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:

Conserved class B GPCR activation by a biased intracellular agonist

Abstract

Class B G-protein-coupled receptors (GPCRs), including glucagon-like peptide 1 receptor (GLP1R) and parathyroid hormone 1 receptor (PTH1R), are important drug targets1,2,3,4,5. Injectable peptide drugs targeting these receptors have been developed, but orally available small-molecule drugs remain under development6,7. Here we report the high-resolution structure of human PTH1R in complex with the stimulatory G protein (Gs) and a small-molecule agonist, PCO371, which reveals an unexpected binding mode of PCO371 at the cytoplasmic interface of PTH1R with Gs. The PCO371-binding site is totally different from all binding sites previously reported for small molecules or peptide ligands in GPCRs. The residues that make up the PCO371-binding pocket are conserved in class B GPCRs, and a single alteration in PTH2R and two residue alterations in GLP1R convert these receptors to respond to PCO371. Functional assays reveal that PCO371 is a G-protein-biased agonist that is defective in promoting PTH1R-mediated arrestin signalling. Together, these results uncover a distinct binding site for designing small-molecule agonists for PTH1R and possibly other members of the class B GPCRs and define a receptor conformation that is specific only for G-protein activation but not arrestin signalling. These insights should facilitate the design of distinct types of class B GPCR small-molecule agonist for various therapeutic indications.

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: Human PTH1R signalling by PCO371 and cryo-EM structure of PTH1R-Gs signalling complex.
Fig. 2: Interactions of PCO371 with PTH1R.
Fig. 3: Conformational changes of TMD helix bundles during receptor activation between PTH-bound and PCO371-bound PTH1R.
Fig. 4: Selectivity of PCO371 for PTH1R and the conservation of the PCO371-binding site in class B GPCRs.
Fig. 5: A mostly conserved PCO371-like-binding pocket in class B GPCRs.

Data availability

A cryo-EM map has been deposited in the Electron Microscopy Data Bank under the accession code EMD-36593 (PCO371-bound PTH1R–Gs complex). The atomic coordinates have been deposited in the PDB under the accession code 8JR9 (PCO371-bound PTH1R–Gs complex).

References

  1. Pal, K., Melcher, K. & Xu, H. E. Structure and mechanism for recognition of peptide hormones by class B G-protein-coupled receptors. Acta Pharmacol. Sin. 33, 300–311 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Cong, Z. et al. Structural perspective of class B1 GPCR signaling. Trends Pharmacol. Sci. 43, 321–334 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Hollenstein, K. et al. Insights into the structure of class B GPCRs. Trends Pharmacol. Sci. 35, 12–22 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. de Graaf, C. et al. Extending the structural view of class B GPCRs. Trends Biochem. Sci. 42, 946–960 (2017).

    Article  PubMed  Google Scholar 

  5. Grigoriadis, D. E., Hoare, S. R., Lechner, S. M., Slee, D. H. & Williams, J. A. Drugability of extracellular targets: discovery of small molecule drugs targeting allosteric, functional, and subunit-selective sites on GPCRs and ion channels. Neuropsychopharmacology 34, 106–125 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Hoare, S. R. Allosteric modulators of class B G-protein-coupled receptors. Curr. Neuropharmacol. 5, 168–179 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Koole, C. et al. Recent advances in understanding GLP-1R (glucagon-like peptide-1 receptor) function. Biochem. Soc. Trans. 41, 172–179 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. 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 

  9. Cong, Z. et al. Structural basis of peptidomimetic agonism revealed by small- molecule GLP-1R agonists Boc5 and WB4-24. Proc. Natl Acad. Sci. USA 119, e2200155119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Griffith, D. A. et al. A small-molecule oral agonist of the human glucagon-like peptide-1 receptor. J. Med. Chem. 65, 8208–8226 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kawai, T. et al. Structural basis for GLP-1 receptor activation by LY3502970, an orally active nonpeptide agonist. Proc. Natl Acad. Sci. USA 117, 29959–29967 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  12. Saxena, A. R. et al. Danuglipron (PF-06882961) in type 2 diabetes: a randomized, placebo-controlled, multiple ascending-dose phase 1 trial. Nat. Med. 27, 1079–1087 (2021).

