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:

Crystal structures of human glycine receptor α3 bound to a novel class of analgesic potentiators

Abstract

Current therapies to treat persistent pain and neuropathic pain are limited by poor efficacy, side effects and risk of addiction. Here, we present a novel class of potent selective, central nervous system (CNS)-penetrant potentiators of glycine receptors (GlyRs), ligand-gated ion channels expressed in the CNS. AM-1488 increased the response to exogenous glycine in mouse spinal cord and significantly reversed mechanical allodynia induced by nerve injury in a mouse model of neuropathic pain. We obtained an X-ray crystal structure of human homopentameric GlyRα3 in complex with AM-3607, a potentiator of the same class with increased potency, and the agonist glycine, at 2.6-Å resolution. AM-3607 binds a novel allosteric site between subunits, which is adjacent to the orthosteric site where glycine binds. Our results provide new insights into the potentiation of cysteine-loop receptors by positive allosteric modulators and hold promise in structure-based design of GlyR modulators for the treatment of neuropathic pain.

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

Figure 1: In vitro profiling of AM-1488 on GlyRs and in vivo reversal of SNI-induced tactile allodynia after oral dosing.
Figure 2: In vitro profiling of AM-3607 on full-length and engineered human GlyR channels.
Figure 3: Architecture of the GlyRα3cryst N38Q in complex with glycine and AM-3607.
Figure 4: Neurotransmitter-binding site occupied by the agonist glycine.
Figure 5: Allosteric binding site occupied by AM-3607.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

Swiss-Prot

References

  1. Dworkin, R.H. et al. Recommendations for the pharmacological management of neuropathic pain: an overview and literature update. Mayo Clin. Proc. 85 (Suppl.), S3–S14 (2010).

    Article  CAS  Google Scholar 

  2. Schofield, P.R. et al. Sequence and functional expression of the GABAA receptor shows a ligand-gated receptor super-family. Nature 328, 221–227 (1987).

    Article  CAS  Google Scholar 

  3. Melzack, R. & Wall, P.D. Pain mechanisms: a new theory. Science 150, 971–979 (1965).

    Article  CAS  Google Scholar 

  4. Foster, E. et al. Targeted ablation, silencing, and activation establish glycinergic dorsal horn neurons as key components of a spinal gate for pain and itch. Neuron 85, 1289–1304 (2015).

    Article  CAS  Google Scholar 

  5. Ahmadi, S., Lippross, S., Neuhuber, W.L. & Zeilhofer, H.U. PGE2 selectively blocks inhibitory glycinergic neurotransmission onto rat superficial dorsal horn neurons. Nat. Neurosci. 5, 34–40 (2002).

    Article  CAS  Google Scholar 

  6. Harvey, R.J. et al. GlyR alpha3: an essential target for spinal PGE2-mediated inflammatory pain sensitization. Science 304, 884–887 (2004).

    Article  CAS  Google Scholar 

  7. Lynch, J.W. & Callister, R.J. Glycine receptors: a new therapeutic target in pain pathways. Curr. Opin. Investig. Drugs 7, 48–53 (2006).

    CAS  PubMed  Google Scholar 

  8. Hejazi, N. et al. Delta9-tetrahydrocannabinol and endogenous cannabinoid anandamide directly potentiate the function of glycine receptors. Mol. Pharmacol. 69, 991–997 (2006).

    Article  CAS  Google Scholar 

  9. Xiong, W. et al. Cannabinoid potentiation of glycine receptors contributes to cannabis-induced analgesia. Nat. Chem. Biol. 7, 296–303 (2011).

    Article  CAS  Google Scholar 

  10. Xiong, W. et al. Cannabinoids suppress inflammatory and neuropathic pain by targeting α3 glycine receptors. J. Exp. Med. 209, 1121–1134 (2012).

    Article  CAS  Google Scholar 

  11. Yevenes, G.E. & Zeilhofer, H.U. Allosteric modulation of glycine receptors. Br. J. Pharmacol. 164, 224–236 (2011).

    Article  CAS  Google Scholar 

  12. Leuwer, M., O′Neill, P., Berry, N. & Haeseler, G. Compounds for use in the treatment of pain. US Patent Application US20120029235 (2012).

  13. Leuwer, M., O′Neill, P., Berry, N. & Pidathala, C. Pharmacologically active compounds. PCT International Patent Application WO2015097475 (2015).

