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:

Inhibitory input directs astrocyte morphogenesis through glial GABABR

Abstract

Communication between neurons and glia has an important role in establishing and maintaining higher-order brain function1. Astrocytes are endowed with complex morphologies, placing their peripheral processes in close proximity to neuronal synapses and directly contributing to their regulation of brain circuits2,3,4. Recent studies have shown that excitatory neuronal activity promotes oligodendrocyte differentiation5,6,7; whether inhibitory neurotransmission regulates astrocyte morphogenesis during development is unclear. Here we show that inhibitory neuron activity is necessary and sufficient for astrocyte morphogenesis. We found that input from inhibitory neurons functions through astrocytic GABAB receptor (GABABR) and that its deletion in astrocytes results in a loss of morphological complexity across a host of brain regions and disruption of circuit function. Expression of GABABR in developing astrocytes is regulated in a region-specific manner by SOX9 or NFIA and deletion of these transcription factors results in region-specific defects in astrocyte morphogenesis, which is conferred by interactions with transcription factors exhibiting region-restricted patterns of expression. Together, our studies identify input from inhibitory neurons and astrocytic GABABR as universal regulators of morphogenesis, while further revealing a combinatorial code of region-specific transcriptional dependencies for astrocyte development that is intertwined with activity-dependent processes.

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: Inhibitory neuron activity regulates astrocyte morphogenesis.
Fig. 2: Gabbr1 is required for astrocyte morphogenesis.
Fig. 3: Gabbr1 regulates astrocyte morphology through Ednrb.
Fig. 4: Region-specific regulation of Gabbr1 by SOX9 and NFIA.
Fig. 5: Regulation of astrocyte morphogenesis by region-specific mechanisms.

Similar content being viewed by others

Data availability

The bulk RNA-seq data from developing astrocytes and Ednrb-cKO astrocytes have been deposited at the NCBI GEO under accession number GSE198632. scRNA-seq data can be found at the NIH GEO (GSE198357, GSE198633). All other data in this Article are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

No custom code was used in this study. The R package limma eBayes function was used to define DEGs. The Bioconductor SVA/Combat package was used for batch correction.

References

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Allen, N. J. Astrocyte regulation of synaptic behavior. Annu. Rev. Cell Dev. Biol. 30, 439–463 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Khakh, B. S. & Deneen, B. The emerging nature of astrocyte diversity. Annu. Rev. Neurosci. 42, 187–207 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Volterra, A. & Meldolesi, J. Astrocytes, from brain glue to communication elements: the revolution continues. Nat. Rev. Neurosci. 6, 626–640 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Baraban, M., Koudelka, S. & Lyons, D. A. Ca2+ activity signatures of myelin sheath formation and growth in vivo. Nat. Neurosci. 21, 19–23 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Osso, L. A., Rankin, K. A. & Chan, J. R. Experience-dependent myelination following stress is mediated by the neuropeptide dynorphin. Neuron 109, 3619–3632 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gibson, E. M. et al. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 344, 1252304 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Bushong, E. A., Martone, M. E., Jones, Y. Z. & Ellisman, M. H. Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains. J. Neurosci. 22, 183–192 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Molofsky, A. V. et al. Astrocytes and disease: a neurodevelopmental perspective. Genes Dev. 26, 891–907 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Seifert, G., Schilling, K. & Steinhäuser, C. Astrocyte dysfunction in neurological disorders: a molecular perspective. Nat. Rev. Neurosci. 7, 194–206 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Pekny, M. et al. Astrocytes: a central element in neurological diseases. Acta Neuropathol. 131, 323–345 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Clarke, L. E. & Barres, B. A. Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 14, 311–321 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Allen, N. J. & Eroglu, C. Cell biology of astrocyte-synapse interactions. Neuron 96, 697–708 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Farhy-Tselnicker, I. et al. Activity-dependent modulation of synapse-regulating genes in astrocytes. eLife 10, e70514 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Müller, C. M. Dark-rearing retards the maturation of astrocytes in restricted layers of cat visual cortex. Glia 3, 487–494 (1990).

