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:

Autoantibody mimicry of hormone action at the thyrotropin receptor

Abstract

Thyroid hormones are vital in metabolism, growth and development1. Thyroid hormone synthesis is controlled by thyrotropin (TSH), which acts at the thyrotropin receptor (TSHR)2. In patients with Graves’ disease, autoantibodies that activate the TSHR pathologically increase thyroid hormone activity3. How autoantibodies mimic thyrotropin function remains unclear. Here we determined cryo-electron microscopy structures of active and inactive TSHR. In inactive TSHR, the extracellular domain lies close to the membrane bilayer. Thyrotropin selects an upright orientation of the extracellular domain owing to steric clashes between a conserved hormone glycan and the membrane bilayer. An activating autoantibody from a patient with Graves’ disease selects a similar upright orientation of the extracellular domain. Reorientation of the extracellular domain transduces a conformational change in the seven-transmembrane-segment domain via a conserved hinge domain, a tethered peptide agonist and a phospholipid that binds within the seven-transmembrane-segment domain. Rotation of the TSHR extracellular domain relative to the membrane bilayer is sufficient for receptor activation, revealing a shared mechanism for other glycoprotein hormone receptors that may also extend to other G-protein-coupled receptors with large extracellular domains.

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: Cryo-EM structures of native human TSH and TR1402 bound to active TSHR complexed with heterotrimeric Gs.
Fig. 2: The activation mechanism of the TSHR revealed by the inactive structure bound to the inverse agonist CS-17.
Fig. 3: Activation of the TSHR by a Graves’ disease autoantibody.
Fig. 4: Membrane bilayer interactions are critical for TSHR activation.
Fig. 5: Model for TSHR activity.

Similar content being viewed by others

Data availability

Coordinates for TSH-bound TSHR–Gs, TR1402-bound TSHR–Gs, M22-bound TSHR–Gs and CS-17-bound TSHR have been deposited in the PDB under accession codes 7T9I, 7UTZ, 7T9N and 7T9M, respectively. Sharpened and unsharpened cryo-EM density maps for TSH-bound TSHR–Gs (composite), TR1402-bound TSHR–Gs (composite), M22-bound TSHR–Gs (composite), CS-17-bound TSHR, and Org 2274179-0-bound TSHR have been deposited in the Electron Microscopy Data Bank under accession codes EMD-25758, EMD-26795, EMD-25763, EMD-25762 and EMD-27640, respectively. Sharpened maps, unsharpened maps, half-maps and masks for each composite map component (ligand-bound ECD or 7TM–G protein), for TSH-bound TSHR–Gs, TR1402-bound TSHR–Gs and M22-bound TSHR-Gs have been deposited in the Electron Microscopy Data Bank under accession codes EMD-27649 (TSH-bound ECD), EMD-27650 (TSH-bound 7TM–Gs), EMD-27647 (TR1402-bound ECD), EMD-27648 (TR1402 7TM–Gs), EMD-27651 (M22-bound ECD) and EMD-27652 (M22-bound 7TM–Gs). Final particle stacks and .star files for TSH-bound TSHR–Gs, TR1402-bound TSHR–Gs and M22-bound TSHR–Gs and CS-17-bound TSHR-containing particle shift and pose assignments have been uploaded to the Electron Microscopy Public Image Archive under the accession codes EMPIAR-11120. Structure files for OPM-based modelling of inactive and active TSHR are available in the Supplementary Information.

References

  1. Oppenheimer, J. H. & Samuels, H. H. Molecular Basis of Thyroid Hormone Action (Academic Press, 1983).

  2. Vassart, G. & Dumont, J. E. The thyrotropin receptor and the regulation of thyrocyte function and growth. Endocr. Rev. 13, 596–611 (1992).

    CAS  PubMed  Google Scholar 

  3. Smith, T. J. & Hegedüs, L. Graves’ disease. N. Engl. J. Med. 375, 1552–1565 (2016).

    Article  PubMed  Google Scholar 

  4. Fekete, C. & Lechan, R. M. Central regulation of hypothalamic–pituitary–thyroid axis under physiological and pathophysiological conditions. Endocr. Rev. 35, 159–194 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Laurent, E., Mockel, J., Van Sande, J., Graff, I. & Dumont, J. E. Dual activation by thyrotropin of the phospholipase C and cyclic AMP cascades in human thyroid. Mol. Cell. Endocrinol. 52, 273–278 (1987).

