scholar 

  65. Adams, P. D. et al. Recent developments in the PHENIX software for automated crystallographic structure determination. J. Synchrotron Radiat. 11, 53–55 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. 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 

  67. Wang, F. I., Ding, G., Ng, G. S., Dixon, S. J. & Chidiac, P. Luciferase-based GloSensor™ cAMP assay: temperature optimization and application to cell-based kinetic studies. Methods 203, 249–258 (2022).

    Article  CAS  PubMed  Google Scholar 

  68. Revel, F. G. et al. Brain-specific overexpression of trace amine-associated receptor 1 alters monoaminergic neurotransmission and decreases sensitivity to amphetamine. Neuropsychopharmacology 37, 2580–2592 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Olsen, R. H. J. 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 

  70. Webb, B. & Sali, A. Comparative Protein Structure Modeling Using MODELLER. Curr. Protoc. Bioinformatics 54, 5.6.1–5.6.37 (2016).

    Article  PubMed  Google Scholar 

  71. Heo, L. & Feig, M. Multi-state modeling of G-protein coupled receptors at experimental accuracy. Proteins 90, 1873–1885 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Olsson, M. H., Søndergaard, C. R., Rostkowski, M. & Jensen, J. H. PROPKA3: consistent treatment of internal and surface residues in empirical pKa predictions. J. Chem. Theory Comput. 7, 525–537 (2011).

    Article  CAS  PubMed  Google Scholar 

  73. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  76. Salomon-Ferrer, R., Götz, A. W., Poole, D., Le Grand, S. & Walker, R. C. Routine microsecond molecular dynamics simulations with AMBER on GPUs. 2. Explicit solvent particle mesh Ewald. J. Chem. Theory Comput. 9, 3878–3888 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Miller, B. R. 3rd et al. MMPBSA.py: an efficient program for end-state free energy calculations. J. Chem. Theory Comput. 8, 3314–3321 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was partially supported by the Ministry of Science and Technology of China (2018YFA0507002 to H.E.X., 2020YFA0509102 to S.W.); the National Natural Science Foundation of China (32071194 to F.X., 32130022 and 82121005 to H.E.X., 52272087 to W.L., 82225025 and 32071197 to S.W.); Shanghai Municipal Science and Technology Major Project (2019SHZDZX02 to H.E.X.); CAS Strategic Priority Research Programme (XDB37030103 to H.E.X.); the Lingang Laboratory, grant no. LG-GG-202204-01 (H.E.X.); and the National Key R&D Program of China (2022YFC2703105 to H.E.X.). The cryo-EM data were collected at the Shanghai Advanced Electron Microscope Center, Shanghai Institute of Material Medica, as well as at the Bio-EM platform of ShanghaiTech University. We thank Q. Yuan, K. Wu, W. Hu and S. Zhang for providing technical support and assistance during data collection at the Shanghai Advanced Electron Microscope Center, Shanghai Institute of Material Medica; we thank Q. Sun, Z. Zhang, L. Wang, D. Liu and Y. Liu at the Bio-EM facility at ShanghaiTech University; and Q. Tan, Q. Shi, J. Liu, N. Chen, S. Hu and W. Xiao from ShanghaiTech University for protein cloning, expression, assay and data collection support. We thank S. Zhao from ShanghaiTech University, J. Wang from Institute of Biophysics, CAS, and Q. Li from Shanghai Jiaotong University to provide initial sets of TAAR1 tool compounds and suggestions for this work. We also thank the support from Shanghai Frontiers Science Center for Biomacromolecules and Precision Medicine at ShanghaiTech University.

