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:

High-resolution crystal structure of parathyroid hormone 1 receptor in complex with a peptide agonist

Abstract

Parathyroid hormone 1 receptor (PTH1R) is a class B multidomain G-protein-coupled receptor (GPCR) that controls calcium homeostasis. Two endogenous peptide ligands, parathyroid hormone (PTH) and parathyroid hormone–related protein (PTHrP), activate the receptor, and their analogs teriparatide and abaloparatide are used in the clinic to increase bone formation as an effective yet costly treatment for osteoporosis. Activation of PTH1R involves binding of the peptide ligand to the receptor extracellular domain (ECD) and transmembrane domain (TMD), a hallmark of class B GPCRs. Here, we present the crystal structure of human PTH1R in complex with a peptide agonist at 2.5-Å resolution, allowing us to delineate the agonist binding mode for this receptor and revealing molecular details within conserved structural motifs that are critical for class B receptor function. Thus, this study provides structural insight into the function of PTH1R and extends our understanding of this therapeutically important class of GPCRs.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: PTH1R–ePTH complex crystal structure.
Fig. 2: Structural and functional details of PTH1R–ePTH interactions.
Fig. 3: Water-mediated extension and conformational changes upon activation in the central polar network.
Fig. 4: Influence of bulky residues in helices I and II on the activation of class B GPCRs.

Similar content being viewed by others

Data availability

Coordinates and structure factors have been deposited in Protein Data Bank (PDB 6FJ3). All other data are available from the corresponding authors upon reasonable request.

References

  1. Gardella, T. J. & Vilardaga, J.-P. International Union of Basic and Clinical Pharmacology. XCIII. The parathyroid hormone receptors–family B G protein-coupled receptors. Pharmacol. Rev. 67, 310–337 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Selye, H. On the stimulation of new bone-formation with parathyroid extract and irradiated ergosterol. Endocrinology 16, 547–558 (1932).

    Article  CAS  Google Scholar 

  3. Cheloha, R. W., Gellman, S. H., Vilardaga, J.-P. & Gardella, T. J. PTH receptor-1 signalling-mechanistic insights and therapeutic prospects. Nat. Rev. Endocrinol. 11, 712–724 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Miller, P. D. et al. Effect of abaloparatide vs placebo on new vertebral fractures in postmenopausal women with osteoporosis: a randomized clinical trial. JAMA 316, 722–733 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Kendler, D. L. et al. Effects of teriparatide and risedronate on new fractures in post-menopausal women with severe osteoporosis (VERO): a multicentre, double-blind, double-dummy, randomised controlled trial. Lancet 391, 230–240 (2017).

    Article  PubMed  Google Scholar 

  6. Pioszak, A. A. & Xu, H. E. Molecular recognition of parathyroid hormone by its G protein-coupled receptor. Proc. Natl Acad. Sci. USA 105, 5034–5039 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bisello, A. et al. Role of glycosylation in expression and function of the human parathyroid hormone/parathyroid hormone-related protein receptor. Biochemistry 35, 15890–15895 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Lee, S.-M. et al. N-Glycosylation of asparagine 130 in the extracellular domain of the human calcitonin receptor significantly increases peptide hormone affinity. Biochemistry 56, 3380–3393 (2017).

    Article  CAS  PubMed  Google Scholar 

  9. Liang, Y.-L. et al. Phase-plate cryo-EM structure of a class B GPCR-G-protein complex. Nature 546, 118–123 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Liang, Y.-L. et al. Phase-plate cryo-EM structure of a biased agonist-bound human GLP-1 receptor–Gs complex. Nature 68, 954 (2018).

