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 pharmacology and therapeutic potential of 5-methoxytryptamines

Abstract

Psychedelic substances such as lysergic acid diethylamide (LSD) and psilocybin show potential for the treatment of various neuropsychiatric disorders1,2,3. These compounds are thought to mediate their hallucinogenic and therapeutic effects through the serotonin (5-hydroxytryptamine (5-HT)) receptor 5-HT2A (ref. 4). However, 5-HT1A also plays a part in the behavioural effects of tryptamine hallucinogens5, particularly 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT), a psychedelic found in the toxin of Colorado River toads6. Although 5-HT1A is a validated therapeutic target7,8, little is known about how psychedelics engage 5-HT1A and which effects are mediated by this receptor. Here we map the molecular underpinnings of 5-MeO-DMT pharmacology through five cryogenic electron microscopy (cryo-EM) structures of 5-HT1A, systematic medicinal chemistry, receptor mutagenesis and mouse behaviour. Structure–activity relationship analyses of 5-methoxytryptamines at both 5-HT1A and 5-HT2A enable the characterization of molecular determinants of 5-HT1A signalling potency, efficacy and selectivity. Moreover, we contrast the structural interactions and in vitro pharmacology of 5-MeO-DMT and analogues to the pan-serotonergic agonist LSD and clinically used 5-HT1A agonists. We show that a 5-HT1A-selective 5-MeO-DMT analogue is devoid of hallucinogenic-like effects while retaining anxiolytic-like and antidepressant-like activity in socially defeated animals. Our studies uncover molecular aspects of 5-HT1A-targeted psychedelics and therapeutics, which may facilitate the future development of new medications for neuropsychiatric 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: Activity of psychedelics at 5-HT1A and 5-HT2A, and cryo-EM structures of 5-MeO-DMT and LSD bound to the 5-HT1A signalling complex.
Fig. 2: Differential pharmacological effects of amine and indole modifications to the 5-MeO-DMT scaffold at 5-HT1A and 5-HT2A.
Fig. 3: Elucidation of potency and selectivity determinants of tryptamines at 5-HT1A and 5-HT2A.
Fig. 4: Structural and functional comparison of 4-F,5-MeO-PyrT and clinical 5-HT1A medications at 5-HT1A.
Fig. 5: Effects of 5-MeO-DMT derivatives on mouse behaviour.

Similar content being viewed by others

Data availability

Density maps and structure coordinates have been deposited into the Electron Microscopy Data Bank (EMDB) and the PDB with the following accession identifiers: EMD-29560 and PDB 8FY8 for 5-MeO-DMT–5-HT1A–Gαi1–Gβ1–Gγ2; EMD-29597 and PDB 8FYT for LSD–5-HT1A–Gαi1–Gβ1-Gγ2; EMD-29571 and PDB 8FYE for 4-F,5-MeO-PyrT–5-HT1A–Gαi1–Gβ1–Gγ2; EMD-29585 and PDB 8FYL for vilazodone–5-HT1A–Gαi1–Gβ1–Gγ2; and EMD-29599 and PDB 8FYX for buspirone–5-HT1A–Gαi1–Gβ1–Gγ2.  Source data are provided with this paper. Additional data from this study are available upon request.

References

  1. Carhart-Harris, R. L. et al. Psilocybin with psychological support for treatment-resistant depression: six-month follow-up. Psychopharmacology 235, 399–408 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Armstrong, S. B. et al. Prospective associations of psychedelic treatment for co-occurring alcohol misuse and posttraumatic stress symptoms among United States Special Operations Forces veterans. Mil. Psychol. https://doi.org/10.1080/08995605.2022.2156200 (2023).

  3. Goodwin, G. M. et al. Single-dose psilocybin for a treatment-resistant episode of major depression. N. Engl. J. Med. 387, 1637–1648 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. Cameron, L. P. et al. 5-HT2ARs mediate therapeutic behavioral effects of psychedelic tryptamines. ACS Chem. Neurosci. https://doi.org/10.1021/acschemneuro.2c00718 (2023).

