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:

Transport mechanism of presynaptic high-affinity choline uptake by CHT1

Abstract

Choline is a vital nutrient and a precursor for the biosynthesis of essential metabolites, including acetylcholine (ACh), that play a central role in fetal development, especially in the brain. In cholinergic neurons, the high-affinity choline transporter (CHT1) provides an extraordinarily efficient reuptake mechanism to reutilize choline derived from intrasynaptical ACh hydrolysis and maintain ACh synthesis in the presynapse. Here, we determined structures of human CHT1 in three discrete states: the outward-facing state bound with the competitive inhibitor hemicholinium-3 (HC-3); the inward-facing occluded state bound with the substrate choline; and the inward-facing apo open state. Our structures and functional characterizations elucidate how the inhibitor and substrate are recognized. Moreover, our findings shed light on conformational changes when transitioning from an outward-facing to an inward-facing state and establish a framework for understanding the transport cycle, which relies on the stabilization of the outward-facing state by a short intracellular helix, IH1.

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

Access options

Buy this article

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

Fig. 1: Architecture of human CHT1.
Fig. 2: Human CHT1 in the HC-3-bound outward-facing state.
Fig. 3: CHT1 in the inward-facing apo and choline-bound inward-facing occluded states.
Fig. 4: Conformational changes between CHT1HC-3/OF and CHT1choline/occ.
Fig. 5: Molecular mechanism of choline transport.

Similar content being viewed by others

Data availability

The cryo-EM density maps of the dimeric CHT1HC-3/OF, monomeric CHT1HC-3/OF, CHT1apo/IF and CHT1choline/occ have been deposited in the Electron Microscopy Data Bank (EMDB) under the accession codes EMD-36025, EMD-36027, EMD-36029 and EMD-36030, respectively. The unsharpened cryo-EM maps, used for model building of the C-terminal region, have also been deposited in the EMDB as additional EM maps within the corresponding deposition entries. The coordinates have been deposited in the Protein Data Bank (PDB) under PDB IDs 8J74, 8J75, 8J76 and 8J77. Molecular dynamics trajectories were deposited into a public repository and are accessible at the following link: https://doi.org/10.5281/zenodo.10526461.

Source data are provided with this paper.

References

  1. Zeisel, S. H. Choline: critical role during fetal development and dietary requirements in adults. Annu. Rev. Nutr. 26, 229–250 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Zeisel, S. H. & Blusztajn, J. K. Choline and human nutrition. Annu. Rev. Nutr. 14, 269–296 (1994).

    CAS  PubMed  Google Scholar 

  3. Sarter, M. & Parikh, V. Choline transporters, cholinergic transmission and cognition. Nat. Rev. Neurosci. 6, 48–56 (2005).

    CAS  PubMed  Google Scholar 

  4. Zeisel, S. Choline, other methyl-donors and epigenetics. Nutrients 9, 445 (2017).

    PubMed  PubMed Central  Google Scholar 

  5. Bekdash, R. A. Neuroprotective effects of choline and other methyl donors. Nutrients 11, 2995 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Tuček, S. Choline acetyltransferase and the synthesis of acetylcholine. In The Cholinergic Synapse (ed. Whittaker, V. P.) 125–165 (Springer Berlin Heidelberg, 1988).

  7. Carlson, A. B. & Kraus, G. P. Physiology, Cholinergic Receptors (StatPearls Publishing, 2022).

  8. Bunge, R., Johnson, M. & Ross, C. D. Nature and nurture in development of the autonomic neuron. Science 199, 1409–1416 (1978).

    CAS  PubMed  Google Scholar 

  9. Everitt, B. J. & Robbins, T. W. Central cholinergic systems and cognition. Annu. Rev. Psychol. 48, 649–684 (1997).

    CAS  PubMed  Google Scholar 

  10. Gallagher, M. & Colombo, P. J. Ageing: the cholinergic hypothesis of cognitive decline. Curr. Opin. Neurobiol. 5, 161–168 (1995).

    CAS  PubMed  Google Scholar 

  11. Brookes, S. J. H., Steele, P. A. & Costa, M. Identification and immunohistochemistry of cholinergic and non-cholinergic circular muscle motor neurons in the guinea-pig small intestine. Neuroscience 42, 863–878 (1991).

    CAS  PubMed  Google Scholar 

  12. Silman, I. & Sussman, J. L. Acetylcholinesterase: ‘classical’ and ‘non-classical’ functions and pharmacology. Curr. Opin. Pharmacol. 5, 293–302 (2005).