    Google Scholar 

  11. Zhang, Y. et al. Cryo-EM structure of the activated GLP-1 receptor in complex with a G protein. Nature 546, 248–253 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, H. et al. Structure of the glucagon receptor in complex with a glucagon analogue. Nature 553, 106–110 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Siu, F. Y. et al. Structure of the human glucagon class B G-protein-coupled receptor. Nature 499, 444–449 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Zhang, H. et al. Structure of the full-length glucagon class B G-protein-coupled receptor. Nature 546, 259–264 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Koth, C. M. et al. Molecular basis for negative regulation of the glucagon receptor. Proc. Natl Acad. Sci. USA 109, 14393–14398 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yang, L. et al. Conformational states of the full-length glucagon receptor. Nat. Commun. 6, 7859 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Jazayeri, A. et al. Crystal structure of the GLP-1 receptor bound to a peptide agonist. Nature 546, 254–258 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Lee, C. et al. Role of the extracellular regions of the parathyroid hormone (PTH)/PTH-related peptide receptor in hormone binding. Endocrinology 135, 1488–1495 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Lee, C. et al. Homolog-scanning mutagenesis of the parathyroid hormone (PTH) receptor reveals PTH-(1-34) binding determinants in the third extracellular loop. Mol. Endocrinol. 9, 1269–1278 (1995).

    CAS  PubMed  Google Scholar 

  20. Zhao, L.-H. et al. Differential requirement of the extracellular domain in activation of class B G protein-coupled receptors. J. Biol. Chem. 291, 15119–15130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Weaver, R. E., Wigglesworth, M. J. & Donnelly, D. A salt bridge between Arg-20 on parathyroid hormone (PTH) and Asp-137 on the PTH1 receptor is essential for full affinity. Peptides 61, 83–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Jin, L. et al. Crystal structure of human parathyroid hormone 1-34 at 0.9-A resolution. J. Biol. Chem. 275, 27238–27244 (2000).

    CAS  PubMed  Google Scholar 

  23. Condon, S. M. et al. The bioactive conformation of human parathyroid hormone. structural evidence for the extended helix postulate. J. Am. Chem. Soc. 122, 3007–3014 (2000).

    Article  CAS  Google Scholar 

  24. Mierke, D. F. et al. Conformational studies of mono- and bicyclic parathyroid hormone-related protein-derived agonists. Biochemistry 36, 10372–10383 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Wootten, D., Simms, J., Miller, L. J., Christopoulos, A. & Sexton, P. M. Polar transmembrane interactions drive formation of ligand-specific and signal pathway-biased family B G protein-coupled receptor conformations. Proc. Natl Acad. Sci. USA 110, 5211–5216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shimizu, M., Carter, P. H., Khatri, A., Potts, J. T. & Gardella, T. J. Enhanced activity in parathyroid hormone-(1-14) and -(1-11): novel peptides for probing ligand-receptor interactions. Endocrinology 142, 3068–3074 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Okazaki, M. et al. Prolonged signaling at the parathyroid hormone receptor by peptide ligands targeted to a specific receptor conformation. Proc. Natl Acad. Sci. USA 105, 16525–16530 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Dean, T., Vilardaga, J.-P., Potts, J. T. & Gardella, T. J. Altered selectivity of parathyroid hormone (PTH) and PTH-related protein (PTHrP) for distinct conformations of the PTH/PTHrP receptor. Mol. Endocrinol. 22, 156–166 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Inooka, H. et al. Conformation of a peptide ligand bound to its G-protein coupled receptor. Nat. Struct. Biol. 8, 161–165 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Neumann, J.-M. et al. Class-B GPCR activation: is ligand helix-capping the key? Trends. Biochem. Sci. 33, 314–319 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Tregear, G. W. et al. Bovine parathyroid hormone: minimum chain length of synthetic peptide required for biological activity. Endocrinology 93, 1349–1353 (1973).

    Article  CAS  PubMed  Google Scholar 

  32. Goltzmann, D., Peytremann, A., Callahan, E., Tregear, G. W. & Potts, J. T. Analysis of the requirements for parathyroid hormone action in renal membranes with the use of inhibiting analogues. J. Biol. Chem. 250, 3199–3203 (1975).

    CAS  PubMed  Google Scholar 

  33. Rosenblatt, M. & Potts, J. T. Analogues of an in vitro parathyroid hormone inhibitor: modifications at the amino terminus. Calcif. Tissue Int. 33, 153–157 (1981).

