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.

Channel opening and gating mechanism in AMPA-subtype glutamate receptors

Abstract

AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-subtype ionotropic glutamate receptors mediate fast excitatory neurotransmission throughout the central nervous system. Gated by the neurotransmitter glutamate, AMPA receptors are critical for synaptic strength, and dysregulation of AMPA receptor-mediated signalling is linked to numerous neurological diseases. Here we use cryo-electron microscopy to solve the structures of AMPA receptor–auxiliary subunit complexes in the apo, antagonist- and agonist-bound states and determine the iris-like mechanism of ion channel opening. The ion channel selectivity filter is formed by the extended portions of the re-entrant M2 loops, while the helical portions of M2 contribute to extensive hydrophobic interfaces between AMPA receptor subunits in the ion channel. We show how the permeation pathway changes upon channel opening and identify conformational changes throughout the entire AMPA receptor that accompany activation and desensitization. Our findings provide a framework for understanding gating across the family of ionotropic glutamate receptors and the role of AMPA receptors in excitatory neurotransmission.

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

Figure 1: GluA2–GSG1L and ion channel structure.
Figure 2: Cryo-EM of an activated GluA2–STZ complex.
Figure 3: Structure of the GluA2–STZ complex.
Figure 4: Ion channel pore in open, closed and desensitized states.
Figure 5: Structural rearrangements during gating.

References

  1. Kumar, J. & Mayer, M. L. Functional insights from glutamate receptor ion channel structures. Annu. Rev. Physiol. 75, 313–337 (2013)

    Article  CAS  PubMed  Google Scholar 

  2. Traynelis, S. F. et al. Glutamate receptor ion channels: structure, regulation, and function. Pharmacol. Rev. 62, 405–496 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bowie, D. Ionotropic glutamate receptors & CNS disorders. CNS Neurol. Disord. Drug Targets 7, 129–143 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chater, T. E. & Goda, Y. The role of AMPA receptors in postsynaptic mechanisms of synaptic plasticity. Front. Cell. Neurosci. 8, 401 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  5. Henley, J. M. & Wilkinson, K. A. Synaptic AMPA receptor composition in development, plasticity and disease. Nat. Rev. Neurosci. 17, 337–350 (2016)

    Article  CAS  PubMed  Google Scholar 

  6. Sobolevsky, A. I. Structure and gating of tetrameric glutamate receptors. J. Physiol. (Lond.) 593, 29–38 (2015)

    Article  CAS  Google Scholar 

  7. Mayer, M. L. Structural biology of glutamate receptor ion channel complexes. Curr. Opin. Struct. Biol. 41, 119–127 (2016)

    Article  CAS  PubMed  Google Scholar 

  8. Sobolevsky, A. I., Rosconi, M. P. & Gouaux, E. X-ray structure, symmetry and mechanism of an AMPA-subtype glutamate receptor. Nature 462, 745–756 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Chen, L., Dürr, K. L. & Gouaux, E. X-ray structures of AMPA receptor-cone snail toxin complexes illuminate activation mechanism. Science 345, 1021–1026 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dürr, K. L. et al. Structure and dynamics of AMPA receptor GluA2 in resting, pre-open, and desensitized states. Cell 158, 778–792 (2014)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Yelshanskaya, M. V., Li, M. & Sobolevsky, A. I. Structure of an agonist-bound ionotropic glutamate receptor. Science 345, 1070–1074 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yelshanskaya, M. V. et al. Structural bases of noncompetitive inhibition of AMPA subtype ionotropic glutamate receptors by antiepileptic drugs. Neuron 91, 1305–1315 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Herguedas, B. et al. Structure and organization of heteromeric AMPA-type glutamate receptors. Science 352, aad3873 (2016)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Meyerson, J. R. et al. Structural mechanism of glutamate receptor activation and desensitization. Nature 514, 328–334 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Twomey, E. C., Yelshanskaya, M. V., Grassucci, R. A., Frank, J. & Sobolevsky, A. I. Elucidation of AMPA receptor-stargazin complexes by cryo-electron microscopy. Science 353, 83–86 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Twomey, E. C., Yelshanskaya, M. V., Grassucci, R. A., Frank, J. & Sobolevsky, A. I. Structural bases of desensitization in AMPA receptor–auxiliary subunit complexes. Neuron 94, 569–580.e5 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhao, Y., Chen, S., Yoshioka, C., Baconguis, I. & Gouaux, E. Architecture of fully occupied GluA2 AMPA receptor–TARP complex elucidated by cryo-EM. Nature 536, 108–111 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Doyle, D. A. et al. The structure of the potassium channel: molecular basis of K+ conduction and selectivity. Science 280, 69–77 (1998)

