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:

Molecular mechanism of GPCR spatial organization at the plasma membrane

Abstract

G-protein-coupled receptors (GPCRs) mediate many critical physiological processes. Their spatial organization in plasma membrane (PM) domains is believed to encode signaling specificity and efficiency. However, the existence of domains and, crucially, the mechanism of formation of such putative domains remain elusive. Here, live-cell imaging (corrected for topography-induced imaging artifacts) conclusively established the existence of PM domains for GPCRs. Paradoxically, energetic coupling to extremely shallow PM curvature (<1 µm−1) emerged as the dominant, necessary and sufficient molecular mechanism of GPCR spatiotemporal organization. Experiments with different GPCRs, H-Ras, Piezo1 and epidermal growth factor receptor, suggest that the mechanism is general, yet protein specific, and can be regulated by ligands. These findings delineate a new spatiomechanical molecular mechanism that can transduce to domain-based signaling any mechanical or chemical stimulus that affects the morphology of the PM and suggest innovative therapeutic strategies targeting cellular shape.

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: Topography-corrected imaging of β1AR density reveals PM domains.
Fig. 2: MFT reveals energetic coupling to shallow mean membrane curvature as the molecular mechanism of domain formation.
Fig. 3: Nanoscopic modulation of PM curvature quantitatively regulates β1AR density.
Fig. 4: Curvature-coupled GPCR domains were identified with high statistical significance for different GPCRs and cell types.
Fig. 5: A general mechanism of spatial organization that is protein specific and can be regulated by ligands.

Similar content being viewed by others

Data availability

The structure of the inactive β1AR is available from the Protein Data Bank with accession code 2YCW. Source data are provided with this paper.

Code availability

Algorithms used by custom analysis code are described in detail in the Methods. Code is available upon reasonable request.

References

  1. Pierce, K. L., Premont, R. T. & Lefkowitz, R. J. Seven-transmembrane receptors. Nat. Rev. Mol. Cell Biol. 3, 639–650 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Weis, W. I. & Kobilka, B. K. The molecular bases of G protein-coupled receptor activation. Annu. Rev. Biochem. 87, 879–919 (2018).

    Article  Google Scholar 

  3. Wootten, D., Christopoulos, A., Marti-Solano, M., Babu, M. M. & Sexton, P. M. Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat. Rev. Mol. Cell Biol. 19, 638–653 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Nikolaev, V. O. et al. β2-Adrenergic receptor redistribution in heart failure changes cAMP compartmentation. Science 327, 1653–1657 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Irannejad, R. et al. Functional selectivity of GPCR-directed drug action through location bias. Nat. Chem. Biol. 13, 799–806 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Briddon, S. J., Kilpatrick, L. E. & Hill, S. J. Studying GPCR pharmacology in membrane microdomains: fluorescence correlation spectroscopy comes of age. Trends Pharmacol. Sci. 39, 158–174 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Insel, P. A. et al. Compartmentation of G-protein-coupled receptors and their signalling components in lipid rafts and caveolae. Biochem. Soc. Trans. 33, 1131-4 (2005).

    Article  PubMed  Google Scholar 

  8. Pontier, S. M. et al. Cholesterol-dependent separation of the β2-adrenergic receptor from its partners determines signaling efficacy: insight into nanoscale organization of signal transduction. J. Biol. Chem. 283, 24659–24672 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lohse, M. J. & Hofmann, K. P. Spatial and temporal aspects of signaling by G-protein-coupled receptors. Mol. Pharmacol. 88, 572–578 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Calebiro, D. & Sungkaworn, T. Single-molecule imaging of GPCR interactions. Trends Pharmacol. Sci. 39, 109–122 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Halls, M. L. et al. Plasma membrane localization of the μ-opioid receptor controls spatiotemporal signaling. Sci. Signal. 9, ra16 (2016).

    Article  PubMed  Google Scholar 

  12. Sungkaworn, T. et al. Single-molecule imaging reveals receptor-G protein interactions at cell surface hot spots. Nature 550, 543–547 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Sezgin, E., Levental, I., Mayor, S. & Eggeling, C. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat. Rev. Mol. Cell Biol. 18, 361–374 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Annibale, P. & Lohse, M. J. Spatial heterogeneity in molecular brightness. Nat. Methods 17, 273–275 (2020).

