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:

Activity-dependent regulation of astrocyte GAT levels during synaptogenesis

Abstract

Astrocytic uptake of GABA through GABA transporters (GATs) is an important mechanism regulating excitatory/inhibitory balance in the nervous system; however, mechanisms by which astrocytes regulate GAT levels are undefined. We found that at mid-pupal stages the Drosophila melanogaster CNS neuropil was devoid of astrocyte membranes and synapses. Astrocyte membranes subsequently infiltrated the neuropil coordinately with synaptogenesis, and astrocyte ablation reduced synapse numbers by half, indicating that Drosophila astrocytes are pro-synaptogenic. Shortly after synapses formed in earnest, GAT was upregulated in astrocytes. Ablation or silencing of GABAergic neurons or disruption of metabotropic GABA receptor 1 and 2 (GABABR1/2) signaling in astrocytes led to a decrease in astrocytic GAT. Notably, developmental depletion of astrocytic GABABR1/2 signaling suppressed mechanosensory-induced seizure activity in mutants with hyperexcitable neurons. These data reveal that astrocytes actively modulate GAT expression via metabotropic GABA receptor signaling and highlight the importance of precise regulation of astrocytic GAT in modulation of seizure activity.

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

Figure 1: Astrocyte infiltration and synaptogenesis are temporally correlated during late metamorphosis.
Figure 2: Genetic ablation of astrocytes during synaptogenesis.
Figure 3: Synapse numbers, but not gross neural architecture, are altered when astrocytes are ablated during late metamorphosis.
Figure 4: GAT is exclusively expressed in astrocytes and activated during synaptogenesis.
Figure 5: GAT expression is sensitive to GABA neurons.
Figure 6: GAT expression is fine-tuned in response to GABA release specifically during synaptogenesis.
Figure 7: GAT expression is modulated through astrocytic metabotropic GABA-Rs.
Figure 8: Regulation of GAT through astrocytic metabotropic GABA-Rs can modulate neurotransmission.

Similar content being viewed by others

References

  1. Ullian, E.M.E., Sapperstein, S.K.S., Christopherson, K.S.K. & Barres, B.A.B. Control of synapse number by glia. Science 291, 657–661 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Schafer, D.P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Allen, N.J. et al. Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors. Nature 486, 410–414 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Eroglu, Ç. et al. Gabapentin receptor α2δ-1 is a neuronal thrombospondin receptor responsible for excitatory CNS synaptogenesis. Cell 139, 380–392 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Christopherson, K.S. et al. Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120, 421–433 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Tasdemir-Yilmaz, O.E. & Freeman, M.R. Astrocytes engage unique molecular programs to engulf pruned neuronal debris from distinct subsets of neurons. Genes Dev. 28, 20–33 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Su, Z.-Z. et al. Insights into glutamate transport regulation in human astrocytes: cloning of the promoter for excitatory amino acid transporter 2 (EAAT2). Proc. Natl. Acad. Sci. USA 100, 1955–1960 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lehre, K.P.K., Levy, L.M.L., Ottersen, O.P.O., Storm-Mathisen, J.J. & Danbolt, N.C.N. Differential expression of two glial glutamate transporters in the rat brain: quantitative and immunocytochemical observations. J. Neurosci. 15, 1835–1853 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Isaacson, J.S., Solís, J.M. & Nicoll, R.A. Local and diffuse synaptic actions of GABA in the hippocampus. Neuron 10, 165–175 (1993).

    Article  CAS  PubMed  Google Scholar 

  10. Rossi, D.J. & Hamann, M. Spillover-mediated transmission at inhibitory synapses promoted by high affinity alpha6 subunit GABAA receptors and glomerular geometry. Neuron 20, 783–795 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Briggs, S.W. & Galanopoulou, A.S. Altered GABA signaling in early life epilepsies. Neural Plast. 2011, 527605 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Dudek, F.E. & Staley, K.J. How does the balance of excitation and inhibition shift during epileptogenesis? Epilepsy Curr. 7, 86–88 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Cope, D.W. et al. Enhanced tonic GABAA inhibition in typical absence epilepsy. Nat. Med. 15, 1392–1398 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jacob, T.C., Moss, S.J. & Jurd, R. GABAA receptor trafficking and its role in the dynamic modulation of neuronal inhibition. Nat. Rev. Neurosci. 9, 331–343 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schousboe, A. Role of astrocytes in the maintenance and modulation of glutamatergic and GABAergic neurotransmission. Neurochem. Res. 28, 347–352 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Sarup, A., Larsson, O.M. & Schousboe, A. GABA transporters and GABA-transaminase as drug targets. Curr. Drug Targets CNS Neurol. Disord. 2, 269–277 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Shigetomi, E., Tong, X., Kwan, K.Y., Corey, D.P. & Khakh, B.S. TRPA1 channels regulate astrocyte resting calcium and inhibitory synapse efficacy through GAT-3. Nat. Neurosci. 15, 70–80 (2012).