  14. Maleeva, G., Buldakova, S. & Bregestovski, P. Selective potentiation of alpha 1 glycine receptors by ginkgolic acid. Front. Mol. Neurosci. 8, 64 (2015).

    Article  Google Scholar 

  15. Shalaly, N.D. et al. Positive modulation of the glycine receptor by means of glycine receptor-binding aptamers. J. Biomol. Screen. 20, 1112–1123 (2015).

    Article  CAS  Google Scholar 

  16. Burgos, C.F., Yévenes, G.E. & Aguayo, L.G. Structure and pharmacological modulation of inhibitory glycine receptors. Mol. Pharmacol. 90, 318–325 (2016).

    Article  CAS  Google Scholar 

  17. Stead, C. et al. Identification of positive allosteric modulators of glycine receptors from a high-throughput screen using a fluorescent membrane potential assay. J. Biomol. Screen. 21, 1042–1053 (2016).

    Article  CAS  Google Scholar 

  18. Beyer, C., Roberts, L.A. & Komisaruk, B.R. Hyperalgesia induced by altered glycinergic activity at the spinal cord. Life Sci. 37, 875–882 (1985).

    Article  CAS  Google Scholar 

  19. Yaksh, T.L. Behavioral and autonomic correlates of the tactile evoked allodynia produced by spinal glycine inhibition: effects of modulatory receptor systems and excitatory amino acid antagonists. Pain 37, 111–123 (1989).

    Article  CAS  Google Scholar 

  20. Sherman, S.E. & Loomis, C.W. Morphine insensitive allodynia is produced by intrathecal strychnine in the lightly anesthetized rat. Pain 56, 17–29 (1994).

    Article  CAS  Google Scholar 

  21. Sivilotti, L. & Woolf, C.J. The contribution of GABAA and glycine receptors to central sensitization: disinhibition and touch-evoked allodynia in the spinal cord. J. Neurophysiol. 72, 169–179 (1994).

    Article  CAS  Google Scholar 

  22. Loomis, C.W., Khandwala, H., Osmond, G. & Hefferan, M.P. Coadministration of intrathecal strychnine and bicuculline effects synergistic allodynia in the rat: an isobolographic analysis. J. Pharmacol. Exp. Ther. 296, 756–761 (2001).

    CAS  PubMed  Google Scholar 

  23. Woolf, C.J. & Salter, M.W. Neuronal plasticity: increasing the gain in pain. Science 288, 1765–1769 (2000).

    Article  CAS  Google Scholar 

  24. Sandkühler, J. Models and mechanisms of hyperalgesia and allodynia. Physiol. Rev. 89, 707–758 (2009).

    Article  Google Scholar 

  25. Kuner, R. Central mechanisms of pathological pain. Nat. Med. 16, 1258–1266 (2010).

    Article  CAS  Google Scholar 

  26. Zeilhofer, H.U., Benke, D. & Yevenes, G.E. Chronic pain states: pharmacological strategies to restore diminished inhibitory spinal pain control. Annu. Rev. Pharmacol. Toxicol. 52, 111–133 (2012).

    Article  CAS  Google Scholar 

  27. Althoff, T., Hibbs, R.E., Banerjee, S. & Gouaux, E. X-ray structures of GluCl in apo states reveal a gating mechanism of Cys-loop receptors. Nature 512, 333–337 (2014).

    Article  CAS  Google Scholar 

  28. Hibbs, R.E. & Gouaux, E. Principles of activation and permeation in an anion-selective Cys-loop receptor. Nature 474, 54–60 (2011).

    Article  CAS  Google Scholar 

  29. Miller, P.S. & Aricescu, A.R. Crystal structure of a human GABAA receptor. Nature 512, 270–275 (2014).

    Article  CAS  Google Scholar 

  30. Hassaine, G. et al. X-ray structure of the mouse serotonin 5-HT3 receptor. Nature 512, 276–281 (2014).

    Article  CAS  Google Scholar 

  31. Huang, X., Chen, H., Michelsen, K., Schneider, S. & Shaffer, P.L. Crystal structure of human glycine receptor-α3 bound to antagonist strychnine. Nature 526, 277–280 (2015).

