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 mechanism of muscle nicotinic receptor desensitization and block by curare

Abstract

Binding of the neurotransmitter acetylcholine to its receptors on muscle fibers depolarizes the membrane and thereby triggers muscle contraction. We sought to understand at the level of three-dimensional structure how agonists and antagonists alter nicotinic acetylcholine receptor conformation. We used the muscle-type receptor from the Torpedo ray to first define the structure of the receptor in a resting, activatable state. We then determined the receptor structure bound to the agonist carbachol, which stabilizes an asymmetric, closed channel desensitized state. We find conformational changes in a peripheral membrane helix are tied to recovery from desensitization. To probe mechanisms of antagonism, we obtained receptor structures with the active component of curare, a poison arrow toxin and precursor to modern muscle relaxants. d-Tubocurarine stabilizes the receptor in a desensitized-like state in the presence and absence of agonist. These findings define the transitions between resting and desensitized states and reveal divergent means by which antagonists block channel activity of the muscle-type nicotinic receptor.

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: Resting-state structure and cholesterol interactions.
Fig. 2: Asymmetric conformational transition between resting and desensitized states.
Fig. 3: Agonist binding site and ion pore profile.
Fig. 4: M4 uncoupling in the desensitized state.
Fig. 5: Structural pharmacology of d-tubo.

Similar content being viewed by others

Data availability

Cryo-EM maps and atomic model coordinates have been deposited in the Electron Microscopy Data Bank and PDB, respectively; apo (EMD-25202 and PDB 7SMM), apo plus cholesterol (EMD-25205 and PDB 7SMQ), carbachol-bound desensitized state (EMD-25206 and PDB 7SMR), d-tubo bound (EMD-25207 and PDB 7SMS) and d-tubo plus carbachol bound (EMD-25208 and PDB 7SMT). Source data are provided with this paper.

References

  1. Gharpure, A., Noviello, C. M. & Hibbs, R. E. Progress in nicotinic receptor structural biology. Neuropharmacology 171, 108086 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Albuquerque, E. X., Pereira, E. F., Alkondon, M. & Rogers, S. W. Mammalian nicotinic acetylcholine receptors: from structure to function. Physiol. Rev. 89, 73–120 (2009).

    Article  CAS  PubMed  Google Scholar 

  3. Karlin, A. Emerging structure of the nicotinic acetylcholine receptors. Nat. Rev. Neurosci. 3, 102–114 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Katz, B. & Thesleff, S. A study of the desensitization produced by acetylcholine at the motor end-plate. J. Physiol. 138, 63–80 (1957).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Raghavendra, T. Neuromuscular blocking drugs: discovery and development. J. R. Soc. Med. 95, 363–367 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bennett, M. R. The concept of transmitter receptors: 100 years on. Neuropharmacology 39, 523–546 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. King, H. Curare. Nature 135, 469–470 (1935).

    Article  CAS  Google Scholar 

  8. King, H. 330. Curare alkaloids. Part I. Tubocurarine. J. Chem. Soc. https://doi.org/10.1039/JR9350001381 (1935).

  9. Rahman, M. M. et al. Structure of the native muscle-type nicotinic receptor and inhibition by snake venom toxins. Neuron 106, 952–962 e955 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Rahman, M. M., Worrell, B. T., Stowell, M. H. B. & Hibbs, R. E. in Methods in Enzymology 653 (eds Minor, D. L. & Colecraft, H. M.) 189–206 (Academic Press, 2021).

  11. Raftery, M. A., Hunkapiller, M. W., Strader, C. D. & Hood, L. E. Acetylcholine receptor: complex of homologous subunits. Science 208, 1454–1456 (1980).

    Article  CAS  PubMed  Google Scholar 

  12. Noda, M. et al. Structural homology of Torpedo californica acetylcholine receptor subunits. Nature 302, 528–532 (1983).

    Article  CAS  PubMed  Google Scholar 

  13. Miyazawa, A., Fujiyoshi, Y., Stowell, M. & Unwin, N. Nicotinic acetylcholine receptor at 4.6 A resolution: transverse tunnels in the channel wall. J. Mol. Biol. 288, 765–786 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Changeux, J. P. The nicotinic acetylcholine receptor: a typical ‘allosteric machine’. Philos. Trans. R. Soc. Lond. B. Biol. Sci. https://doi.org/10.1098/rstb.2017.0174 (2018).

