Main

Astrocytes actively participate in synapse development and function by secreting instructive cues to neurons1. Through their perisynaptic processes, astrocytes maintain ion homeostasis, clear neurotransmitters2 and contribute to neuromodulatory signalling to control circuit activity and behaviour3. These complex functions of astrocytes are reflected in their elaborate structure4,5, with numerous fine processes that interact closely with synapses. Importantly, loss of astrocyte complexity is a common pathological feature in neurological disorders6.

Despite the vital functions of astrocytes in brain development and physiology, it is unclear how their complex morphology is established. Furthermore, we do not know whether disruptions in astrocyte morphogenesis lead to synaptic dysfunction. We investigated these questions in the developing mouse primary visual (V1) cortex during postnatal days 1–21 (P1–P21), when astrocyte morphogenesis occurs concomitantly with synaptic development7,8. Using Aldh1L1–EGFP BAC-transgenic mice, in which all astrocytes express enhanced green fluorescent protein (EGFP)9, we found that astrocytic coverage of the V1 neuropil increased profoundly from P7 to P21 (Fig. 1a–c), coinciding with high rates of synaptogenesis10. This increase was concurrent with the appearance of fine astrocytic processes (Extended Data Fig. 1a), and only became significant between P7 and P14, when eye opening occurs, suggesting that vision drives this growth (Fig. 1b). Indeed, mice reared in the dark showed profoundly stunted astrocyte coverage of V1 but not of the auditory cortex (Extended Data Fig. 1b–d)

Figure 1: Astrocyte morphogenesis occurs in tune with sensory activity.
figure 1

a, V1 cortex images (layers L1–L6) from Aldh1L1–EGFP mice at postnatal days P1–P21. b, Fold change in astrocyte coverage of the neuropil at each cortical layer from P1–P21 (normalized to P1 L1). c, Fold change in astrocyte coverage of the neuropil from P7 and P21 (normalized to P7). b, c, n = 10 regions of interest (ROIs) per layer, more than 3 images per mouse, 3 mice per time point. d, Representative images and NIVs of V1 L4 PALE astrocytes from normal (NR) and dark reared (DR) mice at P7 and P21. Astrocytes were electroporated with EGFP (green) and membrane-tagged mCherry (mCherry–CAAX, red) plasmids. e, Average NIV of P7 and P21 astrocytes from normal and dark-reared mice. n = 3 NIVs per cell, 18–20 cells per condition, 4 mice per condition. One-tailed t-test (c), one-way ANOVA (e). Data are means ± s.e.m. Scale bars, 100 μm (a), 10 μm (d).

PowerPoint slide

Next, we investigated astrocyte growth at the single-cell level using postnatal astrocyte labeling by electroporation (PALE), which sparsely transfects and labels cortical astrocytes (Extended Data Fig. 1e–h). The volume of fluorescently labelled astrocyte processes infiltrating the neuropil (neuropil infiltration volume, NIV) increased markedly between P7 and P21 (Extended Data Fig. 1i–m). Dark rearing decreased NIV at P21, but not at P7 (Fig. 1d, e). V1 astrocytes also increased their territory size by about 1.6-fold between P7 and P21 (Extended Data Fig. 1n, o). Notably, astrocyte territories were significantly reduced by dark rearing at both ages (Extended Data Fig. 1p, q), suggesting that light-induced changes in V1, which occur even before eye opening11, are crucial for astrocyte territory growth. Together, our findings reveal that astrocyte morphogenesis occurs in parallel with the growth and activity of the underlying synaptic circuits of the cortex.

