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 insights into the mechanism of the sodium/iodide symporter

Abstract

The sodium/iodide symporter (NIS) is the essential plasma membrane protein that mediates active iodide (I) transport into the thyroid gland, the first step in the biosynthesis of the thyroid hormones—the master regulators of intermediary metabolism. NIS couples the inward translocation of I against its electrochemical gradient to the inward transport of Na+ down its electrochemical gradient1,2. For nearly 50 years before its molecular identification3, NIS was the molecule at the centre of the single most effective internal radiation cancer therapy: radioiodide (131I) treatment for thyroid cancer2. Mutations in NIS cause congenital hypothyroidism, which must be treated immediately after birth to prevent stunted growth and cognitive deficiency2. Here we report three structures of rat NIS, determined by single-particle cryo-electron microscopy: one with no substrates bound; one with two Na+ and one I bound; and one with one Na+ and the oxyanion perrhenate bound. Structural analyses, functional characterization and computational studies show the substrate-binding sites and key residues for transport activity. Our results yield insights into how NIS selects, couples and translocates anions—thereby establishing a framework for understanding NIS function—and how it transports different substrates with different stoichiometries and releases substrates from its substrate-binding cavity into the cytosol.

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: Structure of NIS.
Fig. 2: Iodide- and sodium-binding sites.
Fig. 3: Perrhenate-binding pocket and localized changes caused by ion binding.
Fig. 4: NIS mechanism.

Similar content being viewed by others

Data availability

Cryo-EM maps and atomic coordinates of the structures presented in this manuscript have been deposited in the Protein Data Bank (PDB) and Electron Microscopy Data Bank (EMDB) under accession codes 7UUY and EMD-26806 (for Apo-NIS), 7UV0 and EMD-26808 (for NIS-I) and 7UUZ and EMD-26807 (for NIS-ReO4). The trajectories and the Jupyter notebook used for molecular dynamics simulations are available at https://codeocean.com/capsule/1458259/tree.

References

  1. Portulano, C., Paroder-Belenitsky, M. & Carrasco, N. The Na+/I symporter (NIS): mechanism and medical impact. Endocr. Rev. 35, 106–149 (2014).

    Article  CAS  Google Scholar 

  2. Ravera, S., Reyna-Neyra, A., Ferrandino, G., Amzel, L. M. & Carrasco, N. The sodium/iodide symporter (NIS): molecular physiology and preclinical and clinical applications. Annu. Rev. Physiol. 79, 261–289 (2017).

    Article  CAS  Google Scholar 

  3. Dai, G., Levy, O. & Carrasco, N. Cloning and characterization of the thyroid iodide transporter. Nature 379, 458–460 (1996).

    Article  ADS  CAS  Google Scholar 

  4. Mullur, R., Liu, Y. Y. & Brent, G. A. Thyroid hormone regulation of metabolism. Physiol. Rev. 94, 355–382 (2014).

    Article  CAS  Google Scholar 

  5. Zhang, Z., Liu, F. & Chen, J. Molecular structure of the ATP-bound, phosphorylated human CFTR. Proc. Natl Acad. Sci. USA 115, 12757–12762 (2018).

    Article  ADS  CAS  Google Scholar 

  6. Reyna-Neyra, A. et al. The iodide transport defect-causing Y348D mutation in the Na(+)/I(-) symporter renders the protein intrinsically inactive and impairs its targeting to the plasma membrane. Thyroid 31, 1272–1281 (2021).

    Article  CAS  Google Scholar 

  7. Tazebay, U. H. et al. The mammary gland iodide transporter is expressed during lactation and in breast cancer. Nat. Med. 6, 871–878 (2000).

    Article  CAS  Google Scholar 

  8. Spitzweg, C. et al. The sodium iodide symporter (NIS): novel applications for radionuclide imaging and treatment. Endocr. Relat. Cancer 28, T193–T213 (2021).

    Article  CAS  Google Scholar 

  9. Kitzberger, C. et al. The sodium iodide symporter (NIS) as theranostic gene: its emerging role in new imaging modalities and non-viral gene therapy. EJNMMI Res. 12, 25 (2022).

