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:

The relaxin receptor RXFP1 signals through a mechanism of autoinhibition

Abstract

The relaxin family peptide receptor 1 (RXFP1) is the receptor for relaxin-2, an important regulator of reproductive and cardiovascular physiology. RXFP1 is a multi-domain G protein-coupled receptor (GPCR) with an ectodomain consisting of a low-density lipoprotein receptor class A (LDLa) module and leucine-rich repeats. The mechanism of RXFP1 signal transduction is clearly distinct from that of other GPCRs, but remains very poorly understood. In the present study, we determine the cryo-electron microscopy structure of active-state human RXFP1, bound to a single-chain version of the endogenous agonist relaxin-2 and the heterotrimeric Gs protein. Evolutionary coupling analysis and structure-guided functional experiments reveal that RXFP1 signals through a mechanism of autoinhibition. Our results explain how an unusual GPCR family functions, providing a path to rational drug development targeting the relaxin receptors.

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 map and model of the RXFP1–Gs complex.
Fig. 2: Regulation of receptor signaling by ECL2 and the ectodomain.
Fig. 3: MD of RXFP1 starting from the inactive-state AlphaFold2 model.
Fig. 4: Cryo-EM and crosslinking MS reveal interactions between relaxin-2 and the leucine-rich repeats.
Fig. 5: Model of RXFP1 activation by relaxin-2.

Similar content being viewed by others

Data availability

The cryo-EM model and maps for RXFP1–Gs–TM are deposited under accession nos. 7TMW (PDB) and EMDB-26003, respectively. The cryo-EM map for RXFP1–Gs–FL is deposited under the accession no. EMDB-26004. The rigid-body docking model of the RXFP1 LRRs bound to relaxin-2 is available on the website for the Kruse lab at Harvard Medical School: https://kruse.hms.harvard.edu/data. The UniRef100 database and UniProt canonical sequence for RXFP1 (Q9HBX9) used for EC analysis can be found at https://www.uniprot.org. Data in Supplementary Fig. 1 are available in the Supplementary Data. Source data are provided with this paper.

References

  1. Petrie, E. J., Lagaida, S., Sethi, A., Bathgate, R. A. D. & Gooley, P. R. In a class of their own—RXFP1 and RXFP2 are unique members of the LGR family. Front. Endocrinol. 6, 1–9 (2015).

    Google Scholar 

  2. Sheau, Y. H. et al. Activation of orphan receptors by the hormone relaxin. Science 295, 671–674 (2002).

    Google Scholar 

  3. Maclennan, A. H., Nicolson, R. & Green, R. C. Serum relaxin in pregnancy. Lancet 328, 241–243 (1986).

    Google Scholar 

  4. Sarwar, M., Du, X. J., Dschietzig, T. B. & Summers, R. J. The actions of relaxin on the human cardiovascular system. Br. J. Pharmacol. 174, 933–949 (2017).

    CAS  PubMed  Google Scholar 

  5. Devarakonda, T. & Salloum, F. N. Heart disease and relaxin: new actions for an old hormone. Trends Endocrinol. Metab. 29, 338–348 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Hsu, S. Y. et al. The three subfamilies of leucine-rich repeat-containing G protein-coupled receptors (LGR): identification of LGR6 and LGR7 and the signaling mechanism for LGR7. Mol. Endocrinol. 14, 1257–1271 (2000).

    CAS  PubMed  Google Scholar 

  7. Scott, D. J. et al. Characterization of novel splice variants of LGR7 and LGR8 reveals that receptor signaling is mediated by their unique low density lipoprotein class A modules. J. Biol. Chem. 281, 34942–34954 (2006).

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  9. Carpenter, B. & Tate, C. G. Engineering a minimal G protein to facilitate crystallisation of G protein-coupled receptors in their active conformation. Protein Eng. Des. Sel. 29, 583–593 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Erlandson, S. C., Wang, J., Jiang, H., Rockman, H. A. & Kruse, A. C. Engineering and characterization of a long half-life relaxin receptor RXFP1 agonist. Preprint at bioRxiv https://doi.org/10.1101/2022.04.19.488796 (2022).