To investigate the mechanisms that link astrocyte morphogenesis to neuronal circuit development, we established a primary rat cortical neuron–astrocyte co-culture system, which takes advantage of the observation that astrocyte complexity is greatly enhanced by co-culture with neurons compared to culturing them alone or on Cos7 cells (Extended Data Fig. 2a–e, h). Neuron-conditioned medium was not sufficient to induce astrocyte elaboration (Extended Data Fig. 2e, h). Furthermore, inhibition of astrocyte glutamate sensing by blocking metabotropic glutamate receptor 5 (mGluR5) only slightly impaired astrocyte elaboration, whereas blocking synaptic network activity with tetrodotoxin (TTX) did not diminish neuron-induced astrocyte elaboration (Extended Data Fig. 2f, i). These findings indicate that contact-mediated mechanisms, rather than secreted factors or synaptic activity, are the primary drivers for astrocyte morphogenesis in vitro. To test this theory, we fixed neurons with methanol to preserve their structure, while eliminating dynamic feedback to astrocytes. Astrocytes fully elaborated by co-culture with methanol-fixed neurons, whereas methanol-fixed Cos7 cells did not induce elaboration (Extended Data Fig. 2g, j–l). Extraction of neuronal structures with urea, while preserving the deposited extracellular matrix, severely reduced astrocyte elaboration (Extended Data Fig. 2g, j, m). Super-resolution imaging of astrocyte processes and synapses showed that astrocyte elaboration occurs near synapses, and astrocytes interact closely with synaptic structures in vitro (Extended Data Fig. 2n). Together, these results show that astrocyte morphogenesis is triggered by direct contact with neurons in vitro.

Astrocytes require neuroligins for complexity

Next, we mined gene expression databases12,13,14 to identify astrocytic cell adhesion molecules (CAMs) that are known to interact with neuronal and synaptic proteins. Notably, astrocytes express three members of the neuroligin family (NL1, NL2, and NL3) at levels comparable to or higher than those seen in neurons (Extended Data Fig. 3a–f). We confirmed that rodent astrocytes express neuroligins by fluorescent in situ hybridization (FISH) in vivo and by PCR with reverse transcription (RT–PCR) and western blotting in vitro (Extended Data Fig. 3g–j).

Neuroligins have been mainly studied in the context of neurons15,16,17, with few exceptions18,19,20. Simultaneous knockdown of all astrocytic neuroligins with short hairpin RNAs (shRNAs) (Extended Data Fig. 4a) completely blocked neuron-induced astrocyte elaboration in vitro (Fig. 2a, b). Silencing each individual astrocytic neuroligin partially but significantly diminished astrocyte arborization, indicating that the neuroligins have non-overlapping roles in astrocyte morphogenesis in vitro (Fig. 2a–e, Extended Data Fig. 4d). Co-transfection of neuroligin-targeting shRNAs with the corresponding RNA interference-resistant neuroligin cDNAs21 (Extended Data Fig. 4b, c) rescued astrocyte elaboration (Fig. 2a, c–e). Knockdown of neuroligins also inhibited astrocyte elaboration induced by co-culture with methanol-fixed neurons (Extended Data Fig. 4e, f). By contrast, knockdown of EphrinA3, a CAM with known roles in astrocyte–neuron interactions22, did not alter astrocyte morphogenesis (Extended Data Fig. 4g, h). Collectively, these results show that astrocytic NL1, NL2, and NL3 are required for the establishment of neuronal contact-induced astrocyte morphogenesis in vitro.

Figure 2: Astrocytic neuroligins control astrocyte morphogenesis through neuronal neurexins.
figure 2

a, Astrocytes (green) were transfected with shRNAs against rat NL1 (shrtNL1), mouse/rat NL2 (shNL2), and/or rat NL3 (shrtNL3) with or without haemagglutinin (HA)-tagged, shRNA-resistant neuroligin plasmids (red) and co-cultured with neurons (not visible). be, Quantification of astrocyte complexity for conditions in a. f, Image of an astrocyte transfected with shrtNL1 (green) and HA-tagged NL1-SWAP (red) in co-culture with neurons (not visible). g, Quantification of astrocyte complexity for f. h, Images of shCtrl-transfected astrocytes (red) in co-culture with neurons transfected with shNrx1, shNrx2 or shScr lentivirus (green). i, Quantification of astrocyte complexity for h. be, i Data represented are from individual experiments with three biological replicates. Similar results were obtained in three independent experiments. n > 20–25 cells per condition per experiment. j, Nanofibres were coated with Fc-tagged proteins to model a neuronal scaffold. Images of EGFP-transfected astrocytes (green) cultured on Fc-protein-coated nanofibres (red). k, Quantification of astrocyte complexity for j. Data represent one experiment with four biological replicates. n > 25 cells per condition. ANCOVA (be, g, i, k). Data are means ± s.e.m. Scale bar, 10 μm.

