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 oxytocin signaling complex reveals a molecular switch for cation dependence

Abstract

Oxytocin (OT) and vasopressin (AVP) are conserved peptide signaling hormones that are critical for diverse processes including osmotic homeostasis, reproduction, lactation and social interaction. OT acts through the oxytocin receptor (OTR), a magnesium-dependent G protein-coupled receptor that is a therapeutic target for treatment of postpartum hemorrhage, dysfunctional labor and autism. However, the molecular mechanisms that underlie OTR activation by OT and the dependence on magnesium remain unknown. Here we present the wild-type active-state structure of human OTR bound to OT and miniGq/i determined by cryo-EM. The structure reveals a unique activation mechanism adopted by OTR involving both the formation of a Mg2+ coordination complex between OT and the receptor, and disruption of transmembrane helix 7 (TM7) by OT. Our functional assays demonstrate the role of TM7 disruption and provide the mechanism of full agonism by OT and partial agonism by OT analogs. Furthermore, we find that the identity of a single cation-coordinating residue across vasopressin family receptors determines whether the receptor is cation-dependent. Collectively, these results demonstrate how the Mg2+-dependent OTR is activated by OT, provide essential information for structure-based drug discovery efforts and shed light on the molecular determinants of cation dependence of vasopressin family receptors throughout the animal kingdom.

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 the OTR–miniGq complex bound to oxytocin.
Fig. 2: OT binding mode and G protein interaction.
Fig. 3: Disruption of TM7 helix underlies receptor activation.
Fig. 4: Architecture of the magnesium cofactor coordination site.
Fig. 5: An aspartate to lysine transition reverses magnesium dependence in the OT/AVP receptor family.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in the Article and its Supplementary Information. Cryo-EM maps of the OT–OTR–Gq complex have been deposited in the Electron Microscopy Data Bank under accession code EMD-24733, including the composite map, global non-uniform refinement, local refinement of TMs with deepEMhancer, and local refinement of G protein. The original micrographs have been deposited in the Electron Microscopy Public Image Archive under accession code EMPIAR-10936. The atomic coordinates have been deposited in the Protein Data Bank under the accession code 7RYC. All other relevant data is available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Fuchs, A. R., Fuchs, F., Husslein, P., Soloff, M. S. & Fernström, M. J. Oxytocin receptors and human parturition: a dual role for oxytocin in the initiation of labor. Science 215, 1396–1398 (1982).

    Article  CAS  PubMed  Google Scholar 

  2. Ferguson, J. N. et al. Social amnesia in mice lacking the oxytocin gene. Nat. Genet. 25, 284–288 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Carcea, I. et al. Oxytocin neurons enable social transmission of maternal behaviour. Nature 596, 553–557 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Augustine, R. A., Seymour, A. J., Campbell, R. E., Grattan, D. R. & Brown, C. H. Integrative neuro-humoral regulation of oxytocin neuron activity in pregnancy and lactation. J. Neuroendocrinol. 30, e12569 (2018).

    Article  CAS  Google Scholar 

  5. Eliava, M. et al. A new population of parvocellular oxytocin neurons controlling magnocellular neuron activity and inflammatory pain processing. Neuron 89, 1291–1304 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. King, C. E., Gano, A. & Becker, H. C. The role of oxytocin in alcohol and drug abuse. Brain Res. 1736, 146761 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kosfeld, M., Heinrichs, M., Zak, P. J., Fischbacher, U. & Fehr, E. Oxytocin increases trust in humans. Nature 435, 673–676 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Patel, S. S. & Ludmir, J. Drugs for the treatment and prevention of preterm labor. Clin. Perinatol. 46, 159–172 (2019).