  5. Krebs-Thomson, K., Ruiz, E. M., Masten, V., Buell, M. & Geyer, M. A. The roles of 5-HT1A and 5-HT2 receptors in the effects of 5-MeO-DMT on locomotor activity and prepulse inhibition in rats. Psychopharmacology 189, 319–329 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Schwelm, H. M. et al. Qualitative and quantitative analysis of tryptamines in the poison of Incilius alvarius (Amphibia: Bufonidae). J. Anal. Toxicol. 46, 540–548 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Hughes, Z. A. et al. Neurochemical evaluation of the novel 5-HT1A receptor partial agonist/serotonin reuptake inhibitor, vilazodone. Eur. J. Pharmacol. 510, 49–57 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Loane, C. & Politis, M. Buspirone: what is it all about? Brain Res. 1461, 111–118 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Winter, J. C., Filipink, R. A., Timineri, D., Helsley, S. E. & Rabin, R. A. The paradox of 5-methoxy-N,N-dimethyltryptamine: an indoleamine hallucinogen that induces stimulus control via 5-HT1A receptors. Pharmacol. Biochem. Behav. 65, 75–82 (2000).

    Article  CAS  PubMed  Google Scholar 

  10. Davis, A. K., So, S., Lancelotta, R., Barsuglia, J. P. & Griffiths, R. R. 5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) used in a naturalistic group setting is associated with unintended improvements in depression and anxiety. Am. J. Drug Alcohol Abuse 45, 161–169 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Reckweg, J. T. et al. The clinical pharmacology and potential therapeutic applications of 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT). J. Neurochem. 162, 128–146 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chauhan, M., Parry, R. & Bobo, W. V. Vilazodone for major depression in adults: pharmacological profile and an updated review for clinical practice. Neuropsychiatr. Dis. Treat. 18, 1175–1193 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Kim, K. et al. Structure of a hallucinogen-activated Gq-coupled 5-HT2A serotonin receptor. Cell 182, 1574–1588.e19 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wacker, D. et al. Crystal structure of an LSD-bound human serotonin receptor. Cell 168, 377–389.e12 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Cameron, L. P. et al. A non-hallucinogenic psychedelic analogue with therapeutic potential. Nature 589, 474–479 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  16. 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 

  17. Carhart-Harris, R. L. & Nutt, D. J. Serotonin and brain function: a tale of two receptors. J. Psychopharmacol. 31, 1091–1120 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Nichols, D. E. Chemistry and structure–activity relationships of psychedelics. Curr. Top. Behav. Neurosci. 36, 1–43 (2018).

    CAS  PubMed  Google Scholar 

  20. Glennon, R. A. et al. Binding of β-carbolines and related agents at serotonin (5-HT2 and 5-HT1A), dopamine (D2) and benzodiazepine receptors. Drug Alcohol Depend. 60, 121–132 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Halberstadt, A. L. & Geyer, M. A. Multiple receptors contribute to the behavioral effects of indoleamine hallucinogens. Neuropharmacology 61, 364–381 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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 

  23. 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. Methods Neurosci. 25, 366–428 (1995).

    Article  CAS  Google Scholar 

  24. Rasmussen, S. G. et al. Crystal structure of the β2 adrenergic receptor–Gs protein complex. Nature 477, 549–555 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wacker, D. et al. Structural features for functional selectivity at serotonin receptors. Science 340, 615–619 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nichols, D. E., Monte, A., Huang, X. & Marona-Lewicka, D. Stereoselective pharmacological effects of lysergic acid amides possessing chirality in the amide substituent. Behav. Brain Res. 73, 117–119 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Speeter, M. E. & Anthony, W. C. The action of oxalyl chloride on indoles—a new approach to tryptamines. J. Am. Chem. Soc. 76, 6208–6210 (1954).

