supporting information - pnas · supporting information ... (nikon a1) and analyzed by ... with a...

11
Supporting Information Clasadonte et al. 10.1073/pnas.1311967110 SI Materials and Methods Animals. We used a line of transgenic micedominant-negative SNARE (dnSNARE) micein which there is an astrocyte- specic expression of an inducible dominant-negative form of synaptobrevin II that inhibits the release of chemical transmitters, including ATP. The generation of the dnSNARE mice has been previously reported (1). Bigenic GFAP.tTA and tetO.dnSNARE transgenic animals (i.e., dnSNARE mice) and their WT litter- mates were used. GFAP.tTA mice and tetO.dnSNARE mice were backcrossed at least 10 generations onto the C57BL/6J background before intercrossing. Mice were bred and housed in a room with controlled photoperiods (12 h light and 12 h darkness) and temperature (2123 °C) with food and water ad libitum. WT and dnSNARE mice were fed a diet containing doxycycline (40 mg/kg) from conception until weaning (3 wk of age). All procedures were in strict accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Tufts University Institutional Animal Care and Use Committee. Pilocarpine-Induced Epilepsy, Video EEG Monitoring, and Seizure Classication. Adult male dnSNARE and WT mice (2 mo old; weight, >20 g) were used in these experiments. WT and dnSNARE mice were maintained on doxycycline containing food until weaning (postnatal day 21), when they were switched to a regular diet to allow the transgene expression in the dnSNARE animals. Thirty minutes before the injections of pilocarpine, methyl- scopolamine (a muscarinic antagonist) was administered in- traperitoneally (1 mg/kg; Sigma) to reduce adverse, peripheral effects. Then, mice were intraperitoneally injected with repeated low-dose pilocarpine hydrochloride (a muscarinic agonist, 100 mg/kg; Sigma) every 20 min until onset of status epilepticus (SE) characterized by continuous tonic-clonic seizures. Ninety mi- nutes after the onset of SE, diazepam (4 mg/kg; Hospira) was administered s.c. to reduce seizure activity. Mice that did not develop SE after the fourth injection of pilocarpine were used as controls. After SE, all animals were intraperitoneally injected with lactated Ringer solution and fed with soaked rodent food. The seizure severity was scored according to the scale of Racine (2): score 0, no signs of motor seizure activity; score 1, immo- bility, rigid posture; score 2, repetitive movements, head bob- bing; score 3, severe seizures with rearing without falling; score 4, severe seizures with rearing and falling, running and jumping, loss of righting ability; and score 5, SE characterized by contin- uous tonic-clonic seizures. The electrographic features of spontaneous recurrent seizures (SRSs) were analyzed with a video EEG monitoring system (Pinnacle Technology). Two days after SE, mice were anes- thetized with isourane and placed into a stereotaxic frame. Skull surface was exposed, and four insulated wire electrodes (Cooner Wire) were placed and screwed as follows: two extradural cortical electrodes were inserted bilaterally in the frontal areas and the two others were inserted bilaterally in the parietal areas. Elec- trodes connected to a microconnector (Pinnacle Technology) were secured at the surface of the skull with dental acrylic. After surgery, mice were intraperitoneally injected with buprenorphine (0.08 mg/kg) and lactated Ringer solution, fed with soaked rodent food, and allowed to recover for 3 d before recording. Freely moving mice were placed into individual Plexiglas circle boxes (Pinnacle Technology) and their microconnectors plugged to an EEG preamplier (Pinnacle Technology). The electrical signal was ltered (pass-band, 1100 Hz), digitized at 200 Hz, and acquired with a computer-based system (Pinnacle Technology). Mice were considered epileptic when more than two SRSs were observed. To detect seizures, EEG recordings were automati- cally screened with SleepSign for Animal software on the basis of 4-s epochs and analyzed using a fast Fourier power spectral analysis. Power was analyzed in ve frequency bands: δ (14 Hz), θ (48 Hz), α (813 Hz), β (1330 Hz), and γ (3040 Hz). An electrographic seizure was characterized by high EEG activity along with a typical increase in the β-band power (Fig. 1B and Fig. S2). The detection of each seizure was visually conrmed by using video recording. EEG interictal spikes were clearly dis- tinguishable from baseline EEG activity by their repetitive oc- currence, large amplitude, and sharp morphology (3). In few cases, spikes with amplitudes that saturated the amplier inputs occurred. They were always associated with movement artifacts and excluded from the analysis. Spike counting was performed with Spike2 software (CED). To examine the electrographic features of SE, mice were implanted with EEG as described earlier 5 d before induction of SE. In these experiments, SE was induced with a single high dose of pilocarpine hydrochloride (333 mg/kg, i.p.; Sigma). Open-Field Test. Eight months after injections of pilocarpine, mice implanted with EEG electrodes (SE and control mice) were subjected to the open-eld test. Mice were allowed to acclimate to the experimental room for 30 min before behavioral testing. The experiment was performed only if no motor seizure was observed for at least 1 h before the test. Mice were placed individually for 30 min in the center of an opaque-sided box (measuring 44 cm long × 44 cm wide × 44 cm high). A video camera was placed above the open eld, and a video tracking system was used to evaluate the exploratory behavior. Before each trial, the eld was cleaned thoroughly with 70% (vol/vol) ethanol solution and dried with paper towels. Mice that experienced SRS during a trial were excluded from the analysis. Immunouorescence Staining. After completion of the behavioral study, mice were deeply anesthetized with a mixture of ketamine hydrochloride and xylazine hydrochloride. They were perfused sequentially via the left ventricle with 20 mL chilled PBS solution (1×) and 100 mL 4% (wt/vol) paraformaldehyde in PBS solution (1×, pH 7.4). Brains were removed and postxed in the same xative for 24 h at 4 °C. After xation, they were soaked 24 h in a solution of 30% (wt/vol) sucrose. Brains were cut serially at 200-μm intervals on a freezing microtome (SM2000R; Leica) into coronal sections at a thickness of 40 μm and then stored in an antifreeze solution at -20 °C until immunostaining. Im- munostaining was conducted on free-oating sections incubated with 5% (vol/vol) normal goat serum blocking solution for 1 h at room temperature and then with primary antibodies overnight at 4 °C on a shaking platform. The following antibodies were used: mouse anti-NeuN (1:1,000; Chemicon), rabbit anti-GFAP (1:1,000; Sigma), and chicken anti-vimentin (1:1,000; Abcam). Sections were rinsed three times with 1× PBS solution and then incubated with secondary antibodies for 2 h at room temperature. The following secondary antibodies were used: goat anti-mouse Alexa 546 (1:1,000; Invitrogen), goat anti-rabbit Alexa 633 (1:1,000; In- vitrogen), and goat anti-chicken Alexa 488 (1:1,000; Invitrogen). For biocytin staining, slices were xed overnight with 4% (wt/vol) paraformaldehyde in PBS solution (1×, pH 7.4) at 4 °C, then rinsed several times with PBS solution and incubated with 0.4% Triton X-100/PBS solution and streptavidin Alexa 546 (1:250; Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 1 of 11