    CAS  PubMed  Google Scholar 

  13. Soreq, H. & Seidman, S. Acetylcholinesterase — new roles for an old actor. Nat. Rev. Neurosci. 2, 294–302 (2001).

    CAS  PubMed  Google Scholar 

  14. Okuda, T. et al. Identification and characterization of the high-affinity choline transporter. Nat. Neurosci. 3, 120–125 (2000).

    CAS  PubMed  Google Scholar 

  15. Apparsundaram, S., Ferguson, S. M., George, A. L. Jr & Blakely, R. D. Molecular cloning of a human, hemicholinium-3-sensitive choline transporter. Biochem. Biophys. Res. Commun. 276, 862–867 (2000).

    CAS  PubMed  Google Scholar 

  16. Okuda, T. & Haga, T. Functional characterization of the human high‐affinity choline transporter. FEBS Lett. 484, 92–97 (2000).

    CAS  PubMed  Google Scholar 

  17. Okuda, T. & Haga, T. High-affinity choline transporter. Neurochem. Res. 28, 483–488 (2003).

    CAS  PubMed  Google Scholar 

  18. Haga, T. Synthesis and release of [14C]acetylcholine in synaptosomes. J. Neurochem. 18, 781–798 (1971).

    CAS  PubMed  Google Scholar 

  19. Kuhar, M. J. & Murrin, L. C. Sodium-dependent, high affinity choline uptake. J. Neurochem. 30, 15–21 (1978).

    CAS  PubMed  Google Scholar 

  20. Ferguson, S. M. et al. Lethal impairment of cholinergic neurotransmission in hemicholinium-3-sensitive choline transporter knockout mice. Proc. Natl Acad. Sci. USA 101, 8762–8767 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Barwick, K. E. et al. Defective presynaptic choline transport underlies hereditary motor neuropathy. Am. J. Hum. Genet. 91, 1103–1107 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Bauché, S. et al. Impaired presynaptic high-affinity choline transporter causes a congenital myasthenic syndrome with episodic apnea. Am. J. Hum. Genet 99, 753–761 (2016).

    PubMed  PubMed Central  Google Scholar 

  23. Neumann, S. A. et al. Polymorphic variation in choline transporter gene (CHT1) is associated with early, subclinical measures of carotid atherosclerosis in humans. Int. J. Cardiovasc. Imaging 28, 243–250 (2012).

    PubMed  Google Scholar 

  24. Hahn, M. K. et al. Multivariate permutation analysis associates multiple polymorphisms with subphenotypes of major depression. Genes Brain Behav. 7, 487–495 (2008).

    CAS  PubMed  Google Scholar 

  25. Bissette, G., Seidler, F. J., Nemeroff, C. B. & Slotkin, T. A. High affinity choline transporter status in Alzheimer’s disease tissue from rapid autopsy. Ann. N. Y. Acad. Sci. 777, 197–204 (1996).

    CAS  PubMed  Google Scholar 

  26. Pascual, J. et al. High-affinity choline uptake carrier in Alzheimer’s disease: implications for the cholinergic hypothesis of dementia. Brain Res. 552, 170–174 (1991).

    CAS  PubMed  Google Scholar 

  27. Ferreira-Vieira, T. H., Guimaraes, I. M., Silva, F. R. & Ribeiro, F. M. Alzheimer’s disease: targeting the cholinergic system. Curr. Neuropharmacol. 14, 101–115 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Ribeiro, F. M. et al. The “ins” and “outs” of the high-affinity choline transporter CHT1. J. Neurochem. 97, 1–12 (2006).

    CAS  PubMed  Google Scholar 

  29. Haga, T. Molecular properties of the high-affinity choline transporter CHT1. J. Biochem. 156, 181–194 (2014).

    CAS  PubMed  Google Scholar 

  30. Uchida, Y. et al. Expression and functional characterization of choline transporter in human keratinocytes. J. Pharm. Sci. 109, 102–109 (2009).

    CAS  Google Scholar 

  31. Mineur, Y. S. et al. Cholinergic signaling in the hippocampus regulates social stress resilience and anxiety-and depression-like behavior. Proc. Natl Acad. Sci. 110, 3573–3578 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Picciotto, M. R., Higley, M. J. & Mineur, Y. S. Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior. Neuron 76, 116–129 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Barker, L. & Mittag, T. Comparative studies of substrates and inhibitors of choline transport and choline acetyltransferase. J. Pharmacol. Exp. Ther. 192, 86–94 (1975).