    Article  CAS  PubMed  Google Scholar 

  34. Gardella, T. J. et al. Determinants of [Arg2]PTH-(1-34) binding and signaling in the transmembrane region of the parathyroid hormone receptor. Endocrinology 135, 1186–1194 (1994).

    Article  CAS  PubMed  Google Scholar 

  35. Gardella, T. J., Luck, M. D., Fan, M. H. & Lee, C. Transmembrane residues of the parathyroid hormone (PTH)/PTH-related peptide receptor that specifically affect binding and signaling by agonist ligands. J. Biol. Chem. 271, 12820–12825 (1996).

    Article  CAS  PubMed  Google Scholar 

  36. Schipani, E. et al. A novel parathyroid hormone (PTH)/PTH-related peptide receptor mutation in Jansen’s metaphyseal chondrodysplasia. J. Clin. Endocrinol. Metab. 84, 3052–3057 (1999).

    CAS  PubMed  Google Scholar 

  37. Hoare, S. R. J. Mechanisms of peptide and nonpeptide ligand binding to Class B G-protein-coupled receptors. Drug Discov. Today 10, 417–427 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Castro, M., Nikolaev, V. O., Palm, D., Lohse, M. J. & Vilardaga, J.-P. Turn-on switch in parathyroid hormone receptor by a two-step parathyroid hormone binding mechanism. Proc. Natl Acad. Sci. USA 102, 16084–16089 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Parthier, C., Reedtz-Runge, S., Rudolph, R. & Stubbs, M. T. Passing the baton in class B GPCRs: peptide hormone activation via helix induction? Trends. Biochem. Sci. 34, 303–310 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Hollenstein, K. et al. Structure of class B GPCR corticotropin-releasing factor receptor 1. Nature 499, 438–443 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Jazayeri, A. et al. Extra-helical binding site of a glucagon receptor antagonist. Nature 533, 274–277 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Odén, A., McCloskey, E. V., Kanis, J. A., Harvey, N. C. & Johansson, H. Burden of high fracture probability worldwide: secular increases 2010-2040. Osteoporos. Int. 26, 2243–2248 (2015).

    Article  PubMed  Google Scholar 

  43. Schütz, M. et al. Directed evolution of G protein-coupled receptors in yeast for higher functional production in eukaryotic expression hosts. Sci. Rep. 6, 21508 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Shimizu, N., Dean, T., Khatri, A. & Gardella, T. J. Amino-terminal parathyroid hormone fragment analogs containing alpha,alpha-di-alkyl amino acids at positions 1 and 3. J. Bone Miner. Res. 19, 2078–2086 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  46. Klenk, C., Schulz, S., Calebiro, D. & Lohse, M. J. Agonist-regulated cleavage of the extracellular domain of parathyroid hormone receptor type 1. J. Biol. Chem. 285, 8665–8674 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yin, J., Mobarec, J. C., Kolb, P. & Rosenbaum, D. M. Crystal structure of the human OX2 orexin receptor bound to the insomnia drug suvorexant. Nature 519, 247–250 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Rosenblatt, M., Goltzman, D., Keutmann, H. T., Tregear, G. W. & Potts, J. T. Chemical and biological properties of synthetic, sulfur-free analogues of parathyroid hormone. J. Biol. Chem. 251, 159–164 (1976).

    CAS  PubMed  Google Scholar 

  49. Neugebauer, W., Barbier, J. R., Sung, W. L., Whitfield, J. F. & Willick, G. E. Solution structure and adenylyl cyclase stimulating activities of C-terminal truncated human parathyroid hormone analogues. Biochemistry 34, 8835–8842 (1995).

    Article  CAS  PubMed  Google Scholar 

  50. Caffrey, M. & Cherezov, V. Crystallizing membrane proteins using lipidic mesophases. Nat. Protoc. 4, 706–731 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kabsch, W. Integration, scaling, space-group assignment and post-refinement. Acta Crystallogr. D Biol. Crystallogr. 66, 133–144 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Collaborative Computational Project, Number 4. The CCP4 suite: programs for protein crystallography. Acta Crystallogr. D Biol. Crystallogr. 50, 760–763 (1994).