    Article  CAS  PubMed  Google Scholar 

  6. Taylor, P. N. et al. Global epidemiology of hyperthyroidism and hypothyroidism. Nat. Rev. Endocrinol. 14, 301–316 (2018).

    Article  PubMed  Google Scholar 

  7. Zimmermann, M. B. & Boelaert, K. Iodine deficiency and thyroid disorders. Lancet Diabetes Endocrinol. 3, 286–295 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Mincer, D. L. & Jialal, I. Hashimoto Thyroiditis. in StatPearls (StatPearls Publishing, 2021); https://www.ncbi.nlm.nih.gov/books/NBK459262/

  9. Lane, L. C., Cheetham, T. D., Perros, P. & Pearce, S. H. S. New therapeutic horizons for Graves’ hyperthyroidism. Endocr. Rev. 41, 873–884 (2020).

    Article  PubMed Central  Google Scholar 

  10. Flack, M. R., Froehlich, J., Bennet, A. P., Anasti, J. & Nisula, B. C. Site-directed mutagenesis defines the individual roles of the glycosylation sites on follicle-stimulating hormone. J. Biol. Chem. 269, 14015–14020 (1994).

    Article  CAS  PubMed  Google Scholar 

  11. Matzuk, M. M., Keene, J. L. & Boime, I. Site specificity of the chorionic gonadotropin N-linked oligosaccharides in signal transduction. J. Biol. Chem. 264, 2409–2414 (1989).

    Article  CAS  PubMed  Google Scholar 

  12. Grossmann, M., Weintraub, B. D. & Szkudlinski, M. W. Novel insights into the molecular mechanisms of human thyrotropin action: structural, physiological, and therapeutic implications for the glycoprotein hormone family. Endocr. Rev. 18, 476–501 (1997).

    Article  CAS  PubMed  Google Scholar 

  13. Jiang, X. et al. Structure of follicle-stimulating hormone in complex with the entire ectodomain of its receptor. Proc. Natl Acad. Sci. USA 109, 12491–12496 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Duan, J. et al. Structures of full-length glycoprotein hormone receptor signalling complexes. Nature 598, 688–692 (2021).

    Article  ADS  PubMed  CAS  Google Scholar 

  15. Chazenbalk, G. D. et al. Evidence that the thyrotropin receptor ectodomain contains not one, but two, cleavage sites. Endocrinology 138, 2893–2899 (1997).

    Article  CAS  PubMed  Google Scholar 

  16. Chen, C.-R., Salazar, L. M., McLachlan, S. M. & Rapoport, B. Deleting the redundant TSH receptor C-peptide region permits generation of the conformationally intact extracellular domain by insect cells. Endocrinology 156, 2732–2738 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nehmé, R. et al. Mini-G proteins: novel tools for studying GPCRs in their active conformation. PLoS ONE 12, e0175642 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Rasmussen, S. G. F. et al. Crystal structure of the β2 adrenergic receptor–Gs protein complex. Nature 477, 549–555 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Szkudlinski, M. W., Teh, N. G., Grossmann, M., Tropea, J. E. & Weintraub, B. D. Engineering human glycoprotein hormone superactive analogues. Nat. Biotechnol. 14, 1257–1263 (1996).

    Article  CAS  PubMed  Google Scholar 

  20. Reinfelder, J. et al. Effects of recombinant human thyroid-stimulating hormone superagonists on thyroidal uptake of 18F-fluorodeoxyglucose and radioiodide. Thyroid 21, 783–792 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Ulloa-Aguirre, A., Timossi, C., Damián-Matsumura, P. & Dias, J. A. Role of glycosylation in function of follicle-stimulating hormone. Endocrine 11, 205–215 (1999).

