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 basis of GABA reuptake inhibition

A Publisher Correction to this article was published on 15 July 2022

This article has been updated

Abstract

γ-Aminobutyric acid (GABA) transporter 1 (GAT1)1 regulates neuronal excitation of the central nervous system by clearing the synaptic cleft of the inhibitory neurotransmitter GABA upon its release from synaptic vesicles. Elevating the levels of GABA in the synaptic cleft, by inhibiting GABA reuptake transporters, is an established strategy to treat neurological disorders, such as epilepsy2. Here we determined the cryo-electron microscopy structure of full-length, wild-type human GAT1 in complex with its clinically used inhibitor tiagabine3, with an ordered part of only 60 kDa. Our structure reveals that tiagabine locks GAT1 in the inward-open conformation, by blocking the intracellular gate of the GABA release pathway, and thus suppresses neurotransmitter uptake. Our results provide insights into the mixed-type inhibition of GAT1 by tiagabine, which is an important anticonvulsant medication. Its pharmacodynamic profile, confirmed by our experimental data, suggests initial binding of tiagabine to the substrate-binding site in the outward-open conformation, whereas our structure presents the drug stalling the transporter in the inward-open conformation, consistent with a two-step mechanism of inhibition4. The presented structure of GAT1 gives crucial insights into the biology and pharmacology of this important neurotransmitter transporter and provides blueprints for the rational design of neuromodulators, as well as moving the boundaries of what is considered possible in single-particle cryo-electron microscopy of challenging membrane proteins.

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: Cryo-EM structure determination of full-length, wild-type human GAT1 in the inward-open conformation.
Fig. 2: Structural details of the GAT1 tiagabine-binding pocket.
Fig. 3: Extracellular gate of GAT1 and comparison to other NSS-fold structures, highlighting rearrangements in the inward-open conformation.
Fig. 4: Proposed two-step mode of tiagabine inhibition.

Similar content being viewed by others

Data availability

The final cryo-EM map has been deposited in the Electron Microscopy Data Bank under accession code: EMD-25170. Corresponding atomic coordinates have been deposited in the PDB under accession code: 7SK2Source data are provided with this paper.

Change history

References

  1. Guastella, J. et al. Cloning and expression of a rat brain GABA transporter. Science 249, 1303–1306 (1990).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Madsen, K. K., White, H. S. & Schousboe, A. Neuronal and non-neuronal GABA transporters as targets for antiepileptic drugs. Pharmacol. Ther. 125, 394–401 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Braestrup, C. et al. (R)-N-[4,4-bis(3-methyl-2-thienyl)but-3-en-1-yl]nipecotic acid binds with high affinity to the brain gamma-aminobutyric acid uptake carrier. J. Neurochem. 54, 639–647 (1990).

    Article  CAS  PubMed  Google Scholar 

  4. Blat, Y. Non-competitive inhibition by active site binders. Chem. Biol. Drug Des. 75, 535–540 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Sieghart, W. Structure, pharmacology, and function of GABAA receptor rubtypes. GABA 54, 231–263 (2006).

    Article  CAS  Google Scholar 

  6. Bowery, N. G. et al. International Union of Pharmacology. XXXIII. Mammalian γ-aminobutyric acidB receptors: structure and function. Pharmacol. Rev. 54, 247–264 (2002).