Upload: voliem

Post on 29-Aug-2018

212 views

Category:

Documents


0 download

TRANSCRIPT

Supporting InformationClasadonte et al. 10.1073/pnas.1311967110SI Materials and MethodsAnimals. We used a line of transgenic mice—dominant-negativeSNARE (dnSNARE) mice—in which there is an astrocyte-specific expression of an inducible dominant-negative form ofsynaptobrevin II that inhibits the release of chemical transmitters,including ATP. The generation of the dnSNARE mice has beenpreviously reported (1). Bigenic GFAP.tTA and tetO.dnSNAREtransgenic animals (i.e., dnSNARE mice) and their WT litter-mates were used. GFAP.tTA mice and tetO.dnSNARE micewere backcrossed at least 10 generations onto the C57BL/6Jbackground before intercrossing. Mice were bred and housed ina room with controlled photoperiods (12 h light and 12 hdarkness) and temperature (21–23 °C) with food and water adlibitum. WT and dnSNARE mice were fed a diet containingdoxycycline (40 mg/kg) from conception until weaning (3 wk ofage). All procedures were in strict accordance with the NationalInstitutes of Health Guide for the Care and Use of LaboratoryAnimals and were approved by the Tufts University InstitutionalAnimal Care and Use Committee.

Pilocarpine-Induced Epilepsy, Video EEG Monitoring, and SeizureClassification. Adult male dnSNARE and WT mice (2 mo old;weight, >20 g) were used in these experiments. WT and dnSNAREmice were maintained on doxycycline containing food untilweaning (postnatal day 21), when they were switched to a regulardiet to allow the transgene expression in the dnSNARE animals.Thirty minutes before the injections of pilocarpine, methyl-scopolamine (a muscarinic antagonist) was administered in-traperitoneally (1 mg/kg; Sigma) to reduce adverse, peripheraleffects. Then, mice were intraperitoneally injected with repeatedlow-dose pilocarpine hydrochloride (a muscarinic agonist, 100mg/kg; Sigma) every 20 min until onset of status epilepticus (SE)characterized by continuous tonic-clonic seizures. Ninety mi-nutes after the onset of SE, diazepam (4 mg/kg; Hospira) wasadministered s.c. to reduce seizure activity. Mice that did notdevelop SE after the fourth injection of pilocarpine were used ascontrols. After SE, all animals were intraperitoneally injectedwith lactated Ringer solution and fed with soaked rodent food.The seizure severity was scored according to the scale of Racine(2): score 0, no signs of motor seizure activity; score 1, immo-bility, rigid posture; score 2, repetitive movements, head bob-bing; score 3, severe seizures with rearing without falling; score 4,severe seizures with rearing and falling, running and jumping,loss of righting ability; and score 5, SE characterized by contin-uous tonic-clonic seizures.The electrographic features of spontaneous recurrent seizures