    Article  CAS  Google Scholar 

  10. Urnauer, S. et al. EGFR-targeted nonviral NIS gene transfer for bioimaging and therapy of disseminated colon cancer metastases. Oncotarget 8, 92195–92208 (2017).

    Article  Google Scholar 

  11. Miller, A. & Russell, S. J. The use of the NIS reporter gene for optimizing oncolytic virotherapy. Expert Opin. Biol. Ther. 16, 15–32 (2016).

    Article  CAS  Google Scholar 

  12. Li, W., Nicola, J. P., Amzel, L. M. & Carrasco, N. Asn441 plays a key role in folding and function of the Na+/I symporter (NIS). FASEB J. 27, 3229–3238 (2013).

    Article  CAS  Google Scholar 

  13. Nicola, J. P. et al. Sodium/iodide symporter mutant V270E causes stunted growth but no cognitive deficiency. J. Clin. Endocrinol. Metab. 100, E1353–E1361 (2015).

    Article  CAS  Google Scholar 

  14. Paroder-Belenitsky, M. et al. Mechanism of anion selectivity and stoichiometry of the Na+/I- symporter (NIS). Proc. Natl Acad. Sci. USA 108, 17933–17938 (2011).

    Article  ADS  CAS  Google Scholar 

  15. Levy, O. et al. N-linked glycosylation of the thyroid Na+/I symporter (NIS). Implications for its secondary structure model. J. Biol. Chem. 273, 22657–22663 (1998).

    Article  CAS  Google Scholar 

  16. Levy, O. et al. Characterization of the thyroid Na+/I symporter with an anti-COOH terminus antibody. Proc. Natl Acad. Sci. USA 94, 5568–5573 (1997).

    Article  ADS  CAS  Google Scholar 

  17. Paroder, V., Nicola, J. P., Ginter, C. S. & Carrasco, N. The iodide transport defect-causing mutation R124H: a δ-amino group at position 124 is critical for maturation and trafficking of the Na+/I- symporter (NIS). J. Cell Sci. 126, 3305–3313 (2013).

    CAS  Google Scholar 

  18. De la Vieja, A., Reed, M. D., Ginter, C. S. & Carrasco, N. Amino acid residues in transmembrane segment IX of the Na+/I symporter play a role in its Na+ dependence and are critical for transport activity. J. Biol. Chem. 282, 25290–25298 (2007).

    Article  Google Scholar 

  19. Chew, T. A. et al. Structure and mechanism of the cation-chloride cotransporter NKCC1. Nature 572, 488–492 (2019).

    Article  ADS  CAS  Google Scholar 

  20. 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  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  22. Niu, Y. et al. Structural basis of inhibition of the human SGLT2-MAP17 glucose transporter. Nature 601, 280–284 (2021).

    Article  ADS  Google Scholar 

  23. Yamashita, A., Singh, S. K., Kawate, T., Jin, Y. & Gouaux, E. Crystal structure of a bacterial homologue of Na+/Cl-dependent neurotransmitter transporters. Nature 437, 215–223 (2005).

    Article  ADS  CAS  Google Scholar 

  24. Faham, S. et al. The crystal structure of a sodium galactose transporter reveals mechanistic insights into Na+/sugar symport. Science 321, 810–814 (2008).

    Article  ADS  CAS  Google Scholar 

  25. Wang, J., Liu, Z., Frank, J. & Moore, P. B. Identification of ions in experimental electrostatic potential maps. IUCrJ 5, 375–381 (2018).

    Article  CAS  Google Scholar 

  26. Zhekova, H. R. et al. Mapping of ion and substrate binding sites in human sodium iodide symporter (hNIS). J. Chem. Inf. Model. 60, 1652–1665 (2020).

    Article  CAS  Google Scholar 

  27. Nicola, J. P., Carrasco, N. & Amzel, L. M. Physiological sodium concentrations enhance the iodide affinity of the Na+/I symporter. Nat. Commun. 5, 3948 (2014).

