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.

Regulating anxiety with extrasynaptic inhibition

A Corrigendum to this article was published on 25 November 2015

This article has been updated

Abstract

Aversive experiences can lead to complex behavioral adaptations including increased levels of anxiety and fear generalization. The neuronal mechanisms underlying such maladaptive behavioral changes, however, are poorly understood. Here, using a combination of behavioral, physiological and optogenetic approaches in mouse, we identify a specific subpopulation of central amygdala neurons expressing protein kinase C δ (PKCδ) as key elements of the neuronal circuitry controlling anxiety. Moreover, we show that aversive experiences induce anxiety and fear generalization by regulating the activity of PKCδ+ neurons via extrasynaptic inhibition mediated by α5 subunit-containing GABAA receptors. Our findings reveal that the neuronal circuits that mediate fear and anxiety overlap at the level of defined subpopulations of central amygdala neurons and demonstrate that persistent changes in the excitability of a single cell type can orchestrate complex behavioral changes.

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: Fear conditioning enhances anxiety.
Figure 2: Bidirectional regulation of fear generalization and anxiety through optogenetic control of CEA PKCδ+ neurons.
Figure 3: Extrasynaptic inhibition mediated by α5-GABAARs controls the spontaneous firing of PKCδ+ neurons.
Figure 4: Experience-dependent reduction of extrasynaptic inhibition predicts fear generalization.
Figure 5: Brain area–specific conditional genetic deletion of α5-GABAAR expression induces fear generalization and anxiety behavior.
Figure 6: Brain area– and cell type–specific knockdown of α5-GABAAR expression induces anxiety behavior and fear generalization.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

Change history

  • 05 October 2015

    In the version of this article initially published, the grant number for U.R. was given as R01MH80006 instead of R01MH080006 and grants to J.M.C. from the National Institute of Neurological Disorders and Stroke (R01NS076517) and the National Institute of Mental Health (R01MH096463), US National Institutes of Health, were missing. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Davis, M., Walker, D.L., Miles, L. & Grillon, C. Phasic vs sustained fear in rats and humans: role of the extended amygdala in fear vs anxiety. Neuropsychopharmacology 35, 105–135 (2010).

    Article  Google Scholar 

  2. Blanchard, D.C. If it reduces stress (hormones), do it!. Horm. Behav. 54, 582–583 (2008).

    Article  Google Scholar 

  3. Lissek, S. et al. Classical fear conditioning in the anxiety disorders: a meta-analysis. Behav. Res. Ther. 43, 1391–1424 (2005).

    Article  Google Scholar 

  4. Duvarci, S., Bauer, E.P. & Paré, D. The bed nucleus of the stria terminalis mediates inter-individual variations in anxiety and fear. J. Neurosci. 29, 10357–10361 (2009).

    Article  CAS  Google Scholar 

  5. LeDoux, J.E. Emotion circuits in the brain. Annu. Rev. Neurosci. 23, 155–184 (2000).

    Article  CAS  Google Scholar 

  6. Tovote, P., Fadok, J.P. & Lüthi, A. Neuronal circuits for fear and anxiety. Nat. Rev. Neurosci. 16, 317–331 (2015).

    Article  CAS  Google Scholar 

  7. Johansen, J.P., Wolff, S.B.E., Lüthi, A. & LeDoux, J.E. Controlling the elements: an optogenetic approach to understanding the neural circuits of fear. Biol. Psychiatry 71, 1053–1060 (2012).

    Article  Google Scholar 

  8. Ciocchi, S. et al. Encoding of conditioned fear in central amygdala inhibitory circuits. Nature 468, 277–282 (2010).

    Article  CAS  Google Scholar 

  9. Haubensak, W. et al. Genetic dissection of an amygdala microcircuit that gates conditioned fear. Nature 468, 270–276 (2010).

    Article  CAS  Google Scholar 

  10. Duvarci, S., Popa, D. & Paré, D. Central amygdala activity during fear conditioning. J. Neurosci. 31, 289–294 (2011).

    Article  CAS  Google Scholar 

  11. Li, H. et al. Experience-dependent modification of a central amygdala fear circuit. Nat. Neurosci. 16, 332–339 (2013).

    Article  CAS  Google Scholar 

  12. Alheid, G.F. & Heimer, L. New perspectives in basal forebrain organization of special relevance for neuropsychiatric disorders: the striatopallidal, amygdaloid, and corticopetal components of substantia innominata. Neuroscience 27, 1–39 (1988).