    Article  CAS  Google Scholar 

  28. Shulgin, A. T. & Shulgin, A. Tihkal: The Continuation (Transform Press, 1997).

  29. Kozell, L. B. et al. Pharmacologic activity of substituted tryptamines at 5-hydroxytryptamine (5-HT)2A receptor (5-HT2AR), 5-HT2CR, 5-HT1AR, and serotonin transporter. J. Pharmacol. Exp. Ther. 385, 62–75 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Blair, J. B. et al. Effect of ring fluorination on the pharmacology of hallucinogenic tryptamines. J. Med. Chem. 43, 4701–4710 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Laban, U., Kurrasch-Orbaugh, D., Marona-Lewicka, D. & Nichols, D. E. A novel fluorinated tryptamine with highly potent serotonin 5-HT1A receptor agonist properties. Bioorg. Med. Chem. Lett. 11, 793–795 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Wang, C. et al. Structural basis for molecular recognition at serotonin receptors. Science 340, 610–614 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gasser, P., Kirchner, K. & Passie, T. LSD-assisted psychotherapy for anxiety associated with a life-threatening disease: a qualitative study of acute and sustained subjective effects. J. Psychopharmacol. 29, 57–68 (2015).

    Article  PubMed  Google Scholar 

  34. Halberstadt, A. L., Koedood, L., Powell, S. B. & Geyer, M. A. Differential contributions of serotonin receptors to the behavioral effects of indoleamine hallucinogens in mice. J. Psychopharmacol. 25, 1548–1561 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Darmani, N. A., Martin, B. R., Pandey, U. & Glennon, R. A. Do functional relationships exist between 5-HT1A and 5-HT2 receptors. Pharmacol. Biochem. Behav. 36, 901–906 (1990).

    Article  CAS  PubMed  Google Scholar 

  36. Arnt, J. & Hyttel, J. Facilitation of 8-OHDPAT-induced forepaw treading of rats by the 5-HT2 agonist DOI. Eur. J. Pharmacol. 161, 45–51 (1989).

    Article  CAS  PubMed  Google Scholar 

  37. Ermakova, A. O., Dunbar, F., Rucker, J. & Johnson, M. W. A narrative synthesis of research with 5-MeO-DMT. J. Psychopharmacol. 36, 273–294 (2022).

    Article  CAS  PubMed  Google Scholar 

  38. Bagot, R. C. et al. Ketamine and imipramine reverse transcriptional signatures of susceptibility and induce resilience-specific gene expression profiles. Biol. Psychiatry 81, 285–295 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Berton, O. et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311, 864–868 (2006).

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Donahue, R. J., Muschamp, J. W., Russo, S. J., Nestler, E. J. & Carlezon, W. A. Jr. Effects of striatal ΔFosB overexpression and ketamine on social defeat stress-induced anhedonia in mice. Biol. Psychiatry 76, 550–558 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Golden, S. A., Covington, H. E. 3rd, Berton, O. & Russo, S. J. A standardized protocol for repeated social defeat stress in mice. Nat. Protoc. 6, 1183–1191 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Krishnan, V. et al. Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell 131, 391–404 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Willner, P., Towell, A., Sampson, D., Sophokleous, S. & Muscat, R. Reduction of sucrose preference by chronic unpredictable mild stress, and its restoration by a tricyclic antidepressant. Psychopharmacology 93, 358–364 (1987).

    Article  CAS  PubMed  Google Scholar 

  44. Li, N. et al. Glutamate N-methyl-d-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol. Psychiatry 69, 754–761 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hesselgrave, N., Troppoli, T. A., Wulff, A. B., Cole, A. B. & Thompson, S. M. Harnessing psilocybin: antidepressant-like behavioral and synaptic actions of psilocybin are independent of 5-HT2R activation in mice. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.2022489118 (2021).

  46. Shao, L. X. et al. Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron 109, 2535–2544.e4 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. de la Fuente Revenga, M. et al. Prolonged epigenomic and synaptic plasticity alterations following single exposure to a psychedelic in mice. Cell Rep. 37, 109836 (2021).

    Article  PubMed  Google Scholar 

  48. Savalia, N. K., Shao, L. X. & Kwan, A. C. A dendrite-focused framework for understanding the actions of ketamine and psychedelics. Trends Neurosci. 44, 260–275 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Bickle, J. G. et al. 5HT1A receptors on dentate gyrus granule cells confer stress resilience. Biol. Psychiatry https://doi.org/10.1016/j.biopsych.2023.10.007 (2023).