    Article  PubMed  Google Scholar 

  9. Peñagarikano, O. et al. Exogenous and evoked oxytocin restores social behavior in the Cntnap2 mouse model of autism. Sci. Transl. Med. 7, 271ra8 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Martins, D., Paduraru, M. & Paloyelis, Y. Heterogeneity in response to repeated intranasal oxytocin in schizophrenia and autism spectrum disorders: a meta-analysis of variance. Br. J. Pharmacol. https://doi.org/10.1111/bph.15451 (2021)

  11. Yoon, S. & Kim, Y.-K. The role of the oxytocin system in anxiety disorders. Adv. Exp. Med. Biol. 1191, 103–120 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Waltenspühl, Y., Schöppe, J., Ehrenmann, J., Kummer, L. & Plückthun, A. Crystal structure of the human oxytocin receptor. Sci. Adv. 6, eabb5419 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Busnelli, M. et al. Functional selective oxytocin-derived agonists discriminate between individual G protein family subtypes. J. Biol. Chem. 287, 3617–3629 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Maeda, S. et al. Development of an antibody fragment that stabilizes GPCR/G-protein complexes. Nat. Commun. 9, 3712 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Israeli, H. et al. Structure reveals the activation mechanism of the MC4 receptor to initiate satiation signaling. Science 372, 808–814 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Wesley, V. J., Hawtin, S. R., Howard, H. C. & Wheatley, M. Agonist-specific, high-affinity binding epitopes are contributed by an arginine in the N-terminus of the human oxytocin receptor. Biochemistry 41, 5086–5092 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Klein, U., Gimpl, G. & Fahrenholz, F. Alteration of the myometrial plasma membrane cholesterol content with beta-cyclodextrin modulates the binding affinity of the oxytocin receptor. Biochemistry 34, 13784–13793 (1995).

    Article  CAS  PubMed  Google Scholar 

  18. Lemel, L. et al. The ligand-bound state of a G protein-coupled receptor stabilizes the interaction of functional cholesterol molecules. J. Lipid Res. 62, 100059 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jafurulla, M., Aditya Kumar, G., Rao, B. D. & Chattopadhyay, A. A critical analysis of molecular mechanisms underlying membrane cholesterol sensitivity of GPCRs. Adv. Exp. Med. Biol. 1115, 21–52 (2019).

    Article  CAS  PubMed  Google Scholar 

  20. Taghon, G. J., Rowe, J. B., Kapolka, N. J. & Isom, D. G. Predictable cholesterol binding sites in GPCRs lack consensus motifs. Structure 29, 499–506.e3 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Ballesteros, J. A. & Weinstein, H. in Methods in Neurosciences Vol. 25 (ed. Sealfon, S. C.) 366–428 (Academic Press, 1995).

  22. Mouillac, B. et al. The binding site of neuropeptide vasopressin V1a receptor. Evidence for a major localization within transmembrane regions. J. Biol. Chem. 270, 25771–25777 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. Kim, K. et al. Structure of a hallucinogen-activated Gq-coupled 5-HT2A serotonin receptor. Cell 182, 1574–1588.e19 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang, L. et al. Cryo-EM structure of the AVP-vasopressin receptor 2-Gs signaling complex. Cell Res. 31, 932–934 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Zhou, F. et al. Molecular basis of ligand recognition and activation of human V2 vasopressin receptor. Cell Res. 31, 929–931 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Bous, J. et al. Cryo-electron microscopy structure of the antidiuretic hormone arginine-vasopressin V2 receptor signaling complex. Sci. Adv. 7, eabg5628 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lamont, R. F. The development and introduction of anti-oxytocic tocolytics. BJOG 110, 108–112 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Zheng, H., Chruszcz, M., Lasota, P., Lebioda, L. & Minor, W. Data mining of metal ion environments present in protein structures. J. Inorg. Biochem. 102, 1765–1776 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Michel, J., Tirado-Rives, J. & Jorgensen, W. L. Prediction of the water content in protein binding sites. J. Phys. Chem. B 113, 13337–13346 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Antoni, F. A. & Chadio, S. E. Essential role of magnesium in oxytocin-receptor affinity and ligand specificity. Biochem. J. 257, 611–614 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jing, Z., Liu, C., Qi, R. & Ren, P. Many-body effect determines the selectivity for Ca2+ and Mg2+ in proteins. Proc. Natl Acad. Sci. USA 115, E7495–E7501 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhu, X., Gilbert, S., Birnbaumer, M. & Birnbaumer, L. Dual signaling potential is common among Gs-coupled receptors and dependent on receptor density. Mol. Pharmacol. 46, 460–469 (1994).