  15. Brannigan, G., Henin, J., Law, R., Eckenhoff, R. & Klein, M. L. Embedded cholesterol in the nicotinic acetylcholine receptor. Proc. Natl Acad. Sci. USA 105, 14418–14423 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Baenziger, J. E., Domville, J. A. & Therien, J. P. D. The role of cholesterol in the activation of nicotinic acetylcholine receptors. Curr. Top. Membr. 80, 95–137 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Hamouda, A. K., Sanghvi, M., Sauls, D., Machu, T. K. & Blanton, M. P. Assessing the lipid requirements of the Torpedo californica nicotinic acetylcholine receptor. Biochemistry 45, 4327–4337 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Unwin, N. Protein-lipid architecture of a cholinergic postsynaptic membrane. IUCrJ 7, 852–859 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Sridhar, A. et al. Regulation of a pentameric ligand-gated ion channel by a semi-conserved cationic-lipid binding site. J. Biol. Chem. https://doi.org/10.1016/j.jbc.2021.100899 (2021).

  20. Zhong, W. et al. From ab initio quantum mechanics to molecular neurobiology: a cation-pi binding site in the nicotinic receptor. Proc. Natl Acad. Sci. USA 95, 12088–12093 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Howard, R. J. Elephants in the Dark: Insights and incongruities in pentameric ligand-gated ion channel models. J. Mol. Biol. https://doi.org/10.1016/j.jmb.2021.167128 (2021).

  22. Nemecz, A., Prevost, M. S., Menny, A. & Corringer, P. J. Emerging molecular mechanisms of signal transduction in pentameric ligand-gated ion channels. Neuron 90, 452–470 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Mitra, A., Cymes, G. D. & Auerbach, A. Dynamics of the acetylcholine receptor pore at the gating transition state. Proc. Natl Acad. Sci. USA 102, 15069–15074 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Grandl, J., Danelon, C., Hovius, R. & Vogel, H. Functional asymmetry of transmembrane segments in nicotinic acetylcholine receptors. Eur. Biophys. J. 35, 685–693 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Grosman, C. & Auerbach, A. Asymmetric and independent contribution of the second transmembrane segment 12’ residues to diliganded gating of acetylcholine receptor channels: a single-channel study with choline as the agonist. J. Gen. Physiol. 115, 637–651 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Unwin, N., Miyazawa, A., Li, J. & Fujiyoshi, Y. Activation of the nicotinic acetylcholine receptor involves a switch in conformation of the alpha subunits. J. Mol. Biol. 319, 1165–1176 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Unwin, N. & Fujiyoshi, Y. Gating movement of acetylcholine receptor caught by plunge-freezing. J. Mol. Biol. 422, 617–634 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Noviello, C. M. et al. Structure and gating mechanism of the alpha7 nicotinic acetylcholine receptor. Cell 184, 2121–2134 e2113 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dwyer, T. M., Adams, D. J. & Hille, B. The permeability of the endplate channel to organic cations in frog muscle. J. Gen. Physiol. 75, 469–492 (1980).

    Article  CAS  PubMed  Google Scholar 

  30. Revah, F. et al. Mutations in the channel domain alter desensitization of a neuronal nicotinic receptor. Nature 353, 846–849 (1991).

    Article  CAS  PubMed  Google Scholar 

  31. Yakel, J. L., Lagrutta, A., Adelman, J. P. & North, R. A. Single amino acid substitution affects desensitization of the 5-hydroxytryptamine type 3 receptor expressed in Xenopus oocytes. Proc. Natl Acad. Sci. USA 90, 5030–5033 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Basak, S. et al. Cryo-EM structure of 5-HT3A receptor in its resting conformation. Nat. Commun. 9, 514 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wilson, G. & Karlin, A. Acetylcholine receptor channel structure in the resting, open, and desensitized states probed with the substituted-cysteine-accessibility method. Proc. Natl Acad. Sci. USA 98, 1241–1248 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, Y. et al. Structural basis of human alpha7 nicotinic acetylcholine receptor activation. Cell Res. 31, 713–716 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kumar, A. et al. Mechanisms of activation and desensitization of full-length glycine receptor in lipid nanodiscs. Nat. Commun. 11, 3752 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Auerbach, A. & Akk, G. Desensitization of mouse nicotinic acetylcholine receptor channels. A two-gate mechanism. J. Gen. Physiol. 112, 181–197 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gharpure, A. et al. Agonist selectivity and ion permeation in the alpha3beta4 ganglionic nicotinic receptor. Neuron, https://doi.org/10.1016/j.neuron.2019.07.030 (2019).