  50. Newman-Tancredi, A. et al. NLX-112, a highly selective 5-HT1A receptor agonist, mediates analgesia and antidepressant-like activity in rats via spinal cord and prefrontal cortex 5-HT1A receptors, respectively. Brain Res. 1688, 1–7 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Becker, A. M. et al. Ketanserin reverses the acute response to LSD in a randomized, double-blind, placebo-controlled, crossover study in healthy subjects. Int. J. Neuropsychopharmacol. https://doi.org/10.1093/ijnp/pyac075 (2022).

  52. Strassman, R. J. Human psychopharmacology of N,N-dimethyltryptamine. Behav. Brain Res. 73, 121–124 (1996).

    Article  CAS  PubMed  Google Scholar 

  53. Pokorny, T., Preller, K. H., Kraehenmann, R. & Vollenweider, F. X. Modulatory effect of the 5-HT1A agonist buspirone and the mixed non-hallucinogenic 5-HT1A/2A agonist ergotamine on psilocybin-induced psychedelic experience. Eur. Neuropsychopharmacol. 26, 756–766 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Reckweg, J. et al. A phase 1, dose-ranging study to assess safety and psychoactive effects of a vaporized 5-methoxy-N,N-dimethyltryptamine formulation (GH001) in healthy volunteers. Front. Pharmacol. 12, 760671 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Shapiro, D. A. et al. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology 28, 1400–1411 (2003).

    Article  CAS  PubMed  Google Scholar 

  56. 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 

  57. 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 

  58. Punjani, A., Zhang, H. & Fleet, D. J. Non-uniform refinement: adaptive regularization improves single-particle cryo-EM reconstruction. Nat. Methods 17, 1214–1221 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  61. Yamashita, K., Palmer, C. M., Burnley, T. & Murshudov, G. N. Cryo-EM single-particle structure refinement and map calculation using Servalcat. Acta Crystallogr. D Struct. Biol. 77, 1282–1291 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  62. The PyMOL Molecular Graphics System v.2.0 (Schrödinger, 2017).

  63. Kroeze, W. K. et al. PRESTO-Tango as an open-source resource for interrogation of the druggable human GPCRome. Nat. Struct. Mol. Biol. 22, 362–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Henke, A. et al. Toward serotonin fluorescent false neurotransmitters: development of fluorescent dual serotonin and vesicular monoamine transporter substrates for visualizing serotonin neurons. ACS Chem. Neurosci. 9, 925–934 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Rodrı́guez, P. et al. A single administration of the atypical psychedelic ibogaine or its metabolite noribogaine induces an antidepressant-like effect in rats. ACS Chem. Neurosci. 11, 1661–1672 (2020).

Download references

Acknowledgements

This work was supported by NIH grant R35GM133504, a Sloan Research Fellowship in Neuroscience, an Edward Mallinckrodt, Jr Foundation Grant, a McKnight Foundation Scholars Award, an Irma T. Hirschl/Monique Weill-Caulier Trust Research Award (all to D.W.); an NIH F31 MH132317 (A.L.W), and T32 Training Grant GM062754 and DA053558 (A.L.W and G.Z.); the G. Harold & Leila Y. Mathers Charitable Foundation, the NIH grant R01DA050613, G.L. Freeman, and Columbia University for support of this work (all to D.S.); and the following NIH grants: R01MH127820 and R01MH104559 (S.J.R.). L.F.P is supported by the Leon Levy Foundation and the Brain and Behavior Research Foundation. Some of this work was performed at the National Center for CryoEM Access and Training (NCCAT) and the Simons Electron Microscopy Center located at the New York Structural Biology Center, supported by the NIH Common Fund Transformative High Resolution Cryo-Electron Microscopy program (U24 GM129539) and by grants from the Simons Foundation (SF349247) and NY State Assembly. We further acknowledge cryo-EM resources at the National Resource for Automated Molecular Microscopy located at the New York Structural Biology Center, supported by grants from the Simons Foundation (SF349247), NYSTAR, and the NIH National Institute of General Medical Sciences (GM103310) with additional support from Agouron Institute (F00316) and NIH (OD019994). For additional data collection, we are grateful to staff at the Laboratory for BioMolecular Structure (LBMS), which is supported by the DOE Office of Biological and Environmental Research (KP160711). This work was supported in part through the computational and data resources and staff expertise provided by Scientific Computing and Data at the Icahn School of Medicine at Mount Sinai and supported by the Clinical and Translational Science Awards (CTSA) grant ULTR004419 from the National Center for Advancing Translational Sciences. We thank B. Bechand for early examination of in vivo pharmacology of the described compounds assisted by V. C. Galicia; C. Hwu for assistance with synthesis and purification of several compounds (all at Columbia University); and F. Zandkarimi from the Columbia University Chemistry Department Mass Spectrometry Core Facility for conducting the high-resolution mass spectrometry experiments.