    Article  Google Scholar 

  53. Evans, P. R. & Murshudov, G. N. How good are my data and what is the resolution? Acta Crystallogr. D Biol. Crystallogr. 69, 1204–1214 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. McCoy, A. J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Horcajada, C., Guinovart, J. J., Fita, I. & Ferrer, J. C. Crystal structure of an archaeal glycogen synthase: insights into oligomerization and substrate binding of eukaryotic glycogen synthases. J. Biol. Chem. 281, 2923–2931 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Murshudov, G. N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D Biol. Crystallogr. 67, 355–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D Biol. Crystallogr. 68, 352–367 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Emami-Nemini, A. et al. Time-resolved fluorescence ligand binding for G protein-coupled receptors. Nat. Protoc. 8, 1307–1320 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Keppler, A. et al. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat. Biotechnol. 21, 86–89 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank B. Blattmann of the Protein Crystallisation Center at the University of Zurich for his support with crystallization, the staff of the X06SA beamline at the Paul Scherrer Institute for support during data collection and I. Berger at the European Molecular Biology Laboratory for providing us with baculovirus transfer vectors. We also thank G. Meier and B. Aebli for support during protein production. This work was supported by Schweizerischer Nationalfonds Grants 31003A_153143 and 31003A_182334 and KTI grant 18022.1 PFLS-LS, all to A.P.; C.K. is the recipient of a fellowship of the German Academy of Sciences Leopoldina (LPDS 2009-48) and a Marie Curie fellowship of the European Commission (FP7-PEOPLE-2011-IEF #299208).

Author information

Authors and Affiliations

Authors

Contributions

C.K., with the help of L.K., performed directed evolution in yeast. J.E. and J.S. carried out the rational mutagenesis for further thermostabilization of the receptor. J.E., J.S. and C.K. characterized expression constructs and optimized protein expression. J.E. and J.S. designed and characterized crystallization constructs; expressed, purified and crystallized PTH1R; and harvested crystals. J.E., J.S. and A.S.D. collected diffraction data. J.E., J.S., M.R. and A.S.D. processed the data and solved and refined the structure. C.K. performed pharmacological characterization. C.K. and A.P. designed the project. Project management was carried out by J.E., J.S., C.K. and A.P. The manuscript was prepared by J.E., J.S., C.K. and A.P. All authors contributed to the final editing and approval of the manuscript.

Corresponding authors

Correspondence to Christoph Klenk or Andreas Plückthun.

Ethics declarations

Competing interests

M.R., L.K. and A.S.D. are employees of Heptares Therapeutics, a company with activities in the GPCR field.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Overview of the crystallized PTH1R construct and thermostability of PTH1R mutant TMDs.

a, Snake plot of the crystallized ECD-PTH1RS-PGS (PTH1RXTAL) construct. The native signal peptide of PTH1R (residues 1–23) was replaced by a melittin signal peptide (dashed circles), followed by a FLAG epitope (red), a His10 tag (purple) and a 3C protease cleavage site (orange). Residues 61–104 within the ECD and 481–593 at the receptor C terminus were deleted. Residues 389–397 of ICL3 were replaced by a PGS fusion. Mutations from directed evolution in S. cerevisiae and the additionally introduced stabilizing mutations are highlighted in green and blue, respectively. Disulfide bonds are indicated as yellow lines. Glycosylation sites observed in the crystal structure are indicated by a trident. The first and last residues of helices I to VIII are labeled with the residue number. Residues that are not resolved in the crystal structure are shown in gray. b, Thermostability assay (CPM) of the TMD of the yeast-evolved PTH1R (PTH1R-SaBRE), the further thermostabilized PTH1R (PTH1RS) and the PTH1RS-PGS fusion (PTH1RS-PGS) bound to [Ac5c1, Aib3, Q10, Hrg11, A12, W14]PTH(1–14) (respective melting temperatures are 53.0 °C, 57.6 °C and 60.9 °C).