    Article  CAS  PubMed  Google Scholar 

  15. Adler, J., Shevchuk, A. I., Novak, P., Korchev, Y. E. & Parmryd, I. Plasma membrane topography and interpretation of single-particle tracks. Nat. Methods 7, 170–171 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Eichel, K. & von Zastrow, M. Subcellular organization of GPCR signaling. Trends Pharmacol. Sci. 39, 200–208 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rosholm, K. R. et al. Membrane curvature regulates ligand-specific membrane sorting of GPCRs in living cells. Nat. Chem. Biol. 13, 724–729 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Paszek, M. J. et al. Scanning angle interference microscopy reveals cell dynamics at the nanoscale. Nat. Methods 9, 825–827 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Elliott, H. et al. Myosin II controls cellular branching morphogenesis and migration in three dimensions by minimizing cell-surface curvature. Nat. Cell Biol. 17, 137–147 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kabbani, A. M. & Kelly, C. V. The detection of nanoscale membrane bending with polarized localization microscopy. Biophys. J. 113, 1782–1794 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. van Rheenen, J. & Jalink, K. Agonist-induced PIP2 hydrolysis inhibits cortical actin dynamics: regulation at a global but not at a micrometer scale. Mol. Biol. Cell 13, 3257–3267 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Clack, N. G. & Groves, J. T. Many-particle tracking with nanometer resolution in three dimensions by reflection interference contrast microscopy. Langmuir 21, 6430–6435 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Calebiro, D. et al. Single-molecule analysis of fluorescently labeled G-protein-coupled receptors reveals complexes with distinct dynamics and organization. Proc. Natl Acad. Sci. USA 110, 743–748 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. May, S. & Ben-Shaul, A. A molecular model for lipid-mediated interaction between proteins in membranes. Phys. Chem. Chem. Phys. 2, 4494–4502 (2000).

    Article  CAS  Google Scholar 

  25. Sperotto, M. M., May, S. & Baumgaertner, A. Modelling of proteins in membranes. Chem. Phys. Lipids 141, 2–29 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Larsen, J. B. et al. Membrane curvature enables N-Ras lipid anchor sorting to liquid-ordered membrane phases. Nat. Chem. Biol. 11, 192–194 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Larsen, J. B. et al. Membrane curvature and lipid composition synergize to regulate N-Ras anchor recruitment. Biophys. J. 113, 1269–1279 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Larsen, J. B. et al. How membrane geometry regulates protein sorting independently of mean curvature. ACS Cent. Sci. 6, 1159–1168 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Moukhametzianov, R. et al. Two distinct conformations of helix 6 observed in antagonist-bound structures of a β1-adrenergic receptor. Proc. Natl Acad. Sci. USA 108, 8228–8232 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lorent, J. H. et al. Plasma membranes are asymmetric in lipid unsaturation, packing and protein shape. Nat. Chem. Biol. 16, 644–652 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Marrink, S. J. et al. Computational modeling of realistic cell membranes. Chem. Rev. 119, 6184–6226 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Latorraca, N. R., Venkatakrishnan, A. J. & Dror, R. O. GPCR dynamics: structures in motion. Chem. Rev. 117, 139–155 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Zhao, W. et al. Nanoscale manipulation of membrane curvature for probing endocytosis in live cells. Nat. Nanotechnol. 12, 750–756 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. McMahon, H. T. & Gallop, J. L. Membrane curvature and mechanisms of dynamic cell membrane remodelling. Nature 438, 590–596 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Aimon, S. et al. Membrane shape modulates transmembrane protein distribution. Dev. Cell 28, 212–218 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Brown, M. F. Curvature forces in membrane lipid–protein interactions. Biochemistry 51, 9782–9795 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Hossein, A. & Deserno, M. Spontaneous curvature, differential stress, and bending modulus of asymmetric lipid membranes. Biophys. J. 118, 624–642 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Soubias, O., Teague, W. E. Jr, Hines, K. G. & Gawrisch, K. Rhodopsin/lipid hydrophobic matching-rhodopsin oligomerization and function. Biophys. J. 108, 1125–1132 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Joyce, G., Robertson, B. D. & Williams, K. J. A modified agar pad method for mycobacterial live-cell imaging. BMC Res. Notes 4, 73 (2011).