Author information

Authors and Affiliations

Authors

Contributions

D.S., A.C.K, M. J. Cunningham and D.L. conceived and initiated the project. A.L.W. designed experiments, expressed and purified protein for grid freezing, collected data, refined structures with help from M. J. Capper, performed signalling and uptake assays, and co-wrote the manuscript. D.L. designed, synthesized, purified and characterized compounds with assistance from V.H., and co-wrote the manuscript. M. J. Cunningham designed, synthesized, purified and characterized compounds. D.L., V.H. and D.S. designed and supervised the pharmacokinetics study. L.F.P. performed the chronic SD stress assay and subsequent behavioural analyses supervised by S.J.R. I.C.S. designed and performed in vivo pharmacology assays, including the open-field and HTR assays, with assistance from P.D. G.Z. prepared grids for structure determination and assisted with data collection. D.S. and D.W. conceptualized the overall project and designed experiments, analysed the data, supervised the project and management, and wrote the manuscript.

Corresponding authors

Correspondence to Dalibor Sames or Daniel Wacker.

Ethics declarations

Competing interests

The authors declare the following competing financial interests: D.S. and A.C.K. are co-founders of Gilgamesh Pharmaceuticals and Kures. M. J. Cunningham is a co-founder of Gilgamesh Pharmaceuticals. A.L.W., D.L., I.C.S., L.F.P., S.J.R., D.S. and D.W. are inventors on a patent application related to the featured compound class. D.W. has consulted for Otsuka Pharmaceutical, Longboard Pharmaceuticals and Ocean Bio on the design of psychedelic-based therapeutics. None of the companies listed herein contributed to the funding or experimental design. All other authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Cryo-EM structure determination of drug-bound 5-HT1A-Gαi1/Gβ1/Gγ2 complexes.

a, Representative structure determination of 5-MeO-DMT-bound 5-HT1A signalling complex. Top Left, Analytical size exclusion chromatography and SDS-PAGE show monodisperse and pure protein of intact complex and its components. Right, representative Cryo-EM micrograph (white bar indicates scale) of 4680 total micrographs and data processing schematic exemplified by 5-MeO-DMT-bound 5-HT1A-Gi1 structure: After particle picking, 2D classification and multiple rounds of 3D classification, the final particle stack was refined using non-uniform refinement. A final map was obtained and resolutions were estimated applying the 0.143 cutoff in GS-FSC. Initial models were built in COOT, and then further refined in PHENIX for the generation of final coordinates shown in this manuscript. b, Local resolution map of a 5-MeO-DMT-bound 5-HT1A-Gi1 complex (left) and FSC curves (right) calculated based on the final reconstruction in cryoSPARC. c, 5-MeO-DMT (yellow) in the orthosteric binding pocket from the side (left) and rotated 45° towards the top of the receptor (right) with the map of ligand and surround residue densities shown at 5σ.