PowerPoint slide

In neurons, neuroligins function by forming trans-synaptic adhesions with neurexins16. Swapping the extracellular cholinesterase (ChoE)-like domain of NL1 with the homologous cholinesterase sequence creates a chimaera, NL1-SWAP, that is expressed and trafficked correctly, but cannot interact with presynaptic neurexins23 (Extended Data Fig. 4i). Co-transfection of NL1-SWAP with an shRNA targeting NL1 (shNL1) failed to rescue astrocyte morphogenesis (Fig. 2f, g), even though shNL1 did not diminish NL1-SWAP expression, and expression of NL1-SWAP alone in astrocytes did not impair astrocyte morphogenesis (Extended Data Fig. 4b, j, k). These results show that contacts mediated by the ChoE-like domain of NL1 are required for astrocyte morphogenesis and suggest that interactions between astrocytic neuroligins and neuronal neurexins regulate astrocyte development.

To test whether neuronal neurexins are required for astrocytic morphogenesis, we silenced neurexin expression in cultured rat neurons using a lentivirus encoding shRNAs against mouse neurexins (Nrx1, Nrx2, and Nrx3 (both α- and β-isoforms24)). In rat neurons, this lentivirus silenced Nrx1 and Nrx2 but not Nrx3, owing to mismatches between the rat and mouse sequences (Extended Data Fig. 5a–d). Silencing Nrx1 and Nrx2 in neurons significantly diminished neuronal contact-induced astrocyte morphogenesis (Fig. 2h, i). Concurrently silencing NL1 in astrocytes did not further reduce astrocyte complexity (Extended Data Fig. 5e–g), indicating that Nrx1 and Nrx2 are the primary neuronal interaction partners for astrocytic NL1. In addition, these findings suggest that interactions between other astrocytic neuroligins and neuronal neurexins are also important for astrocyte morphogenesis in vitro.

Elimination of neuronal neurexins may affect astrocyte morphology independent of the loss of neurexin–neuroligin contacts. To address this possibility, we used methanol-fixed neurons to induce astrocyte morphogenesis and blocked neurexin–neuroligin interactions by applying soluble Fc-tagged Nrxβ ectodomains (Extended Data Fig. 5h–j). Co-application of Nrx1β–Fc, Nrx2β–Fc and Nrx3β–Fc significantly diminished astrocyte complexity compared to Fc-only protein (Extended Data Fig. 5k–m). Application of Nrx1β–Fc or Nrx2β–Fc alone reduced astrocyte elaboration, but Nrx3β–Fc did not (Extended Data Fig. 5n). Meanwhile, silencing astrocytic NL1 did not further diminish astrocyte elaboration (Extended Data Fig. 5o). Next, we tested whether trans-interactions with neurexins are sufficient for astrocyte morphogenesis by coating 3D nanofibres with Nrxβ ectodomains to generate an artificial scaffold mimicking a web of neurites (Fig. 2j). Remarkably, all Nrxβ-coated nanofibres strongly induced astrocyte complexity compared to those coated with Fc only (Fig. 2j, k). Together, these results show that interactions with neuronal neurexins are required and sufficient for contact-induced astrocyte morphogenesis in vitro.

Neuroligins control astrocyte morphology in vivo

To determine whether neuroligins control astrocyte morphogenesis in vivo, we introduced EGFP-expressing shRNA plasmids targeting mouse NL1, NL2 or NL3 into V1 astrocytes by PALE. As a control, we used a scrambled NL1 shRNA sequence (shCtrl, Extended Data Fig. 6a–d). As expected, shCtrl-transfected astrocytes developed main branches by P7 and elaborated finer processes into the neuropil by P21 (Fig. 3a). NL1 knockdown markedly reduced astrocytic NIV at P7, but this was corrected by P21. Conversely, NL3 knockdown did not alter astrocyte morphogenesis at P7, but severely arrested astrocyte growth by P21. Silencing NL2 restricted NIV at both time points (Fig. 3a–f, Extended Data Fig. 6e). These results show that neuroligins are required for astrocyte morphogenesis in vivo and suggest that the three neuroligins have unique temporal roles in astrocyte development.