    Article  CAS  Google Scholar 

  18. Kersanté, F. et al. A functional role for both GABA transporter-1 and GABA transporter-3 in the modulation of extracellular GABA and GABAergic tonic conductances in the rat hippocampus. J. Physiol. (Lond.) 591 (Pt. 10), 2429–2941 (2013).

    Article  CAS  Google Scholar 

  19. Vitellaro-Zuccarello, L. & Calvaresi, N. Expression of GABA transporters, GAT-1 and GAT-3, in the cerebral cortex and thalamus of the rat during postnatal development. Cell Tissue Res. 313, 245–257 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Freeman, M.R. Specification and morphogenesis of astrocytes. Science 330, 774–778 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Enell, L., Hamasaka, Y., Kolodziejczyk, A. & Nässel, D.R. γ-Aminobutyric acid (GABA) signaling components in Drosophila: immunocytochemical localization of GABAB receptors in relation to the GABAA receptor subunit RDL and a vesicular GABA transporter. J. Comp. Neurol. 505, 18–31 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Küppers, B., Sánchez-Soriano, N., Letzkus, J.J., Technau, G.M. & Prokop, A. In developing Drosophila neurones the production of gamma-amino butyric acid is tightly regulated downstream of glutamate decarboxylase translation and can be influenced by calcium. J. Neurochem. 84, 939–951 (2003).

    Article  PubMed  CAS  Google Scholar 

  23. Neckameyer, W.S. & Cooper, R.L. GABA transporters in Drosophila melanogaster: molecular cloning, behavior, and physiology. Invert. Neurosci. 3, 279–294 (1998).

    Article  CAS  PubMed  Google Scholar 

  24. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jefferis, G.S.X.E. et al. Developmental origin of wiring specificity in the olfactory system of Drosophila. Development 131, 117–130 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Marin, E.C., Watts, R.J., Tanaka, N.K., Ito, K. & Luo, L. Developmentally programmed remodeling of the Drosophila olfactory circuit. Development 132, 725–737 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Zhu, S., Chiang, A.-S. & Lee, T. Development of the Drosophila mushroom bodies: elaboration, remodeling and spatial organization of dendrites in the calyx. Development 130, 2603–2610 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. 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).

    Article  CAS  PubMed  Google Scholar 

  29. Madsen, K.K., White, H.S. & Schousboe, A. Neuronal and non-neuronal GABA transporters as targets for antiepileptic drugs. Pharmacol. Ther. 125, 394–401 (2010).

    Article  CAS  PubMed  Google Scholar 

  30. Benediktsson, A.M. et al. Neuronal activity regulates glutamate transporter dynamics in developing astrocytes. Glia 60, 175–188 (2012).

    Article  PubMed  Google Scholar 

  31. Tanaka, K. et al. Epilepsy and exacerbation of brain injury in mice lacking the glutamate transporter GLT-1. Science 276, 1699–1702 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Devaraju, P., Sun, M.-Y., Myers, T.L., Lauderdale, K. & Fiacco, T.A. Astrocytic group I mGluR dependent potentiation of astrocytic glutamate and potassium uptake. J. Neurophysiol. 109, 2404–2414 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Yang, Y. et al. Presynaptic regulation of astroglial excitatory neurotransmitter transporter GLT1. Neuron 61, 880–894 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Charles, K.J., Calver, A.R., Jourdain, S. & Pangalos, M.N. Distribution of a GABAB-like receptor protein in the rat central nervous system. Brain Res. 989, 135–146 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Uwechue, N.M., Marx, M.-C., Chevy, Q. & Billups, B. Activation of glutamate transport evokes rapid glutamine release from perisynaptic astrocytes. J. Physiol. (Lond.) 590, 2317–2331 (2012).

    Article  CAS  Google Scholar 

  36. Porter, J.T. & McCarthy, K.D. Astrocytic neurotransmitter receptors in situ and in vivo. Prog. Neurobiol. 51, 439–455 (1997).

    Article  CAS  PubMed  Google Scholar 

  37. Hamilton, N.B. & Attwell, D. Do astrocytes really exocytose neurotransmitters? Nat. Rev. Neurosci. 11, 227–238 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Padgett, C.L. & Slesinger, P.A. GABAB receptor coupling to G-proteins and ion channels. Adv. Pharmacol. 58, 123–147 (2010).

