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:

Nanobody-based sensors reveal a high proportion of mGlu heterodimers in the brain

Abstract

Membrane proteins, including ion channels, receptors and transporters, are often composed of multiple subunits and can form large complexes. Their specific composition in native tissues is difficult to determine and remains largely unknown. In this study, we developed a method for determining the subunit composition of endogenous cell surface protein complexes from isolated native tissues. Our method relies on nanobody-based sensors, which enable proximity detection between subunits in time-resolved Förster resonance energy transfer (FRET) measurements. Additionally, given conformation-specific nanobodies, the activation of these complexes can be recorded in native brain tissue. Applied to the metabotropic glutamate receptors in different brain regions, this approach revealed the clear existence of functional metabotropic glutamate (mGlu)2–mGlu4 heterodimers in addition to mGlu2 and mGlu4 homodimers. Strikingly, the mGlu4 subunits appear to be mainly heterodimers in the brain. Overall, these versatile biosensors can determine the presence and activity of endogenous membrane proteins in native tissues with high fidelity and convenience.

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: Nanobody-based sensors to detect the expression and activation of endogenous membrane proteins.
Fig. 2: A nanobody-based biosensor to detect the expression and activation of the mGlu2 receptor in both transfected cells and dissociated brain tissues.
Fig. 3: Relative quantification of endogenous mGlu2 and mGlu4 homodimers by FRET.
Fig. 4: Relative quantification of the mGlu2–mGlu4 heterodimer by FRET.
Fig. 5: A conformational biosensor detects activation of the endogenous mGlu2–mGlu4 heterodimer.
Fig. 6: mGlu2–mGlu4 heterodimers are predominant over mGlu4 homodimers outside the cerebellum.

Similar content being viewed by others

Data availability

Materials and protocols are available on request. Source data are provided with this paper.

References

  1. Kniazeff, J., Prezeau, L., Rondard, P., Pin, J. P. & Goudet, C. Dimers and beyond: the functional puzzles of class C GPCRs. Pharmacol. Ther. 130, 9–25 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Lemmon, M. A. & Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 141, 1117–1134 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Paoletti, P., Bellone, C. & Zhou, Q. NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat. Rev. Neurosci. 14, 383–400 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Albizu, L. et al. Time-resolved FRET between GPCR ligands reveals oligomers in native tissues. Nat. Chem. Biol. 6, 587–594 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Rosenbaum, M. I., Clemmensen, L. S., Bredt, D. S., Bettler, B. & Stromgaard, K. Targeting receptor complexes: a new dimension in drug discovery. Nat. Rev. Drug Discov. 19, 884–901 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Schwenk, J. et al. Modular composition and dynamics of native GABAB receptors identified by high-resolution proteomics. Nat. Neurosci. 19, 233–242 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Yu, J. et al. Hippocampal AMPA receptor assemblies and mechanism of allosteric inhibition. Nature 594, 448–453 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Shi, Y. et al. A novel proximity assay for the detection of proteins and protein complexes: quantitation of HER1 and HER2 total protein expression and homodimerization in formalin-fixed, paraffin-embedded cell lines and breast cancer tissue. Diagn. Mol. Pathol. 18, 11–21 (2009).