Figure 3: Neuroligins control the morphological development of astrocytes in vivo.
figure 3

ad, Representative images of shRNA-transfected L4–5 astrocytes (labelled with EGFP) and NIV reconstructions (magenta) at P7 (top) and P21 (bottom). Astrocytes were electroporated with shRNAs against mouse NL1 (b, shmsNL1), NL2 (c), NL3 (d, shmsNL3) or a scrambled control (a, shCtrl). All EGFP+ cells imaged were hevin-positive, confirming their astrocyte identity (red inlay, corresponding to the somatic region). e, f, Fold change in average NIV for P7 (e) and P21 (f) astrocytes (normalized to shCtrl). Three NIVs per cell, 10–20 cells per condition, 3 or more mice per condition. One-way ANOVA (e, f). Data are means ± s.e.m. Scale bars, 10 μm.

PowerPoint slide

To determine how increasing neuroligin expression alters astrocytic morphogenesis, we overexpressed NL1, NL2, NL3 or NL1-SWAP (control) in astrocytes by PALE. Astrocyte territories were greatly enlarged by overexpression of NL1 or NL2 compared to NL1-SWAP (Extended Data Fig. 6f, g). The NIV of NL1-overexpressing astrocytes did not change, whereas NIV decreased slightly for NL2-overexpressing astrocytes (Extended Data Fig. 6f, h). We were unable to find NL3-overexpressing astrocytes at P21, indicating that NL3 overexpression starting at P1 is not compatible with astrocyte survival and/or maturation. Together, these findings show that neuroligin expression in astrocytes controls neuropil infiltration and territory size of these cells.

Astrocytic NL2 controls synaptogenesis

Because NL2 knockdown markedly impaired astrocyte morphogenesis at P7 and P21, we next investigated the specific in vivo functions of NL2 in astrocytes using Nlgn2-floxed mice (in which the gene that encodes NL2, Nlgn2, is flanked with lox sequences)25. To address the cell-autonomous effects of NL2 on astrocyte development, we sparsely deleted NL2 by introducing Cre via PALE into Nlgn2+/+, Nlgn2f/+ or Nlgn2f/f mice (NL2 PALE WT, HET or KO, respectively). These mice also carried a single allele of the Gt(ROSA)26Sortm14(CAG-tdTomato)Hze (RTM) transgene26 to label Cre positive (Cre+) cells with td-Tomato expression. NL2 expression in td-Tomato/Cre+ astrocytes was greatly diminished, and using these mice and PALE, we confirmed the specificity and effectiveness of our shNL2 construct (Extended Data Fig. 7a–f). Similar to our results with shNL2 PALE experiments, deletion of NL2 in V1 layer 4 (L4) astrocytes decreased NIV. Loss of a single allele of Nlgn2 (NL2 PALE HET) was sufficient to cause a partial but significant decrease in astrocyte infiltration (Fig. 4a, b). Furthermore, loss of both Nlgn2 alleles caused a significant reduction in territory size (Fig. 4c, d). Together, our results show that NL2 has an important function in astrocyte morphogenesis in vivo.

Figure 4: Astrocytic NL2 controls astrocyte morphogenesis and excitatory synapse numbers.
figure 4

a, Images of V1 L4 PALE astrocytes (cyan) and NIVs (green) from the P21 Nlgn2+/+, Nlgn2f/+, and Nlgn2f/f mice labelled with td-Tomato from the Cre-dependent RTM transgene. b, Average NIV of td-Tomato/Cre+ PALE astrocytes. Three NIVs per cell, 16–20 cells per condition, four mice per condition. c, Images of V1 L4 td-Tomato/Cre+ P21 PALE astrocytes (cyan) and their territories (red outlines). d, Average territory volumes of td-Tomato/Cre+ PALE astrocytes. Between 16 and 20 cells per condition, four mice per condition. e, Schematic of local synapse density analysis. Synapse numbers were quantified within regions of interest inside the territories of V1 L4 P21 td-Tomato/Cre+ astrocytes (blue) and neighbouring Cre astrocytes (not visible). f, h, j, Images of intracortical excitatory synapses (f, VGluT1 (red) and PSD95 (green)), thalamocortical excitatory synapses (h, VGluT2 (magenta) and PSD95 (green)), and inhibitory synapses (j, VGAT (magenta) and gephyrin (cyan)). Dotted lines show astrocyte territory boundaries and arrows mark co-localized synaptic puncta. g, i, k, Quantification of average intracortical (g), thalamocortical (i), and inhibitory (k) synaptic co-localized puncta within td-Tomato/Cre+ PALE and Cre astrocyte territories of Nlgn2f/+ and Nlgn2f/f mice. One ROI per territory (Cre+ and Cre) per image, five images per cell, three cells per mouse, four mice per genotype. One-way ANOVA (b, d, g, i, k). Data are means ± s.e.m. Scale bars, 10 μm (a, c, e), 2 μm (f, h, j).

