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:

Profiling the proximal proteome of the activated μ-opioid receptor

Abstract

The μ-opioid receptor (μOR) represents an important target of therapeutic and abused drugs. So far, most understanding of μOR activity has focused on a subset of known signal transducers and regulatory molecules. Yet μOR signaling is coordinated by additional proteins in the interaction network of the activated receptor, which have largely remained invisible given the lack of technologies to interrogate these networks systematically. Here we describe a proteomics and computational approach to map the proximal proteome of the activated μOR and to extract subcellular location, trafficking and functional partners of G-protein-coupled receptor (GPCR) activity. We demonstrate that distinct opioid agonists exert differences in the μOR proximal proteome mediated by endocytosis and endosomal sorting. Moreover, we identify two new μOR network components, EYA4 and KCTD12, which are recruited on the basis of receptor-triggered G-protein activation and might form a previously unrecognized buffering system for G-protein activity broadly modulating cellular GPCR signaling.

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: Ligand-dependent proximal proteome changes of the μOR are driven by cellular location.
Fig. 2: Computational framework to model ligand and time-dependent receptor location and deconvoluting receptor location from local interaction network.
Fig. 3: Ligand-dependent proximal interaction network of μOR differs in interactors involved in receptor endocytosis and trafficking.
Fig. 4: Knockouts of VPS35 and COMMD3 change cellular distribution of the μOR.
Fig. 5: EYA4 and KCTD12 are recruited in the μOR proximal interaction network in a Gαi activity-dependent manner.
Fig. 6: EYA4 and KCTD12 modulate G-protein-mediated signaling.

Similar content being viewed by others

Data availability

Shotgun proteomics data access: RAW data and database search results have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository46 with the dataset identifier PXD031415. Targeted proteomics data access: Raw data and SRM transition files can be accessed, queried and downloaded via Panorama47 https://panoramaweb.org/MOR-APEX.url. Source data are provided with this paper.

References

  1. Kieffer, B. L. & Evans, C. J. Opioid receptors: from binding sites to visible molecules in vivo. Neuropharmacology 56, 205–212 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Corder, G., Castro, D. C., Bruchas, M. R. & Scherrer, G. Endogenous and exogenous opioids in pain. Annu. Rev. Neurosci. 41, 453–473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Hilger, D., Masureel, M. & Kobilka, B. K. Structure and dynamics of GPCR signaling complexes. Nat. Struct. Mol. Biol. 25, 4–12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. von Zastrow, M. Proteomic approaches to investigate regulated trafficking and signaling of G protein-coupled receptors. Mol. Pharmacol. 99, 392–398 (2021).

    Article  Google Scholar 

  5. Degrandmaison, J., Rochon-Haché, S., Parent, J.-L. & Gendron, L. Knock-in mouse models to investigate the functions of opioid receptors in vivo. Front. Cell. Neurosci. 16, 807549 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rhee, H.-W. et al. Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science 339, 1328–1331 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hung, V. et al. Proteomic mapping of the human mitochondrial intermembrane space in live cells via ratiometric APEX tagging. Mol. Cell 55, 332–341 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lobingier, B. T. et al. An approach to spatiotemporally resolve protein interaction networks in living cells. Cell 169, 350–360 (2017).

  9. Paek, J. et al. Multidimensional tracking of GPCR signaling via peroxidase-catalyzed proximity labeling. Cell 169, 338–349 (2017).

  10. Manglik, A. et al. Structure-based discovery of opioid analgesics with reduced side effects. Nature 537, 185–190 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. McPherson, J. et al. μ-Opioid receptors: correlation of agonist efficacy for signalling with ability to activate internalization. Mol. Pharmacol. 78, 756–766 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lau, E. K. et al. Quantitative encoding of the effect of a partial agonist on individual opioid receptors by multisite phosphorylation and threshold detection. Sci. Signal. 4, ra52 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Choi, M. et al. MSstats: an R package for statistical analysis of quantitative mass spectrometry-based proteomic experiments. Bioinformatics 30, 2524–2526 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Ehrlich, A. T. et al. Biased signaling of the mu opioid receptor revealed in native neurons. iScience 14, 47–57 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hegde, R. S., Roychoudhury, K. & Pandey, R. N. The multi-functional eyes absent proteins. Crit. Rev. Biochem. Mol. Biol. 55, 372–385 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Fan, X. et al. The alpha subunits of Gz and Gi interact with the eyes absent transcription cofactor Eya2, preventing its interaction with the six class of homeodomain-containing proteins. J. Biol. Chem. 275, 32129–32134 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Embry, A. C., Glick, J. L., Linder, M. E. & Casey, P. J. Reciprocal signaling between the transcriptional co-factor Eya2 and specific members of the Gαi family. Mol. Pharmacol. 66, 1325–1331 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Schwenk, J. et al. Native GABAB receptors are heteromultimers with a family of auxiliary subunits. Nature 465, 231–235 (2010).