    ADS  CAS  PubMed  Google Scholar 

  19. Cao, E., Liao, M., Cheng, Y. & Julius, D. TRPV1 structures in distinct conformations reveal activation mechanisms. Nature 504, 113–118 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Huettner, J. E. Glutamate receptor pores. J. Physiol. (Lond.) 593, 49–59 (2015)

    Article  CAS  Google Scholar 

  21. Wilding, T. J., Lopez, M. N. & Huettner, J. E. Radial symmetry in a chimeric glutamate receptor pore. Nat. Commun. 5, 3349 (2014)

    Article  ADS  PubMed  CAS  Google Scholar 

  22. Lopez, M. N., Wilding, T. J. & Huettner, J. E. Q/R site interactions with the M3 helix in GluK2 kainate receptor channels revealed by thermodynamic mutant cycles. J. Gen. Physiol. 142, 225–239 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Chang, P. et al. Seizure control by decanoic acid through direct AMPA receptor inhibition. Brain 139, 431–443 (2016)

    Article  PubMed  Google Scholar 

  24. Chang, P. et al. Seizure control by derivatives of medium chain fatty acids associated with the ketogenic diet show novel branching-point structure for enhanced potency. J. Pharmacol. Exp. Ther. 352, 43–52 (2015)

    Article  PubMed  CAS  Google Scholar 

  25. Priel, A. et al. Stargazin reduces desensitization and slows deactivation of the AMPA-type glutamate receptors. J. Neurosci. 25, 2682–2686 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Tomita, S. et al. Stargazin modulates AMPA receptor gating and trafficking by distinct domains. Nature 435, 1052–1058 (2005)

    Article  ADS  CAS  PubMed  Google Scholar 

  27. MacLean, D. M., Ramaswamy, S. S., Du, M., Howe, J. R. & Jayaraman, V. Stargazin promotes closure of the AMPA receptor ligand-binding domain. J. Gen. Physiol. 144, 503–512 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Patneau, D. K., Vyklicky, L. Jr & Mayer, M. L. Hippocampal neurons exhibit cyclothiazide-sensitive rapidly desensitizing responses to kainate. J. Neurosci. 13, 3496–3509 (1993)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yelshanskaya, M. V., Saotome, K., Singh, A. K. & Sobolevsky, A. I. Probing intersubunit interfaces in AMPA-subtype ionotropic glutamate receptors. Sci. Rep. 6, 19082 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jiang, Y. et al. The open pore conformation of potassium channels. Nature 417, 523–526 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  31. Long, S. B., Campbell, E. B. & Mackinnon, R. Crystal structure of a mammalian voltage-dependent Shaker family K+ channel. Science 309, 897–903 (2005)

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Sobolevsky, A. I., Yelshansky, M. V. & Wollmuth, L. P. The outer pore of the glutamate receptor channel has 2-fold rotational symmetry. Neuron 41, 367–378 (2004)

    Article  CAS  PubMed  Google Scholar 

  33. Yelshanskaya, M. V., Mesbahi-Vasey, S., Kurnikova, M. G. & Sobolevsky, A. I. Role of the ion channel extracellular collar in AMPA receptor gating. Sci. Rep. 7, 1050 (2017)