    Article  CAS  PubMed  Google Scholar 

  7. Fattorini, G., Melone, M. & Conti, F. A reappraisal of GAT-1 localization in neocortex. Front. Cell. Neurosci. 14, 9 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Roberts, B. M. et al. GABA uptake transporters support dopamine release in dorsal striatum with maladaptive downregulation in a parkinsonism model. Nat. Commun. 11, 4958 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mermer, F. et al. Common molecular mechanisms of SLC6A1 variant-mediated neurodevelopmental disorders in astrocytes and neurons. Brain 144, 2499–2512 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Rosenthal, M. Tiagabine for the treatment of generalized anxiety disorder: a randomized, open-label, clinical trial with paroxetine as a positive control. J. Clin. Psychiatry 64, 1245–1249 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Lyu, S. et al. Blockade of GABA transporter-1 and GABA transporter-3 in the lateral habenula improves depressive-like behaviors in a rat model of Parkinson’s disease. Neuropharmacology 181, 108369 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Fuhrer, T. E. et al. Impaired expression of GABA transporters in the human Alzheimer’s disease hippocampus, subiculum, entorhinal cortex and superior temporal gyrus. Neuroscience 351, 108–118 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Paparrigopoulos, T., Tzavellas, E., Karaiskos, D., Malitas, P. & Liappas, I. An open pilot study of tiagabine in alcohol dependence: tolerability and clinical effects. J. Psychopharmacol. 24, 1375–1380 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. Schwartz, T. L. et al. An open-label study of tiagabine as augmentation therapy for anxiety. Ann. Clin. Psychiatry 17, 167–172 (2005).

    Article  PubMed  Google Scholar 

  15. Carpenter, L. L. et al. Open-label tiagabine monotherapy for major depressive disorder with anxiety. J. Clin. Psychiatry 67, 66–71 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Kragholm, B. et al. Discovery of a subtype selective inhibitor of the human betaine/GABA transporter 1 (BGT-1) with a non-competitive pharmacological profile. Biochem. Pharmacol. 86, 521–528 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Hauke, T. J., Wein, T., Höfner, G. & Wanner, K. T. Novel allosteric ligands of γ-aminobutyric acid transporter 1 (GAT1) by MS based screening of pseudostatic hydrazone libraries. J. Med. Chem. 61, 10310–10332 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Jurik, A. et al. A binding mode hypothesis of tiagabine confirms liothyronine effect on γ-aminobutyric acid transporter 1 (GAT1). J. Med. Chem. 58, 2149–2158 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zafar, S. & Jabeen, I. Molecular dynamic simulations to probe stereoselectivity of tiagabine binding with human GAT1. Molecules 25, 4745 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  20. Skovstrup, S., David, L., Taboureau, O. & Jørgensen, F. S. A steered molecular dynamics study of binding and translocation processes in the GABA transporter. PLoS ONE 7, e39360 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Keynan, S., Suh, Y. J., Kanner, B. I. & Rudnick, G. Expression of a cloned gamma-aminobutyric acid transporter in mammalian cells. Biochemistry 31, 1974–1979 (1992).

    Article  CAS  PubMed  Google Scholar 

  22. Cammack, J. N., Rakhilin, S. V. & Schwartz, E. A. A GABA transporter operates asymmetrically and with variable stoichiometry. Neuron 13, 949–960 (1994).

    Article  CAS  PubMed  Google Scholar 

  23. Lester, H. A., Mager, S., Quick, M. W. & Corey, J. L. Permeation properties of neurotransmitter transporters. Annu. Rev. Pharmacol. Toxicol. 34, 219–249 (1994).