PowerPoint slide

Because astrocytes are key controllers of excitatory and inhibitory synaptogenesis27, we next determined whether astrocytic NL2 is required for proper synaptogenesis. We quantified the synapse density within the territories of NL2 PALE HET and NL2 PALE KO L4 astrocytes and compared it with the density of synapses within the surrounding neuropil infiltrated by WT astrocytes (Fig. 4e). Synapses were labelled by the co-localization of pre- and postsynaptic markers (that is, VGluT1 and PSD95 (intracortical excitatory), VGluT2 and PSD95 (thalamocortical excitatory) and VGAT and gephyrin (inhibitory)). Co-localization of these markers reflects true synapses, as rotating the presynaptic channel by 90° with respect to the postsynaptic channel eliminated most co-localization (Extended Data Fig. 8).

The density of excitatory synapses within the territory of NL2 PALE KO astrocytes was half that of the neighbouring WT astrocytes, whereas loss of a single allele of NL2 did not affect excitatory synapse density within an astrocyte’s domain (Fig. 4f–i). Inhibitory GABA (γ-aminobutyric acid)-ergic synapse density was not altered within the domains of NL2 PALE HET or NL2 PALE KO astrocytes (Fig. 4j, k). These results were unexpected and distinct from the known neuronal role of NL2 as a regulator of inhibitory synapse formation28,29. We found that astrocytic NL2 is essential for local regulation of synapse development in a cell non-autonomous manner by controlling the formation and/or maintenance of excitatory synapses within the territory of a given astrocyte.

Astrocytic NL2 controls synapse function

To investigate how astrocytic NL2 affects the function of cortical synapses, we conditionally deleted Nlgn2 (NL2 cKO) in a large population of astrocytes by combining the Nlgn2 floxed allele containing the RTM transgene with Tg(Slc1a3-cre/ERT)1Nat (GLAST–CreERT2) mice30. Cre-recombination was activated by administering tamoxifen at P10 and P11 and monitored by td-Tomato expression (Extended Data Fig. 9a, b). NL2 (but neither NL1 nor NL3) mRNA was significantly reduced in td-Tomato/Cre+ astrocytes isolated from NL2 cKO cortices by fluorescence-activated cell sorting (FACS) compared to littermate NL2 conditional heterozygous (cHET) mice. However, low levels of NL2 mRNA expression were detected, probably owing to incomplete recombination of both floxed alleles in some Cre+ astrocytes (Extended Data Fig. 9a–e). Notably, GFAP expression did not differ between NL2 cHET and cKO astrocytes (Extended Data Fig. 9e), indicating that NL2 cKO cells retained their astrocyte identity and did not undergo pathological reactivation characterized by enhanced GFAP expression6,31. Genotyping the sorted cells using allele-specific primers verified the recombination of the Nlgn2 locus (Extended Data Fig. 9f–i). In V1 visual cortex essentially all td-Tomato/Cre+ cells expressed GFAP, indicating that Cre expression is restricted to astrocytes. By contrast, 58.7 ± 4.0% (mean ± s.e.m.) of the NL2 cHET or 53.9 ± 4.4% of NL2 cKO astrocytes were td-Tomato/Cre+. Deletion of NL2 did not alter the number or distribution of astrocytes or neurons and the td-Tomato signal was absent from neurons within V1 cortex (Extended Data Fig. 10a–e).