    Article  PubMed  CAS  Google Scholar 

  9. Trifilieff, P. et al. Detection of antigen interactions ex vivo by proximity ligation assay: endogenous dopamine D2–adenosine A2A receptor complexes in the striatum. Biotechniques 51, 111–118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sebastianutto, I. et al. D1–mGlu5 heteromers mediate noncanonical dopamine signaling in Parkinson’s disease. J. Clin. Invest. 130, 1168–1184 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gaborit, N. et al. Time-resolved fluorescence resonance energy transfer (TR-FRET) to analyze the disruption of EGFR/HER2 dimers: a new method to evaluate the efficiency of targeted therapy using monoclonal antibodies. J. Biol. Chem. 286, 11337–11345 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zwier, J. M., Bazin, H., Lamarque, L. & Mathis, G. Luminescent lanthanide cryptates: from the bench to the bedside. Inorg. Chem. 53, 1854–1866 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Nevoltris, D. et al. Conformational nanobodies reveal tethered epidermal growth factor receptor involved in EGFR/ErbB2 predimers. ACS Nano 9, 1388–1399 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Mujic-Delic, A., de Wit, R. H., Verkaar, F. & Smit, M. J. GPCR-targeting nanobodies: attractive research tools, diagnostics, and therapeutics. Trends Pharmacol. Sci. 35, 247–255 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Scholler, P. et al. Allosteric nanobodies uncover a role of hippocampal mGlu2 receptor homodimers in contextual fear consolidation. Nat. Commun. 8, 1967 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Ma, Y. et al. Structure-guided discovery of a single-domain antibody agonist against human apelin receptor. Sci. Adv. 6, eaax7379 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Haubrich, J. et al. A nanobody activating metabotropic glutamate receptor 4 discriminates between homo- and heterodimers. Proc. Natl Acad. Sci. USA 118, e2105848118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Jahnichen, S. et al. CXCR4 nanobodies (VHH-based single variable domains) potently inhibit chemotaxis and HIV-1 replication and mobilize stem cells. Proc. Natl Acad. Sci. USA 107, 20565–20570 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. McMahon, C. et al. Synthetic nanobodies as angiotensin receptor blockers. Proc. Natl Acad. Sci. USA 117, 20284–20291 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Niswender, C. M. & Conn, P. J. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu. Rev. Pharmacol. Toxicol. 50, 295–322 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. El Moustaine, D. et al. Distinct roles of metabotropic glutamate receptor dimerization in agonist activation and G-protein coupling. Proc. Natl Acad. Sci. USA 109, 16342–16347 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Koehl, A. et al. Structural insights into the activation of metabotropic glutamate receptors. Nature 566, 79–84 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Levitz, J. et al. Mechanism of assembly and cooperativity of homomeric and heteromeric metabotropic glutamate receptors. Neuron 92, 143–159 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin, S. et al. Structures of Gi-bound metabotropic glutamate receptors mGlu2 and mGlu4. Nature 594, 583–588 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Seven, A. B. et al. G-protein activation by a metabotropic glutamate receptor. Nature 595, 450–454 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Doumazane, E. et al. A new approach to analyze cell surface protein complexes reveals specific heterodimeric metabotropic glutamate receptors. FASEB J. 25, 66–77 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Moreno Delgado, D. et al. Pharmacological evidence for a metabotropic glutamate receptor heterodimer in neuronal cells. eLife 6, e25233 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Xiang, Z. et al. Input-specific regulation of glutamatergic synaptic transmission in the medial prefrontal cortex by mGlu2/mGlu4 receptor heterodimers. Sci. Signal. 14, eabd2319 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yin, S. et al. Selective actions of novel allosteric modulators reveal functional heteromers of metabotropic glutamate receptors in the CNS. J. Neurosci. 34, 79–94 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee, J. et al. Defining the homo- and heterodimerization propensities of metabotropic glutamate receptors. Cell Rep. 31, 107891 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ferraguti, F. & Shigemoto, R. Metabotropic glutamate receptors. Cell Tissue Res. 326, 483–504 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Wright, R. A. et al. CNS distribution of metabotropic glutamate 2 and 3 receptors: transgenic mice and [3H]LY459477 autoradiography. Neuropharmacology 66, 89–98 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Doumazane, E. et al. Illuminating the activation mechanisms and allosteric properties of metabotropic glutamate receptors. Proc. Natl Acad. Sci. USA 110, E1416–E1425 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Scholler, P. et al. HTS-compatible FRET-based conformational sensors clarify membrane receptor activation. Nat. Chem. Biol. 13, 372–380 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Cao, A. M. et al. Allosteric modulators enhance agonist efficacy by increasing the residence time of a GPCR in the active state. Nat. Commun. 12, 5426 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, J. et al. Allosteric control of an asymmetric transduction in a G protein-coupled receptor heterodimer. eLife 6, e26985 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Asher, W. B. et al. Single-molecule FRET imaging of GPCR dimers in living cells. Nat. Methods 18, 397–405 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Moller, T. C. et al. Oligomerization of a G protein-coupled receptor in neurons controlled by its structural dynamics. Sci. Rep. 8, 10414 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Kinoshita, A. et al. Presynaptic localization of a metabotropic glutamate receptor, mGluR4a, in the cerebellar cortex: a light and electron microscope study in the rat. Neurosci. Lett. 207, 199–202 (1996).