    Article  PubMed  Google Scholar 

  17. Morel, L., Higashimori, H., Tolman, M. & Yang, Y. VGluT1+ neuronal glutamatergic signaling regulates postnatal developmental maturation of cortical protoplasmic astroglia. J. Neurosci. 34, 10950–10962 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Lin, C.-C. J. et al. Identification of diverse astrocyte populations and their malignant analogs. Nat. Neurosci. 20, 396–405 (2017).

    Article  PubMed Central  Google Scholar 

  19. Anthony, T. E. & Heintz, N. The folate metabolic enzyme ALDH1L1 is restricted to the midline of the early CNS, suggesting a role in human neural tube defects. J. Comp. Neurol. 500, 368–383 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Dimidschstein, J. et al. A viral strategy for targeting and manipulating interneurons across vertebrate species. Nat. Neurosci. 19, 1743–1749 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bettler, B., Kaupmann, K., Mosbacher, J. & Gassmann, M. Molecular structure and physiological functions of GABAB receptors. Physiol. Rev. 84, 835–867 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Wu, C. & Sun, D. GABA receptors in brain development, function, and injury. Metab. Brain Dis. 30, 367–379 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Srinivasan, R. et al. New transgenic mouse lines for selectively targeting astrocytes and studying calcium signals in astrocyte processes in situ and in vivo. Neuron 92, 1181–1195 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mariotti, L., Losi, G., Sessolo, M., Marcon, I. & Carmignoto, G. The inhibitory neurotransmitter GABA evokes long-lasting Ca2+ oscillations in cortical astrocytes. Glia 64, 363–373 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Jin, S. et al. Inference and analysis of cell–cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Koyama, Y., Yoshioka, Y., Hashimoto, H., Matsuda, T. & Baba, A. Endothelins increase tyrosine phosphorylation of astrocytic focal adhesion kinase and paxillin accompanied by their association with cytoskeletal components. Neuroscience 101, 219–227 (2000).

    Article  CAS  PubMed  Google Scholar 

  28. Koyama, Y. Endothelin ETB receptor-mediated astrocytic activation: pathological roles in brain disorders. Int. J. Mol. Sci. 22, 4333 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hammond, T. R. et al. Endothelin-B receptor activation in astrocytes regulates the rate of oligodendrocyte regeneration during remyelination. Cell Rep. 13, 2090–2097 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Huang, A. Y.-S. et al. Region-specific transcriptional control of astrocyte function oversees local circuit activities. Neuron https://doi.org/10.1016/j.neuron.2020.03.025 (2020).

  31. Ung, K. et al. Olfactory bulb astrocytes mediate sensory circuit processing through Sox9 in the mouse brain. Nat. Commun. https://doi.org/10.1038/s41467-021-25444-3 (2021).

  32. Deneen, B. et al. The transcription factor NFIA controls the onset of gliogenesis in the developing spinal cord. Neuron 52, 953–968 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Kang, P. et al. Sox9 and NFIA coordinate a transcriptional regulatory cascade during the initiation of gliogenesis. Neuron 74, 79–94 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stolt, C. C. et al. The Sox9 transcription factor determines glial fate choice in the developing spinal cord. Genes Dev. 17, 1677–1689 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Subramanian, L. et al. Transcription factor Lhx2 is necessary and sufficient to suppress astrogliogenesis and promote neurogenesis in the developing hippocampus. Proc. Natl Acad. Sci. USA 108, E265–E274 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kuwaki, T. et al. Physiological role of brain endothelin in the central autonomic control: from neuron to knockout mouse. Prog. Neurobiol. 51, 545–579 (1997).

    Article  CAS  PubMed  Google Scholar 

  37. Haller, C. et al. Floxed allele for conditional inactivation of the GABAB(1) gene. Genesis 40, 125–130 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Akiyama, H., Chaboissier, M.-C., Martin, J. F., Schedl, A. & de Crombrugghe, B. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Gene Dev. 16, 2813–2828 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  40. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  41. Srinivasan, R. et al. Ca2+ signaling in astrocytes from Ip3r2−/− mice in brain slices and during startle responses in vivo. Nat. Neurosci. 18, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ewels, P., Magnusson, M., Lundin, S. & Käller, M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics 32, 3047–3048 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ximerakis, M. et al. Single-cell transcriptomic profiling of the aging mouse brain. Nat. Neurosci. 22, 1696–1708 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tasic, B. et al. Adult mouse cortical cell taxonomy revealed by single cell transcriptomics. Nat. Neurosci. 19, 335–346 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen, T.-W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kim, J.-Y., Grunke, S. D., Levites, Y., Golde, T. E. & Jankowsky, J. L. Intracerebroventricular viral injection of the neonatal mouse brain for persistent and widespread neuronal transduction. J. Vis. Exp. https://doi.org/10.3791/51863 (2014).