    Article  CAS  PubMed  Google Scholar 

  39. Mezler, M., Müller, T. & Raming, K. Cloning and functional expression of GABAB receptors from Drosophila. Eur. J. Neurosci. 13, 477–486 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. 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 

  41. Kaupmann, K. et al. GABAB-receptor subtypes assemble into functional heteromeric complexes. Nature 396, 683–687 (1998).

    Article  CAS  PubMed  Google Scholar 

  42. Dahdal, D., Reeves, D.C., Ruben, M., Akabas, M.H. & Blau, J. Drosophila pacemaker neurons require g protein signaling and GABAergic inputs to generate twenty-four hour behavioral rhythms. Neuron 68, 964–977 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Parker, L., Howlett, I.C., Rusan, Z.M. & Tanouye, M.A. Seizure and epilepsy: studies of seizure disorders in Drosophila. Int. Rev. Neurobiol. 99, 1–21 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Pavlidis, P. & Tanouye, M.A. Seizures and failures in the giant fiber pathway of Drosophila bang-sensitive paralytic mutants. J. Neurosci. 15, 5810–5819 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pavlidis, P., Ramaswami, M. & Tanouye, M.A. The Drosophila easily shocked gene: a mutation in a phospholipid synthetic pathway causes seizure, neuronal failure, and paralysis. Cell 79, 23–33 (1994).

    Article  CAS  PubMed  Google Scholar 

  46. Reynolds, E.R. et al. Treatment with the antiepileptic drugs phenytoin and gabapentin ameliorates seizure and paralysis of Drosophila bang-sensitive mutants. J. Neurobiol. 58, 503–513 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Kuebler, D. & Tanouye, M. Anticonvulsant valproate reduces seizure-susceptibility in mutant Drosophila. Brain Res. 958, 36–42 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Sills, G.J. & Brodie, M.J. Update on the mechanisms of action of antiepileptic drugs. Epileptic Disord. 3, 165–172 (2001).

    CAS  PubMed  Google Scholar 

  49. Kucukdereli, H. et al. Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc. Natl. Acad. Sci. USA 108, E440–E449 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Oka, M., Wada, M., Wu, Q., Yamamoto, A. & Fujita, T. Functional expression of metabotropic GABAB receptors in primary cultures of astrocytes from rat cerebral cortex. Biochem. Biophys. Res. Commun. 341, 874–881 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Doherty, J., Logan, M.A., Taşdemir, O.E. & Freeman, M.R. Ensheathing glia function as phagocytes in the adult Drosophila brain. J. Neurosci. 29, 4768–4781 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ng, M. et al. Transmission of olfactory information between three populations of neurons in the antennal lobe of the fly. Neuron 36, 463–474 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Luo, L., Liao, Y.J., Jan, L.Y. & Jan, Y.N. Distinct morphogenetic functions of similar small GTPases: Drosophila Drac1 is involved in axonal outgrowth and myoblast fusion. Genes Dev. 8, 1787–1802 (1994).

    Article  CAS  PubMed  Google Scholar 

  54. Potter, C.J., Tasic, B., Russler, E.V., Liang, L. & Luo, L. The Q system: a repressible binary system for transgene expression, lineage tracing, and mosaic analysis. Cell 141, 536–548 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lee, T., Lee, A. & Luo, L. Development of the Drosophila mushroom bodies: sequential generation of three distinct types of neurons from a neuroblast. Development 126, 4065–4076 (1999).

    Article  CAS  PubMed  Google Scholar 

  56. Zhang, Y.Q., Rodesch, C.K. & Broadie, K. Living synaptic vesicle marker: synaptotagmin-GFP. Genesis 34, 142–145 (2002).

    Article  CAS  PubMed  Google Scholar 

  57. Grether, M.E., Abrams, J.M., Agapite, J., White, K. & Steller, H. The head involution defective gene of Drosophila melanogaster functions in programmed cell death. Genes Dev. 9, 1694–1708 (1995).

    Article  CAS  PubMed  Google Scholar 

  58. Kitamoto, T. Conditional modification of behavior in Drosophila by targeted expression of a temperature-sensitive shibire allele in defined neurons. J. Neurobiol. 47, 81–92 (2001).

    Article  CAS  PubMed  Google Scholar 

  59. Sweeney, S.T., Broadie, K., Keane, J., Niemann, H. & O'Kane, C.J. Targeted expression of tetanus toxin light chain in Drosophila specifically eliminates synaptic transmission and causes behavioral defects. Neuron 14, 341–351 (1995).