    Article  CAS  Google Scholar 

  32. Du, J., Lü, W., Wu, S., Cheng, Y. & Gouaux, E. Glycine receptor mechanism elucidated by electron cryo-microscopy. Nature 526, 224–229 (2015).

    Article  CAS  Google Scholar 

  33. Gielen, M., Thomas, P. & Smart, T.G. The desensitization gate of inhibitory Cys-loop receptors. Nat. Commun. 6, 6829 (2015).

    Article  CAS  Google Scholar 

  34. Breitinger, H.G., Villmann, C., Becker, K. & Becker, C.M. Opposing effects of molecular volume and charge at the hyperekplexia site alpha 1(P250) govern glycine receptor activation and desensitization. J. Biol. Chem. 276, 29657–29663 (2001).

    Article  CAS  Google Scholar 

  35. Miller, P.S., Da Silva, H.M. & Smart, T.G. Molecular basis for zinc potentiation at strychnine-sensitive glycine receptors. J. Biol. Chem. 280, 37877–37884 (2005).

    Article  CAS  Google Scholar 

  36. Yu, R. et al. Agonist and antagonist binding in human glycine receptors. Biochemistry 53, 6041–6051 (2014).

    Article  CAS  Google Scholar 

  37. Grudzinska, J. et al. The beta subunit determines the ligand binding properties of synaptic glycine receptors. Neuron 45, 727–739 (2005).

    Article  CAS  Google Scholar 

  38. Vandenberg, R.J., Handford, C.A. & Schofield, P.R. Distinct agonist- and antagonist-binding sites on the glycine receptor. Neuron 9, 491–496 (1992).

    Article  CAS  Google Scholar 

  39. Lynch, J.W. et al. Identification of intracellular and extracellular domains mediating signal transduction in the inhibitory glycine receptor chloride channel. EMBO J. 16, 110–120 (1997).

    Article  CAS  Google Scholar 

  40. Berger, M.L. On the true affinity of glycine for its binding site at the NMDA receptor complex. J. Pharmacol. Toxicol. Methods 34, 79–88 (1995).

    Article  CAS  Google Scholar 

  41. Shan, Q., Haddrill, J.L. & Lynch, J.W. Ivermectin, an unconventional agonist of the glycine receptor chloride channel. J. Biol. Chem. 276, 12556–12564 (2001).

    Article  CAS  Google Scholar 

  42. Otwinowski, Z. et al. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  Google Scholar 

  43. McCoy, A.J. Solving structures of protein complexes by molecular replacement with Phaser. Acta Crystallogr. D Biol. Crystallogr. 63, 32–41 (2007).

    Article  CAS  Google Scholar 

  44. Schüttelkopf, A.W. & van Aalten, D.M. PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. D Biol. Crystallogr. 60, 1355–1363 (2004).

    Article  Google Scholar 

  45. Murshudov, G.N., Vagin, A.A. & Dodson, E.J. Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr. D Biol. Crystallogr. 53, 240–255 (1997).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  47. Davis, I.W. et al. MolProbity: all-atom contacts and structure validation for proteins and nucleic acids. Nucleic Acids Res. 35, W375–W383 (2007).

    Article  Google Scholar 

  48. 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  Google Scholar 

  49. DeLano, W.L. The PyMOL Molecular Graphics System (DeLano Scientific, 2002).

  50. Larkin, M.A. et al. Clustal W and Clustal X version 2.0. Bioinformatics 23, 2947–2948 (2007).

    Article  CAS  Google Scholar 

  51. Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals 8th edn. (National Academies Press, 2011).

  52. Pertin, M., Allchorne, A.J., Beggah, A.T., Woolf, C.J. & Decosterd, I. Delayed sympathetic dependence in the spared nerve injury (SNI) model of neuropathic pain. Mol. Pain 3, 21 (2007).

    Article  Google Scholar 

  53. Chaplan, S.R., Bach, F.W., Pogrel, J.W., Chung, J.M. & Yaksh, T.L. Quantitative assessment of tactile allodynia in the rat paw. J. Neurosci. Methods 53, 55–63 (1994).