    CAS  PubMed  Google Scholar 

  33. Theofanopoulou, C., Gedman, G., Cahill, J. A., Boeckx, C. & Jarvis, E. D. Universal nomenclature for oxytocin-vasotocin ligand and receptor families. Nature 592, 747–755 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Donaldson, Z. R. & Young, L. J. Oxytocin, vasopressin, and the neurogenetics of sociality. Science 322, 900–904 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Slodkowicz, G. & Goldman, N. Integrated structural and evolutionary analysis reveals common mechanisms underlying adaptive evolution in mammals. Proc. Natl Acad. Sci. USA 117, 5977–5986 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bharadwaj, V. N. et al. A new hypothesis linking oxytocin to menstrual migraine. Headache 61, 1051–1059 (2021).

    Article  PubMed  Google Scholar 

  37. Towns, J. et al. XSEDE: Accelerating scientific discovery. Comput. Sci. Eng. 16, 62–74 (2014).

    Article  CAS  Google Scholar 

  38. Morales-Perez, C. L., Noviello, C. M. & Hibbs, R. E. Manipulation of subunit stoichiometry in heteromeric membrane proteins. Structure 24, 797–805 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Koehl, A. et al. Structure of the µ-opioid receptor-Gi protein complex. Nature 558, 547–552 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  41. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  43. 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 

  44. 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 

  45. Asarnow, D., Palovcak, E. & Cheng, Y. asarnow/pyem: UCSF pyem v0.5 (Zenodo, 2019); https://doi.org/10.5281/ZENODO.3576630

  46. Zivanov, J., Nakane, T. & Scheres, S. H. W. A Bayesian approach to beam-induced motion correction in cryo-EM single-particle analysis. IUCrJ 6, 5–17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zivanov, J., Nakane, T. & Scheres, S. H. W. Estimation of high-order aberrations and anisotropic magnification from cryo-EM data sets in RELION-3.1. IUCrJ 7, 253–267 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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 

  49. Sanchez-Garcia, R. et al. DeepEMhancer: a deep learning solution for cryo-EM volume post-processing. Commun. Biol. 4, 874 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kooistra, A. J. et al. GPCRdb in 2021: integrating GPCR sequence, structure and function. Nucleic Acids Res. 49, D335–D343 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. 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 

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

    Article  PubMed  CAS  Google Scholar 

  53. Robertson, M. J., van Zundert, G. C. P., Borrelli, K. & Skiniotis, G. GemSpot: a pipeline for robust modeling of ligands into Cryo-EM maps. Structure 28, 707–716.e3 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Robertson, M. J., Meyerowitz, J. G., Panova, O., Borrelli, K., Skiniotis, G. Plasticity in ligand recognition at somatostatin receptors. Preprint at bioRxiv https://doi.org/10.1101/2021.11.02.466988 (2021).

  55. Williams, C. J. et al. MolProbity: more and better reference data for improved all-atom structure validation. Protein Sci. 27, 293–315 (2018).

    Article  CAS  PubMed  Google Scholar 

  56. 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 

  57. Olsen, R. H. J. et al. TRUPATH, an open-source biosensor platform for interrogating the GPCR transducerome. Nat. Chem. Biol. 16, 841–849 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

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

    Article  CAS  Google Scholar 

  60. 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  PubMed  Google Scholar 

  61. Phillips, J. C. et al. Scalable molecular dynamics with NAMD. J. Comput. Chem. 26, 1781–1802 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ryckaert, J.-P., Ciccotti, G. & Berendsen, H. J. C. Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes. J. Comput. Phys. 23, 327–341 (1977).

    Article  CAS  Google Scholar 

  63. Miyamoto, S. & Kollman, P. A. Settle: an analytical version of the SHAKE and RATTLE algorithm for rigid water models. J. Comput. Chem. 13, 952–962 (1992).