  38. Morales-Perez, C. L., Noviello, C. M. & Hibbs, R. E. X-ray structure of the human alpha4beta2 nicotinic receptor. Nature 538, 411–415 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Walsh, R. M. Jr. et al. Structural principles of distinct assemblies of the human alpha4beta2 nicotinic receptor. Nature 557, 261–265 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Polovinkin, L. et al. Conformational transitions of the serotonin 5-HT3 receptor. Nature 563, 275–279 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Basak, S., Gicheru, Y., Rao, S., Sansom, M. S. P. & Chakrapani, S. Cryo-EM reveals two distinct serotonin-bound conformations of full-length 5-HT3A receptor. Nature 563, 270–274 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim, J. J. et al. Shared structural mechanisms of general anaesthetics and benzodiazepines. Nature 585, 303–308 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Masiulis, S. et al. GABAA receptor signalling mechanisms revealed by structural pharmacology. Nature 565, 454–459 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Thompson, M. J., Domville, J. A. & Baenziger, J. E. The functional role of the alphaM4 transmembrane helix in the muscle nicotinic acetylcholine receptor probed through mutagenesis and coevolutionary analyses. J. Biol. Chem. 295, 11056–11067 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Carswell, C. L. et al. Role of the fourth transmembrane alpha helix in the allosteric modulation of pentameric ligand-gated ion channels. Structure 23, 1655–1664 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Butler, A. S. et al. Importance of the C-terminus of the human 5-HT3A receptor subunit. Neuropharmacology 56, 292–302 (2009).

    Article  CAS  PubMed  Google Scholar 

  47. Cory-Wright, J. et al. Aromatic residues in the fourth transmembrane-spanning helix M4 Are important for GABArho receptor function. ACS Chem. Neurosci. 9, 284–290 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Pons, S. et al. Critical role of the C-terminal segment in the maturation and export to the cell surface of the homopentameric alpha 7-5HT3A receptor. Eur. J. Neurosci. 20, 2022–2030 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Jenkinson, D. H. The antagonism between tubocurarine and substances which depolarize the motor end-plate. J. Physiol. 152, 309–324 (1960).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pedersen, S. E. & Papineni, R. V. Interaction of d-tubocurarine analogs with the Torpedo nicotinic acetylcholine receptor. Methylation and stereoisomerization affect site-selective competitive binding and binding to the noncompetitive site. J. Biol. Chem. 270, 31141–31150 (1995).

    Article  CAS  PubMed  Google Scholar 

  51. Moore, M. A. & McCarthy, M. P. Snake venom toxins, unlike smaller antagonists, appear to stabilize a resting state conformation of the nicotinic acetylcholine receptor. Biochim. Biophys. Acta 1235, 336–342 (1995).

    Article  PubMed  Google Scholar 

  52. Sine, S. M. & Steinbach, J. H. Acetylcholine receptor activation by a site-selective ligand: nature of brief open and closed states in BC3H-1 cells. J. Physiol. 370, 357–379 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Steinbach, J. H. & Chen, Q. Antagonist and partial agonist actions of d-tubocurarine at mammalian muscle acetylcholine receptors. J. Neurosci. 15, 230–240 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Neubig, R. R. & Cohen, J. B. Equilibrium binding of [3H]tubocurarine and [3H]acetylcholine by Torpedo postsynaptic membranes: stoichiometry and ligand interactions. Biochemistry 18, 5464–5475 (1979).

