You are on page 1of 10

Neurobiology of Disease 82 (2015) 311–320

Contents lists available at ScienceDirect

Neurobiology of Disease

journal homepage: www.elsevier.com/locate/ynbdi

GABAA currents are decreased by IL-1β in epileptogenic tissue of patients


with temporal lobe epilepsy: implications for ictogenesis
Cristina Roseti b,1, Erwin A. van Vliet c,1, Pierangelo Cifelli a,f, Gabriele Ruffolo a, Johannes C. Baayen h,
Maria Amalia Di Castro a, Cristina Bertollini a, Cristina Limatola a,g, Eleonora Aronica c,d,
Annamaria Vezzani e,⁎⁎, Eleonora Palma a,b,⁎
a
Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
b
IRCCS San Raffaele Pisana, Rome, Italy
c
Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
d
Stichting Epilepsie Instellingen Nederland (SEIN-Heemstede), The Netherlands
e
Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Milano, Italy
f
Ri.MED Foundation, Palermo, Italy
g
IRCCS Neuromed, Pozzilli, Italy
h
Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands

a r t i c l e i n f o a b s t r a c t

Article history: Temporal lobe epilepsy (TLE) is the most prevalent form of adult focal onset epilepsy often associated with drug-
Received 17 April 2015 resistant seizures. Numerous studies suggest that neuroinflammatory processes are pathologic hallmarks of both
Revised 2 July 2015 experimental and human epilepsy. In particular, the interleukin (IL)-1β/IL-1 receptor type 1 (R1) axis is activated
Accepted 6 July 2015
in epileptogenic tissue, where it contributes significantly to the generation and recurrence of seizures in animal
Available online 11 July 2015
models. In this study, we investigated whether IL-1β affects the GABA-evoked currents (IGABA) in TLE tissue from
Keywords:
humans. Given the limited availability of fresh human brain specimens, we used the “microtransplantation”
Cytokine method of injecting Xenopus oocytes with membranes from surgically resected hippocampal and cortical tissue
GABAA receptor from 21 patients with TLE and hippocampal sclerosis (HS), hippocampal tissue from five patients with TLE with-
Neuroinflammation out HS, and autoptic and surgical brain specimens from 15 controls without epilepsy. We report the novel finding
Oocytes that pathophysiological concentrations of IL-1β decreased the IGABA amplitude by up to 30% in specimens from
Pharmacoresistant epilepsy patients with TLE with or without HS, but not in control tissues. This effect was reproduced by patch-clamp re-
cordings on neurons in entorhinal cortex slices from rats with chronic epilepsy, and was not observed in control
slices. In TLE specimens from humans, the IL-1β effect was mediated by IL-1R1 and PKC. We also showed that IL-
1R1 and IRAK1, the proximal kinase mediating the IL-1R1 signaling, are both up-regulated in the TLE compared
with control specimens, thus supporting the idea that the IL-1β/IL-R1 axis is activated in human epilepsy. Our
findings suggest a novel mechanism possibly underlying the ictogenic action of IL-1β, thus suggesting that this
cytokine contributes to seizure generation in human TLE by reducing GABA-mediated neurotransmission.
© 2015 Elsevier Inc. All rights reserved.