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Turecek, R. et al. Auxiliary GABAB receptor subunits uncouple G protein βγ subunits from effector channels to induce desensitization. Neuron 82, 1032–1044 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Zheng, S., Abreu, N., Levitz, J. & Kruse, A. C. Structural basis for KCTD-mediated rapid desensitization of GABAB signalling. Nature 567, 127–131 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. DeWire, S. M. et al. A G protein-biased ligand at the μ-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with morphine. J. Pharmacol. Exp. Ther. 344, 708–717 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Kliewer, A. et al. Morphine‐induced respiratory depression is independent of ß‐arrestin2 signalling. Br. J. Pharmacol. 177, 2923–2931 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Bachmutsky, I., Wei, X. P., Durand, A. & Yackle, K. ß-arrestin 2 germline knockout does not attenuate opioid respiratory depression. eLife 10, e62552 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cullen, P. J. & Steinberg, F. To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat. Rev. Mol. Cell Biol. 19, 679–696 (2018).

    Article  CAS  PubMed  Google Scholar 

  25. Yong, X., Mao, L., Seaman, M. N. J. & Jia, D. An evolving understanding of sorting signals for endosomal retrieval. iScience 25, 104254 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lauffer, B. E. L. et al. SNX27 mediates PDZ-directed sorting from endosomes to the plasma membrane. J. Cell Biol. 190, 565–574 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Temkin, P. et al. SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signalling receptors. Nat. Cell Biol. 13, 715–721 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  28. McGarvey, J. C. et al. Actin-sorting nexin 27 (SNX27)-retromer complex mediates rapid parathyroid hormone receptor recycling. J. Biol. Chem. 291, 10986–11002 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chan, A. S. M. et al. Sorting nexin 27 couples PTHR trafficking to retromer for signal regulation in osteoblasts during bone growth. Mol. Biol. Cell 27, 1367–1382 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. McNally, K. E. et al. Retriever is a multiprotein complex for retromer-independent endosomal cargo recycling. Nat. Cell Biol. 19, 1214–1225 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Healy, M. D. et al. Structure of the endosomal Commander complex linked to Ritscher–Schinzel syndrome. Cell 186, 2219–2237 (2023).

  32. Muntean, B. S. et al. Members of the KCTD family are major regulators of cAMP signaling. Proc. Natl Acad. Sci. USA 119, e2119237119 (2022).

    Article  CAS  PubMed  Google Scholar 

  33. Civciristov, S. et al. Ligand-dependent spatiotemporal signaling profiles of the μ-opioid receptor are controlled by distinct protein-interaction networks. J. Biol. Chem. 294, 16198–16213 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Tsai, T.-H. et al. Selection of features with consistent profiles improves relative protein quantification in mass spectrometry experiments. Mol. Cell. Proteomics 19, 944–959 (2020).

  36. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Korotkevich, G. et al. Fast gene set enrichment analysis. Preprint at bioRxiv https://doi.org/10.1101/060012 (2016).

  38. MacLean, B. et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 26, 966–968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Choi, H. et al. SAINT: probabilistic scoring of affinity purification-mass spectrometry data. Nat. Methods 8, 70–73 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Sowa, M. E., Bennett, E. J., Gygi, S. P. & Harper, J. W. Defining the human deubiquitinating enzyme interaction landscape. Cell 138, 389–403 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Matreyek, K. A., Stephany, J. J., Chiasson, M. A., Hasle, N. & Fowler, D. M. An improved platform for functional assessment of large protein libraries in mammalian cells. Nucleic Acids Res. 48, e1 (2020).