Supplementary Figure 2 Comparison of PTH peptide sequences and in vitro pharmacology of ligands and receptor constructs.

a, Sequence alignment of human PTH (wt PTH) and engineered ePTH. Colored boxes depict sequence changes in comparison to wt PTH, and black boxes denote non-natural amino acids. The topology of PTH1R interaction is indicated on top. Ac5c, aminocyclopentane-1-carboxylic acid; Aib, α-aminoisobutyric acid; Hrg, homoarginine; Nle, norleucine. b, Thermostability assay (CPM) of the crystallized ECD-PTH1RS-PGS (PTH1RXTAL) fusion bound to wt PTH(1–34), wt PTHrP(1–36) and ePTH (respective melting temperatures are 58.0 °C, 58.4 °C and 65.0 °C). Data shown are from a representative experiment. c, Binding of PTH-HL647 to HEK293T cells expressing wt PTH1R in the presence of different concentrations of unlabeled PTH peptides. d, cAMP accumulation measured in HEK293T cells expressing wt PTH1R. e, IP1 accumulation measured in HEK293T cells expressing wt PTH1R. f,g, Binding of PTH-HL647 to HEK293T cells expressing wt PTH1R or the crystallized PTH1R with the PGS fusion (PTH1RXTAL) in the presence of different concentrations of unlabeled ePTH (f) or wt PTH(1–34) (g). h, cAMP accumulation measured in HEK293T cells expressing wt PTH1R or the crystallized PTH1R with the PGS fusion (PTH1RXTAL). Data shown in ch are mean values ± s.e.m. from five (c,f,g), four (d,e) or three (h) independent experiments performed in duplicate. The IC50 values for c, f and g are listed in Supplementary Table 2.

Supplementary Figure 3 PTH1R crystallization.

a, Bright-field image of PTH1R crystals in lipidic cubic phase. b, SDS–PAGE gel of purified PTH1RXTAL (upper band with molecular mass 68.3 kDa) bound to ePTH (lower band with molecular mass 4.3 kDa). c, Size-exclusion profile of PTH1RXTAL-ePTH complex on a Nanofilm SEC-250 column (Sepax Technologies). df, Crystal packing with unit cell indicated in black, axes indicated by arrows (green, TMD; purple, ECD; blue, PGS; orange, ePTH). d, View along a axis. e, View along b axis. f, View along c axis. g, B factor putty of PTH1R (blue, low B factors; red, high B factors). Relatively high B factors are observed within the ECLs and the linker connecting ECD and TMD.

Supplementary Figure 4 Atomic model of the PTH1R-ePTH complex in the electron density map.

ac, The 2Fo – Fc electron density map contoured at 1.0σ and model are shown for all seven transmembrane helices, helix VIII and ePTH (a,b) and the PTH1R-ePTH interacting residues (c). Boxes i–iv illustrate the specific regions of ligand interaction with the receptor.

Supplementary Figure 5 Comparison of the structures of wt PTH and ePTH bound to PTH1R and the role of glycosylation at N161 on PTH binding.

a, Overlay of the crystal structures of the ECDs of PTH1R-ePTH (purple and orange) and of the isolated ECD in complex with wt PTH(1–34) (PDB 3C4M, gray) with ligand residues shown in stick representation. Residues in ePTH differing from wt PTH are highlighted in green. b, Superposition of ePTH with the crystal structure of wt PTH(1–34) (PDB 1ET1, gray). c, ePTH with the FoFc omit density map contoured at 2.5σ. d, ECD of PTH1R with glycosylation (gray) resolved at residue N161 in close proximity to ePTH (orange) shown in sticks. The hydrogen bonds (dashed blue lines) between the amidated peptide C terminus of Y34 and T163 of PTH1R rationalize the reported increase in binding affinity of C-terminal amides over carboxylic acids in PTH(1–34) peptides (Proceedings of the Fifth Parathyroid Conf. 33–39, 1975). The 2Fo – Fc electron density in gray mesh is contoured at 1.0σ. e, Binding of PTH-HL647 to HEK293T cells expressing wt PTH1R and the mutant PTH1R_N161D in the presence of different concentrations of unlabeled PTH peptide. Data are shown as mean values ± s.e.m. from n = 7 independent experiments performed in duplicate. The IC50 values for wt and PTH1R_N161D are listed in Supplementary Table 2.