Download references

Acknowledgements

This work was supported by US National Institutes of Health grants NS071153, AG071687 and NS096096 to B.D. We thank the David and Eula Wintermann Foundation for support; and B. Bettler for providing the Gabbr1-floxed mouse line. scRNA-seq studies were performed at the Single Cell Genomics Core at BCM partially supported by NIH shared instrument grants (S10OD023469, S10OD025240) and P30EY002520. This project was supported by the Cytometry and Cell Sorting Core at Baylor College of Medicine with funding from the CPRIT Core Facility Support Award (CPRIT-RP180672), the NIH (CA125123 and RR024574) and the assistance of J. M. Sederstrom. Research reported in this publication was supported by the Eunice Kennedy Shriver National Institute of Child Health & Human Development of the National Institutes of Health under Award Number P50HD103555 for use of the Microscopy Core facilities and the Animal Phenotyping & Preclinical Endpoints Core facilities.

Author information

Authors and Affiliations

Authors

Contributions

Y.-T.C. and B.D. conceived the project and designed the experiments. Y.-T.C., J.W., Z.-F.L., H.-C.C. and E.L.-F. performed the experiments. J.W. performed the electrophysiology studies. Y.-T.C. and A.S.H. designed and performed the bioinformatics analyses. Y.-T.C. and B.D. wrote the manuscript.

Corresponding author

Correspondence to Benjamin Deneen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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 Normalization of astrocyte morphology in the adult after developmental activation of inhibitory neurons.

a-b. Analysis of SOX9 (e) and Ki67(f) expression within Aldh1l1-GFP astrocytes at P21 after activation of inhibitory neurons (or control); quantification was derived from n = 3 pairs of animals (a, 20,24 images; GLME; b, 12, 11 images; GLME). c. CNO only treatment of Aldh1l1-GFP mice from P7-P21 and analysis of astrocyte morphology at P21. n = 3 animals (39 cells; GLME model with Sidak’s multiple comparisons test). d. Heatmap depicting expression of GABA receptor subunits in developing astrocytes from the cortex (CX), hippocampus (HC), or olfactory bulb (OB) at P1, P7, and P14. See Extended Data Figure 2. d. Example of gating strategy and percentage of GFP+ astrocytes FACS isolated from P1 animal. e. Heatmap depicting expression of GABA receptor subunits in developing astrocytes from the cortex (CX), hippocampus (HC), or olfactory bulb (OB) at P1, P7, and P14. See Extended Data Figure 2. f. Schematic of DREADD-based activation of inhibitory neurons in post-natal Aldh1l1-GFP mice and experimental timeline. g. Imaging of P60 Aldh1l1-GFP astrocytes after hM3Dq activation of inhibitory neurons; quantification of morphological complexity using Sholl analysis, branch number, and total processes at P21; n = 3 pairs of animals (26,35 cells; upper, GLME model with Sidak’s multiple comparisons test; bottom, GLME model). Scale bars, 20 μm (a–c), 30 μm (g). Data represent mean ± s.d. (a–c, g upper), median, minimum value, maximum value and IQR (g bottom).