  12. Erlandson, S. C., McMahon, C. & Kruse, A. C. Structural basis for G protein-coupled receptor signaling. Annu. Rev. Biophys. 47, 1–18 (2018).

    CAS  PubMed  Google Scholar 

  13. Ballesteros, J. A. & Weinstein, H. Integrated methods for the construction of three-dimensional models and computational probing of structure-function relations in G protein-coupled receptors. Methods Neurosci. 25, 366–428 (1995).

    CAS  Google Scholar 

  14. Wang, J., Hua, T. & Liu, Z.-J. Structural features of activated GPCR signaling complexes. Curr. Opin. Struct. Biol. 63, 82–89 (2020).

    CAS  PubMed  Google Scholar 

  15. Venkatakrishnan, A. J. et al. Molecular signatures of G-protein-coupled receptors. Nature 494, 185–194 (2013).

    CAS  PubMed  Google Scholar 

  16. Ring, A. M. et al. Adrenaline-activated structure of β2-adrenoceptor stabilized by an engineered nanobody. Nature 502, 575–579 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wingler, L. M., McMahon, C., Staus, D. P., Lefkowitz, R. J. & Kruse, A. C. Distinctive activation mechanism for angiotensin receptor revealed by a synthetic nanobody. Cell 176, 479–490.e12 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lin, X. et al. Structural basis of ligand recognition and self-activation of orphan GPR52. Nature 579, 152–157 (2020).

    CAS  PubMed  Google Scholar 

  19. 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, 508–520 (2016).

    PubMed  Google Scholar 

  20. Jiang, X., Dias, J. A. & He, X. Structural biology of glycoprotein hormones and their receptors: insights to signaling. Mol. Cell. Endocrinol. 382, 424–451 (2014).

    CAS  PubMed  Google Scholar 

  21. Sethi, A. et al. The complex binding mode of the peptide hormone H2 relaxin to its receptor RXFP1. Nat. Commun. 7, 11344 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Büllesbach, E. E. & Schwabe, C. The trap-like relaxin-binding site of the leucine-rich G-protein-coupled receptor 7. J. Biol. Chem. 280, 14051–14056 (2005).

    PubMed  Google Scholar 

  23. Marks, D. S. et al. Protein 3D structure computed from evolutionary sequence variation. PLoS ONE https://doi.org/10.1371/journal.pone.0028766 (2011).

  24. Sjodt, M. et al. Structure of the peptidoglycan polymerase RodA resolved by evolutionary coupling analysis. Nature 556, 118–121 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Zheng, S. et al. Structure and mutagenic analysis of the lipid II flippase MurJ from Escherichia coli. Proc. Natl Acad. Sci. USA 115, 6709–6714 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, L., Friesner, R. A. & Berne, B. J. Replica exchange with solute scaling: a more efficient version of replica exchange with solute tempering (REST2). J. Phys. Chem. B 115, 9431–9438 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Tunyasuvunakool, K. et al. Highly accurate protein structure prediction for the human proteome. Nature 596, 590–596 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  29. Halls, M. L. et al. Multiple binding sites revealed by interaction of relaxin family peptides with native and chimeric relaxin family peptide receptors 1 and 2 (LGR7 and LGR8). J. Pharmacol. Exp. Ther. 313, 677–687 (2005).

    CAS  PubMed  Google Scholar 

  30. Diepenhorst, N. A. et al. Investigation of interactions at the extracellular loops of the relaxin family peptide receptor 1 (RXFP1). J. Biol. Chem. 289, 34938–34952 (2014).

    PubMed  PubMed Central  Google Scholar 

  31. Mintseris, J. & Gygi, S. P. High-density chemical cross-linking for modeling protein interactions. Proc. Natl Acad. Sci. USA 117, 93–102 (2020).