(SRSs) were analyzed with a video EEG monitoring system(Pinnacle Technology). Two days after SE, mice were anes-thetized with isoflurane and placed into a stereotaxic frame. Skullsurface was exposed, and four insulated wire electrodes (CoonerWire) were placed and screwed as follows: two extradural corticalelectrodes were inserted bilaterally in the frontal areas and thetwo others were inserted bilaterally in the parietal areas. Elec-trodes connected to a microconnector (Pinnacle Technology)were secured at the surface of the skull with dental acrylic. Aftersurgery, mice were intraperitoneally injected with buprenorphine(0.08 mg/kg) and lactated Ringer solution, fed with soaked rodentfood, and allowed to recover for 3 d before recording. Freelymoving mice were placed into individual Plexiglas circle boxes(Pinnacle Technology) and their microconnectors plugged to anEEG preamplifier (Pinnacle Technology). The electrical signalwas filtered (pass-band, 1–100 Hz), digitized at 200 Hz, and

acquired with a computer-based system (Pinnacle Technology).Mice were considered epileptic when more than two SRSs wereobserved. To detect seizures, EEG recordings were automati-cally screened with SleepSign for Animal software on the basis of4-s epochs and analyzed using a fast Fourier power spectralanalysis. Power was analyzed in five frequency bands: δ (1–4 Hz),θ (4–8 Hz), α (8–13 Hz), β (13–30 Hz), and γ (30–40 Hz). Anelectrographic seizure was characterized by high EEG activityalong with a typical increase in the β-band power (Fig. 1B andFig. S2). The detection of each seizure was visually confirmed byusing video recording. EEG interictal spikes were clearly dis-tinguishable from baseline EEG activity by their repetitive oc-currence, large amplitude, and sharp morphology (3). In fewcases, spikes with amplitudes that saturated the amplifier inputsoccurred. They were always associated with movement artifactsand excluded from the analysis. Spike counting was performedwith Spike2 software (CED).To examine the electrographic features of SE, mice were

implanted with EEG as described earlier 5 d before induction ofSE. In these experiments, SE was induced with a single high doseof pilocarpine hydrochloride (333 mg/kg, i.p.; Sigma).

Open-Field Test.Eight months after injections of pilocarpine, miceimplanted with EEG electrodes (SE and control mice) weresubjected to the open-field test. Mice were allowed to acclimate tothe experimental room for 30 min before behavioral testing. Theexperiment was performed only if no motor seizure was observedfor at least 1 h before the test. Mice were placed individually for30 min in the center of an opaque-sided box (measuring 44 cmlong × 44 cm wide × 44 cm high). A video camera was placedabove the open field, and a video tracking system was used toevaluate the exploratory behavior. Before each trial, the fieldwas cleaned thoroughly with 70% (vol/vol) ethanol solution anddried with paper towels. Mice that experienced SRS duringa trial were excluded from the analysis.

Immunofluorescence Staining. After completion of the behavioralstudy, mice were deeply anesthetized with a mixture of ketaminehydrochloride and xylazine hydrochloride. They were perfusedsequentially via the left ventricle with 20 mL chilled PBS solution(1×) and 100 mL 4% (wt/vol) paraformaldehyde in PBS solution(1×, pH 7.4). Brains were removed and postfixed in the samefixative for 24 h at 4 °C. After fixation, they were soaked 24 h ina solution of 30% (wt/vol) sucrose. Brains were cut serially at200-μm intervals on a freezing microtome (SM2000R; Leica)into coronal sections at a thickness of 40 μm and then stored inan antifreeze solution at −20 °C until immunostaining. Im-munostaining was conducted on free-floating sections incubatedwith 5% (vol/vol) normal goat serum blocking solution for 1 h atroom temperature and then with primary antibodies overnight at4 °C on a shaking platform. The following antibodies were used:mouse anti-NeuN (1:1,000; Chemicon), rabbit anti-GFAP (1:1,000;Sigma), and chicken anti-vimentin (1:1,000; Abcam). Sections wererinsed three times with 1× PBS solution and then incubated withsecondary antibodies for 2 h at room temperature. The followingsecondary antibodies were used: goat anti-mouse Alexa 546(1:1,000; Invitrogen), goat anti-rabbit Alexa 633 (1:1,000; In-vitrogen), and goat anti-chicken Alexa 488 (1:1,000; Invitrogen).For biocytin staining, slices were fixed overnight with 4% (wt/vol)paraformaldehyde in PBS solution (1×, pH 7.4) at 4 °C, thenrinsed several times with PBS solution and incubated with 0.4%Triton X-100/PBS solution and streptavidin Alexa 546 (1:250;