    Article  CAS  Google Scholar 

  13. Tye, K.M. et al. Amygdala circuitry mediating reversible and bidirectional control of anxiety. Nature 471, 358–362 (2011).

    Article  CAS  Google Scholar 

  14. Jennings, J.H. et al. Distinct extended amygdala circuits for divergent motivational states. Nature 496, 224–228 (2013).

    Article  CAS  Google Scholar 

  15. Pellow, S., Chopin, P., File, S.E. & Briley, M. Validation of open:closed arm entries in an elevated plus-maze as a measure of anxiety in the rat. J. Neurosci. Methods 14, 149–167 (1985).

    Article  CAS  Google Scholar 

  16. Farrant, M. & Nusser, Z. Variations on an inhibitory theme: phasic and tonic activation of GABAA receptors. Nat. Rev. Neurosci. 6, 215–229 (2005).

    Article  CAS  Google Scholar 

  17. Chadderton, P., Margrie, T.W. & Hausser, M. Integration of quanta in cerebellar granule cells during sensory processing. Nature 428, 856–860 (2004).

    Article  CAS  Google Scholar 

  18. Semyanov, A., Walker, M.C. & Kullmann, D.M. GABA uptake regulates cortical excitability via cell type-specific tonic inhibition. Nat. Neurosci. 6, 484–490 (2003).

    Article  CAS  Google Scholar 

  19. Bai, D. et al. Distinct functional and pharmacological properties of tonic and quantal inhibitory postsynaptic currents mediated by gamma-aminobutyric acid(A) receptors in hippocampal neurons. Mol. Pharmacol. 59, 814–824 (2001).

    Article  CAS  Google Scholar 

  20. McCartney, M.R., Deeb, T.Z., Henderson, T.N. & Hales, T.G. Tonically active GABAA receptors in hippocampal pyramidal neurons exhibit constitutive GABA-independent gating. Mol. Pharmacol. 71, 539–548 (2007).

    Article  CAS  Google Scholar 

  21. Wlodarczyk, A.I. et al. GABA-independent GABAA receptor openings maintain tonic currents. J. Neurosci. 33, 3905–3914 (2013).

    Article  CAS  Google Scholar 

  22. Brickley, S.G. & Mody, I. Extrasynaptic GABAA receptors: their function in the CNS and implications for disease. Neuron 73, 23–34 (2012).

    Article  CAS  Google Scholar 

  23. Herman, M.A., Contet, C., Justice, N.J., Vale, W. & Roberto, M. Novel subunit-specific tonic GABA currents and differential effects of ethanol in the central amygdala of CRF receptor-1 reporter mice. J. Neurosci. 33, 3284–3298 (2013).

    Article  CAS  Google Scholar 

  24. Heldt, S.A. & Ressler, K.J. Training-induced changes in the expression of GABAA-associated genes in the amygdala after the acquisition and extinction of Pavlovian fear. Eur. J. Neurosci. 26, 3631–3644 (2007).

    Article  Google Scholar 

  25. Tasan, R.O. et al. Altered GABA transmission in a mouse model of increased trait anxiety. Neuroscience 183, 71–80 (2011).

    Article  CAS  Google Scholar 

  26. Harris, D. et al. Selective influence on contextual memory: physiochemical properties associated with selectivity of benzodiazepine ligands at GABAA receptors containing the alpha5 subunit. J. Med. Chem. 51, 3788–3803 (2008).

    Article  CAS  Google Scholar 

  27. Savic´, M.M. et al. PWZ-029, a compound with moderate inverse agonist functional selectivity at GABAA receptors containing α5 subunits, improves passive, but not active, avoidance learning in rats. Brain Res. 1208, 150–159 (2008).

    Article  Google Scholar 

  28. Clément, Y. et al. Gabra5-gene haplotype block associated with behavioral properties of the full agonist benzodiazepine chlordiazepoxide. Behav. Brain Res. 233, 474–482 (2012).

    Article  Google Scholar 

  29. Huber, D., Veinante, P. & Stoop, R. Vasopressin and oxytocin excite distinct neuronal populations in the central amygdala. Science 308, 245–248 (2005).

    Article  CAS  Google Scholar 

  30. Cai, H., Haubensak, W., Anthony, T.E. & Anderson, D.J. Central amygdala PKC-delta neurons mediate the influence of multiple anorexigenic signals. Nat. Neurosci. 17, 1240–1248 (2014).

