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.

  • Letter
  • Published:

Structural principles of distinct assemblies of the human α4β2 nicotinic receptor

Abstract

Fast chemical communication in the nervous system is mediated by neurotransmitter-gated ion channels. The prototypical member of this class of cell surface receptors is the cation-selective nicotinic acetylcholine receptor. As with most ligand-gated ion channels, nicotinic receptors assemble as oligomers of subunits, usually as hetero-oligomers and often with variable stoichiometries1. This intrinsic heterogeneity in protein composition provides fine tunability in channel properties, which is essential to brain function, but frustrates structural and biophysical characterization. The α4β2 subtype of the nicotinic acetylcholine receptor is the most abundant isoform in the human brain and is the principal target in nicotine addiction. This pentameric ligand-gated ion channel assembles in two stoichiometries of α- and β-subunits (2α:3β and 3α:2β). Both assemblies are functional and have distinct biophysical properties, and an imbalance in the ratio of assemblies is linked to both nicotine addiction2,3 and congenital epilepsy4,5. Here we leverage cryo-electron microscopy to obtain structures of both receptor assemblies from a single sample. Antibody fragments specific to β2 were used to ‘break’ symmetry during particle alignment and to obtain high-resolution reconstructions of receptors of both stoichiometries in complex with nicotine. The results reveal principles of subunit assembly and the structural basis of the distinctive biophysical and pharmacological properties of the two different stoichiometries of this receptor.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Fab-assisted cryo-EM resolves heterogeneity in nicotinic receptor subunit assembly.
Fig. 2: Interface classes.
Fig. 3: Agonist and pseudo-agonist binding sites.
Fig. 4: Pore and conductance determinants.

Similar content being viewed by others

References

  1. Nemecz, Á., 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 

  2. Lester, H. A. et al. Nicotine is a selective pharmacological chaperone of acetylcholine receptor number and stoichiometry. Implications for drug discovery. AAPS J. 11, 167–177 (2009).

    CAS  PubMed  Google Scholar 

  3. Moroni, M., Zwart, R., Sher, E., Cassels, B. K. & Bermudez, I. α4β2 nicotinic receptors with high and low acetylcholine sensitivity: pharmacology, stoichiometry, and sensitivity to long-term exposure to nicotine. Mol. Pharmacol. 70, 755–768 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Son, C. D., Moss, F. J., Cohen, B. N. & Lester, H. A. Nicotine normalizes intracellular subunit stoichiometry of nicotinic receptors carrying mutations linked to autosomal dominant nocturnal frontal lobe epilepsy. Mol. Pharmacol. 75, 1137–1148 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Weltzin, M. M., Lindstrom, J. M., Lukas, R. J. & Whiteaker, P. Distinctive effects of nicotinic receptor intracellular-loop mutations associated with nocturnal frontal lobe epilepsy. Neuropharmacology 102, 158–173 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Henderson, R. et al. Tilt-pair analysis of images from a range of different specimens in single-particle electron cryomicroscopy. J. Mol. Biol. 413, 1028–1046 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Scheres, S. H. Processing of structurally heterogeneous cryo-EM data in RELION. Methods Enzymol. 579, 125–157 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Morales-Perez, C. L., Noviello, C. M. & Hibbs, R. E. X-ray structure of the human α4β2 nicotinic receptor. Nature 538, 411–415 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nelson, M. E., Kuryatov, A., Choi, C. H., Zhou, Y. & Lindstrom, J. Alternate stoichiometries of α4β2 nicotinic acetylcholine receptors. Mol. Pharmacol. 63, 332–341 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Carbone, A. L., Moroni, M., Groot-Kormelink, P. J. & Bermudez, I. Pentameric concatenated (α4)2(β2)3 and (α4)32)2 nicotinic acetylcholine receptors: subunit arrangement determines functional expression. Br. J. Pharmacol. 156, 970–981 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. DeDominicis, K. E. et al. The (α4)3(β2)2 stoichiometry of the nicotinic acetylcholine receptor predominates in the rat motor cortex. Mol. Pharmacol. 92, 327–337 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Harpsøe, K. et al. Unraveling the high- and low-sensitivity agonist responses of nicotinic acetylcholine receptors. J. Neurosci. 31, 10759–10766 (2011).