    Article  CAS  PubMed  Google Scholar 

  24. Jardetzky, O. Simple allosteric model for membrane pumps. Nature 211, 969–970 (1966).

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Krishnamurthy, H. & Gouaux, E. X-ray structures of LeuT in substrate-free outward-open and apo inward-open states. Nature 481, 469–474 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Penmatsa, A., Wang, K. H. & Gouaux, E. X-ray structure of dopamine transporter elucidates antidepressant mechanism. Nature 503, 85–90 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gotfryd, K. et al. X-ray structure of LeuT in an inward-facing occluded conformation reveals mechanism of substrate release. Nat. Commun. 11, 1005 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Singh, S. K., Yamashita, A. & Gouaux, E. Antidepressant binding site in a bacterial homologue of neurotransmitter transporters. Nature 448, 952–956 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Coleman, J. A., Green, E. M. & Gouaux, E. X-ray structures and mechanism of the human serotonin transporter. Nature 532, 334–339 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Coleman, J. A. et al. Serotonin transporter–ibogaine complexes illuminate mechanisms of inhibition and transport. Nature 569, 141–145 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shahsavar, A. et al. Structural insights into the inhibition of glycine reuptake. Nature 591, 677–681 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Zhou, Z. et al. LeuT-desipramine structure reveals how antidepressants block neurotransmitter reuptake. Science 317, 1390–1393 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kantcheva, A. K. et al. Chloride binding site of neurotransmitter sodium symporters. Proc. Natl Acad. Sci. USA 110, 8489–8494 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Li, F. et al. Ion transport and regulation in a synaptic vesicle glutamate transporter. Science 368, 893–897 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zimmermann, I. et al. Synthetic single domain antibodies for the conformational trapping of membrane proteins. eLife 7, e34317 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Tsutsumi, N. et al. Structure of human Frizzled5 by fiducial-assisted cryo-EM supports a heterodimeric mechanism of canonical Wnt signaling. eLife 9, e58464 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wu, X. & Rapoport, T. A. Cryo-EM structure determination of small proteins by nanobody-binding scaffolds (Legobodies). Proc. Natl Acad. Sci. USA 118, e2115001118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Borden, L. A. et al. Tiagabine, SK&F 89976-A, CI-966, and NNC-711 are selective for the cloned GABA transporter GAT-1. Eur. J. Pharmacol. 269, 219–224 (1994).

    Article  CAS  PubMed  Google Scholar 

  39. Suzdak, P. D., Foged, C. & Andersen, K. E. Quantitative autoradiographic characterization of the binding of [3H]tiagabine (NNC 05-328) to the GABA uptake carrier. Brain Res. 647, 231–241 (1994).

    Article  CAS  PubMed  Google Scholar 

  40. Korkhov, V. M., Farhan, H., Freissmuth, M. & Sitte, H. H. Oligomerization of the γ-aminobutyric acid transporter-1 is driven by an interplay of polar and hydrophobic interactions in transmembrane helix II. J. Biol. Chem. 279, 55728–55736 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. White, H. S. et al. Correlation between anticonvulsant activity and inhibitory action on glial gamma-aminobutyric acid uptake of the highly selective mouse gamma-aminobutyric acid transporter 1 inhibitor 3-hydroxy-4-amino-4,5,6,7-tetrahydro-1,2-benzisoxazole and its N-alkylated analogs. J. Pharmacol. Exp. Ther. 302, 636–644 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Malinauskaite, L. et al. A mechanism for intracellular release of Na+ by neurotransmitter/sodium symporters. Nat. Struct. Mol. Biol. 21, 1006–1012 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ben-Yona, A. & Kanner, B. I. Functional defects in the external and internal thin gates of the γ-aminobutyric acid (GABA) transporter GAT-1 can compensate each other. J. Biol. Chem. 288, 4549–4556 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bismuth, Y., Kavanaugh, M. P. & Kanner, B. I. Tyrosine 140 of the gamma-aminobutyric acid transporter GAT-1 plays a critical role in neurotransmitter recognition. J. Biol. Chem. 272, 16096–16102 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Dhar, T. G. et al. Design, synthesis and evaluation of substituted triarylnipecotic acid derivatives as GABA uptake inhibitors: identification of a ligand with moderate affinity and selectivity for the cloned human GABA transporter GAT-3. J. Med. Chem. 37, 2334–2342 (1994).

    Article  CAS  PubMed  Google Scholar 

  46. Kanner, B. I. Transmembrane domain I of the gamma-aminobutyric acid transporter GAT-1 plays a crucial role in the transition between cation leak and transport modes. J. Biol. Chem. 278, 3705–3712 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Rudnick, G. Forty four years with Baruch Kanner and the chloride ion. Neurochem. Res. 47, 3–8 (2022).