    CAS  PubMed  Google Scholar 

  34. Vickroy, T. W. et al. Quantitative light microscopic autoradiography of [3H] hemicholinium-3 binding sites in the rat central nervous system: a novel biochemical marker for mapping the distribution of cholinergic nerve terminals. Brain Res. 329, 368–373 (1985).

    CAS  PubMed  Google Scholar 

  35. Patrice, G., Pierre, L., Jean, R., Jean Claude, B. & Jacques, G. Inhibition by hemicholinium-3 of [14C] acetylcholine synthesis and [3H] choline high-affinity uptake in rat striatal synaptosomes. Mol. Pharmacol. 9, 630 (1973).

    Google Scholar 

  36. Payette, D. J., Xie, J. & Guo, Q. Reduction in CHT1-mediated choline uptake in primary neurons from presenilin-1 M146V mutant knock-in mice. Brain Res. 1135, 12–21 (2007).

    CAS  PubMed  Google Scholar 

  37. Cummings, M. D., Farnum, M. A. & Nelen, M. I. Universal screening methods and applications of ThermoFluor. SLAS Discov. 11, 854–863 (2006).

    CAS  Google Scholar 

  38. Boivin, S., Kozak, S. & Meijers, R. Optimization of protein purification and characterization using Thermofluor screens. Protein Expr. Purif. 91, 192–206 (2013).

    CAS  PubMed  Google Scholar 

  39. Okuda, T. et al. Transmembrane topology and oligomeric structure of the high-affinity choline transporter. J. Biol. Chem. 287, 42826–42834 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Manaker, S., Wieczorek, C. M. & Rainbow, T. C. Identification of sodium-dependent, high-affinity choline uptake sites in rat brain with [3H]hemicholinium-3. J. Neurochem. 46, 483–488 (1986).

    CAS  PubMed  Google Scholar 

  41. Chatterjee, T. K., Long, J. P., Cannon, J. G. & Bhatnagar, R. K. Methylpiperidine analog of hemicholinium-3: a selective, high affinity non-competitive inhibitor of sodium dependent choline uptake system. Eur. J. Pharmacol. 149, 241–248 (1988).

    CAS  PubMed  Google Scholar 

  42. Han, L. et al. Structure and mechanism of the SGLT family of glucose transporters. Nature 601, 274–279 (2022).

    CAS  PubMed  Google Scholar 

  43. Vickroy, T. W., Roeske, W. R. & Yamamura, H. I. Sodium-dependent high-affinity binding of [3H]hemicholinium-3 in the rat brain: a potentially selective marker for presynaptic cholinergic sites. Life Sci. 35, 2335–2343 (1984).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Wahlgren, W. Y. et al. Substrate-bound outward-open structure of a Na+-coupled sialic acid symporter reveals a new Na+ site. Nat. Commun. 9, 1753 (2018).

    PubMed  PubMed Central  Google Scholar 

  46. Bisignano, P. et al. Inhibitor binding mode and allosteric regulation of Na+-glucose symporters. Nat. Commun. 9, 5245 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Niu, Y. et al. Structural basis of inhibition of the human SGLT2–MAP17 glucose transporter. Nature 601, 280–284 (2022).

    CAS  PubMed  Google Scholar 

  48. Ravera, S. et al. Structural insights into the mechanism of the sodium/iodide symporter. Nature 612, 795–801 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Niu, Y. et al. Structural mechanism of SGLT1 inhibitors. Nat. Commun. 13, 6440 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  51. Dong, Y. et al. Structure and mechanism of the human NHE1–CHP1 complex. Nat. Commun. 12, 3474 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Gao, Y. et al. Molecular insights into the gating mechanisms of voltage-gated calcium channel Ca(V)2.3. Nat. Commun. 14, 516 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Dong, Y. et al. Closed-state inactivation and pore-blocker modulation mechanisms of human Ca(V)2.2. Cell Rep. 37, 109931 (2021).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Schorb, M., Haberbosch, I., Hagen, W. J. H., Schwab, Y. & Mastronarde, D. N. Software tools for automated transmission electron microscopy. Nat. Methods 16, 471–477 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  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).

    CAS  PubMed  Google Scholar 

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

    CAS  Google Scholar 

  59. Ding, K. et al. Observing noncovalent interactions in experimental electron density for macromolecular systems: a novel perspective for protein–ligand interaction research. J. Chem. Inf. Model. 62, 1734–1743 (2022).