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 1 of 11

Invitrogen) for 4 h at room temperature. All sections werecounterstained with DAPI, mounted on slides, and coverslippedwith Vectashield antifade mounting medium. The fluorescentimages were acquired with a confocal laser scanning microscope(Nikon A1) and analyzed by using ImageJ and MetaMorph.Areas CA1 and CA3 and the dentate gyrus (DG) of hippo-campus were independently outlined, and an intensity thresholdwas set to identify pixels that were GFAP- and vimentin-positive.The proportion of total pixels containing GFAP and vimentinimmunoreactivity was then determined. NeuN-positive cells werecounted, and the density of neurons in CA1, CA3, and DG wasplotted and analyzed.

In Vivo Pharmacological Manipulation. Osmotic minipumps (model1002; Alzet; flow rate, 0.25 μL/h; 2-wk duration) were filled withthe NMDA receptor (NMDAR) antagonist D-(-)-2-amino-5-phosphonopentanoate [D-AP5; 5 mM in artificial cerebrospinalfluid (ACSF); Tocris] or vehicle (ACSF for a group of controlmice), secured with the flow moderator, and primed overnight in0.9% saline solution at 37 °C before to be connected to the braincannula (Alzet Brain Infusion Kit 3) by flexible catheter tubing.Two days after SE, mice were implanted with EEG as describedearlier, and the brain cannula was stereotaxically placed into theright lateral ventricle (anteroposterior, −0.2 mm; mediolateral,−1.0 mm; dorsoventral, −2.2 mm). Animals were allowed torecover for 3 d. Osmotic minipumps were externalized, placed ina sealed conic tube of 1 mL filled with 0.9% saline solution, andmaintained at 37 °C with a iBlock Mini Dry Bath (Midsci) placedout of the cage. The catheter tubing was filled with D-AP5 (5 mMin ACSF; Tocris) or vehicle (ACSF for a group of control mice).A small air bubble was inserted into the catheter tubing duringits connection with the pump to monitor the flow rate estimatedat 0.25 μL/h in our conditions. The flexible catheter tubing wascarefully aligned and attached to the EEG cable. The presenceof a swivel (Pinnacle Technology) prevented the torsion of tub-ing and cable.

Slice Preparation. Coronal cortical–hippocampal slices (310–400μm) were obtained from dnSNARE and WT mice at postnataldays 21 to 30. At birth, WT and dnSNARE mice that were to beused for brain slice studies were fed a doxycycline-free diet toallow transgene expression in the dnSNARE mice. Mice wereanesthetized with isoflurane, and, after cervical dislocation, thebrain was rapidly removed and put in ice-cold artificial ACSFcontaining the following (in mM): 120 NaCl, 3.2 KCl, 1 NaH2PO4,26 NaHCO3, 1 MgCl2, 2 CaCl2 and 10 glucose, pH 7.4 (with O295%, CO2 5%). After removal of the cerebellum, the brain wasglued and coronal cortical–hippocampal slices were cut by using aVibratome (VT1200S; Leica). Before recording, slices were incu-bated at 35 °C for a recovery period of 1 h. After recovery, sliceswere placed in a submerged recording chamber (31 °C; WarnerInstruments) and continuously perfused (2 mL/min) with oxygen-ated ACSF.

Extracellular Recordings.Coronal cortical–hippocampal slices (400μm) were visualized with a 16× objective in an upright NikonEclipse FN1 microscope. Glass electrodes (3–4 MΩ) for extra-cellular recording were filled with ACSF. Electrophysiologicalsignals were amplified and filtered at 0.1 Hz and 1 kHz bya Multiclamp 700B amplifier (Axon Instruments), digitized at5 kHz with Digidata 1322A (Axon Instruments), and stored ina personal computer with Clampex 9.2 (Axon Instruments).Epileptiform activity was generated with a proconvulsant ACSFcontaining 0 Mg2+ to relieve the blockade of NMDA receptorsand the GABAA receptor antagonist picrotoxin (100 μM; Sigma),and recorded in the CA1 pyramidal cells layer. Epileptiformevents were detected with Clampfit 9.2 (Axon Instruments) andverified visually.