    Article  CAS  PubMed  Google Scholar 

  13. Ma, H. et al. Structural insights into the activation of GLP-1R by a small molecule agonist. Cell Res. 30, 1140–1142 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Thompson, A., Stephens, J. W., Bain, S. C. & Kanamarlapudi, V. Molecular characterisation of small molecule agonists effect on the human glucagon like peptide-1 receptor internalisation. PLoS ONE 11, e0154229 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Girdhar, K. et al. Novel insights into the dynamics behavior of glucagon-like peptide-1 receptor with its small molecule agonists. J. Biomol. Struct. Dyn. 37, 3976–3986 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Redij, T., Chaudhari, R., Li, Z. Y., Hua, X. X. & Li, Z. J. Structural modeling and in silico screening of potential small-molecule allosteric agonists of a glucagon-like peptide 1 receptor. ACS Omega 4, 961–970 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tamura, T. et al. Identification of an orally active small-molecule PTHR1 agonist for the treatment of hypoparathyroidism. Nat. Commun. 7, 13384 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  18. Zhao, P. et al. Activation of the GLP-1 receptor by a non-peptidic agonist. Nature 577, 432–436 (2020).

    Article  CAS  PubMed  ADS  Google Scholar 

  19. Zhang, X. et al. Evolving cryo-EM structural approaches for GPCR drug discovery. Structure 29, 963–974 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Willard, F. S., Bueno, A. B. & Sloop, K. W. Small molecule drug discovery at the glucagon-like peptide-1 receptor. Exp. Diabetes Res. 2012, 709893 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Zhang, X. et al. Differential GLP-1R binding and activation by peptide and non-peptide agonists. Mol. Cell 80, 485–500 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Arai, Y. et al. Discovery of novel, potent, and orally bioavailable pyrido[2,3-d][1]benzazepin-6-one antagonists for parathyroid hormone receptor 1. Bioorg. Med. Chem. 28, 115524 (2020).

    Article  CAS  PubMed  Google Scholar 

  23. Kobayashi, K. et al. Endogenous ligand recognition and structural transition of a human PTH receptor. Mol. Cell 82, 3468–3483 (2022).

    Article  CAS  PubMed  Google Scholar 

  24. Nishimura, Y. et al. Development of a novel human parathyroid hormone receptor 1 (hPTHR1) agonist (CH5447240), a potent and orally available small molecule for treatment of hypoparathyroidism. J. Med. Chem. 61, 5949–5962 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Nishimura, Y. et al. Lead optimization and avoidance of reactive metabolite leading to PCO371, a potent, selective, and orally available human parathyroid hormone receptor 1 (hPTHR1) agonist. J. Med. Chem. 63, 5089–5099 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Kolb, P. et al. Community guidelines for GPCR ligand bias: IUPHAR review 32. Br. J. Pharmacol. 179, 3651–3674 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Dixon, A. S. et al. NanoLuc complementation reporter optimized for accurate measurement of protein interactions in cells. ACS Chem. Biol. 11, 400–408 (2016).

    Article  CAS  PubMed  ADS  Google Scholar 

  28. Kumari, P. et al. Functional competence of a partially engaged GPCR-beta-arrestin complex. Nat. Commun. 7, 13416 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  29. Zhao, F. et al. Structural insights into hormone recognition by the human glucose-dependent insulinotropic polypeptide receptor. Elife https://doi.org/10.7554/eLife.68719 (2021).

  30. Zhao, L. H. et al. Structure insights into selective coupling of G protein subtypes by a class B G protein-coupled receptor. Nat. Commun. 13, 6670 (2022).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  31. Zhao, L. H. et al. Structure and dynamics of the active human parathyroid hormone receptor-1. Science 364, 148–153 (2019).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  32. Ehrenmann, J. 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).