  40. Bathe-Peters, M. et al. Visualization of β-adrenergic receptor dynamics and differential localization in cardiomyocytes. Proc. Natl Acad. Sci. USA 118, e2101119118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, Y. et al. Cryo-EM structure of the activated GLP-1 receptor in complex with a G protein. Nature 546, 248–253 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Murthy, S. E., Dubin, A. E. & Patapoutian, A. Piezos thrive under pressure: mechanically activated ion channels in health and disease. Nat. Rev. Mol. Cell Biol. 18, 771–783 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Zhou, R., Han, B., Xia, C. & Zhuang, X. Membrane-associated periodic skeleton is a signaling platform for RTK transactivation. Science 365, 929–934 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Antonny, B. Mechanisms of membrane curvature sensing. Annu. Rev. Biochem. 80, 101–123 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Iversen, L., Mathiasen, S., Larsen, J. B. & Stamou, D. Membrane curvature bends the laws of physics and chemistry. Nat. Chem. Biol. 11, 822–825 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Bassereau, P. et al. The 2018 biomembrane curvature and remodeling roadmap. J. Phys. D Appl. Phys. 51, 343001 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Lingwood, D. & Simons, K. Lipid rafts as a membrane-organizing principle. Science 327, 46–50 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Hatzakis, N. S. et al. How curved membranes recruit amphipathic helices and protein anchoring motifs. Nat. Chem. Biol. 5, 835–841 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Huang, W. et al. Structure of the neurotensin receptor 1 in complex with β-arrestin 1. Nature 579, 303–308 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Foerster, F. et al. Targeting the actin cytoskeleton: selective antitumor action via trapping PKCε. Cell Death Dis. 5, e1398 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Roffay, C. et al. Passive coupling of membrane tension and cell volume during active response of cells to osmosis. Proc. Natl Acad. Sci. USA 118, e2103228118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zimmerberg, J. & Kozlov, M. M. How proteins produce cellular membrane curvature. Nat. Rev. Mol. Cell Biol. 7, 9–19 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Wolfenson, H., Yang, B. & Sheetz, M. Steps in mechanotransduction pathways that control cell morphology. Annu. Rev. Physiol. 81, 585–605 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Zimmerberg, J. & Gawrisch, K. The physical chemistry of biological membranes. Nat. Chem. Biol. 2, 564–567 (2006).

    Article  CAS  PubMed  Google Scholar 

  55. Staudt, T. et al. Far-field optical nanoscopy with reduced number of state transition cycles. Opt. Express 19, 5644–5657 (2011).

    Article  PubMed  Google Scholar 

  56. Shelton, E., Serwane, F. & Campas, O. Geometrical characterization of fluorescently labelled surfaces from noisy 3D microscopy data. J. Microsc. 269, 259–268 (2018).

    Article  CAS  PubMed  Google Scholar 

  57. Lohse, M. J., Nuber, S. & Hoffmann, C. Fluorescence/bioluminescence resonance energy transfer techniques to study G-protein-coupled receptor activation and signaling. Pharmacol. Rev. 64, 299–336 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Lambert, N. A. & Javitch, J. A. CrossTalk opposing view: weighing the evidence for class A GPCR dimers, the jury is still out. J. Physiol. 592, 2443–2445 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Barbotin, A. et al. z-STED imaging and spectroscopy to investigate nanoscale membrane structure and dynamics. Biophys. J. 118, 2448–2457 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Uline, M. J. & Szleifer, I. Mode specific elastic constants for the gel, liquid-ordered, and liquid-disordered phases of DPPC/DOPC/cholesterol model lipid bilayers. Faraday Discuss. 161, 177–191 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Keppler, A. et al. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat. Biotechnol. 21, 86–89 (2003).