Patch-Clamp Recordings. CA1 pyramidal neurons from coronalcortical–hippocampal slices (310 μm) were visually identifiedwith a 16× objective and 2× magnification in an upright NikonEclipse FN1 microscope by using IR-differential interferencecontrast. Whole-cell patch-clamp recordings were performed incurrent- and voltage-clamp modes by using a Multiclamp 700Bamplifier (Axon Instruments). Data were filtered at 1 kHz andsampled at 5 kHz with Digidata 1322A interface and pClampsoftware (Axon Instruments).For the AMPA/NMDA ratio measurements, a glass pipette

(3–4 MΩ) was filled with the following internal solution (in mM):120 Cs-MeSO3, 20 CsCl, 0.2 EGTA, Na-phosphocreatine 10, 5QX-314, 4 Mg-ATP, 0.2 Na3-GTP, and 10 Hepes, pH 7.3 withCsOH. Biocytin (2 mg/mL; Sigma) was added to the internalsolution in some experiments. Biocytin was allowed to passivelydiffuse into the cell for at least 10 min. Synaptic AMPA andNMDA currents were evoked by electrostimulation of theSchaffer collateral pathway at 0.033 Hz with a bipolar extracel-lular electrode (FHC) placed 100 to 200 μM apart from therecorded cell. Picrotoxin (100 μM; Sigma) was always added tothe ACSF while measuring AMPA/NMDA ratio. After a stablesynaptic current was obtained, AMPA currents were first re-corded at −70 mV, then the AMPA receptor antagonist 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide (10μM; Tocris) was added to the ACSF, and finally the NMDAcurrent was measured at +40 mV. The NMDAR antagonistD-AP5 (50 μM; Tocris) was added to the perfusion system at theend of the recording to confirm the nature of the evoked synapticcurrent. During AMPA/NMDA ratio measurements, a high-Mg2+

and -Ca2+ (4 mM of MgCl2 and CaCl2) ACSF was always used toincrease membrane stability in the presence of picrotoxin. AMPAand NMDA currents were analyzed with Clampfit 9.2 (Axon In-struments). In some experiments, spontaneous excitatory post-synaptic currents (EPSCs; sEPSCs) were recorded at –70 mVbefore recording evoked AMPA and NMDA currents and ana-lyzed using Minianalysis 6.0 software (Synaptosoft).To measure the AMPA and NMDA components of miniature

EPSCs (mEPSCs), cells were recorded at holding potential −70mV in ACSF containing 0 Mg2+ and 1 μM TTX (Tocris). In-ternal solution contained the following (in mM): 145 K-gluco-nate, 2 MgCl2, 5 EGTA, 2 Na2-ATP, 0.2 Na3-GTP, and 10Hepes, pH 7.2 with KOH. Analysis of mEPSCs was performedby using Minianalysis 6.0 software (Synaptosoft). Events weredetected automatically by using a threshold of 15 pA and verifiedvisually. The average mEPSC waveform of each cell was alignedat their half-maximal rise time. The peak of an average mEPSCwas taken as the AMPA peak. NMDA current amplitude wasmeasured 20 ms after the AMPA peak. Before recording mixedAMPA and NMDA mEPSCs, cells were recorded in normalACSF containing 1 mM Mg2+ and a series of constant currentpulses (from −300 to +300 pA for 300 ms with 20-pA incre-ments, at 1-s intervals) was used to characterize their intrinsicmembrane properties and excitability. Intrinsic membrane prop-erties of CA1 pyramidal cells were not altered in dnSNARE mice(n = 15 cells from five animals) compared with WT mice (n = 11cells from three animals). No difference was found in their restingmembrane potential (WT, −74.00 ± 0.55 mV, vs. −72.46 ± 1.34mV in dnSNARE; Student t test, P > 0.05) and membrane inputresistance (WT, 146.94 ± 11.59 MΩ, vs. 154.16 ± 8.53 MΩ indnSNARE; Student t test, P > 0.05). We also observed nodifference in their excitability and the amplitude of evokedaction potentials measured at rheobase (WT, 88.36 ± 2.28 mV,vs. 83.06 ± 2.80 mV in dnSNARE; Student t test, P > 0.05).Junction potentials were determined to allow correction ofmembrane potential values.

Glutamate and D-Serine Biosensor Recordings. Biosensor micro-electrodes (Sarissa Biomedical) coated with an enzymatic matrix

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 2 of 11

surrounding a platinum electrode (50-μm diameter) were po-larized to +500 mV (vs. Ag/AgCl reference). Electrochemicaldetection occurred via amperometric measurement of hydrogenperoxide produced by the degradation reaction mediated by theenzymes included in the matrix. The current generated wasproportional to the concentration of the analyte of interest.Glutamate release was measured by using a glutamate sensor,comprising glutamate oxidase and using a null sensor (possessingonly the polymeric coat without enzymes), as background signals.Signals from the null sensor were subtracted from the signalsgenerated by the glutamate sensor to give a measure of netglutamate concentrations. D-Serine release was measured byusing a D-serine sensor, comprising D-amino acid oxidase andusing a null sensor, as background. The principles and opera-tion of glutamate and D-serine sensors have been previouslydescribed (4–7).Pairs of sensors (glutamate/null or D-serine/null; 150–200 μm

apart) were inserted sequentially into stratum radiatum of areaCA1 of cortical–hippocampal slices (400 μm) continuously per-fused (2 mL/min) with oxygenated ACSF at 31 °C. Experimentscommenced 30 min after the slices recover and the sensor signalasymptotes to a baseline level. Experiments consisted of col-lecting 20 min of stable baseline, sensor withdrawal, and sensorcalibration. Upon sensor withdrawal, the glutamate and D-serinesignals decreased below the preceding baseline level recorded inthe slices. Subtraction of the pre- and postwithdrawal signals wasused as an indirect measure of glutamate or D-serine tone.Calibration was performed after each experiment by washing in