    Article  CAS  PubMed  Google Scholar 

  40. Neki, A. et al. Metabotropic glutamate receptors mGluR2 and mGluR5 are expressed in two non-overlapping populations of Golgi cells in the rat cerebellum. Neuroscience 75, 815–826 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Chorev, D. S. & Robinson, C. V. The importance of the membrane for biophysical measurements. Nat. Chem. Biol. 16, 1285–1292 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Oliveira, S., Heukers, R., Sornkom, J., Kok, R. J. & van Bergen En Henegouwen, P. M. Targeting tumors with nanobodies for cancer imaging and therapy. J. Control. Release 172, 607–617 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Faklaris, O. et al. Multicolor time-resolved Forster resonance energy transfer microscopy reveals the impact of GPCR oligomerization on internalization processes. FASEB J. 29, 2235–2246 (2015).

    Article  CAS  PubMed  Google Scholar 

  44. Werthmann, R. C. et al. Symmetric signal transduction and negative allosteric modulation of heterodimeric mGlu1/5 receptors. Neuropharmacology 190, 108426 (2021).

    Article  CAS  PubMed  Google Scholar 

  45. Prezeau, L. et al. Functional crosstalk between GPCRs: with or without oligomerization. Curr. Opin. Pharmacol. 10, 6–13 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Grushevskyi, E. O. et al. Stepwise activation of a class C GPCR begins with millisecond dimer rearrangement. Proc. Natl Acad. Sci. USA 116, 10150–10155 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Marek, G. J. Metabotropic glutamate2/3 (mGlu2/3) receptors, schizophrenia and cognition. Eur. J. Pharmacol. 639, 81–90 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Marino, M. J. et al. Allosteric modulation of group III metabotropic glutamate receptor 4: a potential approach to Parkinson’s disease treatment. Proc. Natl Acad. Sci. USA 100, 13668–13673 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rosemond, E. et al. Molecular basis for the differential agonist affinities of group III metabotropic glutamate receptors. Mol. Pharmacol. 66, 834–842 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Niswender, C. M. et al. Context-dependent pharmacology exhibited by negative allosteric modulators of metabotropic glutamate receptor 7. Mol. Pharmacol. 77, 459–468 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Yokoi, M. et al. Impairment of hippocampal mossy fiber LTD in mice lacking mGluR2. Science 273, 645–647 (1996).

    Article  CAS  PubMed  Google Scholar 

  52. Pekhletski, R. et al. Impaired cerebellar synaptic plasticity and motor performance in mice lacking the mGluR4 subtype of metabotropic glutamate receptor. J. Neurosci. 16, 6364–6373 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kubala, M. H., Kovtun, O., Alexandrov, K. & Collins, B. M. Structural and thermodynamic analysis of the GFP:GFP–nanobody complex. Protein Sci. 19, 2389–2401 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lein, E. S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168–176 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Nevoltris and D. Meyer (IRCM) for screening nanobodies and preparing the Fc-DN42 construct, respectively. We thank the Arpège platform of the Institut de Génomique Fonctionnelle for providing facilities and technical support, the imaging facility Montpellier Ressources Imagerie (MRI), member of the national infrastructure France-BioImaging supported by the French National Research Agency (ANR-10-INBS-04, Investments for the Future), the animal facility RAM-iExplore from BioCampus and PerkinElmer Cisbio for providing reagents. We thank C. Goudet and T. Durroux (IGF) for providing the mGlu4-KO mice and for assistance with TR-FRET microscopy, respectively. J.M. and C.X. were supported by the Sino-French Cai Yuanpei program (grant nos. 201604490217 and 201304490188, respectively). P.R. and J.-P.P. were supported by the Centre National de la Recherche Scientifique (CNRS, PICS no. 07030, PRC no. 1403), the Institut National de la Santé et de la Recherche Médicale (INSERM, IRP BrainSignal), the Fondation pour la Recherche Médicale (equipe DEQ20170336747), the Eidos collaborative laboratory with PerkinElmer Cisbio, the Franco–Chinese Joint Scientific and Technological Commission (CoMix) from the French Embassy in China and the LabEx MAbImprove (grant NR-10-LABX-5301). P.R. and J.-P.P. were supported by the ANR (grants ANR-15-CE18-0020-01, ANR-20-CE18-0011-01 and ANR-20-CE44-0006-02). J.L. was supported by the Program of Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory, grant no. 2010A080813001), the Ministry of Science and Technology of China (grant no. 2018YFA0507003), the National Natural Science Foundation of China (grant nos. 81720108031, 31721002 and 81872945), the Program for Introducing Talents of Discipline to the Universities of the Ministry of Education (grant no. B08029), the Key Program of Natural Science Foundation of Hubei Province (grant no. 2019ACA128) and Wuhan (2019020701011481). P.C. was supported by the FUI of the French government (FUI, Cell2Lead project). J.G.-M. was supported by National Institutes of Health grants R01MH084894 and R01MH111940. The mouse pictures in Fig. 1 and Extended Data Fig. 3a were modified from Servier Medical Art (http://smart.servier.com/), licensed under a Creative Commons Attribution 3.0 generic license (https://creativecommons.org/licenses/by/3.0/). The mouse brain in Extended Data Fig. 3a is from Figdraw (https://www.figdraw.com/). Pictures of the microplate in Fig. 1 and Extended Data Fig. 3a are from PerkinElmer Cisbio.