Author information

Authors and Affiliations

Authors

Contributions

F.X. and H.E.X. initiated the project. H.L. designed and screened the expression constructs of TAAR1 and prepared protein samples of TAAR1–METH–Gs, TAAR1–SEP-363856–Gs, 5-HT1AR–SEP-363856–Gi complexes for cryo-EM data collection, performed cryo-EM grids preparation, data acquisition, and structure determination, participated in GloSensor cAMP assay, and prepared the draft of the manuscript and figures. Y.Z. designed and screened the expression constructs of TAAR1 and prepared protein samples of TAAR1–β-PEA-Gs, TAAR1–RO5256390-Gs, complexes for cryo-EM data collection, performed cryo-EM grids preparation, data acquisition and structure determination, and participated in manuscript editing and figure preparation. Y.W. performed GloSensor cAMP assay, G-protein recruitment assay and participated in figure preparation. Y.W. performed the radioligand-binding assay and Gαiβγ dissociation assay. X.H. performed MD simulation analysis. P.X. and S.H. participated in protein sample preparation and structure determination. Q.Y. participated in data acquisition. X.Z. participated in G-protein recruitment assay. L.W. participated in GloSensor cAMP assay. K.J. assisted in some protein preparation work. H.C. assisted in compound preparation and synthesis. Z.L. helped construct the mutations for function assays. W.L. supervised compound preparation and synthesis. S.W. supervised ligand binding and functional assay. F.X. and H.E.X. conceived and supervised the project and participated in manuscript editing. F.X. and H.E.X. wrote the manuscript with input from H.L., Y.Z. and Y.W.

Corresponding authors

Correspondence to Wenbin Liu, Sheng Wang, H. Eric Xu or Fei Xu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Thomas Keck, Tao Che and Harald Sitte for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Functional assessment of TAAR1 by four agonists and structural feature of aminergic receptors.

(a-b) Concentration–response curves of the four agonists activating TAAR1 evaluated by miniGs recruitment assay (a) and radio-ligand-binding assay (b). Data are mean ± S.E.M. from 3 independent experiments (n = 3). (c) Structural comparison of ECL2s from TAAR1 and other aminergic receptors, ECL2 that forms a short helix in the structures is highlighted with red dotted circle.

Extended Data Fig. 2 Purification and structure determination of METH and β-PEA-bound TAAR1-Gs complexes.

(a) Cartoon models of the two TAAR1 constructs used in this study. (b) Representative size exclusion chromatography (SEC) profiles and SDS-PAGE analysis of TAAR1-Gs complex activated by METH. Experiment was repeated at least three times with similar results. (c-d) Representative cryo-EM image from 6,686 movies (c) and 2D classification averages (d) of METH–TAAR1–Gs. (e) Cryo-EM data processing flowcharts of METH-TAAR1-Gs by cyroSPARC 3.2. (f) The Fourier shell correlation (FSC) curves of METH-TAAR1-Gs. The global resolution of the final processed density map estimated at the FSC = 0.143 is 2.8 Å. (g) The global density map of METH-TAAR1-Gs colored by local resolutions. (h) The density maps of helices TM1–TM7 of transmembrane domain, extracellular loop ECL2 of TAAR1, α5 helix of Gαs, and METH in METH-TAAR1-Gs complex. (i) Representative size exclusion chromatography (SEC) profiles and SDS-PAGE analysis of TAAR1–Gs complex activated by β-PEA. Experiment was repeated at least three times with similar results. (j-k) Representative cryo-EM image from 4,371 movies (j) and 2D classification averages (k) of β-PEA-TAAR1-Gs. (l) Cryo-EM data processing flowcharts of β-PEA-TAAR1-Gs by cyroSPARC 3.2. (m) The Fourier shell correlation (FSC) curves of β-PEA-TAAR1-Gs. The global resolution of the final processed density map estimated at the FSC = 0.143 is 3.0 Å. (n) The global density map of β-PEA-TAAR1-Gs colored by local resolutions. (o) The density maps of helices TM1–TM7 of transmembrane domain, extracellular loop ECL2 of TAAR1, α5 helix of Gαs, and β-PEA in β-PEA-TAAR1-Gs complex.

Extended Data Fig. 3 Mutational effects of agonists and MD simulation analysis of TAAR1.