We next performed whole-cell patch-clamp recordings of miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs, respectively) in V1 L5 pyramidal neurons from P21 NL2 cHET and cKO mice (Extended Data Fig. 10f). L5 neurons possess large dendritic trees that project to all cortical layers and receive extensive excitatory and inhibitory synaptic inputs32. The frequency and amplitude of mEPSCs were significantly reduced in NL2 cKO neurons compared to NL2 cHET neurons (Fig. 5a–e). These observations are consistent with our finding that loss of astrocytic NL2 locally decreases excitatory synapse numbers by 50%. Similarly, deletion of NL2 in around 55% of cortical astrocytes reduces the frequency of excitatory synaptic events by about 25%. Together, our data show that astrocytic NL2 is required for the proper formation and function of excitatory synapses in the cortex.

Figure 5: Loss of NL2 in a large population of astrocytes alters excitatory and inhibitory synapse function.
figure 5

a, mEPSC traces from L5 pyramidal neurons in acute V1 slices of P21 NL2 cHET (black) and NL2 cKO (red) mice. b, d, Cumulative distributions of mEPSC frequency (b) and amplitude (d) from NL2 cHET and NL2 cKO pyramidal neurons. c, e, Average neuron mEPSC frequency (c) and amplitude (e) from NL2 cHET and NL2 cKO mice. ae, Between 18 and 19 neurons per genotype. f, mIPSC traces from L5 pyramidal neurons in acute V1 slices of P21 NL2 cHET and NL2 cKO mice. g, i, Cumulative distributions of mIPSC frequency (g) and amplitude (i) from NL2 cHET and NL2 cKO L5 neurons. h, j, Average mIPSC frequency (h) and amplitude (j) from neurons of NL2 cHET and NL2 cKO mice. fj, Between 19 and 20 neurons per genotype. Two sample Kolmogorov–Smirnov test (b, d, g, i), one-sided t-test (c, e, h, j). Data are means ± s.e.m.

PowerPoint slide

We also detected an increase in the frequency of mIPSCs from NL2 cKO neurons compared to cHET neurons, whereas the amplitudes of mIPSCs were indistinguishable (Fig. 5f–j). The elevated frequency of mIPSCs in NL2 cKO neurons might be mediated by an increase in the number of inhibitory synapses. If this is the case, loss of astrocytic NL2 more broadly should enhance inhibitory synaptogenesis. Alternatively, the increase in the frequency of mIPSCs might be due to changes in the presynaptic release properties of inhibitory synapses. Together, these findings reveal a critical and previously unknown role for astrocytic NL2 as an essential governor of excitatory and inhibitory synaptic function in the cortex.

We have uncovered several unknown cell biological aspects of astrocyte–neuron interactions that control the development of cortical astrocytes, and have shown that astrocytic neuroligins have a crucial role in synaptogenesis. Bidirectional signalling via the astrocytic neuroligin and neuronal neurexin adhesions might directly regulate synapse formation and function. Alternatively, astrocytic neuroligins might control synaptic connectivity by altering the expression and/or directed release of synaptogenic factors, such as thrombospondins, SPARCL1 (also known as Hevin) and glypicans33,34,35, from astrocytes. Future studies exploring the link between neuroligin-mediated astrocyte–neuron adhesions and the regulation of astrocyte-induced synaptogenesis are necessary to test these possibilities.

Our findings also challenge the assumption that neuroligins are functional only within neurons in the brain. This is particularly important because gene mutations in neuroligins, including NLGN2, are associated with a number of neurological disorders such as autism and schizophrenia36,37. Neuroligin dysfunction in disease has been postulated to alter the fine balance between inhibition and excitation in the brain38. Here we demonstrate that astrocytic NL2 controls the balance of excitatory and inhibitory synaptic connectivity, indicating that synaptic pathologies associated with neuroligin mutations could originate from astrocytic dysfunction. A recent study found that glial progenitor cells from schizophrenic patients express significantly lower levels of NL1, NL2, and NL3 compared to controls. When these human glial progenitors were injected into mice, they caused neuronal dysfunction, perturbed animal behaviour and yielded abnormal astrocytic morphologies39. In conclusion, our findings reveal how imperative it is to understand the full extent of neuroligin functions in all cell types of the brain to completely comprehend the pathophysiology of these disorders.

Data Availability

The data that support the findings of this study are included in the manuscript. Source Data are provided for Figs 1, 2, 3, 4, 5 and Extended Data Figs 1, 2, 3, 4, 5, 6, 7, 8, 9, 10. Methods are provided within the Supplementary Information.