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:

Chemico-genetic discovery of astrocytic control of inhibition in vivo

Abstract

Perisynaptic astrocytic processes are an integral part of central nervous system synapses1,2; however, the molecular mechanisms that govern astrocyte–synapse adhesions and how astrocyte contacts control synapse formation and function are largely unknown. Here we use an in vivo chemico-genetic approach that applies a cell-surface fragment complementation strategy, Split-TurboID, and identify a proteome that is enriched at astrocyte–neuron junctions in vivo, which includes neuronal cell adhesion molecule (NRCAM). We find that NRCAM is expressed in cortical astrocytes, localizes to perisynaptic contacts and is required to restrict neuropil infiltration by astrocytic processes. Furthermore, we show that astrocytic NRCAM interacts transcellularly with neuronal NRCAM coupled to gephyrin at inhibitory postsynapses. Depletion of astrocytic NRCAM reduces numbers of inhibitory synapses without altering glutamatergic synaptic density. Moreover, loss of astrocytic NRCAM markedly decreases inhibitory synaptic function, with minor effects on excitation. Thus, our results present a proteomic framework for how astrocytes interface with neurons and reveal how astrocytes control GABAergic synapse formation and function.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of the astrocyte–neuron synaptic cleft proteome using in vivo Split-TurboID.
Fig. 2: The astrocyte–neuron synaptic cleft proteome.
Fig. 3: NRCAM controls astrocyte-neuron contacts in vivo.
Fig. 4: Astrocytic NRCAM controls inhibitory synaptic organization and function.

Similar content being viewed by others

Data availability

Proteomics data are available in the MassIVE database under accession MSV000085821. The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Yu, X., Nagai, J. & Khakh, B. S. Improved tools to study astrocytes. Nat. Rev. Neurosci. 21, 121–138 (2020).

    CAS  PubMed  Google Scholar 

  2. Lanjakornsiripan, D. et al. Layer-specific morphological and molecular differences in neocortical astrocytes and their dependence on neuronal layers. Nat. Commun. 9, 1623 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  3. Araque, A., Parpura, V., Sanzgiri, R. P. & Haydon, P. G. Tripartite synapses: glia, the unacknowledged partner. Trends Neurosci. 22, 208–215 (1999).

    CAS  PubMed  Google Scholar 

  4. Khakh, B. S. & Sofroniew, M. V. Diversity of astrocyte functions and phenotypes in neural circuits. Nat. Neurosci. 18, 942–952 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Ma, Z., Stork, T., Bergles, D. E. & Freeman, M. R. Neuromodulators signal through astrocytes to alter neural circuit activity and behaviour. Nature 539, 428–432 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Papouin, T., Dunphy, J., Tolman, M., Foley, J. C. & Haydon, P. G. Astrocytic control of synaptic function. Phil. Trans. R. Soc. Lond. B 372, 20160154 (2017).

    Google Scholar 

  7. Panatier, A. et al. Astrocytes are endogenous regulators of basal transmission at central synapses. Cell 146, 785–798 (2011).

    CAS  PubMed  Google Scholar 

  8. Araque, A. et al. Gliotransmitters travel in time and space. Neuron 81, 728–739 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Stogsdill, J. A. et al. Astrocytic neuroligins control astrocyte morphogenesis and synaptogenesis. Nature 551, 192–197 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  10. Stork, T., Sheehan, A., Tasdemir-Yilmaz, O. E. & Freeman, M. R. Neuron–glia interactions through the Heartless FGF receptor signaling pathway mediate morphogenesis of Drosophila astrocytes. Neuron 83, 388–403 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Sloan, S. A. & Barres, B. A. Mechanisms of astrocyte development and their contributions to neurodevelopmental disorders. Curr. Opin. Neurobiol. 27, 75–81 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Allen, N. J. & Lyons, D. A. Glia as architects of central nervous system formation and function. Science 362, 181–185 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Branon, T. C. et al. Efficient proximity labeling in living cells and organisms with TurboID. Nat. Biotechnol. 36, 880–887 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Schopp, I. M. et al. Split-BioID a conditional proteomics approach to monitor the composition of spatiotemporally defined protein complexes. Nat. Commun. 8, 15690 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. De Munter, S. et al. Split-BioID: a proximity biotinylation assay for dimerization-dependent protein interactions. FEBS Lett. 591, 415–424 (2017).