    Article  ADS  CAS  Google Scholar 

  28. Ravera, S., Quick, M., Nicola, J. P., Carrasco, N. & Amzel, L. M. Beyond non-integer Hill coefficients: a novel approach to analyzing binding data, applied to Na+-driven transporters. J. Gen. Physiol. 145, 555–563 (2015).

    Article  Google Scholar 

  29. Dohan, O. et al. The Na+/I symporter (NIS) mediates electroneutral active transport of the environmental pollutant perchlorate. Proc. Natl Acad. Sci. USA 104, 20250–20255 (2007).

    Article  ADS  CAS  Google Scholar 

  30. Tran, N. et al. Thyroid-stimulating hormone increases active transport of perchlorate into thyroid cells. Am. J. Physiol. Endocrinol. Metab. 294, E802–E806 (2008).

    Article  CAS  Google Scholar 

  31. Zuckier, L. S. et al. Kinetics of perrhenate uptake and comparative biodistribution of perrhenate, pertechnetate, and iodide by NaI symporter-expressing tissues in vivo. J. Nucl. Med. 45, 500–507 (2004).

    CAS  Google Scholar 

  32. Eskandari, S. et al. Thyroid Na+/I- symporter. Mechanism, stoichiometry, and specificity. J. Biol. Chem. 272, 27230–27238 (1997).

    Article  CAS  Google Scholar 

  33. Llorente-Esteban, A. et al. Allosteric regulation of mammalian Na+/I symporter activity by perchlorate. Nat. Struct. Mol. Biol. 27, 533–539 (2020).

    Article  CAS  Google Scholar 

  34. Boutagy, N. E. et al. Noninvasive in vivo quantification of adeno-associated virus serotype 9-mediated expression of the sodium/iodide symporter under hindlimb ischemia and neuraminidase desialylation in skeletal muscle using single-photon emission computed tomography/computed tomography. Circ. Cardiovasc. Imaging 12, e009063 (2019).

    Article  Google Scholar 

  35. Pavelka, A. et al. CAVER: algorithms for analyzing dynamics of tunnels in macromolecules. IEEE/ACM Trans. Comput. Biol. Bioinform. 13, 505–517 (2016).

    Article  Google Scholar 

  36. Ferrandino, G. et al. Na+ coordination at the Na2 site of the Na+/I symporter. Proc. Natl Acad. Sci. USA 113, E5379–E5388 (2016).

    Article  CAS  Google Scholar 

  37. Sun, L. et al. Molecular dynamics simulations of the surface tension and structure of salt solutions and clusters. J. Phys. Chem. 116, 3198–3204 (2012).

    Article  CAS  Google Scholar 

  38. Carugo, O. Buried chloride stereochemistry in the Protein Data Bank. BMC Struct. Biol. 14, 19 (2014).

    Article  Google Scholar 

  39. Kang, B., Tang, H., Zhao, Z. & Song, S. Hofmeister series: insights of ion specificity from amphiphilic assembly and interface property. ACS Omega 5, 6229–6239 (2020).

    Article  CAS  Google Scholar 

  40. Marcus, Y. A simple empirical model describing the thermodynamics of hydration of ions of widely varying charges, sizes, and shapes. Biophys. Chem. 51, 111–127 (1994).

    Article  CAS  Google Scholar 

  41. Yang, D. & Gouaux, E. Illumination of serotonin transporter mechanism and role of the allosteric site. Sci. Adv. 7, eabl3857 (2021).

    Article  ADS  CAS  Google Scholar 

  42. Coleman, J. A. & Gouaux, E. Structural basis for recognition of diverse antidepressants by the human serotonin transporter. Nat. Struct. Mol. Biol. 25, 170–175 (2018).

    Article  CAS  Google Scholar 

  43. Levy, O., Ginter, C. S., De la Vieja, A., Levy, D. & Carrasco, N. Identification of a structural requirement for thyroid Na+/I- symporter (NIS) function from analysis of a mutation that causes human congenital hypothyroidism. FEBS Lett. 429, 36–40 (1998).

    Article  CAS  Google Scholar 

  44. Ferrandino, G. et al. Na+ coordination at the Na2 site of the Na+/I- symporter. Proc. Natl Acad. Sci. USA 113, E5379–E5388 (2016).