    Article  CAS  PubMed  Google Scholar 

  60. Baines, R.A., Uhler, J.P., Thompson, A., Sweeney, S.T. & Bate, M. Altered electrical properties in Drosophila neurons developing without synaptic transmission. J. Neurosci. 21, 1523–1531 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hamada, F.N. et al. An internal thermal sensor controlling temperature preference in Drosophila. Nature 454, 217–220 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ferris, J., Ge, H., Liu, L. & Roman, G. Go signaling is required for Drosophila associative learning. Nat. Neurosci. 9, 1036–1040 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. McGuire, S.E., Le, P.T., Osborn, A.J., Matsumoto, K. & Davis, R.L. Spatiotemporal rescue of memory dysfunction in Drosophila. Science 302, 1765–1768 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to S. Waddell (University of Oxford), G. Miesenböck (University of Oxford), J. Carlson (Yale University), V. Budnik (University of Massachusetts Medical School), B. Ganetsky (University of Wisconsin-Madison) and M. Tanouye (University of California, Berkley) as well as the Vienna Drosophila RNAi Center and the Bloomington Stock Center for generously providing fly stocks. We thank the Transgenic RNAi Project (TRiP) at Harvard Medical School (US National Institutes of Health NIGMS R01-GM084947) for providing transgenic RNAi fly stocks and/or plasmid vectors. The antibodies nc82, anti-PDF, anti-Fasciclin II and anti-Elav, developed respectively by E. Buchner, J. Blau, C. Goodman and G.M. Rubin, were obtained from the Developmental Studies Hybridoma Bank developed under the auspices of the NICHD and maintained by the University of Iowa. We thank the University of Massachusetts Medical School electron microscopy facility, in particular L. Strittmatter, for expert technical assistance with TEM studies. We thank C. Merlin for expert advice on real-time PCR experiments. We thank N. Fox for comments on the manuscript. We thank D. Bergles and all members of the Freeman laboratory for discussion on the manuscript. The project described was supported by Award Number S10RR027897 from the National Center For Research Resources. T.S. was supported by a postdoctoral fellowship from the Deutsche Forschungsgemeinschaft (DFG). This work was supported by NINDS grant R01NS053538 (to M.R.F.). M.R.F. is an Investigator with the Howard Hughes Medical Institute. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Center For Research Resources or the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

A.K.M. performed the experiments; T.S. generated GAT antibodies; M.R.F. supervised the project; A.K.M. and M.R.F. wrote the manuscript.

Corresponding author

Correspondence to Marc R Freeman.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Astrocytes infiltrate the neuropil throughout the central brain during late metamorphosis

(a) Confocal section through central brain showing astrocyte infiltration during late metamorphosis. Astrocyte membranes are labeled by UAS-mCD8::GFP expression using the alrm-GAL4 driver (green), and neuropil is labeled by nc82 antibody staining (red). Scale bar = 50μm. (b) Confocal section through the AL region showing astrocyte morphology at adult stages. Astrocyte membranes are labeled by UAS-mCD8::GFP expression using the alrm-GAL4 driver (green), and neuropil is labeled by nc82 antibody staining (red). Cell bodies, marked by arrow heads, reside along the periphery of neuropil regions while astrocyte processes are present within the neuropil. Scale bar = 10μm.

Supplementary Figure 2 Initial phases of astrocyte infiltration

Confocal section through the central brain, zoomed in on only a few cells, in order to highlight the change in astrocyte morphology during the initial phases of infiltration. Astrocyte membranes are labeled by UAS-mCD8::GFP expression using the alrm-GAL4 driver (green). Scale bar = 10μm.

Supplementary Figure 3 Identification of astrocytes after ablation procedures

(a) Confocal section through AL of adult animals where astrocytes are labeled by anti-GAT antibody staining (green), neuropil is labeled by anti-HRP antibody staining (red), and glial nuclei are labeled with anti-Repo staining (blue). Repo+ nuclei belonging to GAT+ cells were identified as individual astrocytes within neuropil regions of interest. Scale bar = 10μm. (b) High magnification images of Repo+ nuclei belonging to GAT+ cells in our various regions of interest and ablation conditions. Arrows point to examples of cells that are both Repo+ and GAT+ (astrocytes). Arrow heads point to Repo+ nuclei that do not belong to GAT+ cells. Scale bar = 5μm.