    Article  CAS  Google Scholar 

  64. van Zundert, G. C. P., Moriarty, N. W., Sobolev, O. V., Adams, P. D. & Borrelli, K. W. Macromolecular refinement of X-ray and cryoelectron microscopy structures with Phenix/OPLS3e for improved structure and ligand quality. Structure 29, 913–921 (2021).

    Article  PubMed  CAS  Google Scholar 

  65. Jorgensen, W. L. & Tirado-Rives, J. Molecular modeling of organic and biomolecular systems using BOSS and MCPRO. J. Comput. Chem. 26, 1689–1700 (2005).

    Article  CAS  PubMed  Google Scholar 

  66. Edgar, R. C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 32, 1792–1797 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Black, J. W., Leff, P., Shankley, N. P. & Wood, J. An operational model of pharmacological agonism: the effect of E/[A] curve shape on agonist dissociation constant estimation. Br. J. Pharmacol. 84, 561–571 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Theophanopoulou and E. Jarvis at The Rockefeller University for their advice and discussions, E. Montabana for support in data collection and the Stanford cryo-Electron Microscopy center (cEMc). We thank R. Hibbs for gifting the pEZT vector. This work was supported, in part, by National Institute of Health T32-GM089626 (J.G.M.), and the Extreme Science and Engineering Discovery Environment (XSEDE)37 resource comet-gpu and expanse through sdsc-comet/sdsc-expanse allocation TG-MCB190153, which is supported by National Science Foundation grant number ACI-1548562.

Author information

Authors and Affiliations

Authors

Contributions

J.G.M. expressed and purified proteins, prepared cryo-EM samples, processed cryo-EM data, built and refined the structural model, designed and generated the expression and mutagenesis constructs, and performed biochemical assays and analyzed the data. M.J.R. performed molecular dynamics simulations and free energy calculations, model refinement, generation of expression constructs, and protein purification. X.B.-Á. expressed and purified miniGqi. O.P. collected cryo-EM data. R.M.N. assisted with cell surface expression experiments. Y.G. prepared cryo-EM grids. J.G.M. and G.S. wrote the manuscript with input from M.J.R., X.B.-Á., O.P., R.M.N. and Y.G. G.S. supervised the project.

Corresponding author

Correspondence to Georgios Skiniotis.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Florian Ullrich was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

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

Extended data

Extended Data Fig. 1 Oxytocin receptor signaling and purification.

a, Dose−response curves of G protein-specific activation pathways of the oxytocin receptor by oxytocin (orange squares), neurotensin 1 receptor by neurotensin (teal circles), or beta-2 adrenergic receptor by isoproterenol (teal triangles) from simultaneous curve fitting of 3 independent biological replicates with Hill Slope constrained to 1. Error bars are S.E.M. b, Gel filtration chromatography profile and c, protein gel of oxytocin receptor with GFP tag. d, Gel filtration chromatography profile and e, protein gel of oxytocin receptor complex from the protein preparation used for data collection.

Source data

Extended Data Fig. 2 Oxytocin-Oxytocin Receptor-MiniGqi-scFv16 cryoEM data collection and processing.

a, Representative micrograph of OTR complex from 7,727 micrographs. b, Example final 2D classes of OTR complex (128 classes). c, CryoEM data processing workflow. d, Gold-standard FSC curves of global non-uniform refinement of complex, local refinement of transmembrane helices, and local refinement of G protein heterotrimer. e, Overall combined map from two local (TMs and G protein) and global (scFv16) refinements.

Extended Data Fig. 3 Map/model fit and signaling complex architecture.

Fit into cryo-EM map of a, oxytocin, b, the N-terminal domain (NTD), c, extracellular loop 3 (ECL3) and extracellular loop 2 (ECL2), d, cation binding site at high (purple) threshold and low (transparent) threshold. e, the C-terminal alpha-helical domain (α-C) of miniGqi, f, transmembrane (TM) helices 1-7. g, Representative cell cytometry data of surface expression levels of wild-type OTR (WT), OTRR34A, OTRF103A, and OTRR104A detected by Alexa Fluor-647 (AF-647) corresponding to transient transfections used in BRET assays.