Source Data

Extended Data Fig. 2 Transcriptomic RNA-Sequencing analysis of developing astrocytes in the cortex, hippocampus, and olfactory bulb at P1, P7, and P14.

a. Heatmap depicting the expression of neuron-specific and astrocyte-specific genes from P1, P7, and P14 FACS isolated Aldh1l1-GFP astrocytes from the listed brain regions. b. Aldh1l1-GFP astrocytes from the cortex at P1, P7, P14. Principal component (PC) analysis against top 2,000 variable genes across the region and timepoints examined from 3-4 animals in each group. c. Heatmap depicting differential patterns of gene expression in developing astrocytes across brain regions and timepoints. d. Gene Ontology (GO) analysis of the common and region-specific patterns of gene expression. e–f. Ald1l1-CreER; ROSA-LSL-tdTomato mouse line treated with tamoxifen at P1, harvested at P28. Co-immunostaining of tdTomato labelled cells with Sox9, Olig2, NeuN, and Iba1 demonstrating astrocyte-specific activity of Aldh1l1-CreER line. n = 4 animals. Scale bars, 10 μm (b), 40 μm (e–h).

Source Data

Extended Data Fig. 3 Analysis of astrocyte development in the Gabbr1-cKO mouse line.

a. RNA-Scope imaging of Gabbr1 within Aldh1l1-GFP astrocytes from control and Gabbr1-cKO mouse lines; quantification derived from n = 3 pairs of animals (control: OB 18, CX 18, HC 16; cKO: OB 17, CX 19, HC 17 cells; LME model, ***P = 0.00020, ****P = 3.07e-06, **P = 0.0030). Dashed circle denotes astrocyte with Gabbr1. b. Antibody staining for SOX9 in Aldh1l1-GFP astrocytes from cortex of Gabbr1-cKO and control; quantification is derived from n = 3 pairs of animals (35 images; GLME model). c. Pulse-chase EdU-labelling and antibody staining at P28 from all brain regions analysed; quantification is derived from n = 3 pairs of animals (Gabbr1 control: OB 9, CX 9, HC 9, BS 9, CB 9; Gabbr1 cKO: OB 8, CX 9, HC 9, BS 9, CB 9 images; GLME model). d-e. Imaging of Aldh1l1-GFP astrocytes from the brain stem and cerebellum at P28; quantification of morphological complexity was derived from n = 3 pairs of animals (Gabbr1 control: BS 28, CB 29; Gabbr1 cKO: BS 32, CB 29 cells; GLME model with Sidak’s multiple comparisons test, *P = 0.0179, 0.0167). f. Quantitative analysis of branch points and process length from all brain regions analysed; n = 25-38 cells from 3 pairs of animals (Gabbr1 control: OB 38, CX 30, HC 29, BS 28, CB 25; Gabbr1 cKO: OB 33, CX 30, HC 29, BS 32, CB 29 cells; two way ANOVA, **P = 0.0014, *P = 0.0174, P = 0.9040, *P = 0.0132, P = 0.7126, **P = 0.0054, ****P < 0.0001, **P = 0.0066, P = 0.3763). Scale bars, 30 μm (d), 20 μm (c). Data represent mean ± s.d. (b-c, e-g median, minimum value, maximum value and IQR (a).

Source Data

Extended Data Fig. 4 Analysis of Ca2+ activity and sparsely labelled astrocytes in Gabbr1-cKO astrocytes.

a. Schematic describing the experimental timeline and mouse lines rendering astrocyte-specific knockout of Gabbr1 for sparse labelling experiments. b-c. Imaging and quantification of sparsely labelled, tdTomato-expressing astrocytes from Gabbr1-cKO and control mice from the cortex (b) and hippocampus (c); n = 3 pairs of animals (Gabbr1 control: CX 32, HC 30; Gabbr1 cKO: CX 36, HC 38 cells; b,c upper, GLME model with Sidak’s multiple comparisons test, *P = 0.0213, **P = 0.0012; b,c bottom, GLME model, ***P = 0.00043, **P = 0.0027, ***P = 0.00042, ****P < 0.0001). d. Imaging of GCaMP6s activity in control and Gabbr1-cKO astrocytes from the cortex at P28; quantification is derived from n = 3 pairs of animals (24,33 cells; GLME model, P = 0.6361, 0.2239). e. Imaging of GCaMP6s activity in the presence of TTX and baclofen; quantification derived from n = 40 cells from 3 pairs of animals (two-tailed Wilcoxon matched-pairs signed rank test, *P = 0.022, P = 0.89, ***P = 0.0006, P = 0.32). f. Two-photon, slice imaging of GCaMP6s activity in control and Gabbr1-cKO astrocytes from the cortex at P28. Quantification of Ca2+ activity in astrocytic microdomains in the Gabbr1-cKO and control animals, quantification is derived from n = 3 pairs of animals (19, 30 cells; GLME model). Scale bars, 30 μm (b-c), 20 μm (d-f). Data represent mean ± s.d. (b-c, upper), median, minimum value, maximum value and IQR (b-c, lower, d, and f).