    Article  CAS  Google Scholar 

  31. Mitchell, S.J. & Silver, R.A. Shunting inhibition modulates neuronal gain during synaptic excitation. Neuron 38, 433–445 (2003).

    Article  CAS  Google Scholar 

  32. Franklin, K.J.B. & Paxinos, G. The Mouse Brain in Stereotaxic Coordinates (Academic, New York, 1997).

  33. Pirker, S., Schwarzer, C., Wieselthaler, A., Sieghart, W. & Sperk, G. GABAA receptors: immunocytochemical distribution of 13 subunits in the adult rat brain. Neuroscience 101, 815–850 (2000).

    Article  CAS  Google Scholar 

  34. Hörtnagl, H. et al. Patterns of mRNA and protein expression for 12 GABAA receptor subunits in the mouse brain. Neuroscience 236, 345–372 (2013).

    Article  Google Scholar 

  35. Ventura, A. et al. Cre-lox-regulated conditional RNA interference from transgenes. Proc. Natl. Acad. Sci. USA 101, 10380–10385 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of the Lüthi laboratory for discussions and critical comments on the manuscript. In particularly, we thank P. Tovote, S. Wolff, S. Ciocchi, F. Grenier, J. Gruendemann, C. Müller, K. Bylund and A. Loche for comments and for technical support, and W. Sieghart (Medical University of Vienna) and D. Anderson (California Institute of Technology) for antibodies and mice. This work was supported by the Novartis Research Foundation, by the National Center of Competences in Research program 'SYNAPSY — The Synaptic Bases of Mental Diseases' (financed by the Swiss National Science Foundation (SNSF)) as well as by an SNSF core grant to A.L. J.P.F. was supported by a NARSAD fellowship. C.X. was supported by an EMBO Long-Term Fellowship. P.S. and L.X. were supported by the Australian National Health and Medical Research Council. U.R. was supported by grant R01MH080006 from the US National Institute of Mental Health, National Institutes of Health. Funding for J.M.C. was provided by the National Institute of Neurological Disorders and Stroke (R01NS076517) and the National Institute of Mental Health (R01MH096463), US National Institutes of Health. Y.K. and F.F. were supported by Austrian Science Fund (Fonds zur Förderung der Wissenschaftlichen Forschung) Sonderforschungsbereich grant F44-17.

Author information

Authors and Affiliations

Authors

Contributions

The manuscript was prepared by A.L. and P.B. Electrophysiological recordings in vitro, behavioral experiments and viral injections were performed by P.B. Pharmacology combined with single-unit recordings in freely moving animals were designed and performed by L.D. In vivo recordings from optogenetically identified neurons were performed by M.M. and J.P.F. Immunohistochemistry was accomplished by C.X. T.L. cloned the conditional shRNA plasmid. M.M.P. and J.M.C. provided the PWZ-029. L.X. tested the shRNA efficacy on α5GABAAR protein in HEK293T cells and P.S. provided the plasmid expressing shRNA. U.R. provided perfused brains of α5−/− mice and of α5fl/fl mice. All electron microscopy experiments were performed and analyzed by Y.K. and F.F.

Corresponding author

Correspondence to Andreas Lüthi.

Ethics declarations

Competing interests

In the last 3 years, U.R. has received compensation for professional services from Concert Pharmaceuticals.

Integrated supplementary information

Supplementary Figure 1 Bidirectional optogenetic modulation of the tonic activity of CEA PKCδ+ neurons in vitro.

a, Top, Cell-attached voltage recording illustrating the blue light-induced increase in firing rate of a PKCδ+ neuron infected with rAAV 2/7 EF1a::DIO-ChR2(H134R)-2A-NpHR-2A-Venus. Bottom, Enlarged traces showing firing rates at time points indicated by numbers. b, Rate histogram of the cell shown in the panel a. c, Blue light-stimulation reversibly increases the firing rate of ChR2-expressing PKCδ+ neurons in acute CEA slices (n = 5 cells/ 5 slices/ 2 mice). *P = 0.009, Kruskal-Wallis One-Way ANOVA on Ranks followed by Tukey’s multiple comparisons test, H = 9.380, 2 degrees of freedom, lightoff vs. lighton. d, Top, Cell-attached voltage recordings illustrating the yellow light-induced decrease in firing rate of a PKCδ+ neuron infected with rAAV 2/5 CBA::DIO-ARCH-GFP. Bottom, Enlarged traces showing firing rates at time points indicated by numbers. e, Rate histogram of the cell shown in the panel d. f, Yellow light-stimulation of reversibly decreases the firing rate of ARCH-expressing PKCδ+ neurons in acute CEA slices (n = 6 cells/ 6 slices/ 2 mice). *P = 0.006, Kruskal-Wallis One-Way ANOVA on Ranks followed by Tukey’s multiple comparisons test, H = 10.140, 2 degrees of freedom, lightoff vs. lighton. All error bars indicate mean ± s.e.m.