    Article  CAS  PubMed  Google Scholar 

  62. Sungkaworn, T., Rieken, F., Lohse, M. J. & Calebiro, D. High-resolution spatiotemporal analysis of receptor dynamics by single-molecule fluorescence microscopy. J. Vis. Exp. https://doi.org/10.3791/51784 (2014).

  63. Maiellaro, I., Lohse, M. J., Kittel, R. J. & Calebiro, D. Real-time monitoring of GPCR/cAMP signalling by FRET and single-molecule microscopy. Horm. Metab. Res. 46, 827–832 (2014).

    Article  PubMed  Google Scholar 

  64. Curtis, A. The mechanism of adhesion of cell to glass: a study by Interference reflection microscopy. J. Cell Biol. 20, 199–215 (1964).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Limozin, L. & Sengupta, K. Quantitative reflection interference contrast microscopy (RICM) in soft matter and cell adhesion. ChemPhysChem 10, 2752–2768 (2009).

    Article  CAS  PubMed  Google Scholar 

  66. Barr, V. A. & Bunnell, S. C. Interference reflection microscopy. Curr. Protoc. Cell Biol. 4, 4.23 (2009).

  67. Kukulski, W. et al. Precise, correlated fluorescence microscopy and electron tomography of lowicryl sections using fluorescent fiducial markers. Methods Cell. Biol. 111, 235–257 (2012).

    Article  CAS  PubMed  Google Scholar 

  68. Biswas, A., Alex, A. & Sinha, B. Mapping cell membrane fluctuations reveals their active regulation and transient heterogeneities. Biophys. J. 113, 1768–1781 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Vicidomini, G. et al. STED nanoscopy with time-gated detection: theoretical and experimental aspects. PLoS ONE 8, e54421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Manders, E. M. M., Verbeek, F. J. & Aten, J. A. Measurement of co-localization of objects in dual-colour confocal images. J. Microsc. 169, 375–382 (1993).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Novo Nordisk Foundation (grant NNF17OC0028176; D.S.) and NIGMS R35 GM118167 (O.D.W.). We thank S. Hell, C. Eggeling and E. Sezgin for initial assistance with 3D STED and J. T. Groves, R. Dimova, D. Wustner and K. Madsen for helpful discussions on the project. We are also thankful to P. Lappalainen for the GFP–actin plasmid, P. J. Verveer for the EGFR–SNAP plasmid, N. Schmitt for HL-1 cells and K. L. Madsen for COS-7 cells. We thank T. Izard for pcDNA4-GFP-vinculin (Addgene plasmid 107153). We are grateful to D. Gadella for mNeonGreen–Rab7 (Addgene plasmid 129603), 4xmts-mNeonGreen (Addgene plasmid 98876) and lysozyme(1–31)-KDEL–mNeonGreen (Addgene plasmid 137804). We thank Y. Schwab and A. Kolovou for their help and expertise in cryo-EM.

Author information

Authors and Affiliations

Authors

Contributions

D.S. conceived the strategy and was responsible for project management and supervision. L.L. developed and validated the principle of 3D imaging with help from C.G.S. and G.K. G.K. developed and validated the cell compression assay with help from R.M.B. and O.D.W. G.K. and D.S. designed the experiments. G.K. collected data with help from L.L. G.K. analyzed data with help from L.L. M.U. performed all theoretical calculations. C.G.S. was the principal software developer in the project. I.V. performed thin-section cryo-EM experiments. Y.Z. made constructs and provided help with Piezo1 experiments. C.K. developed visualization of color-coded amino acid residues onto the receptor structure in Fig. 2a,b, A.B. and E.K. contributed to conceptualization and preliminary experiments. E.D. provided help with STED microscopy. D.S. and G.K. wrote the main text. G.K. prepared all main and Supplementary figures with the help of L.L. for several Supplementary figures. M.U. and L.L. contributed to the Supplementary Information. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Mark Uline or Dimitrios Stamou.

Ethics declarations

Competing interests

D.S. is the founder of Atomos Biotech. The other authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Paolo Annibale, Juan Vanegas, and Wade Zeno 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 2D imaging reveals spatial variations in receptor intensity that cannot be interpreted without knowledge of membrane topography.