    PubMed  Google Scholar 

  16. Kinoshita, N. et al. Genetically encoded fluorescent indicator GRAPHIC delineates intercellular connections. iScience 15, 28–38 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lee, Y., Messing, A., Su, M. & Brenner, M. GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 56, 481–493 (2008).

    PubMed  Google Scholar 

  18. Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20, 1172–1179 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Uezu, A. et al. Identification of an elaborate complex mediating postsynaptic inhibition. Science 353, 1123–1129 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    CAS  PubMed  Google Scholar 

  22. Sakers, K. & Eroglu, C. Control of neural development and function by glial neuroligins. Curr. Opin. Neurobiol. 57, 163–170 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Incontro, S., Asensio, C. S., Edwards, R. H. & Nicoll, R. A. Efficient, complete deletion of synaptic proteins using CRISPR. Neuron 83, 1051–1057 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Custer, A. W. et al. The role of the ankyrin-binding protein NrCAM in node of Ranvier formation. J. Neurosci. 23, 10032–10039 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Feinberg, K. et al. A glial signal consisting of gliomedin and NrCAM clusters axonal Na+ channels during the formation of nodes of Ranvier. Neuron 65, 490–502 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Demyanenko, G. P. et al. Neural cell adhesion molecule NrCAM regulates Semaphorin 3F-induced dendritic spine remodeling. J. Neurosci. 34, 11274–11287 (2014).

    PubMed  PubMed Central  Google Scholar 

  27. Mohan, V. et al. Temporal regulation of dendritic spines through NrCAM-Semaphorin3F receptor signaling in developing cortical pyramidal neurons. Cereb. Cortex 29, 963–977 (2019).

    PubMed  Google Scholar 

  28. Mauro, V. P., Krushel, L. A., Cunningham, B. A. & Edelman, G. M. Homophilic and heterophilic binding activities of Nr-CAM, a nervous system cell adhesion molecule. J. Cell Biol. 119, 191–202 (1992).

    CAS  PubMed  Google Scholar 

  29. Derouiche, A., Anlauf, E., Aumann, G., Mühlstädt, B. & Lavialle, M. Anatomical aspects of glia-synapse interaction: the perisynaptic glial sheath consists of a specialized astrocyte compartment. J. Physiol. Paris 96, 177–182 (2002).

    CAS  PubMed  Google Scholar 

  30. Lavialle, M. et al. Structural plasticity of perisynaptic astrocyte processes involves ezrin and metabotropic glutamate receptors. Proc. Natl Acad. Sci. USA 108, 12915–12919 (2011).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Scheiffele, P., Fan, J., Choih, J., Fetter, R. & Serafini, T. Neuroligin expressed in nonneuronal cells triggers presynaptic development in contacting axons. Cell 101, 657–669 (2000).

    CAS  PubMed  Google Scholar 

  32. Graf, E. R., Zhang, X., Jin, S. X., Linhoff, M. W. & Craig, A. M. Neurexins induce differentiation of GABA and glutamate postsynaptic specializations via neuroligins. Cell 119, 1013–1026 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Chih, B., Gollan, L. & Scheiffele, P. Alternative splicing controls selective trans-synaptic interactions of the neuroligin–neurexin complex. Neuron 51, 171–178 (2006).

    CAS  PubMed  Google Scholar 

  34. Tremblay, R., Lee, S. & Rudy, B. GABAergic interneurons in the neocortex: from cellular properties to circuits. Neuron 91, 260–292 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Miles, R., Tóth, K., Gulyás, A. I., Hájos, N. & Freund, T. F. Differences between somatic and dendritic inhibition in the hippocampus. Neuron 16, 815–823 (1996).

    CAS  PubMed  Google Scholar 

  36. Wierenga, C. J. & Wadman, W. J. Miniature inhibitory postsynaptic currents in CA1 pyramidal neurons after kindling epileptogenesis. J. Neurophysiol. 82, 1352–1362 (1999).