    Article  CAS  PubMed  Google Scholar 

  48. Wang, K. H., Penmatsa, A. & Gouaux, E. Neurotransmitter and psychostimulant recognition by the dopamine transporter. Nature 521, 322–327 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bulling, S. et al. The mechanistic basis for noncompetitive ibogaine inhibition of serotonin and dopamine transporters. J. Biol. Chem. 287, 18524–18534 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Alberati, D. et al. Glycine reuptake inhibitor RG1678: a pharmacologic characterization of an investigational agent for the treatment of schizophrenia. Neuropharmacology 62, 1152–1161 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. Wang, X., Ratnaraj, N. & Patsalos, P. N. The pharmacokinetic inter-relationship of tiagabine in blood, cerebrospinal fluid and brain extracellular fluid (frontal cortex and hippocampus). Seizure 13, 574–581 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Sandtner, W. et al. Binding mode selection determines the action of ecstasy homologs at monoamine transporters. Mol. Pharmacol. 89, 165–175 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Clausen, R. P. et al. Structure–activity relationship and pharmacology of gamma-aminobutyric acid (GABA) transport inhibitors. Adv. Pharmacol. 54, 265–284 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. Reith, M. E. A. et al. Novel C-1 substituted cocaine analogs unlike cocaine or benztropine. J. Pharmacol. Exp. Ther. 343, 413–425 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Alexandrov, A. I., Mileni, M., Chien, E. Y. T., Hanson, M. A. & Stevens, R. C. Microscale fluorescent thermal stability assay for membrane proteins. Structure 16, 351–359 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  59. Bordoli, L. et al. Protein structure homology modeling using SWISS-MODEL workspace. Nat. Protoc. 4, 1–13 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Goddard, T. D., Huang, C. C. & Ferrin, T. E. Visualizing density maps with UCSF Chimera. J. Struct. Biol. 157, 281–287 (2007).

    Article  CAS  PubMed  Google Scholar 

  61. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D60, 2126–2132 (2004).

    Article  PubMed  Google Scholar 

  62. 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 

  63. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D74, 531–544 (2018).

    Article  CAS  Google Scholar 

  64. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Abraham, M. J. et al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 12, 19–25 (2015).

    Article  ADS  Google Scholar 

  66. Jo, S., Kim, T., Iyer, V. G. & Im, W. CHARMM-GUI: a web-based graphical user interface for CHARMM. J. Comput. Chem. 29, 1859–1865 (2008).

    Article  CAS  PubMed  Google Scholar 

  67. Kim, S. et al. CHARMM-GUI ligand reader and modeler for CHARMM force field generation of small molecules. J. Comput. Chem. 38, 1879–1886 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Leonard, A. N. & Lyman, E. Activation of G-protein-coupled receptors is thermodynamically linked to lipid solvation. Biophys. J. 120, 1777–1787 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  69. Lomize, M. A., Pogozheva, I. D., Joo, H., Mosberg, H. I. & Lomize, A. L. OPM database and PPM web server: resources for positioning of proteins in membranes. Nucleic Acids Res. 40, D370–D376 (2012).

    Article  CAS  PubMed  Google Scholar 

  70. McGibbon, R. T. et al. MDTraj: a modern open library for the analysis of molecular dynamics trajectories. Biophys. J. 109, 1528–1532 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  71. DeLano, W. L. Pymol: an open-source molecular graphics tool. CCP4 http://legacy.ccp4.ac.uk/newsletters/newsletter40/11_pymol.html (2002).

  72. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Villers and C. Hanson for technical support for recombinant protein expression in mammalian and Sf9 cells; C. Cato and R. Oania for general laboratory support; S. Khan for helpful discussions; T. Osinski, J. Chu and B. D. Kim at the USC Center for Advanced Research Computing (CARC) for support with computing resources; and D. J. Slotboom (University of Groningen) and K. Pande (LBNL) for comments and suggestions on the manuscript. This research was supported by a US National Institutes of Health grant R35 GM127086 (to V.C.). We acknowledge the Center of Excellence for Nano Imaging (CNI) at the University of Southern California for microscope time.