Source data

Extended Data Fig. 2 Comparison of different 5-HT1A structures and differences in binding of LSD to 5-HT1A and 5-HT2A.

a, Superposition of herein reported 5-MeO-DMT-bound 5-HT1A-Gi complex with the previously reported 5-HT-bound 5-HT1A-Gi structure (PDB ID: 7E2Y) shows similar conformations. Additional residues in 5-HT1A’s EL2 and G proteins not observed in previous structures are highlighted in red. b, Lipids (blue) and cholesterol hemisuccinate (dark blue) are bound to similar sites as observed before. c, Local resolution map of a LSD-bound 5-HT1A-Gi1 complex (left) and FSC curves (right) calculated the final reconstruction in cryoSPARC. d, LSD (grey) in the orthosteric binding pocket from the side (top) and rotated 45° towards the top of the receptor (bottom) with the map of ligand and surround residue densities shown at 5σ. e, LSD shows distinct binding modes bound to 5-HT1A-Gi signalling complex and 5-HT2A (PDB ID: 6WGT). Left, 5-HT1A-bound LSD (grey) sits deeper in the binding pocket compared to 5-HT2A-bound LSD (orange). Zoom in of LSD in 5-HT1A-Gi structure (middle) and 5-HT2A structure (right) highlights differential stereochemistry and receptor-specific interactions of diethylamide moiety. Hydrogen bonds are indicated as grey dashed lines.

Source data

Extended Data Fig. 3 Global structure-activity landscape of tryptamine psychedelics at 5-HT1A and 5-HT2A receptors and their synthesis.

a, Overview of the cryo-EM structure of the 5HT1A receptor-Gi signalling complex bound to 5-MeO-DMT (center). Classic psychedelics such as the prototypical compounds DMT and LSD are agonists of both 5-HT1A and 5-HT2A receptors (left semi-circle). 5-MeO-DMT (top of the circle), a major psychoactive compound found in toad secretions, shows comparable potency and efficacy at both 5-HT1A and 5-HT2A receptors. Systematic structural mapping via elaboration of the core 5-MeO-DMT structure identifies a class of 5-MeO-tryptamines with increasing 5-HT1A selectivity (right hemi-circle). 5-MeO-DMT can be viewed as a deconstruction of ibogaine, a oneirogen with a complex polycyclic tryptamine structure (bottom of the circle). Iboga compounds show no activity at 5-HT1A and 5-HT2A receptors, but this activity re-emerges by deconstruction of the isoquinuclidine core to simple mono-cyclic tryptamines such as 5-MeO-PipT (5-methoxypiperidinyl-tryptamine) and 4-F,5-MeO-PyrT (4-fluoro, 5-methoxypyrrolidinyl-tryptamine, right hemi-circle). Images of peyote, mushrooms, ayahuasca, and toad are from iStock and ShutterStock, and Tabernanthe iboga schematic is adapted from previous work65b, General synthesis methodology of tryptamines. a. oxalyl chloride, b. MeOH, LAH, c. PPh3, CBr4, d. Amine, TEA, e. Amine, f. LAH.

Extended Data Fig. 4 Structural comparison of 5-MeO-DMT 5-HT1A-selective analog 4-F, 5-MeO-PyrT bound to 5-HT1A.

a, Local resolution map of a 4-F,5-MeO-PyrT-bound 5-HT1A-Gi1 complex (left) and FSC curves (right) calculated from the final reconstruction in cryoSPARC. b, 4-F,5-MeO-PyrT (dark blue) in the orthosteric binding pocket from the side (left) and rotated 45° towards the top of the receptor (right) with the map of ligand and surrounding residue densities shown at 5σ. c, structural side-by-side comparison of 5-HT1A orthosteric site bound to 5-MeO-DMT (yellow) and 4-F,5-MeO-PyrT (dark blue). d, cAMP accumulation assays using wildtype and mutant 5-HT1A, and different drugs. Concentration-response experiments reveal different sensitivities of distinct drugs to F361L mutation. All signalling studies were performed in triplicates and are averaged from two to three independent experiments. Data have been normalized against 5-HT and errors bars denote s.e.m.

Source data

Extended Data Fig. 5 Comparison of 4-F,5-MeO-PyrT binding pose to that of different clinical 5-HT1A drugs.

a, b, Local resolution maps of vilazodone-bound (a) and buspirone-bound (b) 5-HT1A-Gi1 complexes and corresponding FSC curves calculated from the final reconstructions in cryoSPARC. c, d, Vilazodone (c, green) and buspirone (d, teal) in the orthosteric binding pocket from the side (left) and rotated 45° towards the top of the receptor (right) with the density map of ligand and surrounding residues shown at 5σ. e-h, Extracellular view of 4-F,5-MeO-PyrT (e, dark blue), Vilazodone (f, green), Aripiprazole (g, magenta, PDB ID: 7E2Z), and Buspirone (h, teal) binding poses in 5-HT1A’s orthosteric site.