Source Data

Extended Data Fig. 5 RNA-Seq of Gabbr1-cKO astrocytes and single cell RNA-Seq analysis of Gabbr1-cKO cortex.

a. Serut analysis of single cell RNA-Seq (scRNA-Seq) from Gabbr1-cKO and controls from P28 cortex. b. Quantification of cell clusters identifying CNS cell types from scRNA-Seq data. c. CellChat interaction diagram illustrating astrocyte interactions with neurons in the cortex from P28 Gabbr1-cKO mice; width of coloured arrow indicates scale of interaction. See Extended Data Figure 4. d-e. KEGG pathway analysis of neurons from Gabbr1-cKO scRNA-Seq (d, analysed by Enrichr) and dot plot of differentially expressed genes from KEGG (e, analysed by Seurat FindMarkers). f-h. Dot plot summaries demonstrating CellChat interaction profiles and expression patterns of key astrocyte-neuron interaction pathways.

Extended Data Fig. 6 Analysis of cortical neurons in the Gabbr1-cKO mouse line.

a. Antibody staining for BRN2 (Layers II/II). b. CTIP2 (Layers V). c. FOXP2 (Layers VI) layer-specific neuronal markers in the P28 cortex from Gabbr1-cKO and control; quantification is derived from n = 11-12 images from 3 pairs of animals (control 12, cKO 11 images; GLME model). d. Schematic of synaptic markers and cortical layers. e-f. Antibody staining for makers of excitatory synapses Vglut1–PSD95 (e) and Vglut2–PSD95 (f) in layer I of the cortex from Gabbr1-cKO or control mice at P28 (n =3 pairs of animals; GLME model, *P = 0.0490). g. Antibody staining for markers of inhibitory synapses VGAT–Gephyrin at P28; quantification is derived from 3 pairs of animals (GLME model). h-k. Representative traces of action potential in layer II/III excitatory neurons upon varying injected current in Gabbr1-cKO and control (h). Summary data of action potential firing (i; two way ANOVA). Summary data of resting membrane potential (j; two-tailed unpaired Welch’s t-test) and threshold (k; two-tailed unpaired Welch’s t-test) from 3 pairs of animals (n = 13, 12 cells). l-o. Representative traces of action potential in layer II/III inhibitory neurons upon varying injected current in Gabbr1-cKO and control (l). Summary data of action potential firing (m; two way ANOWA). Summary data of resting membrane potential (n; two-tailed unpaired Welch’s t-test, **P = 0.0091) and threshold (o; two-tailed unpaired Welch’s t-test) from 3 pairs of animals (n = 12, 15 cells). Scale bars, 100 μm (a-c), 3 μm (e-f). Data represent mean ± s.d. (a-c, e-g), mean ± s.e.m. (i-k, m-o).

Source Data

Extended Data Fig. 7 Loss of astrocytic Gabbr1 disrupts cortical circuit function.