10 μM L-glutamate (Sigma) or D-serine (Sigma). The θ-burststimulation (TBS) of the Schaffer collaterals was performed witha bipolar extracellular electrode (FHC) positioned in the stratumradiatum 150 to 200 μm from the glutamate sensor. The TBSprotocol was conducted 20 min after a stable baseline signal andconsisted of one single train of five θ-bursts (75–100 μA) at 100Hz separated by 200 ms (8). Measurements were taken at thepeak amplitude of the response. Potentiostat-based recordingswere made by using the ME200+ Duo-Stat (Sycopel Interna-tional) and sampled at 10 kHz with Digidata 1322A interfaceand pClamp software (Axon Instruments). Analysis was performedwith Clampfit 9.2 (Axon Instruments).

Statistical Analysis. The difference between several groups wasanalyzed by one-way ANOVA. Two-way ANOVA was used whenmore than two comparisons (with time and genotype) were made.Repeated-measures ANOVAs were used when multiple mea-surements were made over time in the same groups. ANOVAwasfollowed by a Student–Newman–Keuls post hoc multiple com-parisons test and Bonferroni test for all behavior experiments.Comparisons between two groups were conducted with thepaired or unpaired Student t test or the nonparametric Mann–Whitney test as appropriate. Proportions were compared asappropriate with the Fisher exact test or χ2 test. Cumulativeprobability plots were calculated, and a Kolmogorov–Smirnovtest was used to determine statistical differences. Kaplan–Meieranalysis was performed, and the log-rank test was used to de-termine statistical differences.

1. Pascual O, et al. (2005) Astrocytic purinergic signaling coordinates synaptic networks.Science 310(5745):113–116.

2. Racine RJ (1972) Modification of seizure activity by electrical stimulation. II. Motorseizure. Electroencephalogr Clin Neurophysiol 32(3):281–294.

3. White A, et al. (2010) EEG spike activity precedes epilepsy after kainate-induced statusepilepticus. Epilepsia 51(3):371–383.

4. Tian F, Gourine AV, Huckstepp RT, Dale N (2009) A microelectrode biosensor for realtime monitoring of L-glutamate release. Anal Chim Acta 645(1-2):86–91.

5. Mazzone GL, Nistri A (2011) Electrochemical detection of endogenous glutamaterelease from rat spinal cord organotypic slices as a real-time method to monitorexcitotoxicity. J Neurosci Methods 197(1):128–132.

6. Shigetomi E, Jackson-Weaver O, Huckstepp RT, O’Dell TJ, Khakh BS (2013) TRPA1channels are regulators of astrocyte basal calcium levels and long-term potentiationvia constitutive D-serine release. J Neurosci 33(24):10143–10153.

7. Heinrich A, Andó RD, Túri G, Rózsa B, Sperlágh B (2012) K+ depolarization evokes ATP,adenosine and glutamate release from glia in rat hippocampus: A microelectrodebiosensor study. Br J Pharmacol 167(5):1003–1020.

8. Albensi BC, Oliver DR, Toupin J, Odero G (2007) Electrical stimulation protocols forhippocampal synaptic plasticity and neuronal hyper-excitability: Are they effective orrelevant? Exp Neurol 204(1):1–13.

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 3 of 11

Fig. S1. Behavioral and electrographic features of pilocarpine-induced SE in WT and dnSNARE mice. (A) The seizure-related behavior scored every 5 minaccording to the Racine scale (2) during four successive injections of low-dose pilocarpine (20 min apart) was similar between WT (n = 12) and dnSNARE mice(n = 10; ANOVA, P > 0.05). (B) The latency to onset of SE after the first injection of pilocarpine was comparable in WT mice (n = 12) and dnSNARE mice (n = 10;Student t test, P > 0.05). (C) The percentage of mice that developed and survived SE induced with multiple injections of low-dose pilocarpine was similar in WT(n = 71) and dnSNARE mice (n = 58; χ2 test, P > 0.05). (D and E) EEG recordings before and during SE induced with a single high dose of pilocarpine weresubjected to fast Fourier transformation, and the power in different frequency bands was analyzed in WT (D) and dnSNARE mice (E). Representative analyses