Author information

Authors and Affiliations

Authors

Contributions

J.M. developed the FRET-based detectors and the FRET-based conformational change sensors and performed sensor experiments, IP1 assays, co-immunoprecipitation and immunoblotting. C.X. proposed the idea and set up the protocol for the nanobody-based sensors for the detection of mGlu2 expression and activation in dissociated brain cells and performed the DN1 and DN10 sensor experiments in HEK293 cells and dissociated brain cells and IP1 assays. P.-A.L. performed tissue immunofluorescence assays. S.R. designed and performed experiments with the FRET-based detectors and conformational change sensors. M.M. performed TR-FRET microscopy imaging, nanobody production, purification and labeling. R.Z. performed nanobody production, purification, labeling and co-immunoprecipitation. P.S. and E.B. developed the FRET-based conformational change biosensor for the mGlu2 homodimer in HEK293 cells. J.A.J.B. and J.L.M. provided brain samples and trained J.M. for brain sample preparation. J.G.-M. prepared brain samples for wild-type and mGlu2-KO mice. P.C. screened for anti-mGlu2 and anti-mGlu4 nanobodies and prepared the Fc versions. J.L., J.-P.P. and P.R. conceived experiments, supervised the work and wrote the manuscript.

Corresponding authors

Correspondence to Jianfeng Liu, Jean-Philippe Pin or Philippe Rondard.

Ethics declarations

Competing interests

P.R. and J.-P.P. are funded by PerkinElmer Cisbio through the collaborative laboratory Eidos. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Jonathan Javitch 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 DN1/DN10 FRET-based biosensor detects mGlu2 homodimers on transfected HEK-293 cells.

(a) TR-FRET signal measured on HEK-293 cells transiently transfected with mGlu2 receptors in the presence of donor nanobody DN1-Tb (7.5 nM) and LY379268 (1 μM), with or without acceptor nanobody DN10-d2 (15 nM). (b) Specific TR-FRET signal on HEK-293 cells transiently transfected with mGlu2 receptors in the presence of DN1-Tb (7.5 nM) with increasing concentrations of DN10-d2. For each concentration of acceptor nanobody, specific FRET signal was the difference between samples in the presence of LY379268 (1 μM) and LY341495 (10 μM). Data are mean ± SEM of three independent experiments performed in triplicates and normalized to the maximum. (c) TR-FRET signal measured on HEK-293 cells transiently transfected with indicated mGlu receptors or on mock cells with LY379268 (1 μM). In a and c, data are mean ± SEM of triplicate determinations from one representative out of three experiments.

Source data

Extended Data Fig. 2 TR-FRET confocal imaging on primary cultured hippocampal neurons.

DN10-Tb (80 nM) and DN1-d2 (120 nM) were applied on the primary cultured hippocampal neurons. Fluorescence signals of different channels, TR-FRET channel (left), donor channel (middle) or acceptor channel (right) were obtained under agonist (150 nM LY379268, top) or antagonist (1 μM LY341495, bottom). Scale bar: 20 μm.