Author information

Authors and Affiliations

Authors

Contributions

C.G. designed and supervised the project. Z.M., N.G.A. and H.S. performed sample preparation for the cryo-EM and biochemistry studies. Z.M. and C.G. performed cryo-EM data collection and image processing. G.W.H. performed model building and refinement. H.S., Z.M. and C.G. performed [3H]-GABA uptake and functional experiments. J.H.L. performed molecular dynamics simulations and analysis to validate the ligand-binding pocket, under supervision from V.K. V.C. supervised model building and refinement, and provided suggestions for the manuscript. C.G. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Cornelius Gati.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Azadeh Shahsavar, Harald Sitte and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Protein purification of GAT1 for cryo-EM analysis and [3H]-GABA uptake assay results for quantification of relative uptake rates.

a, Size-exclusion chromatography on a Superose 6 Increase (10/300) column shows a monodisperse peak at a size of approximately 100 kDa, accommodating the protein, as well as the LMNG/CHS detergent micelle. b, SDS-PAGE prior to cryo-EM sample preparation, confirms the high purity of the final sample (> 95%). For gel source data, see Supplementary Fig. 4. c, Thermal stability assay as described in the methods section. Changing the detergent from DDM to LMNG resulted in an increase of Tm by 12 °C. AU, arbitrary units. d, Scintillation proximity assay of GAT1 purified from Sf9 cells, showing specific competition of [3H]-GABA with tiagabine, presented as all data points, overlaid with a nonlinear regression fit, resulting in an IC50 of 290 ± 60 nM. The experiment was performed three times independently with similar results. e, Absolute counts of a representative [3H]-GABA uptake experiment from constructs used for probing tiagabine binding pocket. (Bottom) Comparison of expression levels, based on relative fluorescence of whole cells compared to wildtype GAT1, and maximum uptake values, relative to wildtype GAT1 in above experiment

Source data

Extended Data Fig. 2 Cryo-EM reconstruction of full-length, wild-type human GAT1 in complex with tiagabine.

a, Workflow of cryo-EM processing of GAT1 in complex with tiagabine in the inward-open conformation with representative micrograph, picking templates and 2D classes. Data processing was entirely performed in cryoSPARC57. After motion correction, CTF estimation and particle picking, the dataset was sorted using 2D classification, followed by 3D ab initio reconstruction and heterogenous refinement for further clean-up of the particle stack. Finally, 3D reconstructions were performed using non-uniform refinement58 and local refinement, which yielded a final map of 3.8 Å resolution. b, Data quality of the final reconstruction, illustrated as a local resolution map ranging from 3 - 4.8 Å, gold-standard fourier shell correlation plot (masked and unmasked) and angular sampling of the final reconstruction

Source data

Extended Data Fig. 3 Atomic model and cryo-EM Coulomb potential map of the human GAT1 complex with tiagabine.

The overall structure of the GAT1 (cyan) complex with bound tiagabine (salmon) (right) and magnified views of individual transmembrane helices (left) are shown in the cryo-EM map (grey mesh) at 5 sigma.

Extended Data Fig. 4 Sequence alignment and substrate binding pocket.

a, Sequence alignment of GABA transporters: human GAT1, GAT2, GAT3 (Uniprot: P30531, Q9NSD5, P48066). Green boxes highlight conserved residues observed in the substrate binding pocket and red boxes highlight non-conserved residues. b, AlphaFold55 prediction of GAT1 and GAT2 in the outward-open conformation with tiagabine superimposed with substrate from LeuT (PDB ID: 4HOD33), highlighting non-conserved residues between the two subtypes in the substrate-binding pocket. c, AlphaFold55 prediction of GAT1 outward-open conformation, superimposed with experimentally determined inward-open cryo-EM structure, shows the small relative transition that tiagabine bound to GAT1 would have to undergo for the proposed induced-fit mechanism.