1. Introduction

Temporal lobe epilepsy (TLE) is a form of adult focal onset epilepsy


often associated with pharmacoresistance (Kwan et al., 2010; Blümcke
Abbreviations: Gamma-aminobutyric acid, GABA; GABA type A receptor, GABAA-R; et al., 2013). Pathological changes can be observed in patients with
Hippocampal sclerosis, HS; Interleukin-1β, IL-1β; IL-1 receptor type 1, IL-1R1; IL-1 recep- TLE, the most prominent of which is a loss of neurons in the hippocam-
tor antagonist, IL-1Ra; Interleukin-1 receptor-associated kinase1, IRAK1; N-methyl-D-as-
pus, known as hippocampal sclerosis (HS; Blümcke et al., 2013), and as
partate, NMDA; Protein kinase C, PKC; Temporal lobe epilepsy, TLE.
⁎ Correspondence to: E. Palma, Department of Physiology and Pharmacology, Università many as 75% of patients with mesial TLE are considered to have drug-
di Roma Sapienza, P.le A. Moro 5, 00185 Roma, Italy. resistant epilepsy (Schmidt and Löscher, 2005). The search for mecha-
⁎⁎ Correspondence to: A. Vezzani, Department of Neuroscience, IRCCS Istituto di Ricerche nisms implicated in the pathogenesis of seizures in TLE, with the inten-
Farmacologiche "Mario Negri", Via G. La Masa 19, 20156 Milano, Italy. tion of characterizing new targets for developing novel drugs, revealed a
E-mail addresses: annamaria.vezzani@marionegri.it (A. Vezzani),
eleonora.palma@uniroma1.it (E. Palma).
key role played by neuroinflammation, in particular the activation of the
1
Equal contribution. IL-1β/IL-1 receptor type 1 (IL-1R1) axis, in epileptogenic brain areas
Available online on ScienceDirect (www.sciencedirect.com). (Vezzani et al., 2011a). In addition to IL-1β/IL-1R1 activation, IRAK1, a

http://dx.doi.org/10.1016/j.nbd.2015.07.003
0969-9961/© 2015 Elsevier Inc. All rights reserved.
312 C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320