    CAS  PubMed  Google Scholar 

  32. Büllesbach, E. E., Yang, S. & Schwabe, C. The receptor-binding site of human relaxin II: a dual prong-binding mechanism. J. Biol. Chem. 267, 22957–22960 (1992).

    PubMed  Google Scholar 

  33. Büllesbach, E. E. & Schwabe, C. The relaxin receptor-binding site geometry suggests a novel gripping mode of interaction. J. Biol. Chem. 275, 35276–35280 (2000).

    PubMed  Google Scholar 

  34. van Zundert, G. C. P. et al. The HADDOCK2.2 web server: user-friendly integrative modeling of biomolecular complexes. J. Mol. Biol. 428, 720–725 (2016).

    PubMed  Google Scholar 

  35. Hu, X. et al. Structural insights into the activation of human relaxin family peptide receptor 1 by small-molecule agonists. Biochemistry 55, 1772–1783 (2016).

    CAS  PubMed  Google Scholar 

  36. Duan, J. et al. Hormone- and antibody-mediated activation of the thyrotropin receptor. Nature 609, 854–859 (2022).

    CAS  PubMed  Google Scholar 

  37. Faust, B. et al. Autoantibody mimicry of hormone action at the thyrotropin receptor. Nature 609, 846–853 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Sethi, A. et al. Structural insights into the unique modes of relaxin-binding and tethered-agonist mediated activation of RXFP1 and RXFP2: defining the tethered agonist of RXFP1 and RXFP2. J. Mol. Biol. 433, 167217 (2021).

    CAS  PubMed  Google Scholar 

  39. Staus, D. P. et al. Sortase ligation enables homogeneous GPCR phosphorylation to reveal diversity in β-arrestin coupling. Proc. Natl Acad. Sci. USA 115, 3834–3839 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Zheng, S., Palovcak, E. & Armache, J. P. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Sanchez-Garcia, R. et al. DeepEMhancer: a deep learning solution for cryo-EM volume post-processing. Commun. Biol. 4, 1–8 (2021).

    Google Scholar 

  44. Liebschner, D. et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr. D 75, 861–877 (2019).

    CAS  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Du, Z. et al. The trRosetta server for fast and accurate protein structure prediction. Nat. Protoc. 16, 5634–5651 (2021).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  48. Eswar, N. et al. Comparative protein structure modeling using Modeller. in Current Protocols in Bioinformatics. https://doi.org/10.1002/0471250953.bi0506s15 (John Wiley & Sons, 2006).

  49. García-Nafría, J., Lee, Y., Bai, X., Carpenter, B. & Tate, C. G. Cryo-EM structure of the adenosine A2A receptor coupled to an engineered heterotrimeric G protein. eLife 7, 1–19 (2018).

    Google Scholar 

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

    CAS  PubMed  Google Scholar 

  51. Morin, A. et al. Collaboration gets the most out of software. eLife 2, e01456 (2013).

    PubMed  PubMed Central  Google Scholar 

  52. Potter, S. C. et al. HMMER web server: 2018 update. Nucleic Acids Res. 46, W200–W204 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Suzek, B. E., Wang, Y., Huang, H., McGarvey, P. B. & Wu, C. H. UniRef clusters: a comprehensive and scalable alternative for improving sequence similarity searches. Bioinformatics 31, 926–932 (2015).

    CAS  PubMed  Google Scholar 

  54. Moggridge, S., Sorensen, P. H., Morin, G. B. & Hughes, C. S. Extending the compatibility of the SP3 paramagnetic bead processing approach for proteomics. J. Proteome Res. 17, 1730–1740 (2018).

    CAS  PubMed  Google Scholar 

  55. Eng, J. K., Jahan, T. A. & Hoopmann, M. R. Comet: an open-source MS/MS sequence database search tool. Proteomics 13, 22–24 (2013).