    Article  CAS  Google Scholar 

  45. Ravera, S., Quick, M., Nicola, J. P., Carrasco, N. & Amzel, L. M. Beyond non-integer Hill coefficients: a novel approach to analyzing binding data, applied to Na+-driven transporters. J. Gen. Physiol. 145, 555–563 (2015).

    Article  Google Scholar 

  46. 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  Google Scholar 

  47. Rohou, A. & Grigorieff, N. CTFFIND4: fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    Article  Google Scholar 

  48. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    Article  CAS  Google Scholar 

  49. 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  Google Scholar 

  50. 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  Google Scholar 

  51. Moriya, T. et al. Size matters: optimal mask diameter and box size for single-particle cryogenic electron microscopy. Preprint at bioRxiv https://doi.org/10.1101/2020.08.23.263707 (2020).

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

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  54. Pettersen, E. F. et al. UCSF Chimera-a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  Google Scholar 

  55. Górski, K. M. et al. HEALPix: a framework for high-resolution discretization and fast analysis of data distributed on the sphere. Astrophys. J. 622, 759–771 (2005).

  56. Wang, J., Liu, Z., Frank, J. & Moore, P. B. Identification of ions in experimental electrostatic potential maps. IUCrJ 5, 375–381 (2018).

    Article  CAS  Google Scholar 

  57. Lee, J. et al. CHARMM-GUI input generator for NAMD, GROMACS, AMBER, OpenMM, and CHARMM/OpenMM simulations using the CHARMM36 additive force field. J. Chem. Theory Comput. 12, 405–413 (2016).

    Article  CAS  Google Scholar 

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

    Article  ADS  Google Scholar 

  59. Huang, J. et al. CHARMM36m: an improved force field for folded and intrinsically disordered proteins. Nat. Methods 14, 71–73 (2017).

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  Google Scholar 

  62. Parrinello, M. & Rahman, A. Polymorphic transitions in single crystals: a new molecular dynamics method. J. Appl. Phys. 52, 7182–7190 (1981).

    Article  ADS  CAS  Google Scholar 

  63. Hess, B., Bekker, H., Berendsen, H. J. & Fraaije, J. G. LINCS: a linear constraint solver for molecular simulations. J. Comput. Chem. 18, 1463–1472 (1997).

    Article  CAS  Google Scholar 

  64. Li, P., Song, L. F. & Merz, K. M. Jr. Systematic parameterization of monovalent ions employing the nonbonded model. J. Chem. Theory Comput. 11, 1645–1657 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Mchaourab and members of the Carrasco laboratory for critical reading of the manuscript and insightful discussion, and S. Gabelli for helpful suggestions. This study was supported by National Institutes of Health (NIH) grants GM R01 114250 (to N.C. and L.M.A.), NS021501 (to F.J.S.) and NINDS R21NS108842, and a Pamela Mars Wright Innovator Award (to M.A.B.). We thank S. Wu of the Yale Cryo-EM Resource facility for screening and data collection. This work was conducted in part using the CPU and GPU resources at the Advanced Computing Center for Research and Education at Vanderbilt University and at the Maryland Advanced Research Computing Center. We used the DORS storage system supported by NIH (no. S10RR031634 to J. Smith).

Author information

Authors and Affiliations

Authors

Contributions

S.R., N.C. and L.M.A. conceived the project. S.R. expressed and purified proteins and prepared cryo-grids. S.R. and E.K. processed cryo-EM data. S.R., E.K., L.M.A. and M.A.B. built and refined the atomic models. F.J.S. implemented the protocol for ion identification. S.R. and M.A.B. carried out molecular dynamics simulations. S.R., J.P.N. and G.S.S. generated mutant NIS proteins and carried out functional assays. S.R. and N.C. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Nancy Carrasco.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Jue Chen, Wojciech Kopec, Christine Spitzweg and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Cryo-EM data processing and determination of the structure of NIS.