Extended Data Fig. 3 DN1/DN10 biosensor measures endogenous mGlu2 on cells from brain regions.

(a) Diagram shows flow of the tissue samples experiments. (b) Schematic diagram of adult mouse brain removed from most of the cortex, in top view. Brain regions analyzed are olfactory bulb (blue), prefrontal cortex (red), striatum (green), hippocampus (purple), midbrain (orange) and cerebellum (gray). (c) Relative quantification of the endogenous mGlu2 receptor in the indicated brain regions in the same mouse using the indicate FRET-based biosensor in presence of 1 μM LY379268. Data are mean ± SEM of triplicate determinations from one representative out of three experiments.

Source data

Extended Data Fig. 4 DN1-d2 immunofluorescence staining of a saggital section in wild-type and mGlu2 knock out mouse brains.

The scale bars are indicated.

Extended Data Fig. 5 FRET-based detector mainly detects mGlu2 homodimer.

(a) TR-FRET signal measured on HEK-293 cells transiently transfected with mGlu2 receptors in the presence of DN1-Tb (25 nM) and LY341495 (10 μM), with or without DN1-d2 (25 nM). (b) Specific TR-FRET signal on HEK-293 cells transiently transfected with mGlu2 receptors in the presence of DN1-Tb (25 nM) and LY341495 (10 μM) with increasing concentrations of DN1-d2. For each concentration of acceptor nanobody, specific FRET signal was the difference between samples transfected with mGlu2 receptor or not. Data are mean ± SEM of three independent experiments performed in triplicates and normalized to the maximum. (c and d) TR-FRET intensity and cell surface expression were measured on various expression levels of the indicated cell samples. Experiments were performed with HEK-293 cells transiently transfected with SNAPmGlu2 which fused a SNAP tag at the N terminus of receptors (c) or transiently co-transfected with SNAPmGlu2C1 and mGlu2C2 or mGlu4C2 (d). The surface expression of receptors was measured as the specific Tb emission at 620 nm after labeling by substrate SNAP-Lumi4-Tb and exciting at 337 nm. TR-FRET intensity was measured in the presence of 10 μM LY341495. (e) Relative quantification of mGlu2 receptor in the indicated brain tissues from a same mouse in the presence of 10 μM LY341495. For a and c-e, Data are mean ± SEM of triplicate determinations from one representative out of three experiments.

Source data

Extended Data Fig. 6 Nanobody DN42 specifically targets mGlu4 receptors.

(a) Cartoon illustrating the principle of the TR-FRET binding assay. The receptor fused to a SNAP-tag (dark gray circled labeled ‘S’) is labeled with donor fluorescent dye Lumi4-Tb (blue circled ‘D’) while the nanobody DN42 (dark blue) bearing a 6xHis tag epitope at its C-terminus is labeled with 100 nM of anti-His antibody (bright blue) coupled to d2 fluorophores (red circled ‘A’). Binding of the nanobody to the receptor is then measured by TR-FRET. (b) Specific TR-FRET binding data obtained with the indicated mGlu receptor and either 100 nM Fc-DN42 or a control irrelevant nanobody (100 nM) in cells under indicated drug conditions, in the presence of the agonist 1 μM quisqualic acid (mGlu1 and 5) or 100 μM L-AP4 (mGlu4, 6, 7 and 8) or 100 nM LY379268 (mGlu2 and 3) or the antagonist LY341495 (10 μM). Data are mean ± SEM of triplicate determinations from one representative out of three experiments. (c) Saturation binding curves obtained with DN42 on SNAPmGlu4 receptors under indicated drug conditions, in the presence of the agonist L-AP4 (100 μM), or the antagonist LY341495 (10 μM). Data are mean ± SEM of three independent experiments performed in triplicates and normalized to the maximum. (d) DN42-d2 immunofluorescence staining of a sagittal section in wild-type and mGlu4 KO mouse brains. The scale bars are indicated.

Source data

Extended Data Fig. 7 FRET-based detector mainly detects mGlu4 homodimer.