serine/threonine kinase functionally associated with IL-1R1, has a pivot- study. All surgeries were performed by the same neurosurgeon (J.C.B.)
al role in the activation of this inflammatory pathway (Gottipati e al., at VUMC. In TLE patients, the surgery consisted of an extensive temporal
2008; Iyer et al., 2012). Supportive evidence for a role of IL-1β signaling lobectomy (ETL) including microsurgical resection of the amygdala and
in epilepsy includes (Henshall et al., 2000; Vezzani et al., 2011b): parahippocampal gyrus and en bloc excision of the hippocampal forma-
(i) increased IL-1β biosynthesis and release by astrocytes and microglia tion. The interventions differed in the extent of the neocortical resec-
and induction of IL-1R1 in neurons, both in experimental models and tion. Nondominant ETL included excision of approximately 4–7 cm or
human TLE brain tissue; (ii) powerful anti-ictogenic effects of drugs more cm of the superior, middle and inferior temporal gyrus, depending
blocking the IL-1β/IL-1R1 signaling activation, resulting in delayed sei- on intraoperative EEG measurements, whereas dominant ETL included
zure precipitation and decreased seizure recurrence in various experi- excision of approximately 3–7 cm of the superior, middle and inferior
mental models; (iii) genetic perturbations of the IL-1β system in mice temporal gyrus, depending on language mapping and intraoperative
causing profound changes in their intrinsic seizure susceptibility. This EEG measurements. The predominant seizure types were medically in-
evidence strongly suggests that activation of IL-1R1 signaling by IL-1β tractable complex partial seizures, and all patients had seizures which
is not a by-stander phenomenon but significantly contributes to seizure were resistant to maximal doses of different anti-epileptic drugs
generation and recurrence. Studies of the mechanisms mediating the (Table 1). Epilepsy duration was calculated as the interval in years
ictogenic effects of IL-1β revealed direct neuromodulatory proper- from age at seizure onset to age at surgery; no patients included in
ties of this cytokine (Vezzani and Viviani, 2015). In particular, IL-1β our series had seizures in the 24 h before surgery. Twenty one patients
induces IL-1R1-mediated post-transcriptional signaling in neurons with TLE (#1–21, Table 1) had hippocampal sclerosis (HS) with neuro-
that involves the Src kinase-dependent tyrosine phosphorylation of nal loss within all hippocampal subfields, including CA1 and CA4 (HS,
the NR2B-NMDA receptor, thereby promoting hyperexcitability, sei- ILAE type 1). Five patients with TLE but not HS (#22–26, Table 1)
zures, and excitotoxicity by enhancing neuronal Ca2 + influx (Viviani who underwent resection of the hippocampus for medically intrac-
et al., 2003; Balosso et al., 2008; Vezzani and Viviani, 2015). table TLE were included to provide a comparison group. All cases
It has been proposed since 1958 (Jung et al., 1958) that, seizures were reviewed independently by two neuropathologists, and the
occur as a result of altered excitatory/inhibitory balance; therefore, an diagnosis was confirmed according to the international consensus
altered GABAA receptor (R) function may contribute to epileptogenesis classification (Blümcke et al., 2013).
(Scharfman and Brooks-Kayal, 2014; González et al., 2015). In line with As control tissue, we used specimens from autopsies of patients
this hypothesis, we have shown that GABAA-Rs in the epileptic brain, without any neurological disease and no sign of brain inflammation
when repetitively activated, display a marked current desensitization (#27–41, Table 1; the causes of death included heart failure, respiratory
known as rundown (Ragozzino et al., 2005; Palma et al., 2007), thereby failure, myocardial infarction) and specimens from patients (#42–45,
contributing to the GABAergic impairment in epilepsy. This phenome- Table 1) who underwent surgery for meningioma (WHO grade III).
non is significantly prevented by brain derived neurotrophic factor, The absence of seizures was determined during scheduled neurology
phosphatase inhibitors and the chemokine fractalkine (Palma et al., visits, using 60 min of awake EEG recordings; seizures were classified
2005; Roseti et al., 2013). according to the Engel classification.
Experimental evidence in rodents suggests that the ictogenic effects All autopsies were performed within 10 to 24 h of death. Hippocam-
of IL-1β may involve the GABAA-R since IL-1β reduces the peak ampli- pal tissue specimens were dissected at the Department of Neuropathol-
tude of the GABA-evoked current in rat patch-clamped hippocampal ogy in 5 mm slabs perpendicular to antero-posterior axis. A slice from
neurons (Wang et al., 2000) and decreases synaptic inhibition in CA3 hippocampal mid-body level was fixed overnight in 10% formalin for
pyramidal cells (Zeise et al., 1997). paraffin embedding for histopathology and adjacent tissue was snap-
We therefore tested the hypothesis that there is pathophysiological frozen over liquid nitrogen and stored at −80 °C until use. Frozen tissue
modulation of GABAA-R by IL-1β in human brain specimens, and found was shipped by courier to University of Rome.
that this cytokine significantly reduces GABAA-R mediated currents in Examination of histologically normal tissues obtained at surgery
the hippocampus and the temporal cortex of patients with TLE with or showed a pattern of immunoreactivity (IR) similar to that observed in
without HS, but not in non-epileptic control tissue. The IL-1β effect control tissues from autopsies, thus arguing in favor of antigen preser-
was concomitant with up-regulation of IL-1R1 and its proximal kinase vation in autopsies (Conti et al., 2011; Roseti et al., 2013). Tissue was ob-
IRAK-1 in the same epileptogenic tissue and was mediated by activation tained and used in accordance with the Declaration of Helsinki and the
of IL-1R1 and PKC. AMC Research Code provided by the Medical Ethics Committee and ap-
This novel mechanism may contribute to hyper-excitability in the proved by the science committee of the VUMC Biobank. The Ethics Com-
temporal lobe, thereby underlying seizure generation in TLE, and possi- mittee of the University of Rome “Sapienza” approved the technical
bly in other pharmacoresistant forms of epilepsy associated with neuro- procedures. Informed consent was obtained from all individuals in-
inflammation (Bernard and Shevell, 2008; Vezzani et al., 2011a). volved in this study.