    CAS  PubMed  Google Scholar 

  60. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    CAS  PubMed  Google Scholar 

  61. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    CAS  PubMed  Google Scholar 

  62. Abraham, M. J. et al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 1–2, 19–25 (2015).

    Google Scholar 

  63. Vanommeslaeghe, K. et al. CHARMM general force field: a force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. J. Comput. Chem. 31, 671–690 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. 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).

    CAS  PubMed  Google Scholar 

  65. 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).

    CAS  PubMed  Google Scholar 

  66. Jorgensen, W. L., Chandrasekhar, J., Madura, J. D., Impey, R. W. & Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys. 79, 926–935 (1983).

    CAS  Google Scholar 

  67. Bussi, G., Donadio, D. & Parrinello, M. Canonical sampling through velocity rescaling. J. Chem. Phys. 126, 014101 (2007).

    PubMed  Google Scholar 

  68. Parrinello, M. & Rahman, A. Crystal structure and pair potentials: a molecular-dynamics study. Phys. Rev. Lett. 45, 1196–1199 (1980).

    CAS  Google Scholar 

  69. Essmann, U. et al. A smooth particle mesh Ewald method. J. Chem. Phys. 103, 8577–8593 (1995).

    CAS  Google Scholar 

  70. Hess, B. P-LINCS: a parallel linear constraint solver for molecular simulation. J. Chem. Theory Comput. 4, 116–122 (2008).

    CAS  PubMed  Google Scholar 

  71. Daura, X. et al. Peptide folding: when simulation meets experiment. Angew. Chem. Int. Ed. 38, 236–240 (1999).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank L. Chen (Peking University) and Y. Niu (Peking University) for their helpful discussions. We thank X. Huang, B. Zhu, X. Li and other staff members at the Center for Biological Imaging (CBI), Core Facilities for Protein Science at the Institute of Biophysics, Chinese Academy of Science for their support in cryo-EM data collection; and H. Zhang (Institute of Biophysics, Chinese Academy of Sciences) and T. Sun (Institute of Microbiology, Chinese Academy of Sciences) for their assistance in the [3H]choline-uptake assays. We thank Y. Ren (Institute of Biophysics, Chinese Academy of Sciences) for his assistance in the immunoblotting assays. We thank Y. Wu for his research assistance. This work is funded by Chinese National Programs for Brain Science and Brain-like Intelligence Technology (grant no. 2022ZD0205800 to Y.Z.), the National Key Research and Development Program of China (grant no. 2021YFA1301501 to Y.Z.), the National Natural Science Foundation of China (grant no. 92157102 to Y.Z.) and the Chinese Academy of Sciences Strategic Priority Research Program (grant no. XDB37030304 to Y.Z.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. conceived the project and supervised the research. Y.Q. carried out molecular cloning experiments, expressed and purified protein samples and prepared samples for cryo-EM study. Y.G. and Y.Q. carried out cryo-EM data collection. Y.G. processed the cryo-EM data and built and refined the atomic model. Y.G., Y.Q. and Y.Z. analyzed the structure. B.H. and Q.B. conducted the molecular dynamic simulations. Y.G. wrote the original draft of the manuscript and prepared the figures. Y.Z., Y.G. and Y.Q. edited the manuscript.

Corresponding author

Correspondence to Yan Zhao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Katarzyna Ciazynska, in collaboration with the Nature Structural & Molecular Biology team.

Additional information

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

Extended data

Extended Data Fig. 1 Protein purification of human CHT1.

a. Size-exclusion chromatography of purified human CHT1 protein sample in NaCl/LMNG. Fraction within dashed lines were pooled for cryo-EM sample preparation. b. SDS-PAGE gel of purified human CHT1. Band smears because of heterogeneity caused by N-linked glycosylation. Purification of human CHT1 in the presence (cd) and absence (ef) of 40 mM choline using KCl buffer. c,e. Size-exclusion chromatography of purified human CHT1 protein sample (black) and reconstituted CHT1 in nanodiscs (red). Fraction within dashed lines were pooled for nanodisc reconstitution or cryo-EM sample preparation. d,f. SDS-PAGE gel of purified human CHT1 and MSP1D1E3. Band smears because of heterogeneity caused by N-linked glycosylation. The experiments were repeated independently for more than 3 times with identical results.