    CAS  PubMed  Google Scholar 

  37. Martell, J. D. et al. A split horseradish peroxidase for the detection of intercellular protein-protein interactions and sensitive visualization of synapses. Nat. Biotechnol. 34, 774–780 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Loh, K. H. et al. Proteomic analysis of unbounded cellular compartments: synaptic clefts. Cell 166, 1295–1307 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Cijsouw, T. et al. Mapping the proteome of the synaptic cleft through proximity labeling reveals new cleft proteins. Proteomes 6, E48 (2018).

    Google Scholar 

  40. Li, J. et al. Cell-surface proteomic profiling in the fly brain uncovers wiring regulators. Cell 180, 373–386 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Cho, K. F. et al. Split-TurboID enables contact-dependent proximity labeling in cells. Proc. Natl Acad. Sci. USA 117, 12143–12154 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Elmariah, S. B., Oh, E. J., Hughes, E. G. & Balice-Gordon, R. J. Astrocytes regulate inhibitory synapse formation via Trk-mediated modulation of postsynaptic GABAA receptors. J. Neurosci. 25, 3638–3650 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Hughes, E. G., Elmariah, S. B. & Balice-Gordon, R. J. Astrocyte secreted proteins selectively increase hippocampal GABAergic axon length, branching, and synaptogenesis. Mol. Cell. Neurosci. 43, 136–145 (2010).

    CAS  PubMed  Google Scholar 

  44. Turrigiano, G. G., Leslie, K. R., Desai, N. S., Rutherford, L. C. & Nelson, S. B. Activity-dependent scaling of quantal amplitude in neocortical neurons. Nature 391, 892–896 (1998).

    ADS  CAS  PubMed  Google Scholar 

  45. O’Brien, R. J. et al. Activity-dependent modulation of synaptic AMPA receptor accumulation. Neuron 21, 1067–1078 (1998).

    PubMed  Google Scholar 

  46. Spence, E. F. et al. In vivo proximity proteomics of nascent synapses reveals a novel regulator of cytoskeleton-mediated synaptic maturation. Nat. Commun. 10, 386 (2019).

    ADS  PubMed  PubMed Central  Google Scholar 

  47. Shin, J. H., Yue, Y. & Duan, D. Recombinant adeno-associated viral vector production and purification. Methods Mol. Biol. 798, 267–284 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Takano, T. et al. LMTK1 regulates dendritic formation by regulating movement of Rab11A-positive endosomes. Mol. Biol. Cell 25, 1755–1768 (2014).

    PubMed  PubMed Central  Google Scholar 

  49. Takano, T. et al. Discovery of long-range inhibitory signaling to ensure single axon formation. Nat. Commun. 8, 33 (2017).

    ADS  PubMed  PubMed Central  Google Scholar 

  50. Ippolito, D. M, Eroglu, C. Quantifying synapses: an immunocytochemistry-based assay to quantify synapse number. J. Vis. Exp. 16, 2270 (2010).

    Google Scholar 

  51. Dani, A., Huang, B., Bergan, J., Dulac, C. & Zhuang, X. Superresolution imaging of chemical synapses in the brain. Neuron 68, 843–856 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Katsura for modifying the promoter of the surface and split TurboID plasmids for HEK 293T cell expression, and B. Duncan for technical support. This work was supported by Brain initiative RO1DA047258 from NIH (S.H.S. and C.E.), R01MH113280 from NIH (P.F.M.), Kahn Neurotechnology Award (S.H.S. and C.E.), a Grant-in-Aid for JSPS Fellows (PD) 20153173 from the Japan Society for the Promotion of Science (T.T.), The Uehara memorial Foundation (T.T.), and National Institute of Mental Health Fellowship F30MH117851 (J.L.C).