2. Methods 2.2. Immunohistochemistry

2.1. Patients Tissue was fixed in 10% buffered formalin (autopsy tissue for 2
weeks; surgical specimens for 24 h) and embedded in paraffin. In
The clinical cases and controls included in this study were selected all cases, a representative formalin-fixed, paraffin-embedded tissue
from the Departments of Neuropathology of the Academic Medical Cen- block was studied, selecting large resection specimens containing
ter (AMC, University of Amsterdam) and the VU University Medical normal cortex adjacent to abnormal cortex for comparison, as an in-
Center (VUMC). The clinical characteristics derived from the patients' ternal control. Paraffin-embedded tissue was sectioned at 6 μm,
medical records are summarized in Table 1. In the text, the number mounted on organosilane-coated slides (Star Frost, Waldemar
of patients used in each experiment is reported, and referred to Knittel GmbH, Brunschweig, Germany) and two sections from each
Table 1 using the symbol #. We used 26 surgical epilepsy specimens paraffin block were used for immunohistochemical staining, as de-
(hippocampus and temporal cortex) from patients who underwent scribed previously (Ravizza et al., 2006; Iyer et al., 2010). Sections
surgery for medically intractable epilepsy (#1–26, Table 1). All patients were deparaffinated in xylene, rinsed in ethanol (100%, 95%, 70%)
underwent presurgical evaluation with non-invasive tests. Patients and incubated for 20 min in 0.3% hydrogen peroxide diluted in meth-
who underwent implantation of strip and/or grid electrodes for chronic anol. Antigen retrieval was performed using a pressure cooker in
subdural invasive monitoring before resection were excluded from the 0.1 M citrate buffer pH 6.0 at 120 °C for 10 min. After cooling on
C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320 313

Table 1
Clinical characteristics of patients.

P# Age (yrs)/sex Epilepsy onset Surgical zone Seizure type Number of seizures/month Pathology Medications

#1* 43/M 28 L-T CPS/GTCS 15 HS TPM, PHT, CLB


#2*° 54/F 15 R-T SPS/CPS 24 HS CBZ, VPA, CLB
#3* 39/F 12 R-T CPS/GTCS 10 HS LEV, CBZ, PB, CNP
#4*° 19/M 15 R-T SPS 18 HS CBZ
#5*° 47/M 16 L-T CPS 7 HS VPA, CBZ, CLB
#6* 34/M 4 L-T CPS 4 HS LMT, PB
#7* 29/F 13 R-T SPS/GTCS 32 HS LMT, TPM
#8* 27/M 10 R-T CPS 12 HS CBZ, LCB
#9*° 54/F 12 R-T CPS/GTCS 8 HS CBZ, LMT, PB
#10*° 29/M 18 L-T CPS 14 HS CBZ
#11*° 28/F 14 L-T SPS 120 HS CBZ, LMT
#12*° 37/F 15 R-T CPS 1 HS CBZ, LEV
#13*° 29/M 15 L-T CPS 36 HS CBZ, CLB
#14*° 26/M 6 L-T CPS 5 HS LEV, TPM
#15*° 57/F 47 R-T CPS 1 HS CNP
#16*° 39/M 33 R-T CPS 32 HS CBZ
#17° 42/F 4 R-T CPS/GTCS 1 HS PHT, OXC
#18° 37/F 4 L-T CPS 10 HS CBZ
#19° 66/F 10 R-T CPS 8 HS CBZ, PB, PHT
#20*° 24/F 15 L-T CPS 20 HS TPM, LEV, OXC
#21*° 52/M 10 L-T CPS 1 HS CBZ, PB, VGB
#22*° 21/F 10 L-T SPS 32 non-HS LEV, VPA
#23*° 61/M 36 L-T CPS 2 non-HS CBZ
#24*° 48/M 29 R-T SPS/CPS 32 non-HS LMT, VPA
#25*° 48/F 42 R-T CPS 4 non-HS LEV, LMT
#26*° 63/F 52 R-T CPS 8 non-HS CLB, LMT, TPM
#27* 52/F Absent R-T – – None
#28* 48/M Absent L-T − − None
#29° 81/F Absent R-T – – None
#30*° 68/F Absent L-T − − None
#31*° 86/F Absent R-T − − None
#32*° 30/M Absent L-T − − None
#33*° 65/F Absent L-T − − None
#34*° 71/F Absent R-T − − None
#35° 60/M Absent R-T – – None
#36° 44/F Absent L-T – – None
#37° 63/F Absent R-T – – None
#38*° 25/F Absent L-T – – None
#39*° 31/M Absent L-T – – None
#40*° 63/M Absent L-T – – None
#41*° 56/M Absent L-T – – None
#42* 47/F Absent R-T – – Meningioma
#43* 75/F Absent R-T – – Meningioma
#44* 38/M Absent R-T – – Meningioma
#45* 32/M Absent L-T – – Meningioma