    Article  CAS  PubMed  Google Scholar 

  22. Fan, Q. R. & Hendrickson, W. A. Structure of human follicle-stimulating hormone in complex with its receptor. Nature 433, 269–277 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Costagliola, S. et al. Tyrosine sulfation is required for agonist recognition by glycoprotein hormone receptors. EMBO J. 21, 504 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kosugi, S., Ban, T., Akamizu, T. & Kohn, L. D. Site-directed mutagenesis of a portion of the extracellular domain of the rat thyrotropin receptor important in autoimmune thyroid disease and nonhomologous with gonadotropin receptors. Relationship of functional and immunogenic domains. J. Biol. Chem. 266, 19413–19418 (1991).

    Article  CAS  PubMed  Google Scholar 

  25. Caltabiano, G. et al. The specificity of binding of glycoprotein hormones to their receptors. Cell. Mol. Life Sci. 65, 2484–2492 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Moyle, W. R. et al. Co-evolution of ligand-receptor pairs. Nature 368, 251–255 (1994).

    Article  ADS  CAS  PubMed  Google Scholar 

  27. Lapthorn, A. J. et al. Crystal structure of human chorionic gonadotropin. Nature 369, 455–461 (1994).

    Article  ADS  CAS  PubMed  Google Scholar 

  28. Wu, H., Lustbader, J. W., Liu, Y., Canfield, R. E. & Hendrickson, W. A. Structure of human chorionic gonadotropin at 2.6 Å resolution from MAD analysis of the selenomethionyl protein. Structure 2, 545–558 (1994).

    Article  CAS  PubMed  Google Scholar 

  29. Grossmann, M. et al. Substitution of the seat-belt region of the thyroid-stimulating hormone (TSH) β-subunit with the corresponding regions of choriogonadotropin or follitropin confers luteotropic but not follitropic activity to chimeric TSH. J. Biol. Chem. 272, 15532–15540 (1997).

    Article  CAS  PubMed  Google Scholar 

  30. Dias, J. A., Zhang, Y. & Liu, X. Receptor binding and functional properties of chimeric human follitropin prepared by an exchange between a small hydrophilic intercysteine loop of human follitropin and human lutropin. J. Biol. Chem. 269, 25289–25294 (1994).

    Article  CAS  PubMed  Google Scholar 

  31. Vischer, H. F., Granneman, J. C. M. & Bogerd, J. Opposite contribution of two ligand-selective determinants in the N-terminal hormone-binding exodomain of human gonadotropin receptors. Mol. Endocrinol. 17, 1972–1981 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Chen, F., Wang, Y. & Puett, D. Role of the invariant aspartic acid 99 of human choriogonadotropin β in receptor binding and biological activity. J. Biol. Chem. 266, 19357–19361 (1991).

    Article  CAS  PubMed  Google Scholar 

  33. Smits, G. et al. Glycoprotein hormone receptors: determinants in leucine-rich repeats responsible for ligand specificity. EMBO J. 22, 2692–2703 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. van Koppen, C. J. et al. Mechanism of action of a nanomolar potent, allosteric antagonist of the thyroid-stimulating hormone receptor. Br. J. Pharmacol. 165, 2314–2324 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Van Sande, J. et al. In Chinese hamster ovary K1 cells dog and human thyrotropin receptors activate both the cyclic AMP and the phosphatidylinositol 4,5-bisphosphate cascades in the presence of thyrotropin and the cyclic AMP cascade in its absence. Eur. J. Biochem. 229, 338–343 (1995).

    Article  PubMed  Google Scholar 

  36. Chen, C.-R., McLachlan, S. M. & Rapoport, B. A monoclonal antibody with thyrotropin (TSH) receptor inverse agonist and TSH antagonist activities binds to the receptor hinge region as well as to the leucine-rich domain. Endocrinology 150, 3401–3408 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Mueller, S., Jaeschke, H., Günther, R. & Paschke, R. The hinge region: an important receptor component for GPHR function. Trends Endocrinol. Metab. 21, 111–122 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Mizutori, Y., Chen, C.-R., McLachlan, S. M. & Rapoport, B. The thyrotropin receptor hinge region is not simply a scaffold for the leucine-rich domain but contributes to ligand binding and signal transduction. Mol. Endocrinol. 22, 1171–1182 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brüser, A. et al. The activation mechanism of glycoprotein hormone receptors with implications in the cause and therapy of endocrine diseases. J. Biol. Chem. 291, 508 (2016).