Author information

Authors and Affiliations

Authors

Contributions

T.T., C.E. and S.H.S. designed the study. T.T., J.T.W., A.P., C.E. and S.H.S. wrote the manuscript. T.T., J.T.W., A.U. and E.J.S. performed in vivo BioID-proteomics analysis. T.T., J.T.W., J.L.C., T.S. and P.F.M. produced the constructs. T.T., J.T.W. and K.T.B. performed imaging analysis and the morphological analysis of the astrocytes. A.P. performed electrophysiological analysis. T.T. and K.T.B. performed the biological experiments. All authors discussed the results and commented on the manuscript text.

Corresponding authors

Correspondence to Tetsuya Takano, Cagla Eroglu or Scott H. Soderling.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Thomas Biederer, Peter Scheiffele and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 The reconstituted activity of Split-TurboID in neurons and astrocytes in vitro.

a, Schematics of constructs tested. b, Immunoblot analysis of construct expression and biotinylation activity. c, Schematic of neuron-astrocyte mixed-culture assay for Split-TurboID with cell-type-specific AAVs in vitro. d, Cultured neurons and astrocytes were infected with AAV1/2-GfaABC1D-TurboID-HA-surface, AAV1/2-hSynI-V5-N-TurboID and/or AAV1/2-GfaABC1D-C-TurboID-HA. Representative images of neuron and astrocyte at DIV14 after the treatment of 500 μM biotin for 6h are shown. n = 3 biological repeats.

Extended Data Fig. 2 Split-TurboID maps excitatory and inhibitory perisynaptic proteins.

ad, Representative images demonstrating that proteins biotinylated by astrocytic TurboID-surface or Split-TurboID (cyan) are adjacent to excitatory presynaptic marker VGLUT1 (a), postsynaptic marker HOMER1 (b), inhibitory presynaptic marker VGAT (c), and postsynaptic marker gephyrin (d). Astrocytes were visualized with GfaABC1D-mCherry-CAAX. n = 3 biological repeats.

Extended Data Fig. 3 Brain-wide transduction of astrocytes and neurons.

a, Schematic of AAV PHP.eB viruses for neuronal-EGFP or astrocyte-mCherry-CAAX and retro-orbital injection. b, Sagittal section of mouse brain showing expression throughout the cortex and other structures. c, Representative image from cortex, hippocampus or cerebellum showing high coverage of neuronal and astrocytic expression.

Extended Data Fig. 4 Mapping and identification of tripartite synaptic cleft proteins by Split-TurboID in vivo.

a, Biotinylation activity of Split-TurboID in vivo. Lysates of mouse brain infected with cell-type-specific TurboID-surface-HA, V5-N-TurboID and/or C-TurboID-HA. Brain lysates were analysed by immunoblotting with anti-Streptavidin, anti-V5, anti-HA and anti-Tubulin antibodies. b, The graph indicates the ratio of botinylation activity in vivo (n = 4 brains per each condition). c, d, The biotinylation of Split-TurboID in mouse cortex. e, f, Quantification of average number of excitatory or inhibitory synaptic colocalized puncta in layer 2/3 of the visual cortex. n = 15 slices per each condition from 3 mice. g, Chart summarizing proteomic data set identified by mass spectrometry and filters used to identify top candidates. h, Venn diagram comparing proteome list of Split-TurboID and TurboID-surface. i, Scale-free network of Split-TurboID (green) and TurboID-surface (blue) identified proteins. High-confidence proteins enriched in both Split-TurboID and TurboID-surface fractions are shown in red. Neuronal enriched proteins (RNA-seq expression ratio <1) and astrocyte enriched proteins (RNA-seq expression ratio1.0) are represented as circle or diamond, respectively. At least n = 4 biological repeats. One-way ANOVA (Dunnett’s multiple comparison, P < 0.0001, 0.001). Data are means ± s.e.m.