Patient 1–21: Patients with TLE and hippocampal sclerosis (HS); Patient 22–26: Patients with TLE without hippocampal sclerosis; Patient 27–41: non-epileptic tissues from autopsies (hip-
pocampus and temporal cortex); Patient 42–45: surgical specimens from patients without epilepsy but with meningioma III WHO; T, temporal; L, left; R, right.
Seizures Type: SPS, simple partial seizures; CPS, complex partial seizures; GTCS, generalized tonic/clonic seizures.
Drugs: CLB, Clobazam; CBZ, Carbamazepine; LEV, Levetiracetam; OXC, Oxcabazepine; PB, Phenobarbital; PHT, Phenytoin; VPA, Valproate; LCS, Lacosamide; CNP, Clonazepam; LMT,
Lamotrigine; TPM, Topiramate; VGB Vigabatrin. * Tissues used for electrophysiology, ° tissues used for western blot.

ice, slides were washed with phosphate-buffered saline (PBS; 0.1 M, glycerol; 800 μl Na-orthovanadate (10 mg/ml); 200 μl 0.5 M EDTA
pH 7.4) and incubated overnight in polyclonal goat anti-human IL- pH 8.0; 400 μl protease inhibitors; 200 μl 0.5 M NaF; 11.2 ml H2O. Fifty
1RI (1:50; R&D Systems, Abingdon UK) in PBS at 4 °C. Thereafter, sec- μg total protein per lane, as determined using the bicinchoninic acid
tions were washed in PBS and stained with a polymer based peroxi- method (Smith et al., 1985), was separated by sodium dodecyl
dase immunocytochemistry detection kit (PowerVision Peroxidase sulfate–polyacrylamide gel electrophoresis (10% acrylamide) and trans-
system, ImmunoVision, Brisbane, CA, USA). After washing, sections ferred to immobilon-P membranes (Millipore corporation, Billerica,
were stained with 3,3′-diaminobenzidin tetrahydrochloride (50 mg MA, USA) by electroblotting (BioRad, Transblot SD, Hercules, USA).
DAB, Sigma-Aldrich, Zwijndrecht, The Netherlands) and 5 μl 30% hy- Blots were incubated overnight in Tris buffered saline with Tween
drogen peroxide in a 10 ml solution of Tris–HCl. Sections were coun- (TBST; 20 mM Tris, 150 mM NaCl, 0.1% Tween, pH 7.5)/5% non-fat dry
terstained with hematoxylin, dehydrated in alcohol and xylene, and milk, containing the primary antibody: polyclonal rabbit anti-IRAK-1
coverslipped. Sections incubated without primary antibodies or with (1:1000, SC-7883, Santa Cruz Biotechnology, Heidelberg, Germany) or
pre-immune serum were essentially blank. These experiments were monoclonal mouse anti-ß-actin (1:50,000, clone AC-15, Sigma-Aldrich
performed at University of Amsterdam (E. V. and E. A.). Chemie, Zwijndrecht, The Netherlands). After washing three times for
10 min in TBST, blots were incubated for 1 h in the secondary antibody:
2.3. Western Blot anti-rabbit (1:2500, Invitrogen, Breda, The Netherlands) or anti-mouse
(1:2500, Dako, Glostrup, Denmark) labeled with horseradish peroxi-
Patients with sufficient frozen material available were selected dase. After washing three times for 10 min in TBST, immunoreactivity
for Western blot analysis. Freshly frozen tissue (patients shown in was visualized with WesternBright ECL HRP substrate (Advansta,
Table 1) was homogenized in lysis buffer containing per 20 ml: Menlo Park, CA, USA) and the blots were digitized using a Luminescent
200 μl 1 M Tris pH 8.0; 1 ml 3 M NaCl; 2 ml 10% NP-40; 4 ml 50% Image Analyzer, LAS-4000 (Fuji Film, Japan). The optical density of each
314 C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320