Topology model of the engineered NIS molecule (T-NIS) whose cDNA was used to transduce 293F cells. We engineered an HA tag onto the N-terminus and a HIS and an SBP (streptavidin-binding protein) tag onto the C-terminus for affinity purification. The tags are separated from NIS by a TEV protease site at the N-terminus and a PreScission site at the C-terminus. b. Enrichment of 293F cells expressing NIS by flow cytometry using an anti-HA antibody. After two rounds of sorting, >96% of the cells expressed NIS at the plasma membrane. c. I transport assay. T-NIS transports virtually as much I as wild-type (WT) NIS does, whereas nontransduced (NT) cells transport no I . Results are expressed as pmol of I accumulated/µg DNA ± s.e.m. Values represent averages of the results from two different experiments, each of which was carried out in triplicate (n = 6). d. Size exclusion chromatography (SEC) of the fraction of NIS purified in LMNG/GDN used for cryo-EM imaging detected by Trp-fluorescence. e. Coomassie blue staining of SEC-purified NIS subjected to SDS-PAGE (representative gel (n=3); see also Supplementary Fig. 1). f. Cryo-EM micrograph of apo-NIS (representative of 8025 micrographs that yielded similar results). The scale bar represents 200 nm. g. Selected 2D class averages obtained after 4 rounds of 2D classification using Relion (top image) and data-processing workflow (bottom two rows). h. Fourier shell correlation (FSC) of the locally refined map. i. NIS dimer map colored according to local resolution. j. Fitting of NIS sequence to the electron density map. k. Local refinement.

Extended Data Fig. 2 Cryo-EM densities and model of TMSs and dimeric assembly of NIS.

a. α-helical features are clearly visible in all 13 TMSs. b. NIS structure viewed from the extracellular side of the membrane, with the numbered TMs depicted as cylinders (left panel). NIS embedded in the membrane, top view; an example of 2D classes representing the corresponding view is shown in the black square (middle panel). NIS structure viewed from the intracellular side of the membrane, with the numbered TMs depicted as cylinders (right panel). c. Residues interacting at the dimer interface in apo-NIS.

Extended Data Fig. 3 Cryo-EM data processing and determination of the structures of NIS with substrates bound.

a. NIS-I. b. NIS-ReO4 .

Extended Data Fig. 4 I binds to a partially positively charged cavity.

a. NIS-I side view. The solvent accessible surface is colored according to the electrostatic potential using a double gradient between −2 EV (red) and 0 (white) and between 0 and 2 EV (blue) and cropped to expose the I binding cavity. b. Close-up of the I binding cavity showing the positive nature of its surface.

Extended Data Fig. 5 Effects of single amino acid substitutions at positions 69, 72, 144, 416, 417 on iodide transport.

a–e. NIS-mediated I uptake at steady state. cDNA constructs coding for NIS mutants were transfected into COS7 or HEK cells. I uptake by these NIS mutants was measured at 20 µM (light gray bars) and 200 µM (dark gray bars) I at 140 mM Na+ for 30 min with or without the NIS-specific inhibitor ClO4 to determine NIS-mediated transport (values obtained in the presence of ClO4, which are < 10% of the values obtained in its absence, have already been subtracted). f–j. Kinetic analysis of initial rates of I uptake (2-min time points) determined at 140 mM Na+ and varying concentrations of I. k–o. Kinetic analysis of initial rates of I uptake (2-min time points) determined at varying concentrations of extracellular Na+. All results are expressed as pmol of I accumulated/µg DNA ± s.e.m. Values represent averages of the results from two or three different experiments, each of which was carried out in triplicate (n ≥ 6).

Extended data Fig. 6 Identification of the ions in the NIS-ReO4 structure.

a. The ions transported by NIS were identified by evaluating the map density along 24 lines passing through each site (circled in yellow on images of map slices). Values are plotted for each line and the spherically-averaged mean is plotted in black (lower panels). b. Effects of substitutions in binding-site residues on ReO4 transport at steady state (measured at 3 µM ReO4 and 140 mM Na+ for 30 min (values obtained in the presence of ClO4, which are < 10% of the values obtained in its absence, have already been subtracted). Values are normalized to those obtained with WT NIS. Values represent averages of the results from two or three different experiments, each of which was carried out in triplicate (n ≥ 6) and reported as pmol ReO4 accumulated/µg DNA ± s.e.m.