Extended Data Fig. 5 The validation of candidate proteins with CRISPR-based astrocytic candidate gene depletion strategy.

a, Schematic of CRISPR-based deletion of astrocytic NrCAM in vitro. b, Immunoblots showing loss of NrCAM with sgRNA. AAV1/2-U6-empty sgRNA or AAV1/2-U6-NrCAM sgRNA was co-infected with AAV1/2- GfaABC1D-Cas9 to cultured neurons and astrocytes at DIV14. The cells were subjected to immunoblot analysis with an anti-NrCAM antibody. Tubulin was used as a loading control. c, The bar graph indicates the expression level of NrCAM from 3 independent experiments. d, Schematic of CRISPR-based deletion strategy of candidate gene. e, Experimental timeline of AAV-mediated CRISPR-based astrocytic gene deletion strategy in Flex-TdTomato mice. f, AAV PHP.eB-U6-NrCAM sgRNA was co-infected with AAV PHP.eB-GfaABC1D-Cas9 in Flex-TdTomato mice at P21. Coronal sections were prepared and immunostained with an anti-TdTomato antibody. g, A High-magnification image is shown. h, Images of Tenm2-, Tenm4- or NrCAM-deleted astrocytes (cyan) and their territories (red outlines) in visual cortexes of juvenile mice. i, Average territory volumes at P42 of Tenm2-, Tenm4- or NrCAM-deleted astrocytes. Between 20-25 cells per condition from 3 mice. j, Images of Tenm2-, Tenm4- or NrCAM-deleted astrocytes (cyan) and their NIV reconstructions (orange) in visual cortexes of juvenile mice. k, Average NIV at P42 of Tenm2-, Tenm4- or NrCAM-deleted astrocytes. 51 cells per each condition from 3 mice. n = 3 biological repeats. One-way ANOVA (Dunnett’s multiple comparison, P < 0.0001, 0.01). Data are means ± s.e.m.

Extended Data Fig. 6 NrCAM is a novel tripartite synaptic protein.

a, A high magnification STED image showing that endogenous NrCAM was enriched at biotinylated proteins in vivo. b, Immunoblot analysis of endogenous NrCAM, astrocyte marker GFAP, neuronal marker b-Tubulin III or loading control α-Tubulin from mouse brain or purified astrocyte lysate. c, Schematic of the visualization of astrocytic membrane and endogenous NrCAM in vivo. d, STED images demonstrating the localization of endogenous NrCAM in vivo. Coronal sections were immunostained with anti-NrCAM antibody (cyan). High magnification image was shown (right panel). e, Schematic of the visualization of both astrocytic and neuronal NrCAM in vivo. f, STED images demonstrating that the colocalization of astrocytic NrCAM with neuronal NrCAM in vivo. Coronal sections were prepared and co-immunostained with an anti-V5 (cyan) and anti-HA (magenta) antibody. A high-magnification image is shown in the right. n = 3 biological repeats. Data represent means ± s.e.m.

Extended Data Fig. 7 The role of astrocytic NrCAM in astrocytic morphogenesis in vivo.

a, Schematic of CRISPR-based NrCAM deletion in vivo. b, Schematic of hNrCAM domains and fragments. SP, signal peptide; IG, immunoglobulin; FN, fibronectin; TMD, transmembrane domain; ECD, extracellular domains. c, Immunoblots showing the expression of each NrCAM fragments in HEK293T cells. d, f, h, j, Images of astrocytes following deletion of astrocyte NrCAM alone (NrCAM sgRNA), with coexpression with indicated constructs of sgRNA-resistant human NrCAM, neuronal NrCAM deletion (neuroNrCAM sgRNA), or following neuronal NrCAM deletion alone. Images at indicated ages represent. e, i, Analysis of astrocyte territory, 15–29 cells per each condition from 3 mice; gk, Analysis of neuropil infiltration volume. 50–51 cells per each condition from 3 mice. n = 3 biological repeats. One-way ANOVA (Dunnett’s multiple comparison, P < 0.0001). Data represent means ± s.e.m.