    Article  PubMed  Google Scholar 

  13. Mazzaferro, S. et al. Additional acetylcholine (ACh) binding site at α4/α4 interface of (α4β2)2α4 nicotinic receptor influences agonist sensitivity. J. Biol. Chem. 286, 31043–31054 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shahsavar, A. et al. Acetylcholine-binding protein engineered to mimic the α4-α4 binding pocket in α4β2 nicotinic acetylcholine receptors reveals interface specific interactions important for binding and activity. Mol. Pharmacol. 88, 697–707 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Ahring, P. K. et al. Engineered α4β2 nicotinic acetylcholine receptors as models for measuring agonist binding and effect at the orthosteric low-affinity α4-α4 interface. Neuropharmacology 92, 135–145 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Tapia, L., Kuryatov, A. & Lindstrom, J. Ca2+ permeability of the (α4)32)2 stoichiometry greatly exceeds that of (α4)2(β2)3 human acetylcholine receptors. Mol. Pharmacol. 71, 769–776 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Hansen, S. B., Wang, H. L., Taylor, P. & Sine, S. M. An ion selectivity filter in the extracellular domain of Cys-loop receptors reveals determinants for ion conductance. J. Biol. Chem. 283, 36066–36070 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Imoto, K. et al. Rings of negatively charged amino acids determine the acetylcholine receptor channel conductance. Nature 335, 645–648 (1988).

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Livesey, M. R. et al. Structural determinants of Ca2+ permeability and conduction in the human 5-hydroxytryptamine type 3A receptor. J. Biol. Chem. 283, 19301–19313 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kelley, S. P., Dunlop, J. I., Kirkness, E. F., Lambert, J. J. & Peters, J. A. A cytoplasmic region determines single-channel conductance in 5-HT3 receptors. Nature 424, 321–324 (2003).

    Article  ADS  CAS  PubMed  Google Scholar 

  21. 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 

  22. 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  PubMed  Google Scholar 

  23. Hattori, M., Hibbs, R. E. & Gouaux, E. A fluorescence-detection size-exclusion chromatography-based thermostability assay for membrane protein precrystallization screening. Structure 20, 1293–1299 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Miller, P. S. et al. Structural basis for GABAreceptor potentiation by neurosteroids. Nat. Struct. Mol. Biol. 24, 986–992 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Laverty, D. et al. Crystal structures of a GABAA-receptor chimera reveal new endogenous neurosteroid-binding sites. Nat. Struct. Mol. Biol. 24, 977–985 (2017).

    Article  CAS  PubMed  Google Scholar 

  26. Hamouda, A. K., Chiara, D. C., Sauls, D., Cohen, J. B. & Blanton, M. P. Cholesterol interacts with transmembrane α-helices M1, M3, and M4 of the Torpedo nicotinic acetylcholine receptor: photolabeling studies using [3H]Azicholesterol. Biochemistry 45, 976–986 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Corbin, J., Wang, H. H. & Blanton, M. P. Identifying the cholesterol binding domain in the nicotinic acetylcholine receptor with [125I]azido-cholesterol. Biochim. Biophys. Acta 1414, 65–74 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Morales-Perez, C. L., Noviello, C. M. & Hibbs, R. E. Manipulation of subunit stoichiometry in heteromeric membrane proteins. Structure 24, 797–805 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Grant, T. & Grigorieff, N. Measuring the optimal exposure for single particle cryo-EM using a 2.6 Å reconstruction of rotavirus VP6. eLife 4, e06980 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fernandez-Leiro, R. & Scheres, S. H. W. A pipeline approach to single-particle processing in RELION. Acta Crystallogr. D 73, 496–502 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Scheres, S. H. A Bayesian view on cryo-EM structure determination. J. Mol. Biol. 415, 406–418 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nat. Methods 11, 63–65 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. 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 

  35. Biasini, M. et al. SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information. Nucleic Acids Res. 42, W252–W258, (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  37. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Amunts, A. et al. Structure of the yeast mitochondrial large ribosomal subunit. Science 343, 1485–1489 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Winn, M. D. et al. Overview of the CCP4 suite and current developments. Acta Crystallogr. D 67, 235–242 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. The UniProt Consortium. UniProt: the universal protein knowledgebase. Nucleic Acids Res. 45, D158–D169 (2017).