Extended Data Fig. 7 Effects of single amino acid substitutions at positions 72, 94, 416 and 417 on ReO4 transport.

a-d. NIS-mediated ReO4 uptake at steady state. cDNA constructs coding for NIS mutants in which Q72 is replaced with the residues indicated were transfected into COS7 or HEK cells. ReO4 uptake by these NIS mutants was measured at 3 µM (light gray bars) and 30 µM (dark gray bars) ReO4 at 140 mM Na+ for 30 min with or without the NIS-specific inhibitor ClO4 (values obtained in the presence of ClO4 already subtracted). Results are given as pmols of ReO4 accumulated/µg DNA ± s.e.m. Values represent averages of the results from two or three different experiments, each of which was carried out in triplicate (n ≥ 6). e. Kinetic analysis of initial rates of ReO4 uptake (2-min time points) for Q72 NIS mutants determined at varying concentrations of extracellular ReO4 and varying concentrations of extracellular Na+. Results are given as pmols of ReO4 accumulated/µg DNA ± s.e.m. Values represent averages of the results from two or three different experiments, each of which was carried out in triplicate (n ≥ 6). f. ReO4 KM values determined from (e); error bars represent the standard deviation of the Michaelis-Menten analysis. g. Kinetic analysis of initial rates of ReO4 uptake (2-min time points) for Q72 NIS mutants determined at 100 µM ReO4 and varying concentrations of extracellular Na+. Results are given as pmols of ReO4 accumulated/µg DNA ± s.e.m. Values represent averages of the results from two or three different experiments, each of which was carried out in triplicate (n ≥ 6). h. Na+ KM values determined from (g); error bars represent the standard deviation of the Hill equation analysis.

Extended Data Fig. 8 Entry pathway for the NIS substrates.

a. Surface representation of a side view of the NIS-I structure: the arrow and the dotted square indicate the position of the proposed entry pathway. b. Close-up of the top view of the surface showing the substrates (Na+ ions represented by yellow spheres, I by a magenta sphere), and the positions of F87, L413, and Q414 in the NIS-I structure (magenta) and the models generated from MD simulations corresponding to the opening (wheat) of the substrate-binding cavity to the extracellular milieu. c. Magnification of the top of the substrate-binding cavity. The arrows indicate how the amino acids move away from their original positions as the cavity transitions from closed to open. d. Ramachandran plots of the chi-1 and chi-2 side chain dihedral angles of F87, L413, and Q414 visited during the MD simulations with NIS-I. The dihedral angles selected are the principal determinants of the position of the side chain. The excursions of these dihedral angles (during the MD simulations) away from the conformational basins corresponding to the cryoEM structure (green dots in basins 1, 1, and 2, in F87, L413, and Q414, respectively) and toward conformational basins (blue dots in basins 1, 2, and 3, in F87, L413, and Q414, respectively) open up the entry path (b). In these histograms, the frequency of a given conformational state is indicated by a rainbow gradient from deep purple (0 frequency) to red (highest frequency); the most highly populated conformational basins are numbered in descending order of population.

Extended Data Fig. 9 Perrhenate binding mechanism starting from the apo-NIS structure.

A hydrogen-bond network between F67, S69, Q72, and Y144 and a hydrophobic stacking interaction between F67 and Q72 are disrupted by the binding of the first Na+. This facilitates the binding of a second Na+ and ReO4, which causes the release of one Na+.

Extended Data Fig. 10 Apo-NIS structure with amino acids mutated in patients with iodide transport defects shown as spheres.

Single amino acid substitutions are indicated by red spheres; deleted residues are in blue. The substitutions found in patients are: G18R, V59E, G93R, R124H, Q267E, V270E, D331N, Y348D, T354P, G395R, G543E, S547R, G561E and the deletion of 439-443 (ACNTP).

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–11 and Table 1.

Reporting Summary

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

Ravera, S., Nicola, J.P., Salazar-De Simone, G. et al. Structural insights into the mechanism of the sodium/iodide symporter. Nature 612, 795–801 (2022). https://doi.org/10.1038/s41586-022-05530-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05530-2

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