Legend continued on following page

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 4 of 11

of band power in the β-band are shown. The zero time point corresponds to the time of scopolamine injection 30 min before the pilocarpine injection. Asterisksmark isolated acute seizures before mice enter continuous SE. The horizontal dashed line indicates the death of the animal. (Inset) Traces represent EEGrecordings 15 min before pilocarpine injection (1), during the occurrence of an acute seizure (2), and during continuous SE (3) in WT (D) and dnSNARE mice (E).(Scale bars: 400 μA and 10 s.) (F) As a measure for the severity of acute seizures, the percentage change of power during acute seizures in different frequencybands was calculated relative to the power of the basal EEG recorded before pilocarpine injection. No differences were observed between WT (n = 9) anddnSNARE mice (n = 7; ANOVA, P > 0.05). (G) Similarly, the percentage change in EEG power during continuous SE was not different between WT and dnSNAREmice (n = 10 per genotype; ANOVA, P > 0.05). (H) Average latency to onset of acute seizure after the single injection of high-dose pilocarpine in WT (n = 9) anddnSNARE mice (n = 7; Mann–Whitney test, P > 0.05). (I) Average number of acute seizures occurring between the time of the single injection of high-dosepilocarpine and the beginning of SE in WT (n = 9) and dnSNARE mice (n = 7; Mann–Whitney test, P > 0.05). (J) Average duration of acute seizures occurringafter the single injection of high-dose pilocarpine in WT (n = 9) and dnSNARE mice (n = 7; Student t test, P > 0.05). (K) Average latency of SE induced with thesingle injection of high-dose pilocarpine in WT and dnSNARE mice (n = 10 per genotype; Mann–Whitney test, P > 0.05).

Fig. S2. Normalized EEG spectral power in SE mice during an SRS (Left) and control no-SE mice (Right; 3-min recording). Power spectra were similar in WT anddnSNARE mice during the two different conditions (n = 5 per genotype; ANOVA, P > 0.05).

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 5 of 11

Fig. S3. Seizure and interictal spike development after pilocarpine-induced SE in WT and dnSNARE mice. (A) Average number of seizures per day in seven WT(Upper) and eight dnSNARE mice (Lower) after SE. Horizontal dashed lines represent the maximum number of seizures reached at day 101 and day 126 after SEin WT mice. Note the progressive increased in seizure frequency over time in WT mice (Upper) but not in dnSNARE mice (Lower; ANOVA: genotype, P = 0.002;time, P = 0.089; genotype × time interaction, P = 0.248; post hoc test, *P < 0.05, **P < 0.01, and ***P < 0.001). (B) Individual average number of seizures perday during the fifth month after SE plotted as a function of average number of seizures per day during the first month after SE. Linear regressions indicatea progressive increase in seizure frequency over time in WT mice (dashed line, n = 5) but not in dnSNARE mice (solid line, n = 8). (C) Bar graph illustrating the

Legend continued on following page

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 6 of 11

average duration of seizures for each 1-mo block after SE in WT (n = 4–6) and dnSNARE mice (n = 6–8). Note that no difference was found in the seizureduration over time between genotypes (ANOVA, P > 0.05). (D) Average number of interictal spikes per day in seven WT (Upper) and eight dnSNARE mice(Lower) after SE. Horizontal dashed lines represent the maximum number of interictal spikes reached at day 144 after SE in WT mice. Note the reduced in-terictal spikes frequency in dnSNARE mice (Lower) compared with WT mice (Upper; ANOVA: genotype, P = 0.02; time, P < 0.001; genotype × time interaction,P = 0.960; post hoc test, *P < 0.05, **P < 0.01, and ***P < 0.001).

Fig. S4. Analysis of behavioral parameters of eight WT no-SE mice, seven WT SE mice, seven dnSNARE no-SE mice, and eight dnSNARE SE mice in the openfield over a 30-min test period. (A) Average cumulative distance traveled over the 30-min test period (ANOVA, P > 0.05). (B) Line graph showing the distancetraveled in 1-min bins over the 30-min test period.

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 7 of 11

Fig. S5. NeuN and GFAP staining in WT and dnSNARE mice after pilocarpine treatment. (A and B) Representative hippocampal sections from control no-SEand SE-experienced WT and dnSNARE mice stained with an antibody directed against the neuron-specific epitope NeuN 8 mo after pilocarpine treatment. Apronounced neuronal cell loss in areas CA1 (A, arrowheads) and CA3 (B, arrowheads) was detected in WT and dnSNARE mice that developed SE. Averageneuronal density in CA1 (A) and CA3 (B) are presented under each corresponding section (WT no-SE, n = 3 animals; WT SE, n = 7 animals; dnSNARE no-SE, n = 5animals; dnSNARE SE, n = 4 animals; ANOVA followed by post hoc test, *P < 0.05, **P < 0.01, and ***P < 0.001; ns, nonsignificant). (C and D) Same hippo-campal sections as in A and B but stained with an antibody directed against the astrocyte-specific epitope GFAP. GFAP expression was increased in areas CA1(C) and CA3 (D) in WT mice that entered SE but not in dnSNARE mice. Average GFAP area in CA1 (C) and CA3 (D) are presented under each correspondingsection (ANOVA followed by post hoc test, *P < 0.05 and **P < 0.01; ns, nonsignificant). (Scale bars: 100 μm.)