    CAS  PubMed  Google Scholar 

  43. Hermann, M. et al. Binary recombinase systems for high-resolution conditional mutagenesis. Nucleic Acids Res. 42, 3894–3907 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Abreu, N., Acosta-Ruiz, A., Xiang, G. & Levitz, J. Mechanisms of differential desensitization of metabotropic glutamate receptors. Cell Rep. 35, 109050 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Vivaudou, M. et al. Probing the G-protein regulation of GIRK1 and GIRK4, the two subunits of the KACh channel, using functional homomeric mutants. J. Biol. Chem. 272, 31553–31560 (1997).

    Article  CAS  PubMed  Google Scholar 

  46. Vizcaíno, J. A. et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, 11033 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Sharma, V. et al. Panorama: a targeted proteomics knowledge base. J. Proteome Res. 13, 4205–4210 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by funding from the Defense Advanced Research Projects Agency (DARPA) under the Cooperative Agreements HR0011-19-2-0020 (to B.K.S., N.J.K., M.V.Z. and R.H.) and HR0011-20-2-0029 (to N.J.K. and R.H.). The views, opinions and/or findings contained in this material are those of the authors and should not be interpreted as representing the official views or policies of the Department of Defense or the US Government. This work further received funding from the NIH (R01DA056354 to R.H., W.C.-M., and M.V.Z.; P01HL146366 to N.J.K. and R.H.; 1U01MH115747 to N.J.K. and M.V.Z.; R35GM124731 to J.L; R01DA010711, DA012864 and MH120212 to M.V.Z.) and an NSF Graduate Research Fellowship (to N.A). B.T.L. was a recipient of a K99/R00 (DA043607). E.B. was initially supported by an NIH/NRSA Postdoctoral Fellowship (F32CA260118) and is currently supported by a K99 (K99GM151441). M.K.H. was supported by a training grant from NIH (5T32GM139786). A.G.-H. is funded by the Margarita Salas Fellowship from the Spanish Ministry of Universities. J.L. is also supported by the Rohr Family Research Scholar Award and the Irma T. Hirschl and Monique Weill-Caulier Award. The work was carried out in the Thermo Fisher Scientific Mass Spectrometry Facility for Disease Target Discovery at the J. David Gladstone Institutes and the UCSF Center for Advanced Technology. We thank Luke D. Lavis and Claire Deo (Janelia / HHMI) for critical advice and materials in supporting the development of the Janelia Fluor (JF) dye-based receptor trafficking assay. We thank K. Obernier and M. Eckhardt for reading the manuscript and providing critical feedback and members of both the von Zastrow and Krogan laboratories for helpful advice and comments.

Author information

Authors and Affiliations

Authors

Contributions

B.T.L., B.J.P., M.V.Z. and R.H. conceived and directed the study with input from B.K.S. and N.J.K. B.T.L., Q.L. and P.K. performed APEX proximity labeling. B.J.P. developed the data analysis workflow and performed APEX data analysis with input from R.H. M.K.H., W.C.-M. and R.H. performed HaloTag-based trafficking assay with input from E.E.B. and M.V.Z. Electrophysiology experiments were performed by N.A., A.J.G.-H. and J.L. Constructs were cloned by B.T.L., J.X. and P.K. AP–MS experiments were performed by J.X. with input from R.H. AP–MS data analysis was conducted by Z.Z.C.N. with input from R.H. J.X. and R.H. generated and characterized knockout cell lines. E.E.B., B.T.L. and P.K. performed cAMP measurements with input from M.V.Z. B.N., B.T.L. and M.V.Z. performed imaging.

Corresponding authors

Correspondence to Mark Von Zastrow or Ruth Hüttenhain.

Ethics declarations

Competing interests

The Krogan laboratory has received research support from Vir Biotechnology, F. Hoffmann-La Roche and Rezo Therapeutics. N.J.K. has financially compensated consulting agreements with Maze Therapeutics and Interline Therapeutics. He is on the Board of Directors and is President of Rezo Therapeutics and is a shareholder in Tenaya Therapeutics, Maze Therapeutics, Rezo Therapeutics, GEn1E Lifesciences and Interline Therapeutics. B.K.S. is a founder of Epiodyne Inc., BlueDolphin LLC and Deep Apple Therapeutics, serves on the SAB of Schrodinger LLC and of Vilya Therapeutics, on the SRB of Genentech, and consults for Levator Therapeutics, Hyku Therapeutics and Great Point Ventures. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Nevin Lambert 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 APEX2-tagged μOR remains functional and ligand-dependent proximal interaction networks of μOR are enriched for proteins indicating cellular location.