a. Schematic of viral labelling of inhibitory neurons and experimental timeline. b-e. Representative traces of spontaneous EPSCs and IPSCs from excitatory and inhibitory neurons from cortex of Gabbr1-cKO and controls. Associated cumulative and bar plots demonstrate quantification of sEPSC and sIPSC from 3 pairs of animals (b, n = 13, 15 cells; Kolmogorov-Smirnov test, **** P < 0.0001; two-tailed Mann-Whitney test, P = 0.8207, **P = 0.003; c, n = 15, 12 cells; Kolmogorov-Smirnov test, **** P < 0.0001; two-tailed Mann-Whitney test, P = 0.1995, 0.5888; d, n = 13, 15 cells; Kolmogorov-Smirnov test, **** P < 0.0001; two-tailed Mann-Whitney test, P = 0.7856, 0.0504; e, n = 11, 9 cells; Kolmogorov-Smirnov test, **** P < 0.0001; two-tailed Mann-Whitney test, P = 0.2299, 0.3796). f. Experimental timeline for behavioural analysis. g. 3-chamber social interaction and pre-pulse inhibition studies on Gabbr1-cKO and control mice from 10 animals in control group and 11 animals in cKO group. (left, GLME model with Sidak’s multiple comparisons test, *P = 0.015; right, two-tailed Mann-Whitney test, *P = 0.043). h-m. Summary of behavioural assays conducted on Gabbr1-cKO and control animals including open field (h), elevated plus maze (i), rotarod (j), parallel foot fall (k), contextual fear conditioning (l), and cued fear conditioning (m) on Gabbr1-cKO and control mice from 10 animals in control group and 11 animals in cKO group (two-tailed Mann-Whitney test, ***P = 0.0004) Data represent mean ± s.e.m. (b-e), s.d. (g-m).

Source Data

Extended Data Fig. 8 Transcriptomic analysis of Gabbr1-cKO astrocytes an.

a. Volcano plots from RNA-Seq analysis of control and Gabb1-cKO astrocytes from cortex, hippocampus, and OB. b. Table of the number of differentially expressed genes (DEGs) from each region. c. Gene Ontology (GO) analysis of DEGs performed with Enrichr. d. Quantification of EDNRB expression in virally labelled astrocytes from the P28 cortex of mice where it has been knocked out using guideRNAs in the ROSA-LSL-Cas9-EGFP mouse line; quantification is derived from n = 3 pairs of animals (37, 38 cells; LME model, **P = 0.0068). e. Imaging of virally labelled astrocytes from the P28 cortex of Ednrb-cKO mice where Ednrb has been knocked out using guideRNAs in the ROSA-LSL-Cas9-EGFP mouse line; quantification of morphological complexity via Sholl analysis was derived from n = 3 animals (f, Ednrb-cKO: 22 mcherry-Cas9-EGFP+ cells, 49 mcherry+Cas9-EGFP+ cells; GLME model with Sidak’s multiple comparisons test, *P = 0.0307; GLME model, P = 0.06, **P = 0.008). Data represent mean ± s.d. (e left), median, minimum value, maximum value and IQR (d, e right).

Source Data

Extended Data Fig. 9 Analysis of astrocyte development in the Sox9-cKO and Nfia-cKO mouse lines.

a. Homer transcription factor motif analysis on differentially expressed genes (DEGs) from P1 and P14 timepoints from astrocytes isolated from the cortex, hippocampus, and olfactory bulb. b. Analysis of NFIA expression in Aldh1l1-GFP astrocytes from the Nfia-cKO and control at P7 in the cortex, hippocampus, and olfactory bulb; quantification of knockout efficiency was derived from 3 pairs of animals (two-way ANOVA, ****P < 0.0001). c. Analysis of SOX9 expression in Aldh1l1-GFP astrocytes from the Sox9-cKO and control at P7 in the cortex, hippocampus, and olfactory bulb; quantification of knockout efficiency was derived from 3 pairs of animals (two-way ANOVA, ****P < 0.0001). d. Analysis of NFIA expression in Aldh1l1-GFP astrocytes from the Nfia-cKO and control at P28 in the cortex, hippocampus, cerebellum, and olfactory bulb; quantification of knockout efficiency was derived from 3 pairs of animals (two-way ANOVA, ****P < 0.0001). e. Analysis of SOX9 expression in Aldh1l1-GFP astrocytes from the Sox9-cKO and control at P28 in the cortex, hippocampus, cerebellum, and olfactory bulb; quantification of knockout efficiency was derived from 3 pairs of animals (two-way ANOVA, ****P < 0.0001, *P = 0.0205). f. AAV-based overexpression of NFIA in the developing cortex, analysis of Gabbr1 expression at P28 via RNAscope; n = 3 pairs of animals (19, 18 cells; LME model, *P = 0.023, ***P = 0.00034). g. AAV-based overexpression of SOX9 in the developing olfactory bulb, analysis of Gabbr1 expression at P28 via RNAscope; n = 3 pairs of animals (20,25 cells). h. Chromatin immunoprecipitation of NFIA from P28 cortex or SOX9 from P28 olfactory bulb (OB), followed by PCR detection of association with motif in proximal promoter region of Gabbr1. Scale bars, 50 μm (b–e), 10 μm (f–g). Data represent mean ± s.d. (b–e), median, minimum value, maximum value and IQR (f–g).