(a) Cartoons represent slices of membranes imaged by Confocal or total internal fluorescence microscopy (TIRF) microscopy at the cell equator and at the plasma membrane. (b-f) Confocal (b-d) or TIRF (e-f) images of the PM of HEK293 labeled with CellMask and/or the β1AR. All images are recorded at the basolateral membrane except for (B, left) which is at the cell equator. The heterogenous spatial distribution of intensity (domains of high/low intensity) cannot be interpreted as variations in density without prior knowledge of membrane topography. Color scales show relative intensity that is the smallest intensity present in an image is set to black. Data is from nR = 3. Scalebars: (a-b) 5 µm; (c-f) 500 nm.

Extended Data Fig. 2 Quantitative measurement of protein density and diffusion requires imaging of the plasma membrane in 3D to correct topography-induced artifacts that mimic the appearance of domains in 2D projections.

(a) Experimentally obtained 3D membrane topography map of the adherent plasma membrane of a HEK293 cell labelled using CellMask. (b) Illustration of the confocal excitation volume approximated by an ellipsoid (blue) and the tangent plane at a given point of the membrane (green). The membrane tilt angle θ (angle between the tangent plane and the imaging plane) varies between 0° – 90°. (c) The cross-sectional area between the plane and the ellipsoid (approximated by a cylinder for simplicity) scales with sec θ and serves as an approximation of the tilted membrane area. (d) An experimentally obtained 3D membrane topography map to which we computationally assigned a uniform receptor density. (e) Because of the variations in membrane topography, the 2D density projection of the uniform surface in (d) erroneously suggests the existence of GPCR domains. (f) For similar reasons, the spatially homogeneous diffusion in the 3D surface shown in (d) will erroneously appear to be heterogeneous if projected in 2D. (g) Schematic illustrations. The 2D projection of a uniformly labelled membrane of varying topography (left), can erroneously produce the appearance of 2D domains that cannot a priori be distinguished from bona fide variations in membrane label density (right).

Source data

Extended Data Fig. 3 Generation of high-accuracy topography maps of plasma membranes of living cells.

(a, b) Fluorescence confocal image of HEK293 cells over-expressing SNAP-β1AR (a) and CellMask (b). Data β1AR is representative for nR = 4 and CellMask nR = 3 replicates. (c, d) Illustration of an XZY-stack (dz/dx/dy = 30 nm) of the highlighted area in (a) and (b). (e, f) Extracted intensity Z-profiles of linescans highlighted in (c) and (d) and their corresponding Gaussian fits overlaid. The axial position of the Gaussian peak corresponds to the Z position of the membrane, whereas the amplitude of the Gaussian peak is proportional to protein or Cellmask density. (g) Topography map of the area shown in (b) reconstructed from the Z positions obtained from the Gaussian fitting. Color scale represents membrane height in nm. (h) Local error weighted quadric fit for a 3 × 3 pixel, 90 nm x 90 nm, area using Eq. 1 (see Methods). (i) Recovered denoised topography map after quadric fitting (same area as in (g)). Color scale is same as for (g). (j) Localization precision of the Z positions calculated as the error weighed standard error of the mean for a 3 × 3 pixel, 90 nm × 90 nm, area of the denoised topography maps. (k) Topography map from (i) overlaid with mean membrane curvature.

Source data

Extended Data Fig. 4 Validation of recovered membrane topography with RICM.

(a) Illustration of the overlay of the reconstructed 3D topography map with the corresponding reflection interference contrast microscopy (RICM) image. (b) Recovered membrane height plotted against RICM intensity for a representative cell. As predicted analytically by theoretical models, membrane height and RICM intensity follow a co-sinusoidal relationship (see Methods). The correlation is fitted with a cosine function that describes the data with an R2 = 0.999. Data is binned using an error weighted rolling average (10 ± 10 nm) with error bars showing the s.e.m. Data is from N = 59,280 data points, n = 20 cells from n = 4 experiments.