    CAS  PubMed  Google Scholar 

  56. Eigenbrot, C. et al. X-ray structure of human relaxin at 1.5 Å. Comparison to insulin and implications for receptor binding determinants. J. Mol. Biol. 221, 15–21 (1991).

    CAS  PubMed  Google Scholar 

  57. Schott-Verdugo, S. & Gohlke, H. PACKMOL-Memgen: a simple-to-use, generalized workflow for membrane-protein-lipid-bilayer system building. J. Chem. Inf. Model. 59, 2522–2528 (2019).

    CAS  PubMed  Google Scholar 

  58. Patriksson, A. & van der Spoel, D. A temperature predictor for parallel tempering simulations. Phys. Chem. Chem. Physics 10, 2073–2077 (2008).

    CAS  Google Scholar 

  59. Azzalini, A. W. & Bowman, A. Applied Smoothing Techniques for Data Analysis: The Kernel Approach with S-Plus Illustrations (Oxford Univ. Press, 1997).

  60. Grant, B. J., Rodrigues, A. P. C., ElSawy, K. M., McCammon, J. A. & Caves, L. S. D. Bio3d: an R package for the comparative analysis of protein structures. Bioinformatics 22, 2695–2696 (2006).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Bao for critical reading of the manuscript and the SBGrid Consortium for computational support of structural biology software. Cryo-EM data were collected at the Harvard Center for Cryo-Electron Microscopy, Harvard Medical School, and we thank them for their support and advice during data collection. This work was funded by the National Institutes of Health (NIH) Ruth L. Kirschstein Predoctoral fellowship (no. F31GM128233 awarded to S.C.E.), Damon Runyon Postdoctoral Fellowship (no. DRG: 2489-23 awarded to J.O.-O.), a Blavatnik Biomedical Accelerator grant from Harvard Medical School (to A.C.K.) and NIH grants (nos. R01AR079489 to A.C.K. and R01CA260415 to A.C.K. and D.S.M.). MD simulations were performed using high-performance computing resources from GENCI-TGCC France (grant nos. 2021-2022 Spe00015 and A0100712461).

Author information

Authors and Affiliations

Authors

Contributions

S.C.E. performed the molecular cloning, protein expression and purification, and cryo-EM grid preparation, with supervision from A.C.K. S.C.E. and J.O.-O. performed the flow cytometry and cell-signaling assays, with supervision by A.C.K. S.C.E. and S.R. processed the cryo-EM data. S.C.E. performed model building and refinement, with supervision from A.C.K. K.P.B. performed the EC analysis, with supervision from D.S.M. X.L., J.A.P. and J.M. performed the crosslinking MS, with supervision from S.P.G. X.C. performed the molecular dynamics simulations. S.C.E., X.C. and A.C.K. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Andrew C. Kruse.

Ethics declarations

Competing interests

A.C.K. and S.C.E. are inventors on patent application PCT/US2021/031260 for engineered single-chain relaxin proteins. A.C.K. is a cofounder and consultant for Tectonic Therapeutic and Seismic Therapeutic and for the Institute for Protein Innovation, a nonprofit research institute. D.S.M. is a cofounder of Seismic Therapeutic, a consultant for Tectonic Therapeutic, Dyno Therapeutics, Jura Bio and Octant Bio, and a venture partner at Catalio Capital Management. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Wenqing Shui, Jin-Peng Sun 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

Extended Data Fig. 1 Engineering and purification of the RXFP1–Gs complex.

a, Diagram of the primary structure of RXFP1 domains versus the RXFP1-miniGs399-20res fusion construct. b, Flow cytometry cell surface expression tests in Expi293F tetR cells for RXFP1-miniGs fusion constructs. Data is mean ± s.e.m., n = 3 technical replicates. c, Gs signaling assay comparing the signaling levels of wild type RXFP1 versus RXFP1-miniGs399-20res in response to relaxin-2. Data is mean ± s.e.m., n = 3 technical replicates. d, Size exclusion chromatography profile for the RXFP1–Gs complex. Arrow indicates the peak fractions pooled for RXFP1–Gs. e, Coomassie-stained SDS-PAGE gel for the RXFP1–Gs complex (Representative from 8 purification gels). f, Flow cytometry competition binding assay for SE00111. SE001 competes with 200 nM SE301 for binding to wild type RXFP1. The Ki for SE001 was calculated to be 3.9 nM; data is mean ± s.e.m., n = 3 technical replicates.