Extended Data Fig. 5 Structural comparison of GAT1 with other NSS transporter structures.

a, Superposition of outward-open dDAT26 (PDB ID: 4XP1, yellow, residues F53, Y142, S320 in stick representation) with outward-open SERT30 (PDB ID: 6DZY, red, residues F105, Y121, S336 in stick representation). b, Superposition of outward-open dDAT26 (PDB ID: 4XP1, yellow, residues F53, Y142, S320 in stick representation) with inward-open GAT1 (this study, residues F70, Y86, S295 in stick representation). c, Superposition of inward-open GAT1 structure (this study, residues F70, Y86, S295 in stick representation) with inward-open GlyT131 (PDB ID: 6ZBV, blue, residues F126, Y142, S371 in stick representation). ac are shown to confirm the conformational state (inward-open) of GAT1, together with the conformation of the Na+ and Cl coordinating residues suggesting likely empty ion binding pockets. df, Superimposed structures of inward-open conformation SERT, GlyT1 and GAT1, highlighting differences in the relative orientation of TM1a. g,h, Superposition to GlyT1 and SERT, showing differences in extracellular gate. GlyT1 shows a very similar network of interactions to GAT1, while the inward-open SERT structure shows markedly larger distances between the corresponding residues. Numbers correspond to the shortest atomic distances, between the respective residues, in Å.

Extended Data Fig. 6 Structural comparison of GAT1 (green) with GlyT1 (blue) and SERT (pink) inward-open structures.

a, Superposition of overview models between three structures. b, Focus on the intracellular gate, showing differences in TM6b, TM7, TM1a and TM5. ce, Minor differences between GAT1, GlyT1 and SERT in TM12, TM9, TM10 and TM8 with respective residues highlighted in stick representation: G457 highlights the unique residue in GAT1, potentially leading to additional flexibility of TM10. The interacting residues between ibogaine and SERT TM8 A441-G442-L443 show a subtle helix break, which is unique among the compared sequences and structures.

Extended Data Fig. 7 Summary of MD simulations.

a, Time series showing the shortest observed distance among heavy atoms of the respective GAT1 residues to the 3-methyl-2-thienyl moiety of tiagabine. Numbers in parentheses are the mean and standard deviation of the respective distances in MD simulations. See Supplementary Figs. 1 and 2 for time series of individual trajectories and Supplementary Fig. 3 for histograms of these distances. b, Snapshots of tiagabine binding site for one of the trajectories, taken every 200 ns and spanning 1000 ns. Tiagabine from the cryo-EM structure is colored dark green, MD simulations light green, GAT1 cryo-EM structure in dark pink, GAT1 MD simulations in light pink. c, Root mean square fluctuations (r.m.s.f.) per residue in GAT1. Solid line shows the average of 10 independent trajectories at each residue position; shading refers to 95% confidence interval (n = 10). d, Root mean square deviation (r.m.s.d.) of GAT1 Cα in the 1 µs simulations. The r.m.s.d. are calculated with the protein in trajectories superimposed on the protein in the first frame. e, Root mean square deviation (r.m.s.d.) of tiagabine heavy atoms in the 1 µs simulations. The r.m.s.d. are calculated with the protein in trajectories superimposed on the protein in the first frame.

Extended Data Fig. 8 Structural comparison of known GAT1 substrates and inhibitors.

Blue circle highlights nipecotic acid moiety, sand colored circle highlights bis(3-methyl-2-thienyl) tail.

Extended Data Table 1 Cryo-EM data collection, processing and refinement statistics
Extended Data Table 2 Assembled system for Molecular Dynamics simulation

Supplementary information

Supplementary Information

This file contains Supplementary Table 1 and Supplementary Figures 1–4.

Reporting Summary

Peer Review File

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Motiwala, Z., Aduri, N.G., Shaye, H. et al. Structural basis of GABA reuptake inhibition. Nature 606, 820–826 (2022). https://doi.org/10.1038/s41586-022-04814-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04814-x

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