Source data

Extended Data Fig. 5 Assessment of the membrane stability over time imaged with confocal.

(a) Individual XZ images of a HEK293 acquired on the same location at t = 1 s, 20 s, 40 s, and 60 s. Initial visual assessment shows no major membrane displacements occur over t = 60 s. Data is from nR = 3. Scale bar, 1.5 μm. (b) Map of recovered Z position for the same XZ-slice imaged over t = 60 s. (c) To assess membrane movement, a rolling standard deviation is calculated for each X position over a 6 s time window showing the average movement of the membrane within the time frame needed for stable imaging. This allows us to image the spatial distribution of temporal nanoscopic displacements across the plasma membrane. (d) Histograms of rolling standard deviation (grey) as calculated in (c) and accuracy of retrieving Z position of the membrane after quadric fitting (pink) (Extended Data Fig. 7e). (e) Median membrane displacement, that is median of the rolling standard deviation, as a function of the applied time window. Pink dashed line represents the median accuracy in retrieving the Z position of the membrane after quadric fitting. Error bars show s.d.

Source data

Extended Data Fig. 6 Validation of β1AR density by ratio-metric imaging with a cell membrane probe.

(a) Schematic of experimental approach. HEK293 cells express SNAP-β1AR and are labelled with SS488, while the membrane is labelled with CellMask DeepRed. (b) β1AR density normalized by recovered membrane surface area versus β1AR density normalized by CellMask intensity. The dashed purple line is a linear fit to the data. Data is shown as error-weighted bins with equal number of data points per bin. Error bars, s.e.m for y-axis and s.d. for x-axis. (c) Normalized β1AR density versus mean curvature. Recovery of normalized β1AR density by surface normalization (green) or by CellMask intensity (purple) results in the same density-curvature correlation. Data is binned using an error-weighted rolling average (0.1 ± 0.1 μm−1) with error bars showing s.e.m. Data is from n = 7 cells from n = 1 experiment. (d-e) 2D projection of topography-corrected and normalized density of CellMask and β1AR for the same region at the PM. CellMask density is uniform at the PM (d), whereas β1AR forms domains (e). Scalebar, 500 nm.

Source data

Extended Data Fig. 7 Neither high-density nor low-density actin zones associate with domains of well-defined topography or mean curvature.

(a) Actin intensity at the plasma membrane overlaid with boundaries of high- and low-density zones of actin. Scale bar, 500 nm. (b) Density map of normalized β1AR overlaid with the actin boundaries from (a). Domain 1 in (a) and (b) indicates a low-actin-density region that contains both β1AR-enriched and -depleted domains. (c) Colocalization analysis of high- and low-density zones of actin with β1AR-enriched and -depleted domains. The colocalization percentages are compared to those of randomized actin zones. Quantitative correlations between high-/low-density actin regions and β1AR density patterns were either statistically nonsignificant or had low significance (P = 0.06 n.s., P = 0.03). P values are calculated by a two-sided paired t test. Data are the mean ± s.d. for nC = 23, nR = 2. (d) Membrane topography overlaid with the actin boundaries from (a). High- and low actin density zones are not preferentially colocalizing with membrane peaks or valleys. (e) Mean curvature overlaid with actin boundaries from (a). There is no preferential overlap of high- and low actin zones with positive or negative curvatures. Overlays are representative for n = 23 cells and n = 2 experiments.

Source data

Extended Data Fig. 8 Activation by agonist induces clathrin redistribution from receptor-depleted domains to both receptor-enriched and -depleted domains.