Extended Data Fig. 4 Oxytocin receptor ligand and G protein interactions.

a, View from TM3 of oxytocin (OT) in the oxytocin receptor binding pocket with overlay of cryoEM map at threshold level 0.105 in ChimeraX; receptor in blue, oxytocin in orange, and Mg2+ in green. Superscript is Ballesteros-Weinstein notation. b, Overlay of OT (orange) bound to OTR (blue) and arginine vasopressin (AVP; purple) bound to V2R (tan; PDB: 7KH0). c, Overlay of active state structures of vasopressin 2 receptor-Gs (V2R-Gs; PDB: 7KH0), serotonin receptor 2A-miniGq/i (HTR2A-miniGq; PDB: 5WHA) aligned on TM bundle (transparent) showing placement of G protein N-terminal alpha helical domain (α-N). d, Overlay of same alignment showing slight difference in angular orientation of G protein C terminal helix (α5) between receptors. e, Comparison of G protein engagement between OTR-miniGq (blue) with V2R-Gs (salmon; PDB: 7KH0) aligned on TM bundle.

Extended Data Fig. 5 Activation mechanism of the oxytocin receptor.

a, Overlay of TM7 from OT-bound cryoEM structure (blue) and antagonist-bound crystal structure (mustard) showing broken helix in active state and interaction between Tyr2 of OT (orange) and the backbone carbonyl of L3167.40. b, Overlay of OTR-bound OT with both the L-state and T-state poses (AVP-L and AVP-T) of AVP-bound V2R-Gs (PDB: 7BB6, 7BB7). c, Overlay of OT with OTR-bound retosiban. d, Overlay of TM6 and TM7 from OT-bound (blue) and antagonist-bound (mustard) OTR showing stabilization of TM7 by hydrogen bond between N3217.45 and N325K7.49 in inactive structure, and the rotation of Y3297.53 and displacement of TM6 in the OT-bound structure. Backbone of full transmembrane helix 7 with helical hydrogen bonds and ligand pose for the active-state structures of e, OTR bound to OT and f, HTR2A bound to 25-CN-NBOH (PDB: 6WHA) aligned on TMs and viewed from identical perspective, with receptor TM helices in transparent cartoon.

Extended Data Fig. 6 Oxytocin receptor partial agonists.

a, Gq, G11, and G15 signaling measured by G protein BRET-based biosensors and b, recruitment of β-arrestin 1 and β-arrestin 2 to C-terminal rLuc8-tagged OTR. c, Dose-response curve of 2-(O-methyltyrosine)-oxytocin (meOT) demonstrating antagonism of OTR-dependent Gq signaling in the presence of 100 nM OT, with net BRET normalized to BRET ratio in presence of 100 nM OT. d, Arginine vasopressin (AVP) or Phe2-AVP activation of V1aR or V1bR measured by Gq BRET biosensor, or V2R measured by Gs BRET biosensor. Data points are represented as mean values + /- SEM and curves are simultaneously fit to n = 3 biologically independent replicates.

Extended Data Fig. 7 Magnesium dependence of oxytocin and vasopressin receptors.

a, Recruitment of beta-arrestin 1 (β1) or beta-arrestin 2 (β2) and b, activation of Gq-based BRET biosensor by oxytocin (OT)- or vasopressin (AVP)-stimulated oxytocin receptor (OTR) or vasopressin receptor 1a (V1aR) and mutants in the presence of 2 mM Mg2+ or 0.5 mM EDTA. Data points are represented as mean values + /− SEM and curves are simultaneously fit to n = 3 biologically independent replicates. c, Sequence alignment of cation-coordinating residues in vasopressin 2 receptor (V2R; VTR2C) in primates, with protein accession codes.

Supplementary information

Source data

Source Data Extended Data Fig. 1

Unprocessed SDS-PAGE gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meyerowitz, J.G., Robertson, M.J., Barros-Álvarez, X. et al. The oxytocin signaling complex reveals a molecular switch for cation dependence. Nat Struct Mol Biol 29, 274–281 (2022). https://doi.org/10.1038/s41594-022-00728-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-022-00728-4

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