    Article  CAS  PubMed  Google Scholar 

  33. Zhao, L. H. et al. Molecular recognition of two endogenous hormones by the human parathyroid hormone receptor-1. Acta Pharmacol. Sin. https://doi.org/10.1038/s41401-022-01032-z (2022).

  34. Zhai, X. et al. Molecular insights into the distinct signaling duration for the peptide-induced PTH1R activation. Nat. Commun. 13, 6276 (2022).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  35. Cary, B. P. et al. Molecular insights into peptide agonist engagement with the PTH receptor. Structure https://doi.org/10.1016/j.str.2023.04.002 (2023).

  36. Pace, C. N. & Scholtz, J. M. A helix propensity scale based on experimental studies of peptides and proteins. Biophys. J. 75, 422–427 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yin, Y. et al. Rearrangement of a polar core provides a conserved mechanism for constitutive activation of class B G protein-coupled receptors. J. Biol. Chem. 292, 9865–9881 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  ADS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  40. Chan, P. et al. Purification of heterotrimeric G protein α subunits by GST-Ric-8 association: primary characterization of purified Gαolf. J. Biol. Chem. 286, 2625–2635 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Ma, S. et al. Molecular basis for hormone recognition and activation of corticotropin-releasing factor receptors. Mol. Cell 77, 669–680 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. 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 

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

    Article  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. 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 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  48. 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 

  49. Webb, B. & Sali, A. Comparative protein structure modeling using MODELLER. Curr. Protoc. Bioinformatics 54, 5.6.1–5.6.37 (2016).

    Article  PubMed  Google Scholar 

  50. Smith, R. H. B., Dar, A. C. & Schlessinger, A. PyVOL: a PyMOL plugin for visualization, comparison, and volume calculation of drug-binding sites. Preprint at bioRxiv https://doi.org/10.1101/816702 (2019).

Download references

Acknowledgements

The cryo-EM data were collected at the Advanced Center for Electron Microscopy at the Shanghai Institute of Materia Medica, the Chinese Academy of Sciences (CAS). This work was supported by the CAS Strategic Priority Research Program (XDB37030103 to H.E.X.); the Young Innovator Association of the CAS (2018325 and Y2022078 to L.-H.Z. and 2019282 to K.Wang); the National Natural Science Foundation of China (32071203 to L.-H.Z.; 32130022 and 82121005 to H.E.X.); the National Key R&D Program of China (2019YFA0904200) and the SA-SIBS Scholarship Program to L.-H.Z.; the Shanghai Municipal Science and Technology Major Project (2019SHZDZX02 to H.E.X. and 23ZR1475200 to L.-H.Z.); State Key Laboratory of Drug Research (SKLDR-2023-TT-04).

Author information

Authors and Affiliations

Authors

Contributions

L.-H.Z. designed the expression constructs, purified the complexes, prepared the final samples for collection of cryo-EM data relating to the structure, prepared the cryo-EM grids, participated in model building and function assays, carried out structure and function data analysis, prepared figures and wrote the manuscript; W.H. and K.Wu collected the cryo-EM data, Q.Y. and J.L. carried out map calculations, Q.Y. and H.S. built and refined the structure models; X.H. carried out structure modelling and volume calculation; L.-H.Z., Q.H., Y.G. and Y.L. constructed functional plasmids, Q.H. carried out signalling experiments under the supervision of L.-H.Z.; K.Wang. and J.S. supplied materials; L.-H.Z. and H.E.X. conceived the project and wrote the manuscript.

Corresponding authors

Correspondence to Li-Hua Zhao or H. Eric Xu.

Ethics declarations

Competing interests

H.E.X. is a founder of Cascade Pharmaceuticals. All other authors declare no competing interests.

Peer review

Peer review information

Nature thanks Sudarshan Rajagopal 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 Confocal microscopy of HEK-293 cells expressing PTH1R with β-arrestin-mcherry.