    Article  CAS  PubMed  Google Scholar 

  55. Chiara, D. C. & Cohen, J. B. Identification of amino acids contributing to high and low affinity d-tubocurarine sites in the Torpedo nicotinic acetylcholine receptor. J. Biol. Chem. 272, 32940–32950 (1997).

    Article  CAS  PubMed  Google Scholar 

  56. Sine, S. M. & Claudio, T. Gamma- and delta-subunits regulate the affinity and the cooperativity of ligand binding to the acetylcholine receptor. J. Biol. Chem. 266, 19369–19377 (1991).

    Article  CAS  PubMed  Google Scholar 

  57. Chiara, D. C., Xie, Y. & Cohen, J. B. Structure of the agonist-binding sites of the Torpedo nicotinic acetylcholine receptor: affinity-labeling and mutational analyses identify gamma Tyr-111/delta Arg-113 as antagonist affinity determinants. Biochemistry 38, 6689–6698 (1999).

    Article  CAS  PubMed  Google Scholar 

  58. Sine, S. M. Molecular dissection of subunit interfaces in the acetylcholine receptor: identification of residues that determine curare selectivity. Proc. Natl Acad. Sci. USA 90, 9436–9440 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Strecker, G. J. & Jackson, M. B. Curare binding and the curare-induced subconductance state of the acetylcholine receptor channel. Biophys. J. 56, 795–806 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Colquhoun, D., Dreyer, F. & Sheridan, R. E. The actions of tubocurarine at the frog neuromuscular junction. J. Physiol. 293, 247–284 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Sine, S. M. & Taylor, P. Relationship between reversible antagonist occupancy and the functional capacity of the acetylcholine receptor. J. Biol. Chem. 256, 6692–6699 (1981).

    Article  CAS  PubMed  Google Scholar 

  62. Gielen, M., Barilone, N. & Corringer, P. J. The desensitization pathway of GABAA receptors, one subunit at a time. Nat. Commun. 11, 5369 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. 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).

  64. Frauenfeld, J. et al. A saposin-lipoprotein nanoparticle system for membrane proteins. Nat. Methods 13, 345–351 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  68. Morin, A. et al. Collaboration gets the most out of software. eLife 2, e01456 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Celie, P. H. et al. Nicotine and carbamylcholine binding to nicotinic acetylcholine receptors as studied in AChBP crystal structures. Neuron 41, 907–914 (2004).

    Article  CAS  PubMed  Google Scholar 

  70. Brams, M. et al. A structural and mutagenic blueprint for molecular recognition of strychnine and d-tubocurarine by different cys-loop receptors. PLoS Biol. 9, e1001034 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. 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 

  72. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D. Struct. Biol. 74, 531–544 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Williams, C. J. et al. MolProbity: more and better reference data for improved all-atom structure validation. Protein Sci. 27, 293–315 (2018).

    Article  CAS  PubMed  Google Scholar 

  74. Smart, O. S., Neduvelil, J. G., Wang, X., Wallace, B. A. & Sansom, M. S. HOLE: a program for the analysis of the pore dimensions of ion channel structural models. J. Mol. Graph 14, 354–360 (1996). 376.

    Article  CAS  PubMed  Google Scholar 

  75. Wallace, A. C., Laskowski, R. A. & Thornton, J. M. LIGPLOT: a program to generate schematic diagrams of protein-ligand interactions. Protein Eng. 8, 127–134 (1995).

    Article  CAS  PubMed  Google Scholar 

  76. 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 

  77. 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 

  78. Gao, F. et al. Curariform antagonists bind in different orientations to acetylcholine-binding protein. J. Biol. Chem. 278, 23020–23026 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Basta, T. et al. Self-assembled lipid and membrane protein polyhedral nanoparticles. Proc. Natl Acad. Sci. USA 111, 670–674 (2014).