Supplementary Figure 2 Modulation of open field anxiety, but not freezing, by manipulation of PKCδ+ neurons.

a, No difference in overall EPM track length during the baseline period (before optogenetic stimulation) between the three experimental groups (ChR2, n = 7 mice; Control, n = 9 mice; ARCH, n = 6 mice; P = 0.766 with one-way ANOVA). b, Optogenetic manipulation of PKCδ+ neurons does not induce freezing in naïve animals in the open field arena (P = 0.656; paired t-test between on/off condition in ChR, t = −0.463, 8 degrees of freedom; P = 0.445; paired t-test between on/off condition in Control, t = −0.790, 12 degrees of freedom; P = 0.464; paired t-test between on/off condition in Arch, t = 0.769, 8 degrees of freedom). c, Blue light stimulation of ChR2-expressing PKCδ+ neurons does not induced freezing behavior in a small arena (ChR2, n = 7 mice; P = 0.124; paired t-test, t = 1.789, 6 degrees of freedom). d, Time spent in the center of the open field arena is bi-directionally modulated by optogenetic manipulations of PKCδ+ neurons (n = 7 mice each group; *P = 0.031, Wilcoxon signed rank test for ARCH, ***P < 0.001 for ChR2, paired t-test, t = 7.039, 6 degrees of freedom, for the control group P = 0.895 between lighton and lightoff condition, t = −0.138, 6 degrees of freedom). e, Overall track length in the open field arena is bi-directionally modulated by optogenetic manipulations of PKCδ+ neurons (n = 7 each group; ***P < 0.001 for ChR2, paired t-test, t = 7.383, 6 degrees of freedom, **P = 0.016, Wilcoxon signed rank test for ARCH, but for the control group P = 0.081 between lighton and lightoff condition, t = 2.100, 6 degrees of freedom). f, Time spent in the center of the open field arena is reduced by the expression of α5-shRNA relative to scrambled control (n = 7 mice each group; **P < 0.001 by unpaired t-test for ChR2, t = 7.039, degrees of freedom; P = 0.895, paired t-test, t = −0.138, 6 degrees of freedom; *P = 0.031, Wilcoxon signed rank paired t-test for ARCH). g, Overall track length in the open field arena is reduced by the expression of α5-shRNA relative to scrambled control (n = 7 each group; *P = 0.011, paired t-test, t = 2.629, 12 degrees of freedom). h, Number of center crossings is reduced by the expression of α5-shRNA relative to scrambled control (n = 7 each group; ***P < 0.001, paired t-test, t = 5.564, 12 degrees of freedom). All error bars indicate mean ± s.e.m.

Supplementary Figure 3 Extrasynaptic inhibition controls excitability of PKCδ+ neurons.