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  34. Armstrong, N. & Gouaux, E. Mechanisms for activation and antagonism of an AMPA-sensitive glutamate receptor: crystal structures of the GluR2 ligand binding core. Neuron 28, 165–181 (2000)

    Article  CAS  PubMed  Google Scholar 

  35. Sun, Y. et al. Mechanism of glutamate receptor desensitization. Nature 417, 245–253 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  36. Armstrong, N., Jasti, J., Beich-Frandsen, M. & Gouaux, E. Measurement of conformational changes accompanying desensitization in an ionotropic glutamate receptor. Cell 127, 85–97 (2006)

    Article  CAS  PubMed  Google Scholar 

  37. Salussolia, C. L. et al. Interaction of the M4 segment with other transmembrane segments is required for surface expression of mammalian α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. J. Biol. Chem. 286, 40205–40218 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Salussolia, C. L. et al. A eukaryotic specific transmembrane segment is required for tetramerization in AMPA receptors. J. Neurosci. 33, 9840–9845 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gan, Q., Dai, J., Zhou, H. X. & Wollmuth, L. P. The transmembrane domain mediates tetramerization of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. J. Biol. Chem. 291, 6595–6606 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Shaikh, S. A. et al. Stargazin modulation of AMPA receptors. Cell Reports 17, 328–335 (2016)

    Article  CAS  PubMed  Google Scholar 

  41. Leuschner, W. D. & Hoch, W. Subtype-specific assembly of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor subunits is mediated by their N-terminal domains. J. Biol. Chem. 274, 16907–16916 (1999)

    Article  CAS  PubMed  Google Scholar 

  42. Papadakis, M., Hawkins, L. M. & Stephenson, F. A. Appropriate NR1-NR1 disulfide-linked homodimer formation is requisite for efficient expression of functional, cell surface N-methyl-d-aspartate NR1/NR2 receptors. J. Biol. Chem. 279, 14703–14712 (2004)

    Article  CAS  PubMed  Google Scholar 

  43. Ayalon, G., Segev, E., Elgavish, S. & Stern-Bach, Y. Two regions in the N-terminal domain of ionotropic glutamate receptor 3 form the subunit oligomerization interfaces that control subtype-specific receptor assembly. J. Biol. Chem. 280, 15053–15060 (2005)

    Article  CAS  PubMed  Google Scholar 

  44. Ayalon, G. & Stern-Bach, Y. Functional assembly of AMPA and kainate receptors is mediated by several discrete protein-protein interactions. Neuron 31, 103–113 (2001)

    Article  CAS  PubMed  Google Scholar 

  45. Jin, R. et al. Crystal structure and association behaviour of the GluR2 amino-terminal domain. EMBO J. 28, 1812–1823 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Watson, J. F., Ho, H. & Greger, I. H. Synaptic transmission and plasticity require AMPA receptor anchoring via its N-terminal domain. eLife 6, e23024 (2017)

    Article  PubMed  PubMed Central  Google Scholar 

  47. Cull-Candy, S. G. & Usowicz, M. M. Multiple-conductance channels activated by excitatory amino acids in cerebellar neurons. Nature 325, 525–528 (1987)

    Article  ADS  CAS  PubMed  Google Scholar 

  48. Jahr, C. E. & Stevens, C. F. Glutamate activates multiple single channel conductances in hippocampal neurons. Nature 325, 522–525 (1987)

    Article  ADS  CAS  PubMed  Google Scholar 

  49. Rosenmund, C., Stern-Bach, Y. & Stevens, C. F. The tetrameric structure of a glutamate receptor channel. Science 280, 1596–1599 (1998)

    Article  ADS  CAS  PubMed  Google Scholar 

  50. Smith, T. C. & Howe, J. R. Concentration-dependent substate behavior of native AMPA receptors. Nat. Neurosci. 3, 992–997 (2000)

