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:

G12/13 is activated by acute tethered agonist exposure in the adhesion GPCR ADGRL3

A Publisher Correction to this article was published on 17 August 2020

This article has been updated

Abstract

The adhesion G-protein-coupled receptor (GPCR) latrophilin 3 (ADGRL3) has been associated with increased risk of attention deficit hyperactivity disorder (ADHD) and substance use in human genetic studies. Knockdown in multiple species leads to hyperlocomotion and altered dopamine signaling. Thus, ADGRL3 is a potential target for treatment of neuropsychiatric disorders that involve dopamine dysfunction, but its basic signaling properties are poorly understood. Identification of adhesion GPCR signaling partners has been limited by a lack of tools to acutely activate these receptors in living cells. Here, we design a novel acute activation strategy to characterize ADGRL3 signaling by engineering a receptor construct in which we could trigger acute activation enzymatically. Using this assay, we found that ADGRL3 signals through G12/G13 and Gq, with G12/13 the most robustly activated. Gα12/13 is a new player in ADGRL3 biology, opening up unexplored roles for ADGRL3 in the brain. Our methodological advancements should be broadly useful in adhesion GPCR research.

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: Exposure of the Adgrl3 TA promotes intracellular signaling.
Fig. 2: Adgrl3 CTF signals through Gq and G13.
Fig. 3: Adgrl3 couples to Gα12/13 upon acute exposure of the TA.
Fig. 4: Adgrl3 recruits β-arrestin to the plasma membrane in living cells.

Similar content being viewed by others

Data availability

All cDNA constructs and data are available on request from the authors. Unprocessed full scans are provided for the immunoblots shown in Extended Data Figs. 5 and 8. Source data are provided with this paper.

Change history

References

  1. Purcell, R. H., Hall, R. A. & Adhesion, G. Protein-coupled receptors as drug targets. Annu. Rev. Pharm. Toxicol. 58, 429–449 (2018).

    Article  CAS  Google Scholar 

  2. Hamann, J. et al. International Union of Basic and Clinical Pharmacology. XCIV. Adhesion G protein-coupled receptors. Pharm. Rev. 67, 338–367 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Arcos-Burgos, M. et al. Attention-deficit/hyperactivity disorder in a population isolate: Linkage to loci at 4q13.2, 5q33.3, 11q22 and 17p11. Am. J. Hum. Genet. 75, 998–1014 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Arcos-Burgos, M. et al. ADGRL3 (LPHN3) variants predict substance use disorder. Transl. Psychiatry 9, 42 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. van der Voet, M., Harich, B., Franke, B. & Schenck, A. ADHD-associated dopamine transporter, latrophilin and neurofibromin share a dopamine-related locomotor signature in Drosophila. Mol. Psychiatry 21, 565–573 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Lange, M. et al. The ADHD-susceptibility gene lphn3.1 modulates dopaminergic neuron formation and locomotor activity during zebrafish development. Mol. Psychiatry 17, 946–954 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Lange, M., Froc, C., Grunwald, H., Norton, W. H. J. & Bally-Cuif, L. Pharmacological analysis of zebrafish lphn3.1 morphant larvae suggests that saturated dopaminergic signaling could underlie the ADHD-like locomotor hyperactivity. Prog. Neuropsychopharmacol. Biol. Psychiatry 84, 181–189 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Regan, S. L. et al. Knockout of latrophilin-3 in Sprague–Dawley rats causes hyperactivity, hyper-reactivity, under-response to amphetamine and disrupted dopamine markers. Neurobiol. Dis. 130, 104494 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wallis, D. et al. Initial characterization of mice null for Lphn3, a gene implicated in ADHD and addiction. Brain Res. 1463, 85–92 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Arac, D. et al. A novel evolutionarily conserved domain of cell-adhesion GPCRs mediates autoproteolysis. EMBO J. 31, 1364–1378 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lin, H. H. et al. Autocatalytic cleavage of the EMR2 receptor occurs at a conserved G protein-coupled receptor proteolytic site motif. J. Biol. Chem. 279, 31823–31832 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Paavola, K. J., Stephenson, J. R., Ritter, S. L., Alter, S. P. & Hall, R. A. The N terminus of the adhesion G protein-coupled receptor GPR56 controls receptor signaling activity. J. Biol. Chem. 286, 28914–28921 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Paavola, K. J. & Hall, R. A. Adhesion G protein-coupled receptors: signaling, pharmacology and mechanisms of activation. Mol. Pharm. 82, 777–783 (2012).