a, Representative current trace illustrating the lack of the effect of CGP 52432, a GABAB receptor antagonist, on extrasynaptic inhibition in a PKCδ+ neuron (scale bar: 20 pA, 1 min). b, Bar graph illustrating lack of effect on extrasynaptic inhibition by the application of 1 or 10 μM CGP 52432 (n = 4 cells/ 4 slices/ 2 mice). Extrasynaptic inhibition is fully blocked by the application of 100 μM PTX (n = 4 cells/ 4 slices/ 2 mice; ***P < 0.001, One-Way ANOVA on Ranks followed by Tukey’s pairwise multiple comparison test, F = 22.394, 2 degrees of freedom). c, Top, Representative current trace illustrating the effect of 1 and 50 μM SR-95531 and 100 μM PTX on extrasynaptic inhibition in a PKCδ+ neuron (scale bar: 20 pA, 1 min); Bottom, Enlarged traces for during baseline, or during application of 1 or 50 μM SR-95531 or 100 μM PTX as indicated by the grey rectangles (scale bar: 20 pA, 300 ms). d, Bar graph illustrating effects on extrasynaptic inhibition by the application of 1 or 50 μM SR-95531 or 100 μM PTX (1 μM SR-95531, n = 6 cells/ 6 slices/ 2 mice; 50 μM SR-95531, n = 15 cells/ 15 slices/ 6 mice; 100 μM PTX, n = 12 cells/ 12 slices/ 6 mice; ***P < 0.011, Mann-Whitney rank sum t-test). Sample t-test vs. control 0: P = 0.259 for 1 μM SR-95531, P = 0.130 for 50 μM SR-95531, P < 0.001 for each concentration of SR-95531. e, Left, Complete blockade of sIPSCs by application of 1 or 50 μM SR-95531 (1 μM: n = 6 cells/ 6 slices/ 2 mice; ***P < 0.001, Wilcoxon signed rank paired t-test; 50 μM: n = 15 cells/ 15 slices/ 6 mice; ***P < 0.001, Wilcoxon signed rank paired t-test). f, Representative traces illustrating membrane potential depolarisation and action potentials elicited by current steps of different amplitudes under baseline conditions, or in the presence of SR95531 (1 μM; blue traces) or Picrotoxin (100 μM; red traces). g, Normalized firing rates plotted against the normalized amplitude of the injected current steps reveals a selective increase in the excitability in the presence of PTX (n = 10), but not SR95531 (n = 10). Scale bar: 20 mV, 400 ms. h, Representative trace illustrating membrane potential changes elicited by a 6 pA depolarizing current step before and after the application of SR95531 (1 μM; blue trace) or PTX (100 μM; red trace). Scale bar: 10 mV, 500 ms. i, j, Application of PTX (n = 6), but not SR95531 (n = 4) increases input resistance (left panel: Rin; *P = 0.010, Mann-Whitney rank sum unpaired t-test SR vs. PTX; middle: n = 4; P = 0.188, paired t-test for SR, t = −1.171, 16 degrees of freedom; right: n = 5; *P = 0.031, Wilcoxon signed rank paired t-test for PTX). Sample t-test vs. control 0: P = 0.259 for 1 μM SR-95531, P = 0.130 for 50 μM SR-95531, P < 0.001 for 100 μM PTX. k, Example traces illustrating action potential firing elicited by injection of a Gaussian white noise current before and after the application of SR95531 (1 μM, blue trace) or PTX (100 μM, red trace). Scale bar: 20 mV, 1 s. l, m, Application of PTX (100 μM; n = 10), but not SR95531 (1 μM; n = 10) induces a depolarising shift in the resting membrane potential (left panel: RMP; *P = 0.044, paired t-test, SR vs. PTX, t = −2.270, 11 degrees of freedom; middle: n = 4; P = 0.428, paired t-test for baseline vs. SR, t = −0.841, 7 degrees of freedom; right: n = 5; *P = 0.008, paired t-test for baseline vs. PTX, t = −3.383, 9 degrees of freedom). Sample t-test vs. control 0: P = 0.214 for 1 μM SR-95531, P = 0.008 for 100 μM PTX. All error bars indicate mean ± s.e.m.

Supplementary Figure 4 Activation mechanisms of extrasynaptic GABAARs.

a, Example current trace (left), and bar graphs (right) illustrating the effect of the sequential co-application of bicuculline (20 μM; BIC), SR95531 (20 μM; SR) and picrotoxin (100 μM; PTX)(n = 6 cells/ 6 slices/ 2 mice). Application of BIC blocks both sIPSCs and, due to its inverse agonistic effect, induces a shift in the holding current. This shift is antagonised by subsequent application of SR95531 indicating that ligand-independent activation of extrasynaptic GABAARs contributes to the total extrasynaptic inhibition in PKCδ+ neurons. Finally, application of PTX, which acts on a different site, completely abolishes extrasynaptic inhibition. Scale bar: 5 pA, 2 min. F(5,10) = 21.17, *P < 0.001, One-Way repeated measures ANOVA followed by Dunnett’s multiple comparisons test. b, Top, Example trace showing sIPSCs recorded from a PKCδ+ neuron during application of tetrodotoxin (1 μM; TTX). Scale bar: 20 pA, 1 s. Bottom, TTX significantly decreases sIPSC frequency in PKCδ+ neurons (n = 7 cells/ 7 slices/ 3 mice; *P = 0.010, paired t-test, t = 3.698, 6 degrees of freedom). c, Top, Example trace showing averaged sIPSC waveforms recorded from a PKCδ+ neuron before (baseline) and during application of TTX (1 μM; scale bar: 8 pA, 12 ms). Bottom, TTX has no effect on sIPSC amplitude (n = 7 cells/ 7 slices/ 3 mice; P = 0.904 paired t-test, t = 0.201, 6 degrees of freedom). d, Bar graph illustrating the effect of TTX and PTX on extrasynaptic inhibition in PKCδ+ neurons (*P = 0.016, ***P < 0.001, sample t-test). e, Regression plot reveals significant correlation between TTX-sensitive extrasynaptic inhibition (pS/pF) and TTX-induced reduction in sIPSC frequency (n = 7 cells/ 7 slices/ 3 mice; P = 0.028; linear regression: R = 0.85). All error bars indicate mean ± s.e.m.