sample was measured using Adobe Photoshop. For each sample the op- 2.6. Whole-cell recordings from cortical rat slices from epileptic rats
tical density of IRAK-1 was calculated relative to the optical density of ß-
actin. These experiments were performed at University of Amsterdam Adult male Sprague–Dawley rats (225–250 g; Charles-River, Calco,
(E. V. and E. A.). Italy) were housed at constant temperature (23 °C) and relative humid-
ity (60 ± 5%) with free access to food and water and a fixed 12 h light/
2.4. Membrane preparation and oocytes injection dark cycle. Procedures involving animals and their care were conducted
in conformity with the institutional guidelines that are in compliance
The use of female Xenopus laevis frogs conformed to institutional with national (D.L.n.116, G.U., Suppl. 40, February 18, 1992) and inter-
policies and guidelines of the Italian Ministry of Health. The preparation national laws and policies (EEC Council Directive 86/609, OJ L 358, 1, De-
of human membranes (frozen hippocampus and temporal cortex from cember 12, 1987; Guide for the Care and Use of Laboratory Animals, U.S.
patients shown in Table 1), their injection in X. laevis oocytes, and National Research Council, 1996).
GABA current recordings were carried out as previously described Status epilepticus (SE) was induced in 11 rats by intraperitoneal in-
(Miledi et al., 2006; Eusebi et al., 2009; Roseti et al., 2013). In a set of ex- jection of 10–12 mg/kg kainic acid (KA) in phosphate buffered saline
periments, we performed intranuclear injection in Xenopus oocytes (PBS, pH 7.4). Six control rats were injected with the corresponding vol-
(Miledi et al., 2006) of human complementary DNA (cDNAs) encoding ume of vehicle. As described previously (Röder et al., 1996), KA induced
for α1, β2 and γ2 GABAA subunits (pcDNA3 vector) or co-injected a behavioral syndrome defined as SE which included early staring, “wet
with cDNA encoding human IL-1R1 (pCMV6-XL4 vector). Human dog shakes” and seizures which evolved from mild forehead nodding to
α1β2γ2 cDNA was provided as a gift by Dr. Keith Wafford and human generalized motor convulsions with rearing and falling (stage 4/5 sei-
IL-1R1 cDNA was purchased from Origene 9620 (USA). zures, Racine, 1972). Rats were observed for at least 3 h by two indepen-
dent investigators, and only animals showing stage 4/5 seizures (~80%
2.5. Electrophysiology of injected animals) were selected for further analysis of spontaneous
recurrent seizures (SRS). Three rats died during or immediately after
From 12 to 48 h after injection, membrane currents were recorded SE. Three months after SE induction, remaining rats were video-
from voltage-clamped X. laevis oocytes using two microelectrodes monitored for 4 h/day for one week to detect generalized motor sei-
filled with 3 M KCl as previously described (Miledi et al., 2006). zures. The manifestation of at least two generalized tonic–clonic sei-
The oocytes were placed in a recording chamber (0.1 ml volume) zures during one week of observation was used as a criterion to select
and perfused continuously with oocyte Ringer solution (OR: NaCl 4 epileptic rats for the subsequent electrophysiology experiments. The
82.5 mM; KCl 2.5 mM; CaCl2 2.5 mM; MgCl2 1 mM; Hepes 5 mM, ad- animals were injected and selected for subsequent studies at Mario
justed to pH 7.4 with NaOH) at room temperature (20–22 °C). To Negri Institute (A.V.).
apply GABA or OR we used a gravity driven multi-valve perfusion
system (9–10 ml/min) controlled by computer (Biologique RSC- 2.6.1. Acute slices preparation
200; Claix, France) to ensure the exact duration of each application. Cortical slices were prepared from four epileptic rats and four
Using this system, 0.5 to 1 s are sufficient to reach the complete re- vehicle-injected age-matched controls. Rats were killed by decapitation
placement of applied solution. under deep halothane anesthesia, then the brain was rapidly removed
Unless otherwise indicated, GABA 500 μM (freshly dissolved in OR) and placed in ice-cold oxygenated (95% O2, 5% CO2) sucrose-based arti-