    Article  CAS  Google Scholar 

  14. Liebscher, I. et al. A tethered agonist within the ectodomain activates the adhesion G protein-coupled receptors GPR126 and GPR133. Cell Rep. 9, 2018–2026 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stoveken, H. M., Hajduczok, A. G., Xu, L. & Tall, G. G. Adhesion G protein-coupled receptors are activated by exposure of a cryptic tethered agonist. Proc. Natl Acad. Sci. USA 112, 6194–6199 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lelianova, V. G. et al. Alpha-latrotoxin receptor, latrophilin, is a novel member of the secretin family of G protein-coupled receptors. J. Biol. Chem. 272, 21504–21508 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Rahman, M. A. et al. Norepinephrine exocytosis stimulated by α-latrotoxin requires both external and stored Ca2+ and is mediated by latrophilin, G proteins and phospholipase C. Philos. Trans. R. Soc. Lond. B Biol. Sci. 354, 379–386 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Silva, J. P. et al. Latrophilin 1 and its endogenous ligand Lasso/teneurin-2 form a high-affinity transsynaptic receptor pair with signaling capabilities. Proc. Natl Acad. Sci. USA 108, 12113–12118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Muller, A. et al. Oriented cell division in the C. elegans embryo is coordinated by G-protein signaling dependent on the adhesion GPCR LAT-1. PLoS Genet. 11, e1005624 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Scholz, N. et al. Mechano-dependent signaling by latrophilin/CIRL quenches cAMP in proprioceptive neurons. eLife 6, e28360 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Nazarko, O. et al. A comprehensive mutagenesis screen of the adhesion GPCR latrophilin-1/ADGRL1. iScience 3, 264–278 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rothe, J. et al. Involvement of the adhesion GPCRs latrophilins in the regulation of insulin release. Cell Rep. 26, e1575 (2019).

    Article  Google Scholar 

  23. Liebscher, I. & Schoneberg, T. Tethered agonism: a common activation mechanism of adhesion GPCRs. Handb. Exp. Pharm. 234, 111–125 (2016).

    Article  CAS  Google Scholar 

  24. Martin, A. L., Steurer, M. A. & Aronstam, R. S. Constitutive activity among orphan class-A G protein coupled receptors. PLoS ONE 10, e0138463 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Le Bonniec, B. F. et al. Characterization of the P2′ and P3′ specificities of thrombin using fluorescence-quenched substrates and mapping of the subsites by mutagenesis. Biochemistry 35, 7114–7122 (1996).

    Article  PubMed  Google Scholar 

  26. Okashah, N. et al. Variable G protein determinants of GPCR coupling selectivity. Proc. Natl Acad. Sci. USA 116, 12054–12059 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Green, D. A. & Clark, R. B. Direct evidence for the role of the coupling proteins in forskolin activation of adenylate cyclase. J. Cycl. Nucleotide Res. 8, 337–346 (1982).

    CAS  Google Scholar 

  28. Downs, R. W. Jr. & Aurbach, G. D. The effects of forskolin on adenylate cyclase in S49 wild type and cyc-cells. J. Cycl. Nucleotide Res. 8, 235–242 (1982).

    CAS  Google Scholar 

  29. Hollins, B., Kuravi, S., Digby, G. J. & Lambert, N. A. The C-terminus of GRK3 indicates rapid dissociation of G protein heterotrimers. Cell Signal 21, 1015–1021 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Masuho, I. et al. Distinct profiles of functional discrimination among G proteins determine the actions of G protein-coupled receptors. Sci. Signal. 8, ra123 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Vu, T. K., Wheaton, V. I., Hung, D. T., Charo, I. & Coughlin, S. R. Domains specifying thrombin-receptor interaction. Nature 353, 674–677 (1991).

    Article  CAS  PubMed  Google Scholar 

  32. Hauge Pedersen, M., Pham, J., Mancebo, H., Inoue, A. & Javitch, J. A. A novel luminescence-based β-arrestin membrane recruitment assay for unmodified GPCRs. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/2020.04.09.034520v1 (2020).