Source data

Extended Data Fig. 2 Cryo-EM data processing for the 7TM domain of RXFP1–Gs.

a, Cryo-EM data processing scheme for the 7TM domain of RXFP1 in complex with Gs. Shown are representative processing steps for one of four individual datasets and the steps used for the combined datasets. b, Representative micrograph from the RXFP1–Gs complex datasets (Scale bar = 50 nm; from 13,457 micrographs). c, Two-dimensional class averages for the 7TM domain of RXFP1 and G proteins. d, Angular distribution of particles in the final refinement for the 7TM domain with G proteins. e, Fourier shell correlation (FSC) used to determine the overall map resolution. f, Map to model FSC curve. g, cryoSPARC non-uniform refinement map colored by local resolution.

Extended Data Fig. 3 Cryo-EM data processing for the full−length RXFP1–Gs complex.

a, Cryo-EM data processing scheme for full-length RXFP1 in complex with Gs. Shown are representative processing steps for one of four individual datasets and the steps used for the combined datasets. b, Two-dimensional class averages for the full-length RXFP1–Gs complex. c, FSC used to determine the overall resolution of the map. d, RELION map of the full-length RXFP1 complex colored by local resolution at two different contour levels to display both the TM helices and receptor ectodomain.

Extended Data Fig. 4 Alignments of RXFP1’s ECL2 with GPR52 and family A orthosteric agonists.

Alignment of active-state RXFP1 (green, with ECL2 in magenta) (a), the β2 adrenergic receptor (gray) bound to adrenaline (purple; PDB ID: 4LDO)16 (b), and the angiotensin II type I receptor (gray) bound to the angiotensin II analog S1I8 (purple; PDB ID: 6DO1)17 (c), with GPR52 (gray with ECL2 in purple; PDB ID: 6LI3)18.

Extended Data Fig. 5 Cell surface expression and SE301 binding for RXFP1 constructs.

a-d, Flow cytometry cell surface expression tests with HEK293T cells for RXFP1 ECL2 mutants (a), Leu402 and Leu403 hinge region mutants (b), ectodomain truncation constructs (c), and evolutionary coupling analysis Ile396 and Ser397 hinge mutants (d). Data is mean ± s.e.m., n = 3 technical replicates. Cell surface expression was calculated as a percentage of wild type RXFP1 expression level. e-f, Ratio of SE301 (Fc-tagged relaxin-2) binding to receptor expression for flow cytometry binding assays in Expi293F cells11. The ratio is calculated by dividing the mean fluorescence intensity (MFI) of SE301 binding by the MFI of the expression level. Data is mean ± s.e.m., n = 3 technical replicates. Deletion or mutations to the linker region reduce the ratio of binding to expression, while LDLa deletions and ECL2 mutations retain an ability to bind SE301 (e). Ectodomain deletions (7TM + β2N-term) reduce the ratio of binding to expression, while mutations to the hinge region maintain an ability to bind SE301 (f).