    Article  Google Scholar 

  41. Pei, J., Kim, B. H. & Grishin, N. V. PROMALS3D: a tool for multiple protein sequence and structure alignments. Nucleic Acids Res. 36, 2295–2300 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Krissinel, E. & Henrick, K. Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 372, 774–797 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. 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, 376 (1996).

    Article  CAS  PubMed  Google Scholar 

  44. Hayward, S. & Lee, R. A. Improvements in the analysis of domain motions in proteins from conformational change: DynDom version 1.50. J. Mol. Graph. Model. 21, 181–183 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. 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 

  46. Baker, N. A., Sept, D., Joseph, S., Holst, M. J. & McCammon, J. A. Electrostatics of nanosystems: application to microtubules and the ribosome. Proc. Natl Acad. Sci. USA 98, 10037–10041 (2001).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  48. Venkatachalan, S. P. et al. Optimized expression vector for ion channel studies in Xenopus oocytes and mammalian cells using alfalfa mosaic virus. Pflugers Arch. 454, 155–163 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Galzi, J. L., Bertrand, S., Corringer, P. J., Changeux, J. P. & Bertrand, D. Identification of calcium binding sites that regulate potentiation of a neuronal nicotinic acetylcholine receptor. EMBO J. 15, 5824–5832 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Cawley at OHSU for production of monoclonal antibodies, X. Bai for EM discussion, Y. Jiang for the use of the oocyte rig, C. Noviello for assistance in EM data collection and all members of the Hibbs Laboratory and M. Horvath for discussion. Cryo-EM data were collected at the UT Southwestern Medical Center Cryo-Electron Microscopy Facility, which is funded in part by CPRIT Core Facility Support Award RP170644. We thank D. Nicastro and Z. Chen for support in facility access and data acquisition and W. Chiu for cryo-EM training and resources in the National Center for Macromolecular Imaging (NCMI) at Baylor College of Medicine. NCMI is supported by NIH Grants P41GM103832 and R01GM079429. R.M.W. acknowledges support from the Sara and Frank McKnight Fund for Biochemical Research and the NIH (T32GM008203). R.E.H. is supported by a McKnight Scholar Award, The Welch Foundation (I-1812) and the NIH (DA037492, DA042072, and NS095899).

Reviewer information

Nature thanks R. Aricescu, R. Dutzler and M. Jansen for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

R.M.W. expressed and purified the protein, made the EM samples, collected the EM data, processed the EM data with assistance from S.-H.R., built and refined the models and performed the binding assays. A.G. processed an initial EM dataset revealing the presence of two stoichiometries. C.L.M.-P. performed the mAb and Fab characterization. J.T. performed the electrophysiology. R.M.W. and R.E.H. wrote the manuscript with assistance from S.-H.R.; R.E.H. oversaw all aspects of the project. All authors gave feedback on the manuscript.

Corresponding author

Correspondence to Ryan E. Hibbs.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Biochemistry, binding, electrophysiology and α4β2–Fab interactions.