Extended Data Fig. 8 NrCAM controls inhibitory synaptic specializations through binding the gephyrin.

a, Immunoblot analysis of endogenous NrCAM, astrocyte marker GFAP, Neuroligin 2, Neuroligin 3, Kir4.1 or EAAT2 (GLT1) from purified astrocyte lysate. b, The bar graph indicates the expression level. c, The interaction of NrCAM with PSD95 and gephyrin in HEK293T cells. Cell lysates coexpressing NrCAM-HA with GFP, PSD95-GFP or GFP-gephyrin were incubated with anti-GFP-bound beads. Immunoprecipitated (right) or total (left) NrCAM, GFP, PSD95-GFP or GFP-gephyrin were detected by immunoblotting with anti-HA and anti-GFP antibodies. d, Schematic of HEK293T/neuronal mixed-cultured assay in vitro. eh, Images of in vitro inhibitory synapse formation assays. The graph shows average of the total integrated intensity of VGAT (Cont = 258, NL2 = 222, NrCAM = 242, NrCAM-ΔIG = 288, NrCAM-ΔECD = 303 cells) or GABAA receptor (Cont = 313, NRX1β4(-) = 310, NrCAM = 300, NrCAM-ΔIG = 278, NrCAM-ΔECD = 278 cells) clusters that contact transfected HEK293T cells. il, Images of in vitro excitatory synapse formation assay. The graph shows average of the total integrated intensity of VGLUT1 (Cont = 259, NL2 = 306, NrCAM = 286, NrCAM-ΔIG = 321, NrCAM-ΔECD = 196 cells) or HOMER1 (Cont = 471, NRX1β4(-) = 214, NrCAM = 247, NrCAM-ΔIG = 387, NrCAM-ΔECD = 251 cells) clusters that contact transfected HEK293T cells. n = 3 biological repeats. One-way ANOVA (Dunnett’s multiple comparison, P < 0.0001). Data are means ± s.e.m.

Extended Data Fig. 9 The effect of NrCAM on excitatory synapse formation and function in vivo.

a, Images of postsynapse PSD95 and presynapse VGLUT1 within NrCAM-deletion astrocytes in L1 of the visual cortex. High magnification images (bottom) correspond to boxes (above). b, Quantification of average number of excitatory synaptic colocalized puncta within astrocyte territories. n = 15 cells per each condition from 3 mice. c, mEPSC traces from L2/3 pyramidal neurons following astrocyte control empty sgRNA or NrCAM sgRNA expression. dg, Quantification of mEPSC amplitude (d, e, Cont = 16, NrCAM sgRNA = 14 cells from 4 mice) and frequency (f, g, Cont = 14, NrCAM sgRNA = 17 cells from each of 4 mice). At least n = 3 biological repeats. Student’s t-test (paired, P < 0.05). Data represent means ± s.e.m.

Extended Data Fig. 10 In vivo chemogenetics method, Split-TurboID, reveals a novel astrocytic cell adhesion molecule, NrCAM, that controls inhibitory synaptic organization.

Development of in vivo chemo-affinity codes, Split-TurboID, and a working model of astrocytic NrCAM influencing inhibitory synaptic function. Split-TurboID can map the molecular composition of such intercellular contacts, even within the highly complex structure of the tripartite synapse in vivo. Mapping this interface, we discovered a new molecular mechanism by which astrocytes influence inhibitory synapses within the tripartite synaptic cleft via NrCAM. NrCAM is expressed in cortical astrocytes where it interacts with neuronal NrCAM that is coupled to gephyrin at inhibitory postsynapses. Loss of astrocytic NrCAM dramatically alters inhibitory synaptic organization and function in vivo.

Supplementary information

Supplementary Information

This file contains Supplementary Figure 1 (uncropped blots) and Supplementary Table 4 (Primers used for constructions or sequences).

Reporting Summary

Supplementary Table 1

Split-TurboID Enriched Proteins.

Supplementary Table 2

Astrocyte TurboID-Surface Enriched Proteins.

Supplementary Table 3

High Confidence Tri-partite Synapse Proteome.

Supplementary Table 5

Raw Data of Split-TurboID and Astrocyte TurboID-surface Proteome.

Supplementary Table 6

Contaminating Proteins.

Supplementary Table 7

Summary of Statistical Analysis.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Takano, T., Wallace, J.T., Baldwin, K.T. et al. Chemico-genetic discovery of astrocytic control of inhibition in vivo. Nature 588, 296–302 (2020). https://doi.org/10.1038/s41586-020-2926-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2926-0

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

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