(a) Representative XY micrographs of SNAP-β1AR (magenta) and pmKate2-clathrin (green) and a merge in HEK293 cells. (b-c) XZ micrograph of β1AR, clathrin and a merge. Clathrin colocalizes with depleted domains of β1AR (b), but not with β1AR-enriched domains in the apo state (c). (d-f) XZ micrographs of events of β1AR internalization via clathrin-mediated endocytosis (indicated by arrows) after addition of ISO. These events are removed in our image analysis pipeline for recovering membrane topography and GPCR density. For (b-f) β1AR is imaged in 3D STED (magenta), clathrin in confocal (green) and a merge is shown. (g-h) 2D projection of normalized clathrin (g) and β1AR density (h) in the apo state. Arrows indicate regions of clathrin colocalizing with β1AR-depleted and not with β1AR-enriched domains. (i-j) 2D projection of normalized clathrin (i) and β1AR density (j) after activation by ISO. Arrows indicate regions of clathrin colocalizing both with β1AR-enriched and -depleted domains, however β1AR-enriched and -depleted domains do not always colocalize with clathrin (arrow with asterisk). (k) Colocalization analysis of high-density clathrin regions with β1AR-enriched and -depleted domains before and after activation by isoproterenol (ISO). The colocalization percentages are compared to randomized clathrin zones. Under basal conditions, β1AR and clathrin were anticorrelated (P = 0.01), while after activation with the agonist ISO, clathrin colocalization with GPCR-enriched domains was not statistically significant (P = 0.65). P values are calculated by a two-sided paired t test. Data are mean ± s.d. for nC = 18, nR = 3. Scalebar, (a) 1 µm, (b-f) 200 nm, (g-j) 500 nm.

Source data

Extended Data Fig. 9 Changes in membrane topography, curvature and β1AR density after cell flattening with an agarose pad.

Change in height (a), mean membrane curvature (b) and β1AR density (c) calculated by subtracting the values after flattening from the values before flattening (Fig. 3c,e,g and d,f,h). (d) Quantification of absolute change in height after flattening with an agarose pad from data in (a). The red line indicates the average change in membrane height of 14.5 nm. (e) Comparison of histograms of mean curvature of unperturbed (blue) and flattened (orange) for a single, representative HEK293 cell. After flattening the width of the histogram is smaller compared to the unperturbed cells. (f) Histograms of normalized β1AR density before (blue) and after (orange) flattening of the same cell as in (e). Normalized density of compressed cells shifts towards unity and the width of the histogram decreases. Data is representative for n = 10 cells in n = 5 replicates.

Extended Data Fig. 10 Domains of H-Ras, Piezo1 and EGFR at the PM of live cells.

2D projection of H-Ras density (a), overlay of H-Ras density on super-resolved topography map (b) and mean curvature map (c) of membrane topography shown in (b). H-Ras-enriched domains are formed at negative shallow curvature (see also Fig. 5d). (d-f) 2D projection of Piezo1 density (d), overlay of Piezo1 density on super-resolved topography map (e) and mean curvature map (f). Piezo1-enriched domains have high contrast and strongly couple to positive shallow curvature (see Fig. 5e). (g-i) 2D projection of EGFR density (g), overlay of EGFR density on super-resolved topography map (h) and mean curvature map (i). EGFR density variations couple to positive shallow curvature and are similar to those of β1AR and other studied GPCRs (see Supplementary Fig. 23). Black arrows indicate examples of H-Ras-, Piezo1- and EGFR-enriched domains. Scalebar (a,d,g), 500 nm.

Supplementary information

Supplementary Information

Supplementary Figs. 1–24 and Note.

Reporting Summary

Supplementary Video 1

xzy scanning for the reconstruction of superresolved membrane topography. The movie shows the principle of reconstructing membrane topography. The xz frames are scanned along the y axis, and the blue line represents the recovered membrane height for each frame

Supplementary Video 2

Overlay of recovered z position on an xzy stack imaged with confocal and 3D STED microscopy. The xz frames are shown while scanning in the y direction for confocal (top) and 3D STED (bottom) imaging modes. The confocal xz frames are overlaid with the recovered z position of the membrane in confocal (yellow), whereas the 3D STED xz frames are overlaid with the recovered z position in 3D STED (blue) and confocal (yellow). The recovered z position accurately follows the variations of the PM for both confocal and 3D STED imaging modes.

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. 5

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

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. 8

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

Kockelkoren, G., Lauritsen, L., Shuttle, C.G. et al. Molecular mechanism of GPCR spatial organization at the plasma membrane. Nat Chem Biol 20, 142–150 (2024). https://doi.org/10.1038/s41589-023-01385-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01385-4

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