(a) Concentration–response curves of wild-type/mutants of TAAR1 induced by the four agonists. Every response of mutant was normalized to corresponding wild-type as 100%. Data are mean ± S.E.M. from 3 independent experiments (n = 3). (b-c) RMSD plot of METH (b) or β-PEA (c) in TAAR1 pocket during simulations. (d) The free energy contribution of key residues in TAAR1 pocket for METH or β-PEA binding. Data are mean ± S.E.M. from 6 independent experiments (n = 6).

Source Data

Extended Data Fig. 4 Purification and structure determination of SEP-363856 bound TAAR1-Gs and 5HT1AR-Gi complexes.

(a) Representative size exclusion chromatography (SEC) profiles and SDS-PAGE analysis of TAAR1-Gs complex activated by SEP-363856. Experiment was repeated at least three times with similar results. (b-c) Representative cryo-EM image from 7,262 movies (b) and 2D classification averages (c) of SEP-363856-TAAR1-Gs. (d) Cryo-EM data processing flowcharts of SEP-363856-TAAR1-Gs by cyroSPARC 3.2. (e) The Fourier shell correlation (FSC) curves of SEP-363856-TAAR1-Gs. The global resolution of the final processed density map estimated at the FSC = 0.143 is 2.6 Å. (f) The global density map of SEP-363856-TAAR1-Gs colored by local resolutions. (g) The density maps of helices TM1–TM7 of transmembrane domain, extracellular loop ECL2 of TAAR1, α5 helix of Gαs, and SEP-363856 in SEP-363856-TAAR1-Gs complex. (h) Representative size exclusion chromatography (SEC) profiles and SDS-PAGE analysis of 5HT1AR -Gi complex activated by SEP-363856. Experiment was repeated at least three times with similar results. (i-j) Representative cryo-EM image from 6,222 movies (i) and 2D classification averages (j) of SEP-363856-5HT1AR -Gi. (k) Cryo-EM data processing flowcharts of SEP-363856-5HT1AR-Gi complex by cyroSPARC 3.2. (l) The Fourier shell correlation (FSC) curves of SEP-363856-5HT1AR -Gi complex. The global resolution of the final processed density map estimated at the FSC = 0.143 is 3.0 Å. (m) The global density map of SEP-363856-5HT1AR-Gi colored by local resolutions. (n) The density maps of helices TM1–TM7 of transmembrane domain, α5 helix of Gαi, and SEP-363856 in 5HT1AR -Gi complex.

Extended Data Fig. 5 Comparison of the SEP-363856 bound pockets of TAAR1 and 5HT1AR, and purification, structure determination of RO5256390 bound TAAR1-Gs complex.

(a) Binding pockets of SEP-363856, aripiprazole (PDB ID: 7e2z) and serotonin (PDB ID: 7e2y) in 5HT1AR. (b) Binding pocket of SEP-363856 in TAAR1. (c) Alignment of key residues constituting the ligand-binding pockets in SEP-383656 bound 5HT1AR or TAAR1 structures, and serotonin and aripiprazole bound 5HT1AR structures. The green background indicated 100% conserved residues and orange background indicated residues that are conserved in 5HT1AR but not in TAAR1. (d) Representative size exclusion chromatography (SEC) profiles and SDS-PAGE analysis of TAAR1-Gs complex activated by RO5256390. Experiment was repeated at least three times with similar results. (e-f) Representative cryo-EM image from 5,135 movies (e) and 2D classification averages (f) of RO5256390-TAAR1-Gs. (g) Cryo-EM data processing flowcharts of RO5256390-TAAR1-Gs complex by cyroSPARC 3.2. (h) The Fourier shell correlation (FSC) curves of RO5256390-TAAR1-Gs. The global resolution of the final processed density map estimated at the FSC = 0.143 is 2.8 Å. (i) The global density map of RO5256390-TAAR1-Gs colored by local resolutions. (j) The density maps of helices TM1–TM7 of transmembrane domain, extracellular loop ECL2 of TAAR1, α5 helix of Gαs, and RO5256390 in RO5256390-TAAR1-Gs complex.