Source data

Extended Data Fig. 2 Cryo-EM data processing of dimeric CHT1HC-3/OF.

a. Data processing pipeline for the human dimeric CHT1 in the outward-facing state (CHT1HC-3/OF). A representative micrograph is shown here (Bar = 400 Å). A ‘seed-facilitated’ data processing strategy were used (see Method section for details). Particles were cleaned using multiple rounds of classifications. Monomeric CHT1 were processed separately (see Method section and Extended Data Fig. 3). The final map was reported at 3.6 Å according to GSFSC criterion. b. Angular distribution of the human dimeric CHT1 reconstruction. The height of each spike indicates the particle number at designated orientations. c. Sharpened map of dimeric CHT1, colored according to the estimated value of local resolution. d. Fourier shell correlations (FSC) curves of the dimeric CHT1 and its atomic model. The unmasked (blue) and masked (red) FSC curves was calculated between two independently refined half-maps before and after map sharpening. The model-vs-map FSC (black) was calculated between the full map and the atomic model. e. Representative cryo-EM density map. Critical residues are labeled. f. Different views of the model within the map.

Extended Data Fig. 3 Cryo-EM data processing of monomeric CHT1HC-3/OF.

a. Data processing pipeline for the human monomeric CHT1 in the outward-facing state (CHT1HC-3/OF). A representative micrograph is shown here (Bar = 400 Å). Particles were cleaned using multiple rounds of classifications. The final map was reported at 3.6 Å according to GSFSC criterion. b. Angular distribution of the monomeric CHT1 reconstruction. The height of each spike indicates the particle number at designated orientations. c. Sharpened map of monomeric CHT1, colored according to the estimated value of local resolution. d. Fourier shell correlations (FSC) curves of the monomeric CHT1 and its atomic model. The unmasked (blue) and masked (red) FSC curves was calculated between two independently refined half-maps before and after map sharpening. The model-vs-map FSC (black) was calculated between the full map and the atomic model. e. Representative cryo-EM density map. Critical residues are labeled. f. Different views of the model within the map.

Extended Data Fig. 4 Assembly of the dimeric CHT1HC-3/OF.

a. Cartoon representation of dimeric CHT1HC-3/OF in the side view (up) and the top-down view (down). The two protomers are colored pink and cyan, respectively. N-glycans and CHS are shown as gold sticks. LMNG are shown as green sticks. Dimeric interactions at the extracellular side were indicated using a dashed box and elaborated in (b). b. Zoomed-in view of the interaction at the dimer interface. CHS is shown as gold sticks. LMNG is shown as green sticks, overlaid with transparent grey surface. Residues involved in interactions with LMNG are shown as sticks and labeled. c. Interactions between the two protomers at the intracellular side of dimeric CHT1. Residues that may contribute to CHT1 dimerization were shown as sticks and labeled. d. Superimposition between the dimeric (pink and cyan) and monomeric (grey) CHT1 in the outward-facing state.

Extended Data Fig. 5 Surface expression of CHT1 WT and mutants.

a. Left, β-actin protein was only detected in the cell lysate rather than the surface protein elutes, indicating that the intracellular proteins were not biotinylated. The Na+/K+ ATPase protein was detected in the elution sample from biotinylated cells, indicating that the plasma membrane proteins can be effectively biotinylated and purified using streptavidin beads. FLAG-tagged CHT1 was observed in the lysate of cells expressing CHT1 WT and in the elution from streptavidin beads, but not in cells without CHT1 expression or in unbiotinylated cells, confirming that the biotinylated CHT1 can be detected using the anti-FLAG antibody. Right, immunoblots of biotinylated CHT1 WT and mutants on cell surface. Biotinylated Na+/K+ ATPase on the cell surface was used as the loading control. b. Densitometric quantification of surface expression of CHT1 WT and mutants (glycosylated and non-glycosylated), normalized using density values of Na+/K+ ATPase within a single batch of experiment, and mean density values of all bands (CHT1 WT and mutants) across independent batches of experiments. Each triangular symbol indicates a single data point. Data are represented as mean ± S.E.M. n = 4 biologically independent experiments.

Source data

Extended Data Fig. 6 Cryo-EM data processing of CHT1apo/IF.

a. Data processing pipeline for human CHT1 in the inward-facing apo state (CHT1apo/IF). A representative micrograph is shown here (Bar = 400 Å). Particles were cleaned using multiple rounds of classifications. The final map was reported at 3.7 Å according to GSFSC criterion. b. Angular distribution of the CHT1 reconstruction in the inward-facing apo state. The height of each spike indicates the particle number at designated orientations. c. Sharpened map of CHT1 in the inward-facing apo state, colored according to the estimated value of local resolution. d. Fourier shell correlations (FSC) curves of the inward-facing apo CHT1 and its atomic model. The unmasked (blue) and masked (red) FSC curves was calculated between two independently refined half-maps before and after map sharpening. The model-vs-map FSC (black) was calculated between the full map and the atomic model. e. Representative cryo-EM density map. Critical residues are labeled. f. Different views of the model within the map.