    Article  PubMed  CAS  Google Scholar 

  40. Schulze, A. et al. The intramolecular agonist is obligate for activation of glycoprotein hormone receptors. FASEB J. 34, 11243–11256 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Parma, J. et al. Somatic mutations causing constitutive activity of the thyrotropin receptor are the major cause of hyperfunctioning thyroid adenomas: identification of additional mutations activating both the cyclic adenosine 3′,5′-monophosphate and inositol phosphate-Ca2+ cascades. Mol. Endocrinol. 9, 725–733 (1995).

    CAS  PubMed  Google Scholar 

  42. Vlaeminck-Guillem, V., Ho, S.-C., Rodien, P., Vassart, G. & Costagliola, S. Activation of the cAMP pathway by the TSH receptor involves switching of the ectodomain from a tethered inverse agonist to an agonist. Mol. Endocrinol. 16, 736–746 (2002).

    Article  CAS  PubMed  Google Scholar 

  43. Sanders, J. et al. Human monoclonal thyroid stimulating autoantibody. Lancet 362, 126–128 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Sanders, J. et al. Crystal structure of the TSH receptor in complex with a thyroid-stimulating autoantibody. Thyroid 17, 395–410 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Evans, M. et al. Monoclonal autoantibodies to the TSH receptor, one with stimulating activity and one with blocking activity, obtained from the same blood sample. Clin. Endocrinol. 73, 404–412 (2010).

    Article  CAS  Google Scholar 

  46. Sanders, P. et al. Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J. Mol. Endocrinol. 46, 81–99 (2011).

    CAS  PubMed  Google Scholar 

  47. 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 

  48. Reily, C., Stewart, T. J., Renfrow, M. B. & Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. 15, 346–366 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Erbel, P. J. A., Haseley, S. R., Kamerling, J. P. & Vliegenthart, J. F. G. Studies on the relevance of the glycan at Asn-52 of the α-subunit of human chorionic gonadotropin in the αβ dimer. Biochem. J. 364, 485–495 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Amr, S. et al. Activities of deglycosylated thyrotropin at the thyroid membrane receptor-adenylate cyclase system. J. Endocrinol. Invest. 8, 537–541 (2014).

    Article  Google Scholar 

  51. Fares, F. A., Levi, F., Reznick, A. Z. & Kraiem, Z. Engineering a potential antagonist of human thyrotropin and thyroid-stimulating antibody. J. Biol. Chem. 276, 4543–4548 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Feng, X., Müller, T., Mizrachi, D., Fanelli, F. & Segaloff, D. L. An intracellular loop (IL2) residue confers different basal constitutive activities to the human lutropin receptor and human thyrotropin receptor through structural communication between IL2 and helix 6, via helix 3. Endocrinology 149, 1705–1717 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Zhang, M. et al. Intrinsic differences in the response of the human lutropin receptor versus the human follitropin receptor to activating mutations. J. Biol. Chem. 282, 25527–25539 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Skiba, M. A. & Kruse, A. C. Autoantibodies as endogenous modulators of GPCR signaling. Trends Pharmacol. Sci. 42, 135–150 (2021).

    Article  CAS  PubMed  Google Scholar 

  55. Girdlestone, C. & Hayward, S. The DynDom3D webserver for the analysis of domain movements in multimeric proteins. J. Comput. Biol. 23, 21–26 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Sanders, J. et al. Characteristics of a human monoclonal autoantibody to the thyrotropin receptor: sequence structure and function. Thyroid 14, 560–570 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. Mastronarde, D. N. SerialEM: a program for automated tilt series acquisition on Tecnai microscopes using prediction of specimen position. Microsc. Microanal. 9, 1182–1183 (2003).