a, Size-exclusion chromatogram of α4β2–Fab complex and SDS–PAGE analysis of complex purification (V0, void volume). b, Saturation binding experiments with [3H]-nicotine and a mixture of receptor subunit stoichiometries. Grey squares denote samples with receptor plus Fab and blue circles denote samples with receptor alone. Receptor alone Kd = 7.7 nM (95% confidence interval (CI), 6.9–8.6 nM) and receptor plus Fab Kd = 17.0 nM (95% CI, 10.2–26.6 nM). Receptor alone and receptor plus Fab both exhibited a Hill slope of approximately 1 (1.14 and 1.09, respectively). Plotted results are from a representative experiment performed in triplicate. c, Representative two-electrode voltage clamp (−60 mV) recordings of oocytes injected with cRNAs for the α4 and β2 subunits in ratios to bias assembly,1α:5β (top two traces) and 5α:1β (bottom two traces) for the 2α:3β and 3α:2β assemblies, respectively. The experiments were performed in the presence and absence of Fab to assess the effect of Fab on receptor gating. Each oocyte was perfused with 1 µM nicotine solution for about 2 min, washed with bath solution for 10 min and then perfused for about 2 min with 1 µM nicotine ± Fab. For samples where Fab was included, 1 μM final [Fab] was added directly to the oocyte bath following the first perfusion and allowed to incubate for 1.5 min before perfusing with a second solution containing 1 µM nicotine + 0.1 μM Fab. d, Bar graph quantifying peak currents before and after adding Fab. Currents were normalized to the amplitude of the first nicotine application. Appreciable current rundown was observed (assessed by applying nicotine without Fab as the second application). Change in peak current is similar in the presence and absence of Fab for both stoichiometries, suggesting no substantial effect on gating. n, number of oocytes; error bars, s.d. from mean. eh, Fab–β-subunit interactions at the α–β, β–β, α–β, and α–α interfaces, respectively. With one exception, the Fab molecules interact exclusively with a single β-subunit. The exception is at the β–β interface, where the Fab on the complementary β-subunit forms one potential interaction with the preceding β-subunit. This interaction is displayed as a dashed line (f, inset). The conformation of the principal loop C at the β–β interface is indistinguishable from those where Fab is not interacting, suggesting that Fab does not affect the loop C conformation. Subunits are coloured as in Fig. 1

Extended Data Fig. 2 Cryo-EM image processing procedure.

a, Representative micrograph of the α4β2–Fab complex (scale bar, 50 nm). Boxed regions on the right are magnified to show representative particle images (top/bottom and side views). b, Images of selected 2D classes from reference-free 2D classification by RELION. White arrowheads, Fab fragments. c, Overview of the image processing procedure (see Methods).

Extended Data Fig. 3 Three-dimensional reconstructions of the 2α:3β and 3α:2β assemblies.

a, Angular distribution histogram of 2α:3β assembly projections. b, FSC of 2α:3β assembly maps before (black) and after (blue and red) masking. Two soft masks were used: one (red) mask that included only the receptor and one (blue) mask that included the whole 2α:3β–Fab complex. When a mask was used, the FSC curve was corrected for masking effects during the RELION post-processing procedure. Masks used in FSC calculations are shown in c for the whole 2α:3β–Fab complex (top) and receptor only (bottom), superimposed on the map in question. d, Local resolution of the 2α3β–Fab reconstruction estimated by ResMap33. Shown is the combined map, which has not been sharpened or filtered. eh, As in ad but for the 3α:2β assembly.

Extended Data Fig. 4 3α:2β assembly model-map validation.

a, b, FSC curves for cross-validation between the maps and the models with (a) and without Fab fragments (b) for the 3α:2β assembly. Curves for final model versus summed map (full) in black, for model versus half map (work) in red, and for model versus half map not used for refinement (free) in blue. For validation of receptor alone (without Fab), maps were segmented to exclude the Fab fragments. c, EM density segments of the 3α:2β assembly for a representative α4 subunit. Density map and model for α4 subunit (left). Representative density for extracellular and transmembrane spanning regions (right). Regions are numbered and helices are labelled. Maps were sharpened with a single B factor at −150 Å2. d, As in c, but for a representative β2 subunit.

Extended Data Fig. 5 2α:3β assembly model–map validation.

a, b, FSC curves for cross-validation between the maps and the models with (a) and without Fab fragments (b) for the 2α:3β assembly. Curves for final model versus summed map (full) in black, for model versus half map (work) in red, and for model versus half map not used for refinement (free) in blue. For validation of receptor alone (without Fab) maps segmented to exclude the Fab fragments were used. c, EM density segments of the 2α:3β assembly for a representative α4 subunit. Density map and model for α4 subunit (left). Representative density for extracellular and transmembrane spanning regions (right). Regions are numbered and helices are labelled. Maps were sharpened with a single B factor at −170 Å2. d, As in c, but for a representative β2 subunit.