Supplementary Figure 5 Immunohistochemical staining of α5-GABAARs in CEA.

a, Example anti α5-GABAAR staining in CEA of a wild type mouse (Gabra5(+/+)). b, Anti- α5 staining is completely absent in slices from α5 deficient knock-out animals (Gabra5(-/-)). c–e, Double-staining against α5-GABAARs and CFP expressed in PKCδ neurons of PKCδ-Cre animals (PKCδ+ (CFP)) reveals co-localisation. Scale bar: 20 µm. f, Pie chart illustrating the percentage of PKCδ+ neurons expressing α5-GABAARs (n = 5 sections from 5 animals). All error bars indicate mean ± s.e.m.

Supplementary Figure 6 Pharmacology of extrasynaptic inhibition in PKCδ+ neurons.

a, Representative current traces recorded from PKCδ+ neurons showing the effect of the α5-GABAAR inverse agonist ethyl (13aS)-7-methoxy-9-oxo-11,12,13,13a-tetrahydro-9H-imidazo[1,5-a]pyrrolo[2,1-c][1,4]benzodiazepine-1-carboxylate (L-655,708, 50 nM, left trace), of the neurosteroid 3α,5α- tetrahydrodeoxycorticosterone (THDOC; 10 – 100 nM, middle trace), a modulator of δ-containing GABAARs, and of the agonist of α1-containing GABAARs benzodiazepine, N,N-dimethyl-2-(6-methyl-2-p-tolylimidazo[1,2-a]pyridin-3-yl)acetamide (Zolpidem, 20 −300 nM, right trace) on extrasynaptic inhibition. b, Extrasynaptic inhibition (expressed in pS/pF) is reduced by the application of the α5-GABAAR inverse agonist (L-655,708; 50 nM, 5 and 50 μM; n = 6 – 9 cells/ 6 – 9 slices/ 4 – 5 mice). THDOC, 10 – 100 nM, and Zolpidem, 20 to 300 nM, have no significant effect (n = 8 cells/ 8 slices/ 3 mice). The total extrasynaptic inhibition is revealed by the application of picrotoxin (PTX, 100 μM; n = 14 cells/ 14 slices/ 6 mice). *P = 0.019 for PWZ (100 µM), P = 0.024 for L-655,708 (50 nM), **P = 0.004 – 0.009, ***P < 0.001, sample t-test versus the control baseline. P = 0.012, for 50 μM L-655,708 vs. 100 nM THDOC. F(5,48) = 12.90, P < 0.001 for 100 μM PTX vs. all other conditions, Kruskal-Wallis One-Way ANOVA on Ranks followed by Tukey’s multiple comparisons test, H = 53.167, 10 degrees of freedom. c, Top, Representative traces showing sIPSCs recorded from PKCδ+ neuron before (baseline) and during application of PWZ-029 (1 μM). Scale bar: 20 pA, 400 ms. Bottom, Two different inverse agonists at α5-GABAARs (L-655,708 and PWZ-029) have no effect on sIPSC frequency in PKCδ+ neurons (L-655,708; 50 nM, 5 and 50 μM: n = 6 – 9 cells/ 6 – 9 slices/ 4 – 5 mice, P = 0.325 – 0.593, sample t-test; PWZ-029, 1 nM and 1 µM: n = 4 – 5 cells/ 4 – 5 slices/ 2 mice, P = 0.325 – 0.902, sample t-test). d, Top, Representative trace of averaged sIPSC waveforms recorded from PKCδ+ a neuron before (baseline) and during application of PWZ-029. Scale bar: 8 pA, 8 ms. Bottom, Two different inverse agonists at α5-GABAARs (L-655,708 and PWZ-029) have no effect on sIPSC amplitude in PKCδ+ neurons (L-655,708; 50 nM, 5 and 50 μM, n = 6 – 9 cells/ 6 – 9 slices/ 4 – 5 mice, P = 0.144 – 0.208, sample t-test; PWZ-029, 1 nM and 1 μM, n = 4 – 5 cells/ 4 – 5 slices/ 2 mice; P = 0.240 and P = 0.532, sample t-test). All error bars indicate mean ± s.e.m.

Supplementary Figure 7 α5-GABAAR-mediated extrasynaptic inhibition in PKCδ+ versus PKCδ neurons.

a, Top, Representative trace of sIPSCs recorded from a PKCδ neuron before (baseline) and during application of 100 nM or 1 μM PWZ-029. Scale bar: 30 pA, 300 ms. Bottom, α5-GABAAR-mediated extrasynaptic inhibition (expressed in pS/pF) in PKCδ neuron was significantly greater compared to PKCδ+ neurons (n = 5 – 6 cells/ 5 – 6 slices/ 2 mice; *P = 0.021, paired t-test, t = 3.406, 6 degrees of freedom). b, Pre-embedding double-labeling electron microscopy detected α5-GABAAR expression in both PKCδ+ and PKCδ neurons of the CEl. PKCδ+ neurons were revealed by HRP-DAB immunoreactivity whereas immunometal (gold/silver) particles identified α5-GABAAR. Immunometal particles present at the plasma membrane (arrows) were in extrasynaptic locations. In both PKCδ+ and PKCδ neurons, immunometal particles were more frequently observed within the cytoplasmic area, and in particular around mitochondria. Synapses are indicated by arrowheads. Scale bars: upper left panel 1 μm; all others 500 nm. c, Bar graphs show quantification for plasma membrane-associated (PKCδ+: 0.17 ± 0.02, n = 70; PKCδ: 0.48 ± 0.08, n = 58; ***P < 0.001 Mann-Whitney rank sum unpaired t-test) and intracellular (PKCδ+: 3.4 ± 0.3, n = 70; PKCδ: 8.4 ± 0.8, n = 58; ***P < 0.001 Mann-Whitney rank sum unpaired t-test) immunometal particles in PKCδ+ and PKCδ dendrites collected from at least 2 tissue blocks/mouse (n = 2). All error bars indicate mean ± s.e.m.

Supplementary Figure 8 α5-GABAAR-mediated extrasynaptic inhibition controls the firing of CEA neurons in vivo.

a, Schematic representation of the technical procedure. Electrodes and glass pipette or a cannula were implanted in CEA allowing the recording of single units and application of PWZ-029 (PWZ) or vehicle in freely moving animals. b, Example time course histogram of the tonic firing of a CEA single unit (expressed in Hz) before, during and after vehicle application. Light grey bar marks the 10 min application of vehicle. Time bin: 1 min. c, Same as b but for the application of PWZ-029 application (light pink bar). d, Bar graph illustrating the change in firing rate as compared to pre-drug baseline (expressed in Hz) for application of vehicle (n = 8 units from 2 animals) or PWZ-029 (n = 6 units from 2 animals). ***P < 0.001, Mann-Whitney rank sum unpaired t-test. All error bars indicate mean ± s.e.m.

Supplementary Figure 9 Specificity of fear conditioning–induced changes in extrasynaptic inhibition.