Source data

Extended Data Fig. 6 Evolutionary coupling analysis of RXFP1.

a, Evolutionary couplings for RXFP1 residues 405–689 (black) compared to the active-state structure contacts (green) show close agreement between predicted contacts from ECs and the cryo-EM model. b, Evolutionary couplings for RXFP1 residues 120–757 (black) compared to the active-state 7TM structure and LRR AlphaFold227 model contacts (blue), highlighting ECL2 evolutionary couplings that provide insight into two potential loop conformations in magenta (T559ECL2–Ile396, Gly561ECL2–Ser397, Phe564ECL2–Ile396, Phe564ECL2–Val6667.38). c, Diagram of RXFP1 domains. Stars indicate regions of RXFP1 containing residues with the highest scoring ECs with ECL2, TM7 and the hinge region. d, The Phe564ECL2 and Val6667.38 residues from evolutionary coupling analysis are in close contact in the RXFP1 active-state structure.

Extended Data Fig. 7 Molecular dynamics of RXFP1 7TM deactivation.

a, The truncated 7TM domain is deactivated by adding a sodium in the conserved sodium-binding site. This leads to an inactive state during the simulations that closely resembles the AlphaFold2 model. b-c, Ionic lock distance and Cα RMSD of the TM region with respect to the AlphaFold2 model. Shown here are probability density distributions during 50 ns of REST2-MD simulations (10 frames/ns, n = 500). SE, integrated squared error between the density estimate and a standard normal density function. Vertical dashed lines indicated the values in the AlphaFold2 model and in the initial model based on cryo-EM.

Extended Data Fig. 8 Principal component analysis.

Principal component analysis of halfLRRs-7TM WT, S397A, and S397A/L402A/L403A are presented together. a, b, Projection of the simulation frames onto the 2D and 3D spaces defined by the 3 largest global PCs, covering 38.58%, 22.03% and 11.17% of the total variance, respectively. Dashed circle indicates (roughly) the active-like conformations of halfLRRs-7TM S397A. c, Conformational variance associated with PC3, reflecting TM6 opening of the S397A mutant upon activation. The same PC was found in the analysis of the S397A mutant alone (PC2 in Fig. 3d). For clarity, only 50 frames per system (1 frame/ns) are shown here.

Extended Data Fig. 9 Comparison of LHCGR and RXFP1 structures.

a, b, The hybrid model of active-state RXFP1 (based on our 7TM domain structure, cryo-EM maps, and AlphaFold2 model of the LRRs with docked relaxin-2 hormone) compared to active-state LHCGR (PDB ID: 7FIG) and inactive-state LHCGR (PDB ID: 7FIJ)8. The receptors are aligned on the 7TM domain. b, c, Side views (b) and top view (c) of the hybrid model of active-state RXFP1 compared to active-state LHCGR (PDB ID: 7FIG). The receptors are aligned on the 7TM domain and the ligands (relaxin-2 and chorionic gonadotropin) not displayed in order to highlight differences in active-state LRR orientations.

Extended Data Fig. 10 Signaling of RXFP1 constructs by the small molecule agonist ML290.

a, Gs signaling at wild type RXFP1 and ECL2 mutants at basal levels and in response to 490 nM ML290. Data is mean ± s.e.m., n = 3 technical replicates. b, Gs signaling at constructs of the RXFP1 7TM domain fused to the β2 adrenergic receptor N-terminus at basal levels and in response to 490 nM ML290. Data is mean ± s.e.m., n = 3 technical replicates.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–5, Tables 1–12 and legend for Supplementary Video 1.

Reporting Summary

Supplementary Data

Source Data for Supplementary Fig. 1.

Supplementary Video 1

Supplementary Video 1, related to Fig. 4 and Extended Data Fig. 3 Dynamics of RXFP1’s active-state ectodomain. Continuous heterogeneity within the final cryo-EM particle stack used for refinement of the full-length RXFP1–Gs complex. Flexibility of the ectodomain with respect to the 7TM domain and G proteins was visualized using 3D variability analysis in cryoSPARC2.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed gel.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Erlandson, S.C., Rawson, S., Osei-Owusu, J. et al. The relaxin receptor RXFP1 signals through a mechanism of autoinhibition. Nat Chem Biol 19, 1013–1021 (2023). https://doi.org/10.1038/s41589-023-01321-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01321-6

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