    Article  CAS  PubMed  Google Scholar 

  51. Poon, K., Ahmed, A. H., Nowak, L. M. & Oswald, R. E. Mechanisms of modal activation of GluA3 receptors. Mol. Pharmacol. 80, 49–59 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Prieto, M. L. & Wollmuth, L. P. Gating modes in AMPA receptors. J. Neurosci. 30, 4449–4459 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Swanson, G. T., Kamboj, S. K. & Cull-Candy, S. G. Single-channel properties of recombinant AMPA receptors depend on RNA editing, splice variation, and subunit composition. J. Neurosci. 17, 58–69 (1997)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kohda, K., Wang, Y. & Yuzaki, M. Mutation of a glutamate receptor motif reveals its role in gating and δ2 receptor channel properties. Nat. Neurosci. 3, 315–322 (2000)

    Article  CAS  PubMed  Google Scholar 

  55. Goehring, A. et al. Screening and large-scale expression of membrane proteins in mammalian cells for structural studies. Nat. Protocols 9, 2574–2585 (2014)

    Article  CAS  PubMed  Google Scholar 

  56. Kawate, T. & Gouaux, E. Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins. Structure 14, 673–681 (2006)

    Article  CAS  PubMed  Google Scholar 

  57. Russo, C. J. & Passmore, L. A. Electron microscopy: ultrastable gold substrates for electron cryomicroscopy. Science 346, 1377–1380 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Suloway, C. et al. Automated molecular microscopy: the new Leginon system. J. Struct. Biol. 151, 41–60 (2005)

    Article  CAS  PubMed  Google Scholar 

  59. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kimanius, D., Forsberg, B. O., Scheres, S. H. & Lindahl, E. Accelerated cryo-EM structure determination with parallelisation using GPUs in RELION-2. eLife 5, e18722 (2016)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Scheres, S. H. & Chen, S. Prevention of overfitting in cryo-EM structure determination. Nat. Methods 9, 853–854 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chen, S. et al. High-resolution noise substitution to measure overfitting and validate resolution in 3D structure determination by single particle electron cryomicroscopy. Ultramicroscopy 135, 24–35 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. 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 

  65. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D 68, 352–367 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Tang, G. et al. EMAN2: an extensible image processing suite for electron microscopy. J. Struct. Biol. 157, 38–46 (2007)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Kao for computational support, A. des Georges, I. S. Fernandez, M. Fislage and A. K. Singh for processing advice. E.C.T. is supported by National Institutes of Health (NIH) F31 NS093838. A.I.S. is supported by the NIH (R01 NS083660, R01 CA206573), the Pew Scholar Award in Biomedical Sciences, and the Irma T. Hirschl Career Scientist Award. J.F. is supported by the Howard Hughes Medical Institute and the NIH (R01 GM029169). Cryo-EM data were collected at the Columbia University Medical Center cryo-EM facility and at the Simons Electron Microscopy Center and National Resource for Automated Molecular Microscopy located at the New York Structural Biology Center, supported by grants from the Simons Foundation (349247), NYSTAR, and the NIH National Institute of General Medical Sciences (GM103310).

Author information

Authors and Affiliations

Authors

Contributions

E.C.T. designed the constructs, prepared the protein samples, carried out cryo-EM data collection and processing, built models, analysed data and wrote the manuscript. M.V.Y. carried out electrophysiology experiments, assisted in protein production and edited the manuscript. R.A.G. assisted in cryo-EM data collection. J.F. advised on the cryo-EM workflow and provided funding. A.I.S. supervised the project, built models, analysed data, wrote the manuscript and provided funding. E.C.T. and A.I.S. designed the project.

Corresponding author

Correspondence to Alexander I. Sobolevsky.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks M. Mayer, S. Traynelis and the other anonymous reviewer(s) for their contribution to the peer review of this work.