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 8 of 11

Fig. S6. Vimentin staining in WT and dnSNARE mice after pilocarpine treatment. (A–C) Representative hippocampal sections fromWT and dnSNARE mice thatentered or did not enter SE stained with an antibody directed against the astrocyte-specific epitope vimentin 8 mo after pilocarpine treatment. Note theincreased expression of vimentin in areas CA1 (A) and CA3 (B) and in the hilus of the DG (C) in WT mice that entered SE but not in dnSNARE mice. Averagevimentin area in CA1 (A), CA3 (B), and hilus of the DG (C) are presented under each corresponding section (WT no-SE, n = 11 animals; WT SE, n = 11 animals;dnSNARE no-SE, n = 9 animals; dnSNARE SE, n = 7 animals; ANOVA followed by post hoc test, *P < 0.05, **P < 0.01, and ***P < 0.001; ns, nonsignificant). (Scalebars: 100 μm.)

Fig. S7. Representative whole-cell recordings of CA1 pyramidal cells depicting mEPSCs in the presence of 1 μM TTX and 0 Mg2+ in WT (Upper) and dnSNAREmice (Lower).

Fig. S8. (A) D-AP5 (1 μM) reduced the amplitude of evoked synaptic NMDA currents by 32% in CA1 pyramidal neurons from WT slices (n = 4 slices from fouranimals). (B) Average of 15 consecutive synaptic NMDA currents before (control) and during application of 1 μM D-AP5.

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 9 of 11

Fig. S9. Glutamate and D-serine microelectrode biosensor measurements in hippocampal slices from WT and dnSNARE mice. (A) Response of the glutamatesensor (Glu; Upper) to different concentrations of glutamate (10, 5, 1, 0.5, and 0.2 μM) applied through the perfusion of the recording chamber. (Bottom) Trace(Glu – Null) obtained by subtracting the signal from the null sensor (Middle) from the glutamate trace (Glu, Upper). (B) Plot depicting the calibration of theglutamate sensor performed in A (linear regression: y = 0.1041x − 0.0009, r2 = 0.9990). (C) Picture showing the typical placement of microelectrodes at thesurface of hippocampal slices. Glutamate and null sensors were placed closed together into the stratum radiatum (s.r.). (Scale bar: 200 μm.) (D) Example oftraces illustrating basal tone measurements of glutamate in WT (Left) and dnSNARE slices (Right). Subtracted signals (Glu – Null) are presented. Glutamate and

Legend continued on following page

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 10 of 11

null sensors were removed from the slices after a baseline recording of 20 min. Subtraction of the pre- and postremoval signals was used to determine the basaltone of glutamate that was calculated by using a calibration of 10 μM glutamate at the end of the experiment. Dashed lines indicate the baseline signal beforesensors removal. (E) Average glutamate tone in WT (n = 11 slices from six animals) and dnSNARE slices (n = 10 slices from five animals; Mann–Whitney test, P >0.05). (F) Representative traces showing the response of the glutamate sensor to TBS (arrow, one train of five bursts at 100 Hz separated by 200 ms) in WT(Upper) and dnSNARE slices (Lower). Subtracted signals (Glu – Null) are presented. The dashed line indicates the baseline signal before TBS. (G) Averageconcentration of glutamate detected in response to TBS in WT (n = 9 slices from five animals) and dnSNARE slices (n = 9 slices from six animals; Mann–Whitneytest, P > 0.05). (H) Response of the D-serine sensor (D-ser; Upper) to different concentrations of D-serine (10, 5, 1, 0.5, and 0.2 μM). (I) Plot depicting the cal-ibration of the D-serine sensor performed in H (linear regression: y = 0.1090x – 0.0082, r2 = 0.9994). (J) Example of traces illustrating basal tone measurementsof D-serine in the stratum radiatum of WT (Left) and dnSNARE hippocampal slices (Right). Subtracted signals (D-ser – Null) are presented. The D-serine signalwas calibrated with 10 μM D-serine at the end of the experiment. (K) Average D-serine tone in WT (n = 9 slices from six animals) and dnSNARE slices (n = 10 slicesfrom five animals; Mann–Whitney test, P > 0.05).

Clasadonte et al. www.pnas.org/cgi/content/short/1311967110 11 of 11