Extended Data Fig. 6 Molecular mechanism of the high potency and selectivity of RO5256390.

(a) Interactions of RO5256390 and key residues in TAAR1 pocket. (b) RO5256390 inserted deep into the cavity formed by D1033x32, S1073x36, W2646x48, Y2947x43. (c-d) Concentration–response curves for mutations of key residues in TAAR1 induced by RO5256390 using Glo-sensor assay. Data are mean ± S.E.M. from 3 independent experiments (n = 3). (e) and (g) Evaluation of RO5256390 and SEP-363856 activating 5HT1AR using Gi protein dissociation assay (e) or Glo-sensor assay (g), 5HT was used as reference. Data are mean ± S.E.M. from 3 independent experiments (n = 3). (f) and (h) Evaluation of RO5256390 activating 5HT1AR with C120 substituted with alanine or serine, using Gi protein dissociation assay (f) or Glo-sensor assay (h), 5HT was used as reference. Data are mean ± S.E.M. from 3 independent experiments (n = 3).

Source Data

Extended Data Fig. 7 Conservation of key residues for TAAR1 activating and role of ECL2 in aminergic receptors.

(a) Sequence alignment of key residues in ECL2 from representative aminergic receptors. (b) Display of residues in ECL2s from TAAR1 and other aminergic receptors that may take part in ligand recognition and binding pocket formation.

Extended Data Fig. 8 Functional assessment of four agonists activating human TAAR family subtypes.

Human TAAR family subtypes (hTAAR1/2/5/6/8/9) were cloned into pcDNA3.1 vector and the activity was measured by Glo-Sensor assay. The negative control was pcDNA3.1 vector and DMSO solvent. The results showed that only TAAR1 can be activated by the four agonist. Data are mean ± S.E.M. from 3 independent experiments (n = 3).

Source Data

Extended Data Fig. 9 Sequence alignment of human TAAR family, TAAR1 activation mechanism, and analysis of METH binding in human and rodent TAAR1.

(a-b) Sequence alignment of residues important for TAAR1-ligand binding (a) or activating (b) from human TAAR family and mTAAR9. (c-d) Superposition of activated TAAR1 (dark cyan) with active mTAAR9 (light green; PDB ID: 8iw7), β2AR (orange; PDB ID: 6kr8) and inactive β2AR (gray; PDB ID: 5jqh). Notable conformational changes occur at extracellular end of TM1 and intracellular ends of TM6, TM7 and H8 upon receptor activation, side view (c) and bottom view (d). (e) The “toggle switch”, W2646.48, of TAAR1 display relative rotameric change when sensing agonist. (f–h) The key P-I-F6.44 (f), D-R3.50-Y (g), and N-P7.50-xx-Y7.53 (h) motifs displayed conformational rearrangement in activated TAAR1 structure. (i–k) RMSD plot of METH binding in rat, mouse, or human TAAR1 pocket during simulations. (l) Bar graph showing RMSD differences for METH binding in rat, mouse, or human TAAR1 pocket during simulations. Data are mean ± S.E.M. of RMSD from 3 independent experiments (n = 3) for rat and mouse TAAR1 and 6 independent experiments (n = 6) for human TAAR1. (m) Sequence alignment for residues that are important for ligand binding to TAAR1 from rat, mouse or human.

Source Data

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Supplementary Information

Supplementary Tables 1–6 and Supplementary Fig. 1.

Reporting Summary

Peer Review File

Source data

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, H., Zheng, Y., Wang, Y. et al. Recognition of methamphetamine and other amines by trace amine receptor TAAR1. Nature 624, 663–671 (2023). https://doi.org/10.1038/s41586-023-06775-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06775-1

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: 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