    Article  CAS  PubMed  Google Scholar 

  80. McWilliam, H. et al. Analysis tool web services from the EMBL-EBI. Nucleic Acids Res. 41, W597–W600 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Crooks, G. E., Hon, G., Chandonia, J. M. & Brenner, S. E. WebLogo: a sequence logo generator. Genome Res. 14, 1188–1190 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Sine for Torpedo receptor cDNAs, and C. Noviello and S. Zhu for assistance in cryo-EM sample screening. We are grateful to S. Burke, J.J. Kim, C. Noviello, S. Sine and M. Klymkowsky for critical feedback on the manuscript, D. Borek for model building discussion and A. Sobolevsky for helpful discussion related to measuring recovery from desensitization. Single-particle cryo-EM grids were screened at the University of Texas Southwestern Medical Center Cryo-Electron Microscopy Facility, which is supported by the CPRIT Core Facility Support award no. RP170644. We thank H. Scott for cryo-EM data collection at the PNCC under user proposal nos. 50839 and 51574. A portion of this research was supported by National Institutes of Health (NIH) grant no. U24GM129547 and performed at the PNCC at the Oregon Health & Science University and accessed through EMSL (grid.436923.9), a Department of Energy Office of Science User Facility sponsored by the Office of Biological and Environmental Research. M.M.R. acknowledges a postdoctoral fellowship from the American Heart Association (no. 827474). This work was supported by grants from the NIH (nos. DA042072 to R.E.H., AG061829 to M.H.B.S. and NS120496 to R.E.H. and M.H.B.S.) and the MCDB Neurodegenerative Disease Fund to M.H.B.S.

Author information

Authors and Affiliations

Authors

Contributions

M.M.R. performed the sample preparation and data processing for cryo-EM, structural analysis and drafted the manuscript with R.E.H. J.T. performed the electrophysiology. B.T.W. synthesized the ATM affinity reagent. T.B. and M.L. performed the lipid quantification. M.H.B.S. and R.E.H. assisted in structural analysis and model validation and directed the project. M.M.R., R.E.H. and M.H.B.S. revised the manuscript with input from all other authors.

Corresponding authors

Correspondence to Michael H. B. Stowell or Ryan E. Hibbs.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural and Molecular Biology thanks Michaela Jansen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Florian Ullrich was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team. 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

Extended Data Fig. 1 Relion 3.1 workflow for data processing.

Representative processing approach for apo dataset.

Extended Data Fig. 2 Local and global resolution estimates for cryo-EM maps.

Local resolution is illustrated by variation in map surface color and was estimated using RELION. Global resolution was estimated at FSC = 0.143 (dotted line) from half maps.

Extended Data Fig. 3 Lipid-receptor interactions.

a, Cholesterol (red) and phospholipid (yellow) binding sites in the receptor. α subunit - green, β subunit - khaki, γ subunit - blue, δ subunit – violet. High-affinity cholesterol binding sites are near the MX helices and low-affinity binding sites are near outer membrane leaflet. Among the five subunit interfaces, the high affinity cholesterols occupy the same positions in three interfaces: α/γ, α/δ, and β/α, but not γ/αδ and δ/β. The environment of that site is more polar in γ and δ subunits, where an asparagine replaces I291/α or L297/β (in M3). b, Conservation of the residues in the cholesterol high affinity binding site, residues numbers are according to T. californica α-subunit. Accession numbers for sequences used to determine conservation are given in the Methods.

Extended Data Fig. 4 Transition between resting and desensitized states.

a, Two electrode voltage clamp (TEVC) recording show activation and desensitization of the Torpedo nicotinic receptor by carbachol; 5 mM carbachol was supplemented into the cryo-EM sample for structure determination of the desensitized state. b, Asymmetry in the TMD conformational change between resting and desensitized states; residues at the 16′ position are shown as sticks. c, Conformational changes in loop C of an α-subunit and Loop F of a complementary subunit after agonist and antagonist binding compared to resting state. Carbachol is shown as green spheres. d, Conformational differences in a representative coupling region; representative α and γ subunits.

Extended Data Fig. 5 Permeation pathway and pore profile.

a, Pore diameter comparison of Torpedo in resting and desensitized states vs. other Cys-loop receptors: resting, α7 nAChR (PDB ID: 7EKI), 5-HT3AR (PDB ID: 6BE1), GlyR (PDB ID: 6UBS); and desensitized, α7 nAChR (PDB ID: 7KOQ), α4β2 nAChR (PDB ID: 6CNJ), α3β4 nAChR (PDB ID: 6PV7), 5-HT3AR (PDB ID: 6HIQ), GlyR (PDB ID: 6UBT). b, Permeation pathway cutaway colored by electrostatic potential. c, Water molecules at the 2′ gate of the desensitized state structure. d, −1′ residue orientation in the Torpedo receptor in resting and desensitized states. Transparent surface is experimental density map. e, Two α-subunits superposition of resting state structure. f, Dose response parameters measured by TEVC of WT and mutants. Nonlinear regression was carried out using GraphPad Prism 8. Replicate measurements are from independent oocytes.