Supplementary Figure 6 Conserved residues and implication of R440 in ligand binding and receptor activation.

a, Extracellular view on the conserved tryptophan in ECL2 of superposed structures of PTH1R-ePTH (green-orange), GCGR-NNC1702 (yellow; PDB 5YQZ), GLP1R-ExP5 (blue; PDB 6B3J) and GLP1R-GLP1 (gray; PDB 5VAI) complexes. While this tryptophan is positioned between transmembrane helices III and IV in the structures of the other peptide-bound class B GPCRs, it is found in a different orientation packed against the ligand in the PTH1R-ePTH complex. b, The distinct orientation of W352 in PTH1R is defined by the clear side chain electron density in the 2Fo – Fc map contoured at 1.0σ. c, The stabilizing mutation R4407.38 (Q4407.38 in wt PTH1R) forms a hydrogen bonding network to ePTH via E4447.42 and to the backbone oxygen of F4246.56 at the extracellular end of helix VI. d, Binding of PTH-HL647 to HEK293T cells expressing wt or mutated PTH1R in the presence of different concentrations of unlabeled PTH peptides. Mutation of Q4407.38 to arginine largely retains ligand binding, whereas disruption of the hydrogen bond to E4447.42 by introduction of small, apolar side chain residues severely reduces ligand binding (Mol. Endocrinol. 9, 1269–1278, 1995). e,f, Normalized concentration–response curves for cAMP (e) or IP1 (f) accumulation, measured in HEK293T cells expressing wt or mutant PTH1R. In contrast to Q4407.38 of wt PTH1R, the extended sidechain of R4407.38 stabilizes the top of helix VI by an additional hydrogen bond to F4246.56 and thus strongly reduces receptor activation, which requires conformational rearrangements at the extracellular end of helix VI. g, Amino acid alignment of specific conserved residues of 15 human class B GCPRs grouped into those of the central polar network, those involved in receptor activation, the P-x-x-G motif and in the orthosteric binding pocket (Wootten numbering is given as a superscript). Data in d and e are shown as mean values ± s.e.m. from seven (d), five (e) or three (f) independent experiments performed in duplicate. The IC50 values of d are listed in Supplementary Table 2.

Supplementary Figure 7 Binding of PTH(1–34) to wild-type and mutant variants of PTH1R.

Binding of PTH-HL647 to HEK293T cells expressing wild-type and mutated PTH1R variants in the presence of different concentrations of unlabeled wt PTH(1–34) as a competitor. Data are shown as mean values ± s.e.m. from 4–12 independent experiments performed in duplicate. The corresponding IC50 values and numbers of independent experiments for wt and mutant PTH1Rs are listed in Supplementary Table 2.

Supplementary Figure 8 Jansen’s metaphyseal chondrodysplasia–related mutations at the conserved HETX motif result in constitutive PTH1R activation.

a, Extracellular view on the class B conserved HETX motif of superposed PTH1R (green) and activated GLP1R (blue; PDB 5VAI). In the active state receptor, T6.42 is removed from the network owing to rearrangements in transmembrane helix VI. Residues associated with Jansen’s metaphyseal chondrodysplasia are highlighted in red. b,c, The thermostabilizing mutation I4587.56A in the crystallized PTH1R and the Jansen’s Metaphyseal Chondrodysplasia-related mutation I4587.56R facing towards S4096.41 are located close to the HETX motif (view from the membrane). The bulky side chain of I4587.56R clashes (indicated by red disks) with helix VI in the inactive receptor state (representative rotamer from PyMOL Mutagenesis Wizard shown).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1 and 2

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ehrenmann, J., Schöppe, J., Klenk, C. et al. High-resolution crystal structure of parathyroid hormone 1 receptor in complex with a peptide agonist. Nat Struct Mol Biol 25, 1086–1092 (2018). https://doi.org/10.1038/s41594-018-0151-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0151-4

This article is cited by

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