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 Figure 1 FSC curves for cryo-EM reconstructions.

FSC curves calculated between half-maps for GluA2–GSG1LZK-1, GluA2–GSG1LZK-2, GluA2–GSG1Lapo-1, GluA2–GSG1Lapo-2 and GluA2–STZGlu+CTZ cryo-EM reconstructions, as well as for the GluA2–STZGlu+CTZ TMD reconstruction from directed refinement. The dashed line indicates FSC = 0.143.

Extended Data Figure 2 Local resolution and fitting of cryo-EM maps.

ap, Local resolution calculated using Resmap and two unfiltered halves of the reconstruction for GluA2–GSG1LZK-1, GluA2–GSG1LZK-2, GluA2–GSG1Lapo-1, GluA2–GSG1Lapo-2 and GluA2–STZGlu+CTZ structures viewed parallel to the membrane as a surface (a, d, h, k, n) and slice through the centre of the receptor (b, e, i, l, o), with the cross-validation FSC curves for the refined model versus unfiltered half maps (one used in the refinement, work, and another one not, free) and the unfiltered summed maps shown on the right (c, f, j, m, p).

Extended Data Figure 3 Closed state 1 cryo-EM density and comparison of ZK200775-bound and apo states.

ad, Fragments of GluA2–GSG1LZK-1 and GluA2–GSG1Lapo-1 with the corresponding cryo-EM density. a, d, ATD and LBD of subunit A in GluA2–GSG1LZK-1 (a) and GluA2–GSG1Lapo-1 (d) with density for ZK200775 indicated in the GluA2–2×GSG1LZK-1 structure. b, c, M2 helix (b) and selectivity filter (c) with the Q/R-site Gln586 side chains pointing towards the centre of the pore in GluA2–GSG1LZK-1. e, Superposition of GluA2–GSG1LZK-1 (blue) and GluA2–GSG1Lapo-1 (red) viewed parallel to the membrane. Note that the structures are almost indistinguishable (r.m.s.d. = 0.526 Å). Densities are shown at 6σ.

Extended Data Figure 4 Closed state 2 structure and digitonin-binding pocket.

a, b, Structures of GluA2–GSG1LZK-1 (a) and GluA2–GSG1LZK-2 (b) viewed parallel to the membrane. The GluA2 subunits A and C are coloured purple, B and D in green and GSG1L in red. The competitive antagonist ZK200775 and digitonin are shown as space-filling models. In b, inset shows expanded view of the boxed region, demonstrating cryo-EM density for digitonin (blue mesh, 4σ). Digitonin and the surrounding residues in the inset are shown in stick representation. ch, Top down views along the axis of the overall two-fold rotational symmetry on the ATD (c, d), LBD (e, f) and TMD (g, h) layers. Rigid-body rotation of the ATD tetramer in d and rotation of LBD dimers in f are indicated by red arrows. i, j, Superposition of GluA2–GSG1LZK-2 (blue) and GluA2–GSG1Lapo-2 (red) viewed parallel to the membrane. Note that the structures are almost indistinguishable (r.m.s.d. = 0.701 Å).

Extended Data Figure 5 Cryo-EM density for the open state.

ae, Fragments of GluA2–STZGlu+CTZ with the corresponding cryo-EM density. a, Zoomed view of the glutamate-binding pocket. b, c, The ion channel pore with a central density at the selectivity filter, probably for a sodium ion that is hydrated based on the pore diameter, viewed from the top of the selectivity filter looking down into the cytoplasm (b) or parallel to the membrane with two (front and back) GluA2 subunits removed (c). d, Density for CTZ. e, Transmembrane domain segments for GluA2 (top row) and STZ (bottom row).