(a–b) The cells expressing GFP-PTH1R (green) with β-arrestin-mcherry (red) and treated with PTH or PCO371 (5 min) were shown with significant recruitment of both β-arrestin 1 and β-arrestin 2 to the cells membranes after PTH stimulation (top row), whereas PCO371 and vehicle control did not stimulate membrane association of both β-arrestin1 and β-arrestin 2 (middle and bottom row). (c–d) HEK-293 cells coexpressing untagged PTH1R and β-arrestin -GFP and treated with PTH (5 min) had significant recruitment of both β-arrestin 1 and β-arrestin 2 to cell membranes (top row). While β-arrestin-GFP (green) appeared diffuse in cells treated with PCO371 stimulation and untreated control (vehicle) (middle and bottom row). Data are from three independent experiments (n = 3). Scale bar, 10 μm.

Extended Data Fig. 2 Construct of receptor and purification of the PCO371-PTH1R-Gs complex.

(a) Snake plot diagram of the PTH1R-LgBiT construct. (b) The size-exclusion chromatography elution profile on Superdex200 Increase 10/300GL (left panel) and SDS-PAGE analysis (right panel) of the PCO371-PTH1R-Gs complex.

Extended Data Fig. 3 Single particle cryo-EM data analysis of the PCO371-PTH1R-Gs complex.

(a) Representative cryo-EM micrograph from 5364 movies of the PCO371-PTH1R-Gs complex and representative 2D class averages with distinct secondary structure features from different views. (b) Data processing flowchart of PCO371-PTH1R-Gs complex by CryoSPARC and Relion. (c) Color cryo-EM map of the PCO371-PTH1R-Gs complex, showing local resolution (Å) calculated using Relion. (d) “Gold-standard” FSC curve of the PCO371-PTH1R-Gs complex, with the global resolution defined at the FSC = 0.143 is 2.57 Å.

Extended Data Fig. 4 Cryo-EM density maps of the PCO371-PTH1R-Gs protein structures.

Cryo-EM density map and the model of the PCO371-PTH1R-Gs structure are shown for all transmembrane helices and helix 8 of PTH1R, PCO371, and Gαs-α5 helix. The model is shown in stick representation. All of them display good density.

Extended Data Fig. 5 Structure comparisons of the active state and inactive state of PTH1R induced by different ligands.

(a–d) Structure comparisons are between the active state complexes with PTH and with PCO371. Superimposition of PTH1R from PDB: 8HA0 (PTH1R: royal blue, PTH: light coral) and the PCO371-bound PTH1R structure (PTH1R: light sea green, PCO371: crimson) reveals different peptide- and PCO371-binding sites. (a–b) Side view of different binding pockets and conformational changes in receptors; (c) Extracellular view and (d) intracellular view of PTH1R conformational changes. (e–h) Superimposed structures of PCO371-bound PTH1R in the active state, and ePTH-bound PTH1R in the inactive state from PDB: 6FJ3 (PTH1R: dark gray, ePTH: dark khaki) and the PCO371-bound PTH1R structure (PTH1R: light sea green, PCO371: crimson). (e–f) Side view of different binding pockets and conformational changes in receptors; (g) Extracellular view and (h) intracellular view of PTH1R conformational changes.

Extended Data Fig. 6 Comparisons of peptide hormone binding pockets and small molecule agonist binding sites of class B GPCRs.