  33. Soh, U. J. & Trejo, J. Activated protein C promotes protease-activated receptor-1 cytoprotective signaling through β-arrestin and dishevelled-2 scaffolds. Proc. Natl Acad. Sci. USA 108, E1372–E1380 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Demberg, L. M., Rothemund, S., Schoneberg, T. & Liebscher, I. Identification of the tethered peptide agonist of the adhesion G protein-coupled receptor GPR64/ADGRG2. Biochem. Biophys. Res. Commun. 464, 743–747 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Worzfeld, T., Wettschureck, N. & Offermanns, S. G12/G13-mediated signalling in mammalian physiology and disease. Trends Pharmacol. Sci. 29, 582–589 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Iguchi, T. et al. Orphan G protein-coupled receptor GPR56 regulates neural progenitor cell migration via a Gα12/13 and Rho pathway. J. Biol. Chem. 283, 14469–14478 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Ohta, S. et al. Agonistic antibodies reveal the function of GPR56 in human glioma U87-MG cells. Biol. Pharm. Bull. 38, 594–600 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Zhang, D. L. et al. Gq activity- and β-arrestin-1 scaffolding-mediated ADGRG2/CFTR coupling are required for male fertility. eLife 7, e33432 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Peeters, M. C. et al. The adhesion G protein-coupled receptor G2 (ADGRG2/GPR64) constitutively activates SRE and NFκB and is involved in cell adhesion and migration. Cell Signal. 27, 2579–2588 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Kishore, A., Purcell, R. H., Nassiri-Toosi, Z. & Hall, R. A. Stalk-dependent and Stalk-independent signaling by the adhesion G protein-coupled receptors GPR56 (ADGRG1) and BAI1 (ADGRB1). J. Biol. Chem. 291, 3385–3394 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Okajima, D., Kudo, G. & Yokota, H. Brain-specific angiogenesis inhibitor 2 (BAI2) may be activated by proteolytic processing. J. Recept. Sig. Transduct. Res. 30, 143–153 (2010).

    Article  CAS  Google Scholar 

  42. O’Sullivan, M. L. et al. FLRT proteins are endogenous latrophilin ligands and regulate excitatory synapse development. Neuron 73, 903–910 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Sando, R., Jiang, X. & Sudhof, T. C. Latrophilin GPCRs direct synapse specificity by coincident binding of FLRTs and teneurins. Science 363, eaav7969 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, J. et al. Structural basis for teneurin function in circuit-wiring: a toxin motif at the synapse. Cell 173, 735–748 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cruz-Ortega, J. S. & Boucard, A. A. Actin cytoskeleton remodeling defines a distinct cellular function for adhesion G protein-coupled receptors ADGRL/latrophilins 1, 2 and 3. Biol. Open 8, bio039826 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Southern, C. et al. Screening β-arrestin recruitment for the identification of natural ligands for orphan G-protein-coupled receptors. J. Biomol. Screen. 18, 599–609 (2013).

    Article  PubMed  Google Scholar 

  47. Lefkowitz, R. J. Arrestins come of age: a personal historical perspective. Prog. Mol. Biol. Transl. Sci. 118, 3–18 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Spiess, K. et al. Arrestin-independent constitutive endocytosis of GPR125/ADGRA3. Ann. N. Y. Acad. Sci. 1456, 186–199 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Petersen, S. C. et al. The adhesion GPCR GPR126 has distinct, domain-dependent functions in Schwann cell development mediated by interaction with laminin-211. Neuron 85, 755–769 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Scholz, N. et al. The adhesion GPCR latrophilin/CIRL shapes mechanosensation. Cell Rep. 11, 866–874 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Promel, S. et al. The GPS motif is a molecular switch for bimodal activities of adhesion class G protein-coupled receptors. Cell Rep. 2, 321–331 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Stoveken, H. M. et al. Dihydromunduletone is a small-molecule selective adhesion G protein-coupled receptor antagonist. Mol. Pharmacol. 90, 214–224 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Stoveken, H. M., Larsen, S. D., Smrcka, A. V. & Tall, G. G. Gedunin- and khivorin-derivatives are small-molecule partial agonists for adhesion G protein-coupled receptors GPR56/ADGRG1 and GPR114/ADGRG5. Mol. Pharmacol. 93, 477–488 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Grundmann, M. et al. Lack of β-arrestin signaling in the absence of active G proteins. Nat. Commun. 9, 341 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Stallaert, W. et al. Purinergic receptor transactivation by the β2-adrenergic receptor increases intracellular Ca2+ in nonexcitable cells. Mol. Pharmacol. 91, 533–544 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants MH112156 (J.A.J.), GM130142 (N.A.L.), GM131672 (N.O.) and T32-GM007315 (A.V.), and by the Hope for Depression Research Foundation (J.A.J.). A.I. was funded by PRIME 18gm5910013 and LEAP 18gm0010004 from the Japan Agency for Medical Research and Development (AMED) and KAKENHI 17K08264 from the Japan Society for the Promotion of Science (JSPS). T.L. was funded by the Deutsche Forschungsgemeinschaft through FOR2149 project P01 [LA2861/4-2] and CRC 1423, project number 421152132, subprojects A06, B06. We thank L. Lavis and J.B. Grimm (Janelia Research Campus) for generously providing the JF-525 and JF-646 fluorophores.