Source data

Extended Data Fig. 6 Selectivity of different 5-MeO-DMT analogs and off-target activity of 4-F,5-MeO-PyrT.

a, 5-HT1A-Gi and 5-HT2A-Gq BRET of 5-HT, 5-MeO-DMT, 5-MeO-MET, and 4-F,5-MeO-PyrT with respective potencies and 5-HT1A > 5-HT2A selectivities. b, Off-target inhibition of transporters SERT, PMAT, OCT1, and OCT2 by 4-F,5-MeO-PyrT and known inhibitors. SERT uptake was performed in triplicates and data was averaged from two independent experiments showing data as mean+s.e.m. PMAT, OCT1, and OCT2 uptake was performed once in quadruplicate. c, Arrestin-recruitment of 5-HT and 4-F,5-MeO-PyrT at all human 5-HT receptor subtypes except for 5-HT7A, whose activation was measured via cAMP stimulation. All functional studies were performed in triplicates and are averaged from two to three independent experiments. Data have been normalized against 5-HT, Citalopram, and Decynium-22, and errors bars denote the s.e.m.

Source data

Extended Data Fig. 7 Effects of 5-MeO-DMT derivatives on rodent behavior.

a, Evaluation of 4-F,5-MeO-PyrT in open field for two hours (n = 3-4/group). b, Exemplary traces of the ambulatory distance traveled in open field following 4-F,5-MeO-PyrT (1 mg/kg, s.c.) administration and with and without WAY-100635 pre-treatment (1 mg/kg, s.c., 15 min prior). Panel was created with BioRender.com. c, Effect of WAY-100635 (1 mg/kg, s.c., 15 min prior) on 4-F,5-MeO-PyrT’s and 5-MeO-MET’s effect on total locomotion (n = 7 - 8/group, 30 min). d, Determination of optimal inhibitory WAY-100635 dose via administration of 1 mg/kg and 2 mg/kg WAY-100635 prior to studying 4-F,5-MeO-PyrT’s effects on total locomotion (n = 7 - 8 /group) and HTR (n = 6/group). Analysis was done using one-way ANOVA with multiple comparisons with Tukey’s post hoc test, and exact p values have been denoted in the Figure. e-g, Effects of saline or 4-F,5-MeO-PyrT administration on control (Control) or chronically defeated mice (Stress). Determination of e, distance moved as a measure of locomotor activity, f, social interaction as a measure of anxiety- and depression-related phenotype, g, corner time as a measure of anxiety-like behavior. Analysis was done in a sub-cohort of the animals reported in Fig. 5d. Number of mice for each group is indicated below the data for each respective cohort. Differences were determined by two-way ANOVA with multiple comparisons using Fisher’s LSD post hoc test, and exact p values have been denoted in the Figure. h, Vehicle- and drug-treated stressed mice shown in Fig. 5d were divided into susceptible (SI ratio<1) and resilient (SI ratio>1) populations. Significance in the population shift was determined by a two-sided Fisher’s exact test and p value and number of mice have been denoted in the Figure. Error bars denote the s.e.m.

Source data

Extended Data Table 1 Cryo-EM Data Collection, Model Refinement and Validation
Extended Data Table 2 AUClast and Cmax of 4-F,5-MeO-PyrT at 1 mg/kg SC administration, and Plasma and Brain Tissue Binding of 4-F,5-MeO-PyrT

Supplementary information

Supplementary Information

This supplementary information file contains supplementary data, Supplementary Tables 1–3 and supplementary references.

Reporting Summary

Peer Review File

Supplementary Data

Source data for Supplementary Tables 1 and 3.

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

Warren, A.L., Lankri, D., Cunningham, M.J. et al. Structural pharmacology and therapeutic potential of 5-methoxytryptamines. Nature (2024). https://doi.org/10.1038/s41586-024-07403-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-024-07403-2

Comments

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

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing