Reducing excessive GABA-mediated tonic inhibition promotes functional recovery after stroke

Andrew N Clarkson, Ben S Huang, Sarah E Macisaac, Istvan Mody, S Thomas Carmichael, Andrew N Clarkson, Ben S Huang, Sarah E Macisaac, Istvan Mody, S Thomas Carmichael

Abstract

Stroke is a leading cause of disability, but no pharmacological therapy is currently available for promoting recovery. The brain region adjacent to stroke damage-the peri-infarct zone-is critical for rehabilitation, as it shows heightened neuroplasticity, allowing sensorimotor functions to re-map from damaged areas. Thus, understanding the neuronal properties constraining this plasticity is important for the development of new treatments. Here we show that after a stroke in mice, tonic neuronal inhibition is increased in the peri-infarct zone. This increased tonic inhibition is mediated by extrasynaptic GABA(A) receptors and is caused by an impairment in GABA (γ-aminobutyric acid) transporter (GAT-3/GAT-4) function. To counteract the heightened inhibition, we administered in vivo a benzodiazepine inverse agonist specific for α5-subunit-containing extrasynaptic GABA(A) receptors at a delay after stroke. This treatment produced an early and sustained recovery of motor function. Genetically lowering the number of α5- or δ-subunit-containing GABA(A) receptors responsible for tonic inhibition also proved beneficial for recovery after stroke, consistent with the therapeutic potential of diminishing extrasynaptic GABA(A) receptor function. Together, our results identify new pharmacological targets and provide the rationale for a novel strategy to promote recovery after stroke and possibly other brain injuries.

Figures

Figure 1. Elevated tonic inhibition in peri-infarct…
Figure 1. Elevated tonic inhibition in peri-infarct cortex
a, Images showing the peri-infarct recording site. Whole-cell patch-clamp recordings were made from post-stroke brain slices, within 200μm of infarct (top left), from layer-2/3 (top right) pyramidal neurons (bottom panels). b, Box-plot (boxes: 25–75%, whiskers:10–90%, lines: median) showing significantly elevated tonic inhibition in peri-infarct cortex (asterisk: P<0.05; see Supplementary Fig. 2 for additional analyses). c,d, Representative traces showing tonic inhibitory currents in control and peri-infarct neurons, respectively. Tonic currents were revealed by the shift in holding currents after blocking all GABAARs with gabazine (>100μM). Cells were voltage-clamped at +10mV.
Figure 2. Post-stroke impairment in GABA transport…
Figure 2. Post-stroke impairment in GABA transport and effect of blocking α5-GABAARs
a, Blocking GAT-1 (NO-711) produced a higher % increase in Itonic after stroke; combined blockade of GAT-1 and GAT-3/4 (NO-711 + SNAP-5114) produced a substantial Itonic increase in controls but only an increase equivalent to blocking GAT-1 alone after stroke. b,c, Itonic in sequential drug applications. Note the lack of response to SNAP-5114 application in the post-stroke slice. d, L655,708 reduced Itonic. e, L655,708 significantly decreased post-stroke Itonic. f, Drug treatment reverted post-stroke Itonic to near-control level (asterisk: P<0.05; n.s.: no significance, bar graphs represent mean ± s.e.m.).
Figure 3. Behavioral recovery after stroke with…
Figure 3. Behavioral recovery after stroke with L655,708 treatment and in Gabra5−/−and Gabrd−/− animals
ac, L655,708 treatment starting from 3-days post-stroke resulted in a dose-dependent improvement in functional recovery post-stroke. df, Gabra5−/− and Gabrd−/− mice also showed decreased motor deficits post-stroke. Functional recovery was assessed with forelimb (a, d) and hindlimb foot-faults (b, e), and on the forelimb asymmetry (c, f). Low-dose L655,708 = 200μg/kg/day per animal; high-dose L655,708 = 400μg/kg/day per animal. Data are ± s.e.m. *** = P≤0.001 stroke + vehicle vs Sham; + = P≤0.05, ## = P≤0.01, # = P≤0.001 vs stroke + vehicle.
Figure 4. Inflection point in L655,708 treatment…
Figure 4. Inflection point in L655,708 treatment effect on infarct size
Representative Nissl stained sections at 7-days post-stroke from stroke + vehicle-treatment (a), stroke + L655,708-treatment starting at the time of stroke (b) and stroke + L655,708-treatment starting from 3-days post-insult (c). Quantification of the stroke volume is shown in panel (d). Data are mean ± s.e.m. for n=4 per group, * = P≤0.05.

References

    1. Cramer SC. Repairing the human brain after stroke: I. Mechanisms of spontaneous recovery. Ann Neurol. 2008;63:272–287.
    1. Brown CE, Aminoltejari K, Erb H, Winship IR, Murphy TH. In vivo voltage-sensitive dye imaging in adult mice reveals that somatosensory maps lost to stroke are replaced over weeks by new structural and functional circuits with prolonged modes of activation within both the peri-infarct zone and distant sites. J Neurosci. 2009;29:1719–1734.
    1. Dijkhuizen RM, et al. Correlation between brain reorganization, ischemic damage, and neurologic status after transient focal cerebral ischemia in rats: a functional magnetic resonance imaging study. J Neurosci. 2003;23:510–517.
    1. Dobkin BH. Training and exercise to drive poststroke recovery. Nat Clin Pract. 2008;4:76–85.
    1. Carmichael ST. Cellular and molecular mechanisms of neural repair after stroke: making waves. Ann Neurol. 2006;59:735–742.
    1. Nudo RJ. Mechanisms for recovery of motor function following cortical damage. Curr Opin Neurobiol. 2006;16:638–644.
    1. Alonso-Alonso M, Fregni F, Pascual-Leone A. Brain stimulation in poststroke rehabilitation. Cerebrovasc Dis. 2007;24(Suppl 1):157–166.
    1. Di Lazzaro V, et al. Motor Cortex Plasticity Predicts Recovery in Acute Stroke. Cereb Cortex. 2009;20:1523–1528.
    1. Hensch TK. Critical period plasticity in local cortical circuits. Nat Rev Neurosci. 2005;6:877–888.
    1. Donoghue JP, Suner S, Sanes JN. Dynamic organization of primary motor cortex output to target muscles in adult rats. II. Rapid reorganization following motor nerve lesions. Exp Brain Res. 1990;79:492–503.
    1. Foeller E, Celikel T, Feldman DE. Inhibitory sharpening of receptive fields contributes to whisker map plasticity in rat somatosensory cortex. J Neurophysiol. 2005;94:4387–4400.
    1. Hess G, Aizenman CD, Donoghue JP. Conditions for the induction of long-term potentiation in layer II/III horizontal connections of the rat motor cortex. J Neurophysiol. 1996;75:1765–1778.
    1. Glykys J, Mody I. Hippocampal network hyperactivity after selective reduction of tonic inhibition in GABA A receptor alpha5 subunit-deficient mice. J Neurophysiol. 2006;95:2796–2807.
    1. Walker MC, Semyanov A. Regulation of excitability by extrasynaptic GABA(A) receptors. Results Probl Cell Differ. 2008;44:29–48.
    1. Collinson N, et al. Enhanced learning and memory and altered GABAergic synaptic transmission in mice lacking the alpha 5 subunit of the GABAA receptor. J Neurosci. 2002;22:5572–5580.
    1. Atack JR, et al. L-655,708 enhances cognition in rats but is not proconvulsant at a dose selective for alpha5-containing GABAA receptors. Neuropharmacology. 2006;51:1023–1029.
    1. Keros S, Hablitz JJ. Subtype-specific GABA transporter antagonists synergistically modulate phasic and tonic GABAA conductances in rat neocortex. J Neurophysiol. 2005;94:2073–2085.
    1. Glykys J, Mody I. Activation of GABAA receptors: views from outside the synaptic cleft. Neuron. 2007;56:763–770.
    1. Yoshiike Y, et al. GABA(A) receptor-mediated acceleration of aging-associated memory decline in APP/PS1 mice and its pharmacological treatment by picrotoxin. PLoS One. 2008;3:e3029.
    1. Cui Y, et al. Neurofibromin regulation of ERK signaling modulates GABA release and learning. Cell. 2008;135:549–560.
    1. Ginsberg MD. Neuroprotection for ischemic stroke: past, present and future. Neuropharmacology. 2008;55:363–38.
    1. Que M, Schiene K, Witte OW, Zilles K. Widespread up-regulation of N-methyl-D-aspartate receptors after focal photothrombotic lesion in rat brain. Neurosci Lett. 1999;273:77–80.
    1. edecker C, Luhmann HJ, Hagemann G, Fritschy JM, Witte OW. Differential downregulation of GABAA receptor subunits in widespread brain regions in the freeze-lesion model of focal cortical malformations. J Neurosci. 2000;20:5045–5053.
    1. Frahm C, et al. Regulation of GABA transporter mRNA and protein after photothrombotic infarct in rat brain. J Comp Neurol. 2004;478:176–188.
    1. Neumann-Haefelin T, et al. Immunohistochemical evidence for dysregulation of the GABAergic system ipsilateral to photochemically induced cortical infarcts in rats. Neuroscience. 1998;87:871–879.
    1. Kharlamov EA, Downey KL, Jukkola PI, Grayson DR, Kelly KM. Expression of GABA A receptor alpha1 subunit mRNA and protein in rat neocortex following photothrombotic infarction. Brain Res. 2008;1210:29–38.
    1. Lee JK, Kim JE, Sivula M, Strittmatter SM. Nogo receptor antagonism promotes stroke recovery by enhancing axonal plasticity. J Neurosci. 2004;24:6209–6217.
    1. Tanaka Y, Furuta T, Yanagawa Y, Kaneko T. The effects of cutting solutions on the viability of GABAergic interneurons in cerebral cortical slices of adult mice. J Neurosci Methods. 2008;171:118–125.
    1. Baskin YK, Dietrich WD, Green EJ. Two effective behavioral tasks for evaluating sensorimotor dysfunction following traumatic brain injury in mice. J Neurosci Methods. 2003;129:87–93.
    1. Ohab JJ, Fleming S, Blesch A, Carmichael ST. A neurovascular niche for neurogenesis after stroke. J Neurosci. 2006;26:13007–13016.
Methods References
    1. Glykys J, Mann EO, Mody I. Which GABA(A) receptor subunits are necessary for tonic inhibition in the hippocampus? J Neurosci. 2008;28:1421–6.
    1. Verheugen JA, Fricker D, Miles R. Noninvasive measurements of the membrane potential and GABAergic action in hippocampal interneurons. J Neurosci. 1999;19:2546–2555.
    1. Barlow R. Cumulative frequency curves in population analysis. Trends Pharmacol Sci. 1990;11:404–406.
    1. Baskin YK, Dietrich WD, Green EJ. Two effective behavioral tasks for evaluating sensorimotor dysfunction following traumatic brain injury in mice. J Neurosci Methods. 2003;129:87–93.
    1. Schallert T, Fleming SM, Leasure JL, Tillerson JL, Bland ST. CNS plasticity and assessment of forelimb sensorimotor outcome in unilateral rat models of stroke, cortical ablation, parkinsonism and spinal cord injury. Neuropharmacology. 2000;39:777–787.
    1. Moore CS, Hebb AL, Blanchard MM, Crocker CE, Liston P, Korneluk RG, Robertson GS. Increased X-linked inhibitor of apoptosis protein (XIAP) expression exacerbates experimental autoimmune encephalomyelitis (EAE) J Neuroimmunol. 2008;203:79–93.

Source: PubMed

3
订阅