Author information

Authors and Affiliations

Authors

Contributions

S.M. and J.A.J. designed the overall project strategy and experiments, which were performed by S.M., T.P. and N.A.P. GTPγS and dual luciferase SRE experiments were designed by G.G.T. and performed by H.M.S., D.P.M. and A.V. GTP-depleted BRET experiments were designed by N.A.L. and performed by N.O. A.I. provided CRISPR KO cell lines. T.L. provided Adgrl3 cDNA. S.M. and J.A.J. wrote the manuscript. T.L., A.I., N.A.L., G.G.T., S.M. and J.A.J. discussed the experimental findings, interpretation of results and edited the manuscript.

Corresponding author

Correspondence to Jonathan A. Javitch.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 TA-enhanced signaling effect is also observed in SRE, NFAT, and NFκB gene expression assays.

a, SRE b, NFκB and c, NFAT. For all gene response elements (SRE, NFAT, and NFκB) signaling was increased significantly when the entire N-terminal fragment up to the GPS cleavage site (CTF) was removed; FL receptor also showed some activity in SRE (comparable to CRE in Fig. 1). Luminescence in (a-c) was measured for a range of increased receptor cDNA concentrations 24 h after transfection in HEK293T cells. All data points are normalized to an empty vector control. Data are presented as mean values ±SEM from 3 independent experimental replicates.

Source data

Extended Data Fig. 2 Successive truncation of the first three ADGRL3 tethered agonist residues dramatically blunts SRE-Luciferase gene reporter activation.

SRE gene expression assay for ADGRL3 (human homolog) CTF, Δ1-CTF, Δ2-CTF and Δ3-CTF. Luminescence was measured for a range of increased receptor cDNA amounts (ng) 24 h after transfection in HEK293T cells. For the dual luciferase assay, data are presented as Firefly/Renilla luciferase units, and all data points are normalized to the corresponding ratio for the empty vector control15,52,53. Data are from one representative experiment performed 3 times. Data are presented as mean ±SD from triplicate technical replicates.

Source data

Extended Data Fig. 3 Screen of Adgrl3 (FL, CTF, and Δ5-CTF constructs) signaling in the 4 major G protein signaling pathways utilizing a HEK293 CRISPR knockout cell line (HEKΔ7) and a panel of gene expression assays.

a, CRE b, NFκB c, SRE. Each Gα protein species was reintroduced one at a time (see color legend for specification) at optimized cDNA concentrations and luminescence signals were evaluated for empty vector control and receptor constructs 24 h after transfection. All data points are normalized to corresponding empty vector control. Bars indicate mean values ±SEM from 4 (a) and 5 (b-c) independent experimental replicates. Bars for Gαolf and12 are presented as mean values ±SEM from 3 independent experimental replicates.

Source data

Extended Data Fig. 4 CTF Gαq signaling is detected both in CRE and NFκB.

CTF signaling in CRE, NFκB, and SRE was evaluated after 18 h of treatment with either vehicle or a potent Gαq inhibitor (YM-254890, 1 μM). Data was collected in regular HEK293T cells. Data points are normalized to empty vector control and displayed as the fold decrease with YM-254890. Bars show mean ±SEM from 4 independent experimental replicates.