    Article  ADS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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 

  60. 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 

  61. Scheres, S. H. W. A Bayesian view on cryo-EM structure determination. J. Mol. Biol. 415, 406–418 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  65. Zhuang, Y. et al. Mechanism of dopamine binding and allosteric modulation of the human D1 dopamine receptor. Cell Res. 31, 593–596 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  67. Croll, T. I. ISOLDE: a physically realistic environment for model building into low-resolution electron-density maps. Acta Crystallogr. D 74, 519–530 (2018).

    Article  CAS  Google Scholar 

  68. 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 

  69. 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 

  70. Bligh, E. G. & Dyer, W. J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Physiol. 37, 911–917 (1959).

    Article  CAS  PubMed  Google Scholar 

  71. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Marty, M. T. et al. Bayesian deconvolution of mass and ion mobility spectra: from binary interactions to polydisperse ensembles. Anal. Chem. 87, 4370–4376 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Zhang, L. & Hermans, J. Hydrophilicity of cavities in proteins. Proteins 24, 433–438 (1996).

    Article  CAS  PubMed  Google Scholar 

  74. Betz, R. Dabble. https://doi.org/10.5281/zenodo.836914 (2017).

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

    Article  CAS  PubMed  Google Scholar 

  76. Klauda, J. B. et al. Update of the CHARMM all-atom additive force field for lipids: validation on six lipid types. J. Phys. Chem. B 114, 7830–7843 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Case, D. A. et al. Amber 2021 (Univ. California, San Francisco, 2021).

  78. Salomon-Ferrer, R., Götz, A. W., Poole, D., Le Grand, S. & Walker, R. C. Routine microsecond molecular dynamics simulations with AMBER on GPUs. 2. Explicit solvent particle mesh Ewald. J. Chem. Theory Comput. 9, 3878–3888 (2013).

    Article  CAS  PubMed  Google Scholar 

  79. Hopkins, C. W., Le Grand, S., Walker, R. C. & Roitberg, A. E. Long-time-step molecular dynamics through hydrogen mass repartitioning. J. Chem. Theory Comput. 11, 1864–1874 (2015).

    Article  CAS  PubMed  Google Scholar 

  80. Roe, D. R. & Cheatham, T. E. III. PTRAJ and CPPTRAJ: software for processing and analysis of molecular dynamics trajectory data. J. Chem. Theory Comput. 9, 3084–3095 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 27–28 (1996).

    Article  Google Scholar 

  82. Latorraca, N. R. et al. How GPCR phosphorylation patterns orchestrate arrestin-mediated signaling. Cell 183, 1813–1825.e18 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Asarnow, D., Palovcak, E. & Cheng, Y. UCSF pyem v0. 5. https://doi.org/10.5281/zenodo.3576630 (2019).

Download references

Acknowledgements

We thank R. Yan and D. Toso for help in microscope operation and data collection; C. Bracken for mass spectrometry troubleshooting; and H. Shan for sample preparation for lipid mass spectrometry. This work was supported by National Institutes of Health (NIH) grants DP5OD023048 (A.M.), 1R35GM140847 (Y.C.), P30CA014195, R01GM102491 (A.S.), R01GM127359 (R.O.D.), 5R44CA224376 (M.W.S.) and a Human Frontier Science Program Long-Term Fellowship LT000916/2018-L (C.-M.S.). Cryo-EM equipment at UCSF is partially supported by NIH grants S10OD020054 and S10OD021741. Some of this work was performed at the Stanford-SLAC Cryo-EM Center (S2C2), which is supported by the National Institutes of Health Common Fund Transformative High-Resolution Cryo-Electron Microscopy program (U24 GM129541). This research was, in part, supported by the National Cancer Institute’s National Cryo-EM Facility at the Frederick National Laboratory for Cancer Research under contract HSSN261200800001E. Some of this work was supported by the Mass Spectrometry Core of the Salk Institute with funding from NIH-NCI CCSG: P30CA014195. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. A.M. acknowledges support from the Pew Charitable Trusts, the Esther A. & Joseph Klingenstein Fund, the Searle Scholars Program, the Edward Mallinckrodt Jr Foundation, and the Vallee Foundation. Y.C. is an Investigator of Howard Hughes Medical Institute. A.M. is a Chan Zuckerberg Biohub investigator.