was applied for 4 s to oocytes to elicit inward currents (IGABA). To deter- ficial cerebrospinal fluid (ACSF) containing 87 mM NaCl, 75 mM su-
mine the effect of IL-1β on IGABA, oocytes were first exposed to GABA crose, 2 mM KCl, 7 mM MgCl2, 0.5 mM CaCl2, 25 mM NaHCO3, 1.2 mM
alone (two repetitive applications at 4 min intervals). Only the oocytes NaH2PO4 and 10 mM glucose, pH 7.4, 300–305 mOsm. Transverse slices
where IGABA was stable (less than 5% of peak amplitude after the second of the entorhinal cortex (350 μm, ThermoScientific HM 650 V) were cut
application) were subsequently used either for co-application of GABA and placed in a slice incubation chamber at room temperature with ox-
and IL-1β (2.5, 25, 250 and 400 ng/ml), or for GABA application after ygenated ACSF containing 125 mM NaCl, 2 mM KCl, 1.2 mM MgCl2,
4 min, 10 min or 60–250 min pre-incubation with the various IL-1β con- 2 mM CaCl2, 25 mM Na HCO3, 1.2 mM NaH2PO4 and 10 mM glucose,
centrations. The outcome measure was the percentage change in IGABA with pH and osmolarity as above, and transferred to a recording cham-
amplitude with respect to the current evoked before IL-1β application. ber within 1 to 6 h of slice preparation.
The IGABA decay was defined as the time required for the current to
decay from its peak to the half-peak value (T0.5). GABA current rundown 2.6.2. Electrophysiology
was defined as the percentage decrease of the current peak amplitude Whole-cell patch clamp recording was performed on layer V pyra-
after six 10s-applications of GABA at 40 s intervals (Eusebi et al., 2009; midal neurons at 24–25 °C. Cell capacitance was constantly monitored,
Roseti et al., 2013). Unless otherwise indicated, numbers (n) refer either and experiments where access resistance changed more than 20% were
to oocytes or neuronal cells used in each experiment. discarded. Glass electrodes (3–4 MΩ) were filled with 140 mM Cs-
In some experiments, the following drugs were incubated for vari- Methanesulfonate, 10 mM HEPES, 0.5 mM EGTA, and 2 mM Mg-ATP,
ous times before and during IL-1β application: 10 μM IL-1 receptor an- 0.3 mM Na3-GTP, 2 mM MgCl2. IGABA currents were recorded at 0 mV
tagonist (IL-1Ra), 10 μM PP2, a Src inhibitor (Viviani et al., 2003), to avoid spurious contributions of mechanosensitive Na+ currents
1 μM BEZ 235, a PI3K inhibitor (Chi et al., 2014), 50 μM capsazepine, a (Mazzuferi et al., 2010; Ragozzino et al., 2005), with glutamatergic cur-
TRPV1 channel blocker (Musumeci et al., 2011), 5 μM staurosporine rents at their reversal potential. Under these experimental conditions,
and 1 μM bisindolylmaleimide, two PKC inhibitors (Palma et al., with inactivated voltage-gated channels, cells were stable and healthy
2005), and 50 nM phorbol 12-myristate 13-acetate (PMA), a PKC activa- for at least 1 h. Spontaneous inhibitory postsynaptic currents were
tor (Bright and Smart, 2013). All drugs were purchased from Sigma Ita- present but did not affect the quantification of GABA-induced currents.
lia with the exception of GABA (Tocris, Bristol, UK), PP2 (Calbiochem, GABA (100 μM) was delivered to cells by pressure applications
UK), bisindolylmaleimide (Santa Cruz, USA), human recombinant IL- (5–10 psi; 100 ms; Picospritzer II; General Valve) from glass micropi-
1β and IL-1Ra (R&D Systems, Minnesota, USA). Drugs were dissolved pettes positioned close to whole-cell voltage-clamped neurons. After
in sterile buffered saline and used fresh. Staurosporine, PMA, and current amplitude stabilization had occurred (less than 5% of peak am-
bisindolylmaleimide were dissolved in DMSO (final concentration plitude decrease after the second GABA application), IL-1β (25 ng/ml)
≥1:3000) and stored at −20 °C as stock solution. All the electrophysio- was applied to the bath using a gravity-driven perfusion system
logical experiments were done at University of Rome Sapienza (P.C., (speed 1.5 ml/min); this was washed out after 15 min. In all recordings
G.R., E.P.) and at San Raffaele Pisana in Rome (C.R.). IGABA was evoked every 5 min in the control period, and during IL-1β
C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320 315