Source data

Extended Data Fig. 5 Urea-mediated ADGRL3 N-terminal Fragment dissociation.

For the membrane urea treatment experiments presented in this figure, a FLAG- (N-terminal) and His8- (C-terminal) tagged ADGRL3 construct that was truncated N-terminally to the HormR domain was used21 (See Fig. 1 for Adgrl3 architecture). Insect cell membranes (High-Five) with expressed ADGRL3 were mock treated or extracted with urea. The presence of the ADGRL3 NTF and CTF in the membrane (Mem) and extract (Soluble, Sol) fractions was determined by immunoblotting with an anti-FLAG antibody to detect the NTF and an anti-penta-His antibody to detect the CTF. The NTF (apparent MW ~50 kDa) was partially solubilized with the urea, whereas the CTF (apparent MW ~27 kDa) was not. The penta-His blot panels are from one contiguous blot, but broken to avoid oversaturation of the ~70 kDa band (unprocessed receptor) and to show a higher exposure of low MW panel (~27 kDa CTF). Data from one representative experiment that was repeated three times.

Source data

Extended Data Fig. 6 CRE, NFκB, and SRE gene expression assays for PAR1-CTF and corresponding T923S/ΔN924-CTF control construct.

CRE a, NFκB b, and SRE c, signaling was increased significantly for T923S/ΔN924-CTF to levels comparable with CTF, whereas PAR1-CTF signals were comparable to FL levels. CTF and FL are replotted from Fig. 1c and Extended Data Fig. 1 for direct comparison. Luminescence was measured for a range of receptor cDNA concentrations 24 h after transfection in HEK293T cells. All data points are normalized to an empty vector control. Data are shown as mean ±SEM from 3 independent experimental replicates.

Source data

Extended Data Fig. 7 TA-exposed Adgrl3 does not activate the Gαi/o family.

a, Gβγ release assay testing D2R activation of Gαi1, Gαi2, Gαi3, GαoA and GαoB in HEK full G protein KO cells. In comparison to the HEKΔ7 CRISPR knockout, this cell line also lacks the full Gαi/o family. Luminescence was read 10 min after stimulation with 10 μM quinpirole. b, Gβγ release assay testing the T923S/ΔN924-CTF, PAR1-CTF, and PAR1 activation of Gαi1, Gαi2, Gαi3, GαoA and GαoB in HEK full G protein KO cells. Luminescence was read 10 min after stimulation with 1 μM thrombin. All data are normalized to buffer controls and show the BRET effect induced by ligands. Bars show mean ±SEM from 3 independent experimental replicates. One-way ANOVA with Dunnett’s multiple-comparison post-hoc test was performed for each cDNA construct individually, (no receptor (empty vector), T923S/ΔN924-CTF, PAR1-CTF, and PAR1) to determine statistical significance between the No Gα control and each Gα subtype (For Gαi3 **p = 0.0064, for GαoB **p = 0.0032). See Supplementary Data for the full set of p-values.

Source data

Extended Data Fig. 8 β-arrestin-2 decreases G protein-dependent ERK1/2 phosphorylation.

HEK Δβarr1/2 cells were transfected with PAR1-CTF or PAR1-CTF with β-arrestin-2. After 48 hr, the cells were acutely activated with 1 μM thrombin over a time course of 45 min. a, Representative immunoblotting analysis with antibodies against phosphoERK1/2 (#9101 S), total ERK1/2 (#9102 S), and HA (#2367 S). Each sample was derived from the same experiment and the blots were processed in parallel. The HA blot was used as a sample processing control to ensure uniform β-arrestin-2 expression. b, The level of phosphoERK1/2 was normalized to total ERK and the baseline at 0 min was subtracted to produce the time-dependent change in pERK1/2. Data are presented as mean ± SEM from 3 (PAR1-CTF) 4 (PAR1-CTF, β-Arrestin-2) independent experimental replicates.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–8 and Table 1.

Reporting Summary

Supplementary Data

Source Data for Supplementary Figs. 1–8 and P values for relevant figures.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots and/or gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mathiasen, S., Palmisano, T., Perry, N.A. et al. G12/13 is activated by acute tethered agonist exposure in the adhesion GPCR ADGRL3. Nat Chem Biol 16, 1343–1350 (2020). https://doi.org/10.1038/s41589-020-0617-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0617-7

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