Source Data

Extended Data Fig. 10 Analysis of astrocyte morphogenesis in the Sox9-cKO and Nfia-cKO mouse lines.

a–b. Two-photon, slice imaging of spontaneous GCaMP6s activity in control and Nfia-cKO astrocytes from the cortex at P28 (a) or control and Sox9-cKO astrocytes from the olfactory bulb at P28 (b); quantification is derived from 3 pairs of animals (two-tailed Mann-Whitney test). c–d. Pulse-chase EdU-labelling and antibody staining at P28 from the cortex of Nfia-cKO (c) and olfactory bulb of Sox9-cKO (d); quantification is derived from 3 pairs of animals (two-tailed Mann-Whitney test). e–f. Quantification of the number of Aldh1l1-GFP astrocytes in the cortex of the Nfia-cKO (e) or olfactory bulb from Sox9-cKO (f) and associated controls; quantification is derived from 3 pairs of animals (two-tailed Mann-Whitney test). g–h. Imaging of Aldh1l1-GFP astrocytes from the hippocampus, brainstem, and cerebellum at P28 from the Nfia-cKO (g) or Sox9-cKO (h) and associated controls; quantification of morphological complexity via Sholl analysis was derived from n = 3 pairs of animals (g, Nfia control: HC 64, BS 28, CB 47; Nfia cKO: HC 65, BS 43, CB 55 cells; GLME model with Sidak’s multiple comparisons test, **P = 0.0015; h, Sox9 control: HC 32, BS 36, CB 32; Sox9 cKO: HC 24, BS 24, CB 37; GLME model with Sidak’s multiple comparisons test). i–j. Quantification of astrocytic branch number and process length from Nfia-cKO (i) or Sox9-cKO (j) across cortex, olfactory bulb, hippocampus, brainstem, and cerebellum; derived from n = 3 pairs of animals (i, Nfia control: OB 59, CX 43, HC 54, BS 27, CB 48; Nfia cKO: OB 50, CX 56, HC 59, BS 38, CB 54 cells; two-way ANOVA, **P = 0.0054, ****P <0.0001; j, Sox9 control: OB 29, CX 31, HC 32, BS 37, CB 33; Sox9 cKO: OB 40, CX 27, HC 33, BS 21, CB 39; two-way ANOVA, **P = 0.0025, ****P < 0.0001, ***P = 0.0002). Scale bars, 10 μm (a–b), 50 μm (c–f), 30 μm (g–h). Data represent mean ± s.d. (a–f,i–j).

Source Data

Extended Data Fig. 11 Electrophysiological and behavioural analysis of the Nfia-cKO mouse line.