Supplementary Figure 4 Ultrastructure of neuropil after astrocyte ablation

Low magnification image of AL neuropil tissue ultrastructure following the 30°C astrocyte ablation procedure. Asterisks mark mitochondrial structures. Many mitochondrial structures looked unhealthy and ruptured in adult tissue following astrocyte ablations. However, this trend was not observed at earlier stages. Arrows point to structures that we suspect are astrocyte membranes. These structures become extremely difficult to identify following astrocyte ablations. Scale bar = 1μm.

Supplementary Figure 5 Constitutive ablation of astrocytes during late metamorphosis

Confocal section through AL of animals 60 and 84 h APF that were undergoing the 30°C astrocyte ablation procedure. Astrocytes are successfully ablated during development. Astrocytes are labeled by anti-GAT staining (green) and neuropil is labeled by anti-HRP staining (red). Scale bar = 10μm.

Supplementary Figure 6 Astrocyte ablations result in higher frequency of immature synaptic structures at 84 h APF

(a) Quantification of the number of immature synaptic structures in the AL of 84 h APF animals (n = 19 sections, Control; n = 20 sections, Hid) (b) Stacked representation of the number of synapses with the number of immature synaptic structures in the AL of 84 h APF animals (n = 19 sections, Control; n = 20 sections, Hid). (c) Sum of the number of synapses and immature synaptic structures in the AL of 84 h APF animals and adult animals (n = 19 sections, Control 84 h APF; n = 20 sections, Astro> Hid 84 h APF; n = 23 sections, Control Adult; n = 27 sections, Astro>Hid Adult. ***P≤0.001, unpaired Student’s t-test. Error bars, s.e.m.

Supplementary Figure 7 Morphology of cortex glia and ensheathing glia are grossly unaltered following astrocyte ablations

Confocal section through region surrounding AL, showing cortex and ensheathing glia morphology by anti-Draper stain, and location of glial nuclei by anti-Repo stain. Scale bar = 10μm

Supplementary Figure 8 GAT is not expressed in neurons

The pan-neuronal driver, elav-GAL4, was used to express UAS-gat RNAi. Adult brains were stained with anti-GAT and anti-Elav antibodies. Confocal section through region surrounding AL shows that GAT+(red) cells are not Elav+ (green), and that gat knockdown in neurons has no effect on GAT expression. Scale bar = 10μm

Supplementary Figure 9 Temperature shift scheme for GABA neuron ablations

The gad-GAL4 driver and tub-GAL80ts were used to conditionally express UAS-hid in GABA neurons specifically during metamorphosis.

Supplementary Figure 10 Distribution of GABA release sites and astrocyte morphology are grossly unaltered following inhibition of GABA neuron activity during synaptogenesis

The gad-GAL4 driver was used to either co-express UAS-syt::eGFP and UAS-shits or express UAS-syt::eGFP alone. Dominant negative Shits was conditionally expressed during synaptogenesis, and 84 h APF animals were dissected and stained with anti-GAT antibody. Confocal section through AL shows astrocyte morphology (red) and distribution of GABAergic pre-synaptic sites (GABA neuron>Syt::eGFP). Scale bar = 10μm

Supplementary Figure 11 Temperature shift scheme for adult specific inhibition of GABABR1/2 signaling in astrocytes

Tub-GAL80ts with UAS-GABABR1 RNAi or UAS-GABABR2 RNAi were expressed under the control of the alrm-GAL4 driver. RNAi expression was induced in 1 day old adult animals for 7-10 days.

Supplementary Figure 12 Full length blots from the main figures

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 (PDF 9108 kb)

Supplementary Methods Checklist (PDF 396 kb)

Typical behavior of animals following severe astrocyte ablations: control

One day old control animals after undergoing severe ablation temperature shifts (30°C). Animals do not display any overt behavioral defects. (MOV 3049 kb)

Typical behavior of animals following severe astrocyte ablations: fully eclosed

Example of behavior displayed by 1 day old animals following severe (30°C) astrocyte ablation. Flies are fallen over with non-inflated wings. Movement is mostly in the legs and sometimes in the proboscis. This is in striking contrast to control animals (Supplementary Video 1). (MOV 3768 kb)

Typical behavior of animals following severe astrocyte ablations: partially eclosed

Example of an animal that partially eclosed after severe astrocyte ablation. This animal is still alive, demonstrated by its ability to extend its proboscis. (MOV 3287 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Muthukumar, A., Stork, T. & Freeman, M. Activity-dependent regulation of astrocyte GAT levels during synaptogenesis. Nat Neurosci 17, 1340–1350 (2014). https://doi.org/10.1038/nn.3791

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3791

This article is cited by

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