    Article  CAS  Google Scholar 

  54. Dixon, W.J. Efficient analysis of experimental observations. Annu. Rev. Pharmacol. Toxicol. 20, 441–462 (1980).

    Article  CAS  Google Scholar 

  55. Berry, L.M., Roberts, J., Be, X., Zhao, Z. & Lin, M.H. Prediction of V(ss) from in vitro tissue-binding studies. Drug Metab. Dispos. 38, 115–121 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G. Ranieri and R. Walter at Shamrock Structures and the staff at beamline 22-ID at the Advanced Photon Source for data collection. We also thank V. Berry, L. Berry, M. Chaves, J. Hu, P. Pegman, N.K. Yelleswarapu, the Comparative Animal Research staff and the large team responsible for toxicology data acquisition for technical support. We are grateful to Z. Wang, A. Guzman-Perez, M. Chu-Moyer, T. Kornecook, Z. Kaprielian and K. Wild for critical review of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The authors jointly contributed to project design, data analysis and manuscript preparation. P.L.S. performed initial construct design and purification experiments, structure solution, model building, structural analysis and ITC binding titrations; X.H. performed construct design, protein purifications, crystallization, structural solution, model building and structural analysis; H.C. performed cloning and expression experiments; H.B., S.Y. and E.F.D. designed and synthesized AM-3607 and AM-1488; S.A. purified AM-3607; J.R.S., S.S., J.A.L., D.J.M., S.I.M., M.Z., S.G.L. and J.G. performed functional studies; Y.T. performed the pharmacokinetic analysis; K.M. and M.H.P. performed SPR binding experiments; X.H., P.L.S., E.F.D. and J.G. wrote the manuscript with assistance from the other coauthors.

Corresponding authors

Correspondence to Xin Huang, Paul L Shaffer, Erin F DiMauro or Jacinthe Gingras.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 In vitro electrophysiological and binding characterization of AM-1488 and AM-3607 through IonFlux HT and SPR.

(a) AM-1488 potentiates currents evoked by EC20 glycine in HEK293T cells stably expressing human GlyRα3β (EC50 = 0.26 ± 0.02 μM), as shown by the dose response plotted as percent of maximal evoked current amplitude (1 mM glycine). (b) AM-3607 potentiates currents evoked by EC20 glycine in HEK293T cells stably expressing human GlyRα3β (EC50 = 0.051 ± 0.016 μM), as shown by the dose response plotted as percent of maximal evoked current amplitude (1 mM glycine). All values are calculated EC50 ± s.e.m. and each data point is mean ± s.e.m. with N≥5 for (a) and (b). (c) Representative binding trace of AM-1488 to GlyRα3cryst measured by SPR with kinetic fit overlaid. (d) Representative binding trace of AM-3607 to GlyRα3cryst measured by SPR with kinetic fit overlaid. Values for KD are mean ± s.d. determined from N=3 experiments for (c) and (d). (e) Representative trace of AM-1488 not binding to GlyRα3cryst preincubated with AM-3607 measured by SPR. Trace is representative of N=4 total trials on two different flow cells.

Supplementary Figure 2 In vivo evaluation of AM-1488 in a mouse open field assay.

(a) AM-1488 had no significant effect on the total basic movements (5708 ± 482 cm, N=10), when compared to the vehicle treated group (5052 ± 131 cm, N=10; t18 = 1.3, p > 0.05) at 60 min post oral dosing in naïve mice. (b) AM-1488 had slight reduction on total rearing counts (354 ± 137), when compared to the vehicle treated group (498 ± 149; t18 = 2.2, p < 0.05). All values are mean ± s.e.m. except Cu values which are mean ± s.d..

Supplementary Figure 3 Crystallographic packing of GlyRα3cryst.

(a) Packing of the GlyRα3–glycine–AM-3607 complex. Receptor in the assymmetric unit is colored by subunit, with subunit A in pale green and B in cyan. AM-3607 molecules are shown as slate spheres. Symmetry related receptors are colored grey. (b) Packing of the GlyRα3N38Q–glycine–AM-3607 complex. Receptor in the assymmetric unit is colored by subunit, with subunit A in pale green and B in cyan. AM-3607 molecules are shown as slate spheres. Symmetry related receptors are colored grey.

Supplementary Figure 4 Omit maps of the orthosteric binding site and the allosteric binding site occupied by the agonist glycine and AM-3607, respectively.