a, Representative current traces recorded from PKCδ neurons before (baseline) and during application of 100 μM PTX in slices obtained from animals exposed to the CS only or from fear conditioned animals (CS-US). Scale bar: 20 pA, 5 s. b, Fear conditioning increased extrasynaptic inhibition in PKCδ neurons relative to control animals (CS only: n = 8 cells/ 8 slices/ 5 mice; CS-US: n = 12 cells/ 12 slices/ 7 mice, **P = 0.008 Mann-Whitney rank sum unpaired t-test). c, There was no significant correlation between extrasynaptic inhibition in PKCδ neurons and behavioral fear generalization in fear conditioned animals (n = 7; P = 0.188, R = 0.1). d, Representative current traces recorded from PKCδ+ neurons before (baseline) and during the application of 10 and 100 nM THDOC and 100 μM PTX in CS only and fear conditioned (CS-US) animals. e, THDOC had no effect on extrasynaptic inhibition in PKCδ+ neurons (n = 5 cells / 5 slices/ 2 mice for each group, **P = 0.001, ***P < 0.001, repeated measures One-Way ANOVA, PTX vs. THDOC). f, Fear conditioning does not change the frequency or amplitude spontaneous inhibitory postsynaptic currents (sIPSCs) in PKCδ+ neurons (n = 23 cells/ 8 mice each group; sIPSCs amplitude: P = 0.495, t = 0.689, 44 degrees of freedom, unpaired t-test between CS only and CS-US groups; sIPSCs frequency: P = 0.762, Mann-Whitney rank sum unpaired t-test between CS only and CS-US groups). g, Contextual fear conditioning increases EPM anxiety behaviour. Mice were subjected to five unsignaled USs (n = 11 mice) on day 1 and tested on the EPM on day 2. Similar to animals subjected to cued fear conditioning (CS-US), animals subjected to contextual fear conditioning spent less time on the EPM open arms. h, Following the EPM test, US-only animals were tested for context discrimination (5 min in each context). Contextual fear generalization was quantified as the freezing ratio in a novel context (CTX) vs. the conditioning context (CTX+). Animals exhibited an inverse correlation between the time spent in the open arms (expressed in %) and contextual fear generalisation (CTX/CTX+) (n = 11 mice; P > 0.001, R = 0.8, linear regression). i, Similar to animals subjected to cued fear conditioning (CS-US, n = 26 cells/ 26 slices/ 8 mice, Fig. 4), PKCδ+ neurons recorded in slices obtained from animals subjected to contextual fear conditioning (US only, n = 6 cells/ 6 slices/ 3 mice) exhibited a reduction in the total PTX-sensitive extrasynaptic inhibition compared to PKCδ+ neurons recorded in control animals (P < 0.001, Kruskal-Wallis One-Way ANOVA on Ranks followed by Dunn multiple comparison post hoc test, H = 23.935, 2 degrees of freedom, CS only vs. CS-US or vs. US only). j, Similar to animals subjected to cued fear conditioning (CS-US, 28 cells/ 28 slices/ 8 mice, Fig. 4), PKCδ+ neurons recorded in slices obtained from animals subjected to contextual fear conditioning (US only, n = 6 cells/ 6 slices/ 3 mice) exhibited a reduction in α5-GABAAR-mediated L-655,708-sensitive extrasynaptic inhibition compared to PKCδ+ neurons recorded from control animals (CS only, n = 14 cells/ 14 slices/ 8 mice; F(2,42) = 10.66, **P = 0.004, Kruskal-Wallis One-Way ANOVA on Ranks followed by Dunn multiple comparison post hoc test, H = 11.597, 2 degrees of freedom, CS only vs. CS-US or vs. US only). All error bars indicate mean ± s.e.m.

Supplementary Figure 10 Cre-regulated knockdown of α5 subunits.

a, structure of the plasmid expressing shRNA in a Cre-dependent manner packed into a pAAV virus. The arrow indicates the direction of expression. The right expression of shRNA is observed with Tdtomato expression. b, Bottom, Picture of transiently transfected P19 cells in cell culture with a plasmid expressing GFP and Cre (CMV-CRE-eGFP, green), TdTomato and shRNA (EF1-DIO-U6-Tdtomato-shRNA-KD, red) and the merge picture (yellow). c, Western blots from HEK293T cells transfected with rat α5 subunit and four different RNAi oligonucleotides (RNAi 5.1 to RNAi 5.4; lanes 2-5). dsRed was co-transfected to mark transfected cells. Lane1 show cells transfected with α5 alone, lane6 shows cells transfected with dsRed alone and lane7 are non-transfected cells. Alpha tubulin was used as the loading control. Blots were probed with a α5 antibody. d, RNAi5.2 or a scrambled oligo (Scr) was cloned into a floxed lentilox and HEK 293 cells with transfected with or without Cre-recombinase. Transfected HEK293T cells were harvested 3 days after transfection and western blots probed with an anti-α5 antibody (Novus).

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Botta, P., Demmou, L., Kasugai, Y. et al. Regulating anxiety with extrasynaptic inhibition. Nat Neurosci 18, 1493–1500 (2015). https://doi.org/10.1038/nn.4102

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4102

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