application and wash-out. Signals were acquired (sampling 10 kHz,


low-pass filtered 2 kHz) with DigiData-1440A using pCLAMP-v10 soft-
ware (Axon Instruments). These experiments have been performed at
University of Rome (M.DC, C.B. and C.L.).

2.7. Statistical analysis of data

Data are presented as mean ± s.e.m. The number of the patients' or


non-epileptic controls' specimens used in each experiment for oocyte
transplantation is reported in Table 1. The experiments and data analy-
sis were performed by different researchers (one of them operating
with blind procedures), and each tissue specimen was used several
times in different sets of experiments. The normality of data sets was
tested according to the Shapiro Wilk test. Statistical significance was
assessed by Student's test or by Kruskal–Wallis one-way ANOVA on
ranks when data set were not normally distributed. Statistical tests
were performed using Sigmaplot Systat (Systat Software GmbH, CA,
USA) or Origin 8 software (Origin Lab Corporation, MA, USA) using
raw data. Only differences considered significant (p b 0.05) are indicat-
ed in the text and figures legends.

3. Results

3.1. IL-1β signaling increases in TLE

To show whether IL-1β signaling was induced in the specimens from


patients with TLE, we measured IRAK-1, a recognized marker of IL-1β/
IL-1R1 signaling activation (Gottipati et al., 2008; Iyer et al., 2012)
using western blot. Fig. 1 shows a 4-fold increase on average in IRAK-1
levels in the hippocampus (Fig. 1A, B) and temporal cortex (Fig. 1C) of
16 patients with TLE and HS and five without HS compared with 13 con-
trols from autopsy without epilepsy (see Table 1).
We pooled specimens from patients with TLE with and without HS
in a single group since we found a similar increase in IRAK-1, thus sug-
gesting that the activation of this signaling is independent of HS, and is
most likely induced by recurrent seizures. In accordance with the IRAK-
1 up-regulation, immunohistochemical analysis of the temporal cortex
of patients with TLE showed an increased IL-1R1 immunoreactivity in
neurons and glia (Fig. 2C, D) compared with control cortex (Fig. 2A,
B), as previously reported in the hippocampus of patients with TLE
(Ravizza et al., 2006).

3.2. IL-1β reduces GABA currents in tissue from patients with TLE but not in
controls Fig. 1. IRAK-1 level is increased in tissue from patients with TLE. (A) Representative bands of
IRAK1 in western blot from tissue homogenates of controls (autopsy specimens without
To investigate whether IL-1R1 activation affects GABA-mediated epilepsy) and from patients with TLE with and without HS (B,C). Densitometric analysis
(mean ± s.e.m. of optical density units relative to corresponding β-actin) shows a 4-fold
currents, we microtransplanted Xenopus oocytes with hippocampal
increase on average of IRAK1 level in the hippocampus and cortex of patients with TLE
membranes from a randomly chosen cohort of 23 patients with TLE, in- (patients # 2, 4–5, 9–26, Table 1) compared with control tissue (patients # 29 to 41,
cluding the five patients without HS, and 15 control specimens (includ- Table 1). *p b 0.05, Student's t-test.
ing those used in Fig. 1; see Table 1), and exposed them to IL-1β