a. Receptor signaling was measured using a commercial cAMP biosensor (pGloSensor-20F). cAMP accumulation was measured after ~10 minutes of DAMGO/isoproterenol incubation and normalized to isoproterenol alone. Data from three independent experiments are presented as mean ± SEM. b. Comparison of agonist-stimulated receptor internalization as assayed by loss of cell surface immunoreactivity and measured by flow cytometry comparing untreated (control) and treated samples (10 μM DAMGO, 30 min). Data from four independent experiments are presented as mean ± SEM. c. Comparison of cell surface recovery of receptors (‘recycling’) following 30 min of DAMGO application (10 μM), agonist removal, and a 30 min recovery period in the presence of antagonist (10 μM). Data from four independent experiments are presented as mean ± SEM. d. The heatmap shows all significantly enriched gene ontology terms (adjusted P value < 0.05) among the proteins that significantly change in the proximal protein environment of the μOR upon activation with DAMGO, morphine, or PZM21 including the number of proteins that match the gene ontology terms. Cluster 1-4 refer to the clustering of the heatmap in Fig. 1b. e. Colocalization of μOR with endosomes to monitor receptor trafficking following activation. HEK293 cells stable expressing the μOR with an N-terminal Flag-tag were activated with 10 μM DAMGO, morphine, or PZM21 for 10 min. The receptor was imaged using anti-Flag. Endosomes were marked with anti-EEA1. n = 3 independent biological replicates, representative example shown, Scale bar is 10μm.

Source data

Extended Data Fig. 2 Validation of spatial reference cell lines and μOR trafficking.

a. Colocalization of location markers with APEX2 spatial references. HEK293 cells stably expressing PM-APEX2, Endo-APEX2, or Lyso-APEX2 were imaged with either Cell Mask to mark the plasma membrane, anti-EEA1 to mark endosomes, or Lyso-Tracker to mark lysosomes. n = 3 independent biological replicates, representative example shown, scale bar is 5μm. b. Colocalization of biotin with APEX2 spatial reference constructs. Localization of biotin following APEX-mediated proximity labeling was probed with Neutravidin and APEX2 constructs were detected with anti-APEX. n = 3 independent biological replicates, representative example shown, scale bar is 5μm.

Extended Data Fig. 3 Prediction of expected protein intensities based on location coefficients.

The heatmap shows the location specific proteins that were selected by pairwise comparison of the spatial reference data and their scaled intensity measured across the spatial references (left side of the heatmap). Agonist and time point dependent expected protein intensities were estimated by summing the spatial reference protein intensities that were weighted with their respective location coefficient. Observed protein intensities are shown as comparison (right side of the heatmap). Data from three independent experiments are presented as mean.

Source data

Extended Data Fig. 4 Effect of data detrending for AP2 complex subunits.

Data detrending process to dissect localization specific effect from effect of interaction with the receptor for AP2A1 (a) and AP2B1 (b), members of the adaptor protein complex. Three different temporal profiles are depicted for each protein, ligand, and replicate: the initial observed intensities, the expected intensities based on the location specific references, and the intensities after detrending. Data from three independent experiments are presented.

Source data

Extended Data Fig. 5 Correlation between ARRB2 engagement upon μOR activation with receptor trafficking.

Correlation between the minimum location coefficient calculated for the plasma membrane (PM) and the maximum ARRB2 log2FC over the time course of μOR activation with DAMGO (red), morphine (yellow) and PZM21 (green).

Source data

Extended Data Fig. 6 Comparing the DAMGO-dependent proximal proteome changes of the μOR in HEK293 and SH-SY5Y cells.

a. Comparison of μOR-APEX2 experiment upon activation with DAMGO in HEK293 and SH-SY5Y neuroblastoma cells. Heatmap focuses on all proteins significant for DAMGO in HEK293 data depicted in Fig. 3a. Data from three biological replicates are presented as mean. b. Temporal profile for selected proximal interactors of the μOR in HEK293 and SH-SY5Y cells. Line charts represent the log2FC over the time course of receptor activation with DAMGO in HEK293 (black) and SH-SY5Y (red) cells. Data from three biological replicates are presented.