Author information

Authors and Affiliations

Authors

Contributions

B.F. cloned, expressed, and biochemically optimized the purification of all TSHR constructs for structural studies. B.F. expressed and purified Gβ1γ2, Nb35, M22 Fab, CS-17 IgG and K1-70 IgGs, and performed enzymatic digestion and further purification to form CS-17 and K1-70 Fab fragments. B.F. performed complexing and identified optimal cryo-EM grid preparation procedures, screened samples, and collected 300 keV datasets. B.F. determined high-resolution cryo-EM maps by extensive image processing under the guidance of A.M. and Y.C. B.F. and A.M built and refined models of TSHR complexes. B.F. and C.B.B. generated receptor constructs and determined expression levels by flow cytometry and performed signalling studies and analysed the data. C.B.B. assisted with cloning of TSHR constructs, expression, purification and labelling of CS-17 and M22 Fab fragments, and cloning and generation of baculoviruses for expression of Gβ1γ2. C.-M.S. performed and analysed molecular dynamics simulations under the supervision of R.O.D. I.S. performed initial biochemical optimization of TSHR complexes with M22 Fab and worked with K.Z. to collect negative stain and cryo-EM data. N.H. prepared samples for, performed, and analysed native mass spectrometry experiments and prepared control samples for lipid identification experiments. J.K.D. analysed native human TSH glycosylation by mass spectrometry. Y.M. generated TSHR and M22 Fab expression constructs and performed pilot biochemical purification of TSHR complexes. A.F.M.P. performed and analysed data from the lipid identification experiments with guidance from A.S. M.W.S. led production of highly pure TR1402 agonist. Figures were generated and the manuscript was written by B.F., C.B.B. and A.M., with edits from Y.C., R.O.D., C.-M.S., and M.W.S. and with approval from all authors. The overall project was supervised by Y.C. and A.M.

Corresponding authors

Correspondence to Yifan Cheng or Aashish Manglik.

Ethics declarations

Competing interests

A.M. and R.O.D. are consultants for and stockholders in Septerna Inc. Y.C. is a consultant and advisor of Shuimu BioScience Ltd.

Peer review

Peer review information

Nature thanks Marvin Gershengorn, Patrick M. Sexton 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 Signaling studies for TSHR.

a, b) DNA titrations of TSHR mutants tested in cAMP production assays. Median fluorescence intensity ± SD (n = 3) of anti-FLAG-A647 staining is plotted. Filled in bars represent the DNA concentration used for co-transfection experiments with pGlo cAMP biosensor plasmid. Horizontal red line indicates the mean wild-type (WT) TSHR fluorescence intensity. Flow cytometry experiments were performed in triplicate, with identical gating across all cell lines tested. c) cAMP production of TSHR cysteine mutants comparing the basal level (-) to 100 nM of TSH, M22, or CS-17 for untreated cells (left panel) and cells treated with 500 µM TCEP (right panel). Data points are means ± SD from three biological replicates. d, e) cAMP production curves comparing TSH and M22-mediated activation of WT and C-peptide deleted TSHR constructs. f) cAMP production curves for TSH and TR1402-mediated WT TSHR activation. g) cAMP production curves for TSH-mediated WT and TSHR mutant cell line (Y385F, Y385A) activation. h) cAMP production curves for M22-mediated WT and lipid-displacing TSHR mutant cell line (A644K, A647K) activation. Plotted data points in panels c-g are means of triplicate measurements ± SD from a representative experiment of n = 3 biological replicates.

Extended Data Fig. 2 Cryo-EM data processing for TSH-bound TSHR-Gs complex.

a) Representative image from 15,345 micrographs. Scale bar, 50 nm. b) Selected 2D class averages. c) Processing approach used for reconstruction of TSH-bound TSHR-Gs complex. A local resolution map was calculated from cryoSPARC using masks from indicated local refinement, then visualized with the composite map in the same scale. A viewing distribution plot was generated using scripts from the pyEM software suite83 and visualized in ChimeraX. GS-FSC and Directional FSC (dFSC, shown as purple lines) curves were generated in cryoSPARC and as previously described in Dang, S. et al. Nature 552, 426–429 (2017).