Extended Data Fig. 7 Cryo-EM data processing of CHT1choline/occ.

a. Data processing pipeline for human CHT1 in the choline-bound inward-facing occluded state (CHT1choline/occ). A representative micrograph is shown here (Bar = 400 Å). Particles were cleaned using multiple rounds of classifications. The final map was reported at 3.7 Å according to GSFSC criterion. b. Angular distribution of the CHT1 reconstruction in the inward-facing occluded state. The height of each spike indicates the particle number at designated orientations. c. Sharpened map of CHT1 in the inward-facing occluded state, colored according to the estimated value of local resolution. d. Fourier shell correlations (FSC) curves of the inward-facing occluded CHT1 and its atomic model. The unmasked (blue) and masked (red) FSC curves was calculated between two independently refined half-maps before and after map sharpening. The model-vs-map FSC (black) was calculated between the full map and the atomic model. e. Representative cryo-EM density map. Critical residues are labeled. f. Different views of the model within the map.

Extended Data Fig. 8 Structural comparison between CHT1apo/IF and CHT1choline/occ and choline binding site.

ab. Superimposition between the CHT1apo/IF (blue) and CHT1choline/occ (red). cd. Binding pocket of choline in the inward-facing CHT1 structures, overlaid with their corresponding density shown in blue mesh. Choline and critical residues that may contribute to choline recognition are labeled. e. Backbone R.M.S.D. of CHT1 (excluding choline and high flexibility loop residue 27–49 and 501–517) from the initial structure and the heavy atom R.M.S.D. of choline from its initial position are plotted over time. f. The representative structure obtained from the clustering analysis involving three 120 ns independent trajectories. Only one cluster was observed considering choline and the pocket (i. e. protein residues within 5 Å of the choline) and using 1.5 Å as the R.M.S.D. cut-off. The hydrogen bonds between the hydroxyl group of choline and residues W62, W406 are indicated with dashed lines. g. Statistical analysis of hydrogen bonds between choline and the residues in the choline binding pocket for Trajectory 1–3 and the concatenated trajectory. Hydrogen bonds were detected with a polarity heavy atom donor-acceptor distance of 3.5 Å and an angle cut-off of 40°.

Source data

Extended Data Fig. 9 Structural analysis of CHT1 and SMCT1.

a. Superimposition between CHT1apo/IF (blue) and the inward-facing SMCT1 (grey). b. ThermoFluor curves of the IH1/(GS)3 substitution (CHT1IH1/GS) mutant. CHT1IH1/GS respond to choline but failed to exhibit sensitivity to HC-3 in the NaCl or KCl buffers.

Source data

Extended Data Fig. 10 Structure analysis of the sodium binding sites.

ab. Structural comparison between NIS (gray) and CHT1choline/occ (red). Na1 and the non-canonical NIS Na2 sites of NIS were labeled. The sodium ions and the iodide ion were shown as spheres and labeled. Residues interacting with the ions are shown as sticks and labeled in (b). cd. Structural comparison between the outward-facing SiaT (grey) and CHT1HC-3/OF (green). Sodium ions at the Na2 (c) and Na3 (d) sites of SiaT were shown as spheres and labeled. Critical residues within the sodium binding sites are shown as sticks or spheres (G64 Cα atom) and labeled. Residues constituting the sodium binding sites via backbone carbonyl were labeled within square brackets. e. Sequence alignment of canonical Na2 and canonical Na3 sites between CHT1 homologues. f. Structures of CHT1HC-3/OF (green) and CHT1choline/occ (red), overlaid with transparent grey surfaces. Canonical sodium binding sites Na2 and Na3 sites (SiaT nomination) are labeled. Residues constituting the binding sites were shown as sticks. The Na3 site are occluded in the outward-facing open CHT1HC-3/OF (left) and exposed to the cytosol in the inward-facing occluded CHT1choline/occ (right).

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qiu, Y., Gao, Y., Huang, B. et al. Transport mechanism of presynaptic high-affinity choline uptake by CHT1. Nat Struct Mol Biol 31, 701–709 (2024). https://doi.org/10.1038/s41594-024-01259-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-024-01259-w

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