Extended Data Fig. 6 Comparison of map and model of 2α3β by X-ray crystallography at 3.9 Å resolution and by cryo-EM at 3.4 Å resolution, and comparison of subunit backbone conformations.

a, The X-ray electron density map (left) and cryo-EM map (right) are displayed in a linear representation (unwrapped) to illustrate the overall resolvability of all five subunits: two α4 subunits (green) and three β2 subunits (blue). Red circles indicate the locations of crystal contacts on the X-ray electron density map (left) or Fab binding sites on cryo-EM density map (right). b, X-ray structure of the 2α:3β assembly (PDB accession 5KXI8) after alignment to the cryo-EM structure (this study) shown in ribbon representation and coloured according to Cα r.m.s.d. values (from blue (low) to red (high)). While the areas involved in crystal contacts displayed relatively higher r.m.s.d. values for Cα ( > 2 Å), the Fab binding location shows r.m.s.d. values for Cα of < 1 Å. This result implies that zones of protein–protein contacts in the crystal lattice may substantially affect local structure on the pentamer, but Fab binding in the cryo-EM study did not result in substantial structural change, consistent with the binding assays and electrophysiological results (Extended Data Fig. 1). c, Fit of X-ray model (5KXI) to X-ray electron density map (left) and cryo-EM model to cryo-EM density map (middle) shown as selected areas for high r.m.s.d. (loop C) and low r.m.s.d. (M2 helix), respectively. Right column shows superposition of X-ray and cryo-EM model at the selected areas. dh, Subunit superpositions. d, Comparison of all α4 subunits in the 2α:3β and 3α:2β assemblies reveals no substantial conformational differences in α-carbon backbones. e, Superposition of all β2 subunits in the two assemblies reveals a domain motion of the TMD relative to the ECD of the subunits that comprise the β–β interface. The rotation of 4.4° between the two β-subunits that compose the β–β interface is denoted by black double-headed arrows. The principal β-subunit (β2 + ) is coloured magenta while the complementary β-subunit (β2 −) is shown in cyan. fh, Superposition of α4 (green) with the three distinct β2 conformations: principal β–β (magenta) complementary β–β (cyan) and α–β–α (grey). The conformation of the principal β–β (magenta) subunit is distinct from every other subunit in the 2α:3β and 3α:2β assemblies. The conformation of the complementary (−) subunit of the β–β interface is rotated towards the pore axis, accommodating the marked conformational change of the principal β2 subunit away from the pore axis. This conformational change results in the complementary β–β (cyan) subunit adopting a backbone conformation similar to α4 subunits.

Extended Data Fig. 7 Fenestration at β–β interface.

Extracellular fenestration unique to this class of interface. Inset of fenestration with distances indicated by dashed lines and side chains surrounding fenestration shown as sticks. Two conserved glutamic acid residues in close proximity to this gap at the β–β interface (yellow) have been implicated in Ca2+ potentiation of nicotinic receptors49.

Extended Data Fig. 8 Basis of α4β2 heteromeric assembly.

a, Cartoon representation of top view of observed (2α:3β and 3α:2β) and computational (4α:1β, 1α:4β, 5α and 5β) α4β2 pentameric assemblies. Assemblies on the top row (3α:2β, 4α:1β, and 5α) are arranged by increasing α4 composition. Assemblies on the bottom row (2α:3β, 1α:4β and 5β) are arranged by increasing β2 composition. Agonist binding sites are denoted by red circles. Buried interface areas (Å2) for the interfaces analysed in b, c are listed below each pentameric assembly. Subunits are coloured in a as described below for b, c. b, Superposition of α–α from 3α:2β and final α–α interface in 5α homopentamer. Principal subunits (grey) were superimposed to highlight differences at the interface. Complementary (−) subunits are coloured light green for α–α from 3α:2β and dark green for the α–α interface in the 5α homopentamer. Sticks are displayed for amino acid clashes that have greater than 1.5 Å overlap assessed by Molprobity. c, Superposition of the β–β interface from 2α:3β and final β–β interface in the 5β homopentamer. Principal subunits (grey) were superimposed to highlight differences at the interface. Complementary (−) subunits are coloured light blue for the β–β interface from the 2α:3β assembly and dark blue for the final β–β interface in the 5β homopentamer.