Extended Data Fig. 3 Model map correlation coefficients for cryo-EM structures.

Correlation values for resolved residues in each modeled chain are shown. Low values indicate regions that are poorly resolved, e.g. the 7TM domain of TSHR in CS-17-TSHR complex or the constant domains of the Fab fragments for CS-17 and M22.

Extended Data Fig. 4 Cryo-EM data processing for TR1402-bound TSHR-Gs complex.

a) Representative image from 14,277 micrographs. Scale bar, 50 nm. b) Selected 2D class averages from final reconstruction. c) Processing approach used for reconstruction of TR1402-bound TSHR-Gs complex. A local resolution map was calculated from cryoSPARC using masks from indicated local refinement, then visualized with the composite map in the same scale. A viewing distribution plot was generated using scripts from the pyEM software suite and visualized in ChimeraX. GS-FSC and dFSC curves were generated in cryoSPARC and as previously described in Dang, S. et al. Nature 552, 426–429 (2017).

Extended Data Fig. 5 Glycosylation status of native human TSH.

a) Cryo-EM density map of TSH-bound TSHR showing resolved glycan density for Asn52. b) Peptide coverage of Asn52 from native human TSH GPHα chain in mass spectrometry experiments. c) Peptide spectra match (PSM) of glycans detected on Asn52 of GPHα chain. d,e) Representative MS/MS spectra showing Asn52 with various length glycans.

Extended Data Fig. 6 Receptor:hormone interaction comparisons across the glycoprotein hormone receptor family.

a) Comparison of TSHR, FSHR (PDB: 4AY9), and LH/CGR ECD:hormone (PDB: 7FIH) interactions shows the comparable concave binding-interface and lateral-ECD contacts made by GPHα and receptor-specific β subunits. b) Alignment of the GPHR ECDs reveals the variability in modeled poses of hinge-hormone contacts observed in prior GPHR:hormone structures. c) The N-terminal hinge-hormone disulfide-linked alpha helix presents a hydrophobic face to the α-L1 and α-L3 loops of the TR1402 α-chain and is stabilized by multiple phenylalanine contacts in the GPHα and TSHβ chains. C-terminal hinge-region α helix hidden for visualization. d) Comparison of the TSHR and LHCGR (PDB: 7FIH) transmembrane pockets highlights the similarity in the DPPC and the allosteric agonist Org43554 binding sites. TM4/5 and ECL2 not shown. e) Lipid:protein molar ratio comparison between the M22:TSHR:Gs complex and a non-GPHR Class A receptor control. Data points represent individual measurements from technical replicates (n = 3 for non-GPHR control and n = 4 for M22:TSHR:Gs complex) of the ratio of pmol of lipid DPPC per pmol of protein. **P = 0.0088; Unpaired two-tailed t test was used to calculate statistical differences in lipid:protein molar ratios.

Extended Data Fig. 7 Cryo-EM data processing for Org 274179-0 bound TSHR.

a) Representative image from 10,003 micrographs. Scale bar, 50 nm. b) Selected 2D class averages generated from curated particles. c) Processing approach used for low resolution reconstruction. Despite starting with a similar or larger sized dataset as for other TSHR samples, the TSHR–Org 274179-0 complex did not yield high resolution reconstruction of the 7TM domain. A low-resolution reconstruction of the TSHR ECD was observed. This suggests potential flexibility between the TSHR 7TM domain and the ECD.

Extended Data Fig. 8 Cryo EM data processing for the CS-17 bound TSHR:Org 274179-0 complex.

a) Representative image from 4,952 micrographs. Scale bar, 50 nm. b) Selected 2D class averages generated from the final reconstruction. c) Processing approach used for reconstruction of the complex. A viewing distribution plot was generated using scripts from the pyEM software suite and visualized in ChimeraX. Local resolution map generated from non-uniform refinement mask in cryoSPARC. GS-FSC and dFSC curves were generated in cryoSPARC and as previously described in Dang, S. et al. Nature 552, 426–429 (2017).