a. Schematic of synaptic markers and cortical layers. b–c. Antibody staining for makers of excitatory synapses Vglut1–PSD95 (b) and Vglut2–PSD95 (c) in layer I of the cortex from Nfia-cKO or control mice at P28, quantification is derived from 3 pairs of animals (GLME model). d. Antibody staining for markers of inhibitory synapses VGAT–Gephyrin at P28; quantification is derived from 3 pairs of animals (GLME model). e–f. Representative traces of spontaneous EPSCs and IPSCs from excitatory (e) and inhibitory (f) neurons from cortex of Nfia-cKO and controls. Associated cumulative and bar plots demonstrate quantification of amplitude and frequency of sEPSC and sIPSC from 3 pairs of animals (e, Kolmogorov-Smirnov test, **** P < 0.0001, *P = 0.0181, two-tailed Mann-Whitney test; f, Kolmogorov-Smirnov test, **P = 0.0026, ****P < 0.0001, two-tailed Mann-Whitney test, *P = 0.0149). g–h. Representative traces of action potential in layer II/III neurons upon varying injected current in Nfia-cKO and control. Summary data of action potential firing, resting membrane potential, and threshold from excitatory neurons (g) and inhibitory neurons (h); quantification is derived from at least 3 pairs of animals (two-way ANOVA and two-tailed Mann-Whitney test). i-j. 3-chamber social interaction and prepulse inhibition behavioural studies on Nfia-cKO and control mice from 11-13 animals in each group (i, n = 13 pairs of animals, two-way ANOVA, ***P = 0.0002; j, n = 13, 11 animals, two-tailed Mann-Whitney test, **P = 0.0050). k–p. Summary of behavioural tests from NFIA-cKO and control, including open field; n = 13, 11 animals (k), elevated plus maze; n = 10 animals (l), rotarod; n = 5, 8 animals (m), parallel footfall; n = 13, 11 animals (n), contextual fear conditioning; n = 12, 10 animals (o), and cued fear conditioning; n = 12, 10 animals (p) (two-tailed Mann-Whitney test, *P = 0.0265, 0.0136). Scale bars, 3 μm (b–d). Data represent mean ± s.d. (b–d, i, o–p), mean ± s.e.m. (e–h, j–n).

Source Data

Extended Data Fig. 12 Analysis of cortical neurons in the Nfia-cKO mouse line and co-immunoprecipitation with Lhx2 and NPAS3.

a. Antibody staining for BRN2 (Layers II/II), CTIP2 (Layers V), and FOXP2 (Layers VI) layer-specific neuronal markers in the P7 cortex from Nfia-cKO and control; quantification is derived from n = 3 pairs of animals (6 images, GLME model). b. CX-specific transcription factor DEGs increased between P7-P14. c. OB-specific transcription factor DEGs increased between P7-P14. d. Immunoprecipitation of LHX2 and immunoblot of LHX2 and NFIA from the P28 cortex; arrow heads label the proteins of interest. e. Immunoprecipitation of NPAS3 and immunoblot of NPAS3 and SOX9 from P28 cortex; arrow heads label the proteins of interest. f. Quantification of LHX2 expression in virally labelled astrocytes from the P28 cortex of mice where it has been knocked out using guideRNAs in the ROSA-LSL-Cas9-EGFP mouse line; quantification is derived from n = 3 pairs of animals (37, 39 cells; GLME model, ***P = 0.00099). g. Imaging of virally labelled astrocytes from the P28 cortex of Lhx2-cKO mice where Lhx2 has been knocked out using guideRNAs in the ROSA-LSL-Cas9-EGFP mouse line; quantification of morphological complexity via Sholl analysis was derived from n = 3 animals (Lhx2-cKO: 13 mcherry-Cas9-EGFP+ cells, 40 mcherry+Cas9-EGFP+ cells; GLME model with Sidak’s multiple comparisons test, **P = 0.0037; GLME model, **P = 0.006, 0.003). h. RNAscope for Gabbr1 expression in Cas9-EGFP cortical astrocytes lacking Lhx2 and controls at P28; quantitative analysis demonstrating reduction of Gabbr1 expression is derived from n = 3 pairs of animals (51,55 cells; GLME model, *P = 0.015; LME model, **P = 0.0003). Scale bars, 100 μm (a), 20 μm (m). Data represent mean ± s.d. (a, g right), median, minimum value, maximum value and IQR (f, g left, h).

Source Data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

P1–P14 astrocyte transcriptomic data, showing Gene Ontology analysis across timepoints and regions.

Supplementary Table 2

RNA-seq transcriptomic data from Gabbr1-cKO and control astrocytes, showing DEGs between Gabbr1-cKO and controls.

Supplementary Table 3

scRNA-seq data from Gabbr1-cKO and control cortex, comparing DEGs.

Supplementary Table 4

P1–P14 astrocyte transcriptomic data, showing DEGs across timepoints and regions.

Supplementary Table 5

Transcription factor expression across the OB and cortex from P1 to P14.

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

Cheng, YT., Luna-Figueroa, E., Woo, J. et al. Inhibitory input directs astrocyte morphogenesis through glial GABABR. Nature 617, 369–376 (2023). https://doi.org/10.1038/s41586-023-06010-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06010-x

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