Extended Data Fig. 9 Ligand binding site comparisons.

a, Stereo images of nicotine bound at the α–α interface. Subunits are coloured as in Fig. 1. Density (purple mesh) is displayed at a threshold of 0.0306 in Chimera. Electrostatic interactions denoted as dashed magenta lines. Binding pocket residues and nicotine displayed as sticks. b, Ligplot of nicotine bound at the α–α and α–β interfaces (left and right, respectively). Residue contacts within 4.5 Å are displayed in red, residue displayed in purple (His116) is within 5.5 Å. c, Stereo images of nicotine bound at the α–β interface. Density (purple mesh) is displayed at a threshold of 0.0306 in Chimera. Hydrogen bonds denoted as dashed magenta lines. Binding pocket residues and nicotine displayed as sticks. d, e, Stereo image overlays of nicotine bound at the α–α (magenta) and α–β interfaces (cyan). Ligand density is displayed as transparent surfaces. Density is displayed at a threshold of 0.406 for the α–α (magenta) and 0.500 for the α–β (cyan) interface in Chimera. e, Ribbon removed for clarity, rotated 180° relative to d. Key residues implicated in the difference in sensitivity between the classical α–β neurotransmitter binding site (high sensitivity V111/F119/L121) and the unique α–α binding site (low sensitivity H116/Q124/T126) are labelled.

Extended Data Fig. 10 Comparison of ions located in the pore above the constriction point.

a, Top view of EM density map of ion in the pore of the 3α:2β assembly. Density (blue mesh) is displayed at a threshold of 0.027 in Chimera. Subunits are coloured as in Fig. 1. Modelled Na+ ion is represented as a purple sphere. Nearest residues on the M2 α-helices are indicated. b, As a, but for the 2α:3β assembly. c, Side view of EM density map of ion in the pore of the 3α:2β assembly. Two subunits (one α and one β) removed for clarity. Colours and residues indicated as in a. d, As c, but for the 2α:3β assembly. e, Superposition of constriction region of 2α:3β (light blue) and 3α:2β (light green) assemblies.

Extended Data Fig. 11 Comparison of putative cholesterol binding orientations.

a, b, EM density map showing cholesterol sites from all TMD interfaces in the 3α:2β assembly and 2α:3β assembly, respectively. Pentagons (top) are coloured to illustrate subunits composing the displayed interface. c, Stereo image of representative cholesterol binding site with α-subunit on principal side ( + ). When the principal side ( + ) is an α4 subunit, both molecules tilt towards the principal face. One residue from the complementary side β2 (−), Y232, contacts both cholesterol molecules at the α–β interface (at the α–α interface the equivalent residue Y240 in the complementary (−) α4 contacts both cholesterol molecules at the interface; not shown). d, Stereo image of representative cholesterol binding site with β-subunit on principal side ( + ). When the principal side ( + ) is a β2 subunit, both cholesterol molecules are oriented orthogonal to the plane of the membrane. One residue from the principal ( + ) β2 subunit, V288, makes contacts with both cholesterol molecules at the β–α and β–β (not shown) interfaces. Cholesterol and interacting side chains are shown as sticks. Density maps in panels ad are displayed at a threshold of 0.025 in Chimera. e, Alignment of nicotinic receptor subunits for region encompassing putative cholesterol sites. Residues in cholesterol binding pocket are boxed and amino acid position implicated in differential cholesterol binding is highlighted in magenta. Uniprot accession IDs are provided40. Residues mapped using a photoreactive cholesterol analogue in Torpedo californica (TC P02710)26,27 are highlighted in cyan. All other sequences are from Homo sapiens (HS P43681, P11230, P17787, Q05901, P30926 for α4 and β1–4, respectively). f, Superposition of β–α (yellow) from α4β2 and α1–α1 (blue) GABAA–GLIC chimaera (PDB accession 5OSC25) TMD interface to compare putative binding sites. Pregnenolone sulfate, CHS and cholesterol are shown as sticks.

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

Supplementary information

Supplementary Information

The uncropped gel shown in Extended Data Fig. 1

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Walsh, R.M., Roh, SH., Gharpure, A. et al. Structural principles of distinct assemblies of the human α4β2 nicotinic receptor. Nature 557, 261–265 (2018). https://doi.org/10.1038/s41586-018-0081-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0081-7

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

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