(a) TR-FRET signal measured on HEK-293 cells transiently transfected with mGlu4 receptors in the presence of DN42-Tb (1.6 nM) and LY341495 (10 μM), with or without DN42-d2 (1.6 nM). (b) Specific TR-FRET signal on HEK-293 cells transiently transfected with mGlu4 receptors in the presence of DN42-Tb (1.6 nM) and LY341495 (10 μM), with increasing concentrations of DN42-d2. For each concentration of acceptor nanobody, specific FRET signal was the difference between samples transfected with mGlu4 receptors or not. Data are mean ± SEM of three independent experiments performed in triplicates and normalized to the maximum. (c and d) TR-FRET intensity and cell surface expression were measured on various expression levels of the indicated cell samples. Experiments were performed with HEK-293 cells transiently transfected with SNAPmGlu4 which fused a SNAP tag at the N terminus of receptors (c) or transiently co-transfected with SNAPmGlu4C2 and mGlu4C1 or mGlu2C1 (d). The surface expression of receptors was measured as the specific Tb emission at 620 nm after labeling by substrate SNAP-Lumi4-Tb and exciting at 337 nm. TR-FRET intensity was measured in the presence of 10 μM LY341495. (e) Relative quantification of mGlu4 receptor in the indicated brain tissues from a same mouse in the presence of 10 μM LY341495. For a and c-e, Data are mean ± SEM of triplicate determinations from one representative out of three experiments.

Source data

Extended Data Fig. 8 FRET-based detector only detects mGlu2-4 heteromer.

(a) TR-FRET signal measured on HEK-293 cells transiently expressing mGlu2C1-4C2 heterodimer in the presence of DN42-Tb (1.6 nM) and LY341495 (10 μM), with or without DN1-d2 (25 nM). (b) Specific TR-FRET signal on HEK-293 cells transiently expressing mGlu2C1-4C2 heterodimer in the presence of DN42-Tb (1.6 nM) and LY341495 (10 μM) with increasing concentrations of DN1-d2. For each concentration of acceptor nanobody, specific FRET signal was the difference between samples expressing mGlu2C1-4C2 heterodimer or not. Data are mean ± SEM of three independent experiments performed in triplicates and normalized to the maximum. (c) TR-FRET intensity and cell surface expression were measured for various expression levels of the indicated cell samples. Experiments were performed with HEK-293 cells transiently co-transfected with SNAPmGlu2C1 and mGlu4C2 (red) or SNAPmGlu2C1 and mGlu2C2 (blue). The surface expression of receptors was measured as the specific Tb emission at 620 nm after labeling by substrate SNAP-Lumi4-Tb and exciting at 337 nm. TR-FRET intensity was measured in the presence of 10 μM LY341495. (d) TR-FRET signal measured on HEK-293 cells transiently transfected with indicated the mGlu receptors or on mock cells with LY341495 (10 μM). (e and g) Relative quantification of mGlu2-4 heterodimer in the indicated brain tissues in the presence of 10 μM LY341495. (f) mGlu2 and mGlu4 are co-immunoprecipitated from mouse olfactory bulb. FcDN42 was used as probe antibody in pull down experiments. FcGFP was used as a negative control (IgG control). Results are one representative out of three independent experiments performed. For a, c-e and g, Data are mean ± SEM of triplicate determinations from one representative out of three experiments.

Source data

Extended Data Fig. 9 Nanobody DN10 allows to develop a FRET-based conformational biosensor for mGlu2-4 heterodimer.