Extended Data Fig. 6 Receptor activation and antagonism by carbachol and d-tubo; d-tubo binding sites with corresponding density maps.

a, Two-electrode voltage clamp (TEVC) recording illustrates receptor antagonism by d-tubo. b, d-Tubo at α/γ interface (site 1). c, d-Tubo at α/δ interface (site 2). d, d-Tubo in the pore (site 3). e, d-Tubo at junction of M1, M3 and M4 helices of the αγ subunit (site 4). Corresponding d-tubo densities are shown as semitransparent surfaces. The d-tubo density was very clear at sites 1 and 4. Site 2 density suggested two different orientations of d-tubo and we modeled the best fitted one. The density at site 3 is poorly resolved suggesting multiple orientations of d-tubo in the pore; we used the unsharpened map to model d-tubo there. Density map was contoured at a threshold of 0.02 for site 1, 2 & 4 and 0.013 for site 3 in UCSF chimera.

Extended Data Fig. 7 Orthosteric binding-sites details and carbachol vs. d-tubo complex superposition.

a, b, Electrostatic potential of the two orthosteric ligand-binding sites in the apo form; binding pockets are indicated by asterisks. c, Residue differences between two orthosteric ligand-binding sites; αγ/γ as colored (α- green, γ- blue) and αδ/δ as gray. d, Superposition of desensitized and d-tubo bound structures; d-Tubo model is colored (α, green; β, khaki; γ, blue; δ, violet) and desensitized structure is in gray. (e) Conformational difference in M4 of two α-subunits in resting, desensitized and pure d-tubo bound structures.

Supplementary information

Reporting Summary

Peer Review Information

Supplementary Video 1

Structural transition between resting and desensitized states. Morphing video illustrates changes from resting state to desensitized state to resting state in the top view. α subunit, green; β subunit, khaki; γ subunit, blue and δ subunit, violet.

Supplementary Video 2

Structural transition between resting and desensitized states. Morphing video illustrates changes from resting state to desensitized state to resting state in the side view. α subunit, green; β subunit, khaki; γ subunit, blue and δ subunit, violet.

Supplementary Video 3

d-Tubo interactions at the α–γ interface (site 1). d-Tubo is shown as sticks (orange) and corresponding densities are shown as semitransparent surfaces. Interacting residues are also shown as sticks and colored by subunits: α subunit, green and γ subunit, blue.

Supplementary Video 4

d-Tubo interactions at the α–δ interface (site 2). d-Tubo is shown as sticks (orange) and corresponding densities are shown as semitransparent surfaces. Interacting residues are also shown as sticks and colored by subunits: α subunit, green and δ subunit, violet.

Supplementary Video 5

d-Tubo interactions at the pore (site 3). d-Tubo is shown as sticks (orange) and corresponding densities are shown as semitransparent surfaces. Interacting residues are also shown as sticks and colored by subunits: α subunit, green; β subunit, khaki; γ subunit, blue and δ subunit, violet.

Supplementary Video 6

d-Tubo interactions at the M1, M3 and M4 helices of αγ subunit (site 4). d-Tubo is shown as sticks (orange) and corresponding densities are shown as semitransparent surfaces. Interacting residues are also shown as sticks and colored by subunit: α subunit, green.

Source data

Source Data Fig. 1

Cholesterol and phospholipid assay.

Source Data Fig. 4

Dose–response curve and desensitization recovery.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rahman, M.M., Basta, T., Teng, J. et al. Structural mechanism of muscle nicotinic receptor desensitization and block by curare. Nat Struct Mol Biol 29, 386–394 (2022). https://doi.org/10.1038/s41594-022-00737-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00737-3

This article is cited by

Search

Quick links

Nature Briefing

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

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