Extended Data Fig. 9 Comparison of Org 274179-0 bound- and CS-17 bound TSHR EM maps.

a) Org 274179-0 bound-TSHR EM density map (dark) fit into the CS-17-bound TSHR map (colored by TSHR-CS-17 model). b) CS-17-bound TSHR model fit into Org 274179-0 bound-TSHR map, suggesting that inactive state TSHR ECD orientations are similar between CS-17 and Org 274179-0-bound states.

Extended Data Fig. 10 Comparison of TSHR activation with other GPCRs.

a) Ribbon diagram of TSHR in active and inactive conformations. b) Ribbon diagram of LH/CGR in active (PDB:7FIH) and inactive (PDB:7FIJ) conformations. A similar reorientation of the ECD is shared between TSHR and LH/CGR upon activation. c) Comparison of active TSHR to active β2-adrenoceptor (β2AR, PDB:3SN6). d) Comparison of inactive TSHR to inactive β2AR, PDB:5JQH). e) Comparison of active conformations of TSHR and LH/CGR reveals similar overall structures of the 7TM domain and the p10 peptide.

Extended Data Fig. 11 Cryo-EM data processing for M22-bound TSHR-Gs complex.

a) Representative image from 25,030 micrographs. Scale bar, 50 nm. b) Selected 2D class averages from final reconstruction. c) Processing approach used for reconstruction of M22-bound TSHR-Gs complex. A local resolution map was calculated from cryoSPARC using masks from indicated local refinement, then visualized with the composite map in the same scale. A viewing distribution plot was generated using scripts from the pyEM software suite and visualized in ChimeraX. GS-FSC and dFSC curves were generated in cryoSPARC and as previously described in Dang, S. et al. Nature 552, 426–j429 (2017).

Extended Data Fig. 12 Extent of ECD activation for all individual simulations across four simulation conditions.

Dashed lines indicate the projection metric values (see Methods) in the inactive (0.0 Å) and active (33.5 Å) state cryo-EM structures. Thick traces indicate the moving average smoothed over a 25-ns window, and thin traces represent unsmoothed data. Each column represents a distinct simulation condition: started from the active structure, with M22 bound (brown traces, first column); started from the active structure, with M22 removed (grey traces, second column); started from the inactive structure, with CS-17 bound (blue traces, third column); started from the inactive structure, with CS-17 removed (grey traces, fourth column).

Extended Data Fig. 13 Glycosylation of engineered K1-70glyco construct.

a) Native mass spectrum (nMS) of K1-70, K1-70glyco, and K1-70glyco(N16Q) Fab fragments. b) Accurate mass assignment of Fab fragments. nMS demonstrates ~1.7 kDa increased mass for K1-70glyco and more heterogeneity consistent with N-linked glycosylation. The smaller mass and increased homogeneity of the K1-70glyco(N16Q) construct further supports glycosylation at the engineered N16 position. c) Crystal structure of K1-70 TSHR-ECD complex (PDB: 2XWT) aligned to CS-17 bound TSHR. The membrane-proximal N229 residue (of the “GlycoB” glycosylation motif) is highlighted in red. d) cAMP production comparing K1-70 WT, K1-70 GlycoB and M22 Fab fragment-mediated activation of TSHR. Plotted data points are means of triplicate measurements ± SD from one of three biological replicates.

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

Supplementary information

Supplementary Table 1

Summary of TSHR signalling studies. Values are expressed as mean pEC50 or mean Emax ± s.e.m. from (n) biological replicates. ND, Not determined.

Reporting Summary

Supplementary Data 1

Structure files for static modelling of TSHR in active and inactive conformations bound to TSH, M22 CS-17, and K1-70.

Peer Review File

Supplementary Video 1

3D variability analysis of TR1402-bound TSHR–Gs complex.

Rights and permissions

Springer Nature or its licensor 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

Faust, B., Billesbølle, C.B., Suomivuori, CM. et al. Autoantibody mimicry of hormone action at the thyrotropin receptor. Nature 609, 846–853 (2022). https://doi.org/10.1038/s41586-022-05159-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-05159-1

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