Source data

Extended Data Fig. 7 Changes in proximity labeling of EYA4 upon μOR activation.

a. Volcano plot depicting log10 P value and log2FC comparing biotin labeled proteins in the proximity of EYA4 in the presence and absence of μOR activation by treatment with 10 μm DAMGO for 10 min. Data from three biological replicates are presented as mean. b. Volcano plot depicting log10 P value and log2FC comparing biotin labeled proteins in the proximity of EYA4 in the presence and absence of μOR activation by treatment with 10 μm DAMGO for 10 min and treatment with PTX. Data from three independent replicates are presented as mean.

Source data

Extended Data Fig. 8 Generation and validation of EYA4 and KCTD12 KO cell lines.

KO was validated by PCR and sequencing (EYA4) or Western blot analysis (KCTD12). n = 2 independent biological replicates, representative examples shown.

Extended Data Fig. 9 EYA4 and KCTD12 functional validation.

a. cAMP activity in control non-targeting (NT) (closed) and EYA4 KO (open) HEK293 cells stably expressing the μOR. Change in fluorescence intensity of cAMP biosensor upon stimulation with 100 nM isoproterenol (Iso) is plotted. n = 3, *p = 0.0275. b. cAMP activity EYA4 KO cells upon stimulation with Iso without (black) and with co-application of 1 μM somatostatin (SST, green) or 10 μM DAMGO (blue) is plotted. Iso curve is repeated from panel A. n = 3. c. Percent inhibition of Iso-stimulated cAMP with co-application of DAMGO (left) and integrated Iso-stimulated cAMP (right) in control non-targeting (NT) and EYA4 KO cell lines stably expressing the μOR pretreated with PTX. n = 3, *p = 0.0252. d. cAMP activity in control (closed) and KCTD12 KO (open) HEK293 cells stably expressing the μOR upon stimulation with Iso. Control curve is repeated from panel A. n = 3, **p = 0.007. e. cAMP activity in control and KCTD12 KO cells upon stimulation with Iso and SST. Percent inhibition data for control is repeated from panel B. n = 3, **p = 0.0093. f. cAMP activity in control and KCTD12 KO cells upon stimulation with Iso and DAMGO for clones used in main figure (circles) with the main text control curve repeated from Fig. 5a. n = 3. g. cAMP activity in control and KCTD12 KO cells upon stimulation with Iso and DAMGO for clones used in supplemental figures (diamonds). n = 3. h. cAMP activity in WT cells stably overexpressing μOR-APEX2 and transiently overexpressing KCTD12 or an empty vector control upon stimulation with Iso and DAMGO. n = 3. For all panels, data represent biological replicates, shown as individual data points or mean ± SD, and significance was determined by unpaired, two-tailed t-test.

Source data

Extended Data Fig. 10 Electrophysiology measurements for KCTD12 KO.

a. Summary bar graphs showing the average peak amplitudes of μOR-mediated GIRK currents over 60 s 10 µM DAMGO application, in control and KCTD12 KO HEK cells. Each point represents an individual cell. Error bars represent SEM. b. Left, summary bar graphs showing the average peak amplitudes and percent desensitization of μOR-mediated GIRK currents over 60 s 100 nM DAMGO, in control and KCTD12 KO HEK cells. Each point represents an individual cell. Unpaired, two-tailed t-test, * p = 0.0116. Error bars represent SEM. Right, representative whole cell patch clamp recordings of μOR-mediated GIRK currents in response to 60 s 100 nM DAMGO, in control and KCTD12 KO cells. c. Left, Quantification of the tau of desensitization of μOR-mediated GIRK currents over 60 s 10 µM DAMGO application, without (control) and with KCTD12 overexpression in HEK 293 T cells. Each point represents an individual cell. Unpaired t-test, ** p = 0.0038. Error bars represent SEM. Right, representative whole cell patch clamp recordings showing GIRK currents mediated by μOR activation over 60 s 10 µM DAMGO.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Table 8.

Reporting Summary

Supplementary Tables 1–7

Supplementary Tables 1–7. See sheet 1 for table descriptions.

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 Fig. 4 and Extended Data Fig. 8

Uncropped immunoblot images.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

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

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 9

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

Polacco, B.J., Lobingier, B.T., Blythe, E.E. et al. Profiling the proximal proteome of the activated μ-opioid receptor. Nat Chem Biol (2024). https://doi.org/10.1038/s41589-024-01588-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41589-024-01588-3

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