Extended Data Figure 6 Overview of single-particle cryo-EM and stoichiometry for GluA2–STZ and GluA2–GSG1L solubilized in digitonin.

a, b, Two-dimensional class averages for GluA2–STZGlu+CTZ (a) and GluA2nGSG1LZK (b) indicating three-layer architecture of the particles. c, d, Final densities for GluA2–STZGlu+CTZ (c) and GluA2–GSG1LZK-1 (d) with the GluA2 subunits A and C coloured purple, B and D in green, STZ in cyan and GSG1L in red. Insets show 2D slices made parallel to the membrane through the refined, nonfiltered map. Note, although four STZ molecules bind one receptor, only two copies of GSG1L can bind per GluA2 tetramer.

Extended Data Figure 7 Conformational differences between the closed, open and desensitized states.

ac, Structures of GluA2–GSG1LZK-1 in the closed state (a), GluA2–STZGlu+CTZ in the open state (b) and GluA2–2×GSG1LQuis in the desensitized state (c), viewed parallel to the membrane. The GluA2 subunits A and C are coloured purple, B and D in green, GSG1L in red and STZ in cyan. The competitive antagonist ZK200775, agonists Glu and Quis and positive allosteric modulator CTZ are shown as space-filling models. dl, Top down views along the axis of the overall two-fold rotational symmetry on the layers of ATD (df), LBD (gi) and TMD (jl). Rigid-body rotation of the ATD tetramers in e and f, broadening of LBD layer in h and rotation of subunit A/C LBDs in i are indicated by red arrows. Note the dramatic opening in the middle of the LBD layer (h) and pore dilation (k) in the open state.

Extended Data Figure 8 iGluR gating mechanism.

Two out of four iGluR subunits are shown with the ATDs omitted. Four basic states of iGluR gating are illustrated: resting, represented by apo (GluA2–GSG1Lapo-1) or antagonist-bound closed state (GluA2–GSG1LZK-1) structures; closed, agonist-bound (pre-active state crystal structures10,11); open (GluA2–STZGlu+CTZ); and desensitized (GluA2–2×GSG1LQuis complex16). Transitions between the states are indicated by black arrows, conformational rearrangements by blue arrows, and ionic current through the open channel by an orange arrow. Upper and lower gates are indicated by one and two red asterisks, respectively, with red sticks at the upper gate representing channel occluding residues at the bundle crossing and the Q/R site at the lower gate. Glutamate molecules are illustrated by orange wedges. The receptor sits in a resting, closed state, with its LBD clamshells in the maximally open conformations, unoccupied by the neurotransmitter glutamate. Upon glutamate binding, the LBD clamshells close, as described in the pre-activated crystal structures, to an intermediate state that does not put enough strain on the LBD–TMD linkers to open the channel. The LBDs then transition to their maximally closed state, which strains the LBD–TMD linkers, causing the channel pore to open and conduct ions. Most AMPARs, however, quickly desensitize, transitioning to the desensitized state from the open state via the agonist-bound, closed state. Desensitization is accompanied by the rupture of the upper LBD interfaces, with the LBDs adapting their maximally closed clamshell conformations, as described in the desensitized-state GluA2–GSG1L complex.

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

Supplementary information

Reporting Summary (PDF 67 kb)

Conformational changes between GluA2 closed, open and desensitized states

A morph between GluA2 closed, open and desensitized stated represented by the GluA2-GSG1LZK-1, GluA2-STZGlu-CTZ and GluA2-2xGSG1LQuis structures, respectively. The GluA2 subunits A and C are colored purple and B and D green. The agonists Glu are represented by space-filling models. Note, the pre-M4 helices in subunits A and C unfold during channel opening, while the entire TMD becomes wider. The receptor undergoes a corkscrew twist and becomes shorter while progressing from the closed to open to desensitized states. (MP4 9845 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Twomey, E., Yelshanskaya, M., Grassucci, R. et al. Channel opening and gating mechanism in AMPA-subtype glutamate receptors. Nature 549, 60–65 (2017). https://doi.org/10.1038/nature23479

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature23479

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