The PCO371 binding pocket is different from both peptide binding pockets and other small molecule binding pockets of class B GPCRs. (a–d) Comparisons the ligand-binding pockets between PCO371 and peptides of PTH1R. The receptor is shown in surface representation and colored in light sea green and PCO371 in crimson is shown as sticks. In three PTH-, PTHrP- and LA-PTH-bound PTH1R-Gs complex structures, the receptors are shown in surface representation and colored in royal blue, dark khaki and dark orchid, respectively. PTH, PTHrP and LA-PTH are colored in light coral, medium violet red and forest green, respectively. They are shown as sticks and ribbon (PDB: 8HA0, 8HAF and 6NBF). G proteins and Nb35 are omitted for clarity. (e–l) Comparisons of small molecule agonist binding sites of class B GPCRs. Superimposition of the PTH1R (light sea green) in complex with Gs bound to PCO371 (crimson) with the GLP1R in complexes with Gs bound to different non-peptidic ligands, including small molecule agonists: TT-OAD2(PDB: 6ORV; TT-OAD2: dark orchid, GLP1R: burly wood); RGT1383 (PDB: 7C2E; RGT1383: green, GLP1R: light salmon); PF-06882961(PDB: 6X1A; PF-06882961: dark orange, GLP1R: thistle); CHU-128 (PDB: 6X19; CHU-128: yellow, GLP1R: rosy brown); LY3502970 (PDB: 6XOX; LY3502970: sandy brown, GLP1R: silver); Boc5 (PDB: 7X8R; Boc5:blue, GLP1R: dark sea green) and WB4-24 (PDB:7X8S; WB4-24: indigo, GLP1R: light steel blue) and with an allosteric ligand, Compound 2, (PDB: 7EVM; Compound 2: goldenrod, GLP1R: tomato), G proteins and Nb35 are omitted for clarity.

Extended Data Fig. 7 Chemical structure of PCO371 and PCO371-mediated cAMP production by receptors containing alanine mutants of key residues in PCO371 binding pocket.

(a) The chemical structure of PCO371 is comprised of the head imidazolidinone, the middle dimethylphenyl, the sulfonamide linker, the piperidine motif, the middle spiro-imidazolone, and the tail trifluoromethoxy phenyl. (b) PCO371-mediated cAMP production by receptors containing alanine mutants of key residues within TM2, TM3, TM6, TM7 and H8. Data from three independent experiments (n = 3) performed in technical triplicate are presented as mean ± s.e.m.

Extended Data Fig. 8 The similarity and the difference of PTH1R in G protein-coupling by hormone peptide and small molecule agonist.

(a) Structural comparison of G protein in different ligands bound PTH1R-Gs complex structures. (b) Close up of the αN and Gαs-α5 helix of Gαs, which form interactions with ICL2 and TMD helix bundles in all G protein bound complex structures, showing similar G protein conformation, but the noteworthy difference is that the C-terminal of Gαs-α5 helix makes additional interactions with the small molecule agonist. (c) Good cryo-EM density supports ligand interact with Gαs. (d-f) The similar set of interactions between the C-terminal of Gαs-α5 helix with the receptor. E392 shifts outward due to steric clash. Y391, E392, and L393 form additional interactions with PCO371.

Extended Data Fig. 9 Key residues for PCO371 selectivity among class B GPCRs.

(a) Structural comparison of receptors and ligands among PCO371-PTH1R-Gs and TT-OAD2-GLP1R-Gs and TIP39-PTH1R-Gs complexes. (a-b) Structural comparison of the cytoplasmic regions of PTH1R, PTH2R and GLP1R during the receptor activation. (c) Structural comparison of P4156.47b and L3706.47b in PTH receptors. (d-e) Different conformations of residues in the active PTH1R, and GLP1R that are involved the interface of PCO371 in receptor activation. (f) Stimulation of cAMP production by PCO371 in the WT and mutants of GLP1R. Data from three independent experiments (n = 3) performed in technical triplicate are presented as mean ± s.e.m. (g) Stimulation of cAMP production by the cognate ligands of PTH1R, PTH2R and GLP1R in mutants of receptors. Data from three independent experiments (n = 3) performed in technical triplicate are presented as mean ± s.e.m.

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics
Extended Data Table 2 Effects of mutations on PCO371-induced activation of PTH1R WT and mutants

Supplementary information

Supplementary Fig. 1

Raw SDS–polyacrylamide gel electrophoresis images of the PCO371–PTH1R–Gs complex.

Reporting Summary

Peer Review File

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

Zhao, LH., He, Q., Yuan, Q. et al. Conserved class B GPCR activation by a biased intracellular agonist. Nature 621, 635–641 (2023). https://doi.org/10.1038/s41586-023-06467-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06467-w

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research