(a) 2Fo-Fc omit map of the orthosteric binding site viewed perpendicular to the plasma membrane contoured at 1σ. Glycine was omitted from the electron density map calculation and is shown as grey sticks. The principal or (+) subunit is colored in pale green and the complementary or (-) subunit is colored cyan. Important residues are noted. Black dashed line indicates hydrogen bonds. (b) 2Fo-Fc omit map of the allosteric binding site viewed parallel to the plasma membrane contoured at 1σ. AM-3607 was omitted from the electron density map calculations and is shown as slate sticks. (c) Fo-Fc omit map of the orthosteric binding site in same view as in (a) contoured at ±4σ. (d) Fo-Fc omit map of the allosteric binding site with the view rotated ~75° along the vertical axis from (b) contoured at ±4σ.

Supplementary Figure 5 The ion channel of the GlyRα3–glycine–AM-3607 complex in a desensitized state.

(a) Solvent contours of the transmembrane pore of the AM-3607 bound GlyRα3 pore showing the M2 helices of subunits A and C. Side chains of pore lining residues are shown. Numbering is according to the protein sequence and position in the M2 helix. Small purple, green and red spheres define a radius of >3.3 Å, 1.8-3.3 Å and <1.8 Å, respectively. (b), Contours of the glycine–ivermectin bound GlyRα1 pore, similar to panel (a). (c) Plot of pore radii as a function of distance along the pore axis for AM-3607 bound GlyRα3, glycine–ivermectin bound GlyRα1, ivermectin-bound GluClα, and benzamidine-bound GABAAR-β3. (d) Electrophysiological response to 30 μM glycine applied to HEK293T cells expressing human GlyRα3. Black (control) and blue (wash) traces are glycine application alone before and after co-application with 0.2 μM AM-3607 (red trace), showing potentiation of the glycine-induced current. (e) Desensitization of GlyRα3 upon stimulation with 1 mM glycine is not affected by subsequent co-application of 0.2 μM AM-3607.

Supplementary Figure 6 Putative sites of Cl and Zn2+.

(a) The putative Cl- site with Cl- shown as green sphere and the side chains of Ala254 and Thr258 shown in sticks. The Fo-Fc omit map is contoured at 5σ. Subunit B has been removed for clarity. Distances from side chain carbons to Cl- are 5.4 – 6.4 Å. (b) The putative binding site of Zn2+ with Zn2+ shown as grey sphere and the side chains of Glu192, Asp194, and His215 shown as sticks. The NCS-averaged anomalous difference map is contoured at 5σ. Distances from Zn2+ to side chain oxygen atoms average 2.2 Å and to His215 nitrogen atoms average 2.6 Å across all five subunits.

Supplementary Figure 7 Binding of glycine to GlyRα3cryst without and with bound AM-3607.

(a) Determination of glycine affinity to apo GlyRα3cryst measured by SPR. KD value is mean ± s.e.m., determined from N=2 experiments. (b) Determination of glcyine affinity to AM-3607 bound GlyRα3cryst by ITC. Orthosteric ligand glycine was titrated into purified GlyRα3cryst preincubated with AM-3607. A representative titration is shown with non-linear best fit. Binding parameters were determined from N=2 experiments and are means ± s.d..

Supplementary Figure 8 Sequence alignment of GlyRα3cryst with representative eukaryotic Cys-loop-receptor family members.

Residue conservation is indicated by grey and black highlights. Residues involved in binding of AM-3607 are indicated by pale green (the principal subunit) and cyan (the complementary subunit) dots. Residues involved in binding of glycine are indicated by pale green (the principal subunit) and cyan (the complementary subunit) squares. Secondary structure elements are denoted by cylinders (helices) and arrows (strands) above the alignment. The alignment was generated using ClustalW. Protein sequences are from the following entires: Zebrafish GlyRα1 (PDB 3JAF), Human GlyRβ (Uniprot P48167), GluClcryst (PDB 4TNV), GABAAR-β3cryst (PDB 4COF), 5HT3A (PDB 4PIR), and T. marmorata nAchRα (Uniprot P02711).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8, Supplementary Tables 1–4 and Supplementary Notes 1–5 (PDF 3386 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, X., Shaffer, P., Ayube, S. et al. Crystal structures of human glycine receptor α3 bound to a novel class of analgesic potentiators. Nat Struct Mol Biol 24, 108–113 (2017). https://doi.org/10.1038/nsmb.3329

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3329

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research