(a) Schematic representation of the TR-FRET binding assay for the mGlu2-4 controlled heterodimer. The receptor fused to a SNAP-tag (dark gray circled labeled ‘S’) at the N-terminus of mGlu4 subunit is labeled with donor fluorescent dye Lumi4-Tb (blue circled ‘D’), while the nanobody DN10 (purple) coupled to a d2 fluorophore (red circled ‘A’). Binding of the nanobody to the receptor is then measured by TR-FRET. (b) Saturation binding experiments with DN10-d2 on the cell surface mGlu2-4 controlled heterodimer using the constructs mGlu2C1 and SNAPmGlu4C2 transiently co-transfected in HEK-293 cells. Binding of DN10-d2 to the receptor was measured by TR-FRET under the indicated conditions, buffer (gray, n = 5) or in the presence of the mGlu2 agonist LY379268 (1 μM, blue, n = 5), the mGlu4 agonist L-AP4 (100 μM, red, n = 3), or the antagonist LY341495 (10 μM, black, n = 5). Data are mean ± SEM of at least three individual experiments each performed in triplicates and normalized to the maximum. (c and d) Dose-dependent effects of the indicated ligands on TR-FRET signal between SNAP-tag labeled with donor fluorescent dye Lumi4-Tb (blue circled ‘D’) and 25 nM DN10-d2. Signals were measured on HEK-293 cells transiently co-transfected with mGlu2C1 and SNAPmGlu4C2 (c, n = 3) or co-transfected with mGlu2C1 and SNAPmGlu2C2 (d, n = 4). Data are mean ± SEM of at least three independent experiments performed in triplicate and normalized to the response of LY379268. (e) The FRET potencies (pEC50) in the indicated conditions. Data are mean ± SEM from HEK-293 cells transiently co-transfected with mGlu2C1 and SNAPmGlu4C2 (c, n = 3) or with mGlu2C1 and SNAPmGlu2C2 (d, n = 4). One-way ANOVA with Tukey’s multiple comparisons test (mGlu2 under the LY379268 is not included), with **P = 0.0041. (f) TR-FRET signal measured on HEK-293 cells transiently transfected with mGlu2C1 and mGlu4C2 or on mock cells in presence of DN42-Tb (1.6 nM) and DN10-d2 (25 nM) with 1 μM LY379268 (LY37) or 10 μM LY341495 (LY34). (g) TR-FRET signal measured on HEK-293 cells transiently transfected with the indicated mGlu receptors or on mock cells with LY379268 (1 μM). For f - g, Data are mean ± SEM of triplicate determinations from one representative out of three experiments.

Source data

Extended Data Fig. 10 IP1 accumulation of mGlu2-4 heterodimer shows its specific pharmacological fingerprints.

(a and b) Dose-dependent effects of the indicated ligands on IP1 accumulation. Signals were measured on HEK-293 cells transiently co-transfected with mGlu2C1 and mGlu4C2 (a) or with mGlu2 and mGlu4 (b) and EAAC1 and the chimeric G protein Gqi9. Data are mean ± SEM of three independent experiments performed in triplicate and normalized to the response of LY379268. (c) IP1 accumulation potencies (pEC50) of the indicated drug on the receptor conditions in a and b. Data are mean ± SEM of three independent experiments. Two-way ANOVA with Tukey’s multiple comparisons test, with ***P < 0.001 and *P ≤ 0.05. (d) TR-FRET potencies (pEC50) for the indicated ligands on the conformational sensor with the constructs mGlu2C1 and mGlu4C2, or when both wild-type mGlu2 and mGlu4 subunits are co-transfected. Data are presented as the mean ± SEM of four independent experiments. Two-way ANOVA followed by a Tukey’s post-hoc test, with ***P < 0.001, **P ≤ 0.01, and ns P > 0.05. (e) TR-FRET potencies (pEC50) of the ligands on the conformational sensor measured in dissociated olfactory bulb cells. Data are presented as the mean ± SEM of four independent experiments. One-way ANOVA followed by a Tukey’s post-hoc test, with ****P ≤ 0.0001 and ***P ≤ 0.001. (f) Correlation between the potencies (pEC50) determined with the indicated agonist conditions by the heterodimer conformational sensor and IP1 assay. Data are mean ± SEM of at least three independent experiments (TR-FRET of olfactory bulb (n = 4), mGlu2 + mGlu4 (n = 3) and mGlu2C1 + mGlu4C2 (n = 3) and IP1 accumulation of mGlu2C1 + mGlu4C2 (n = 3)). LY37 means LY379268. All exact P values are indicated in the panels c-e.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–3

Reporting Summary

Supplementary Data 1

Source data for Supplementary Information.

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 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. 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.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meng, J., Xu, C., Lafon, PA. et al. Nanobody-based sensors reveal a high proportion of mGlu heterodimers in the brain. Nat Chem Biol 18, 894–903 (2022). https://doi.org/10.1038/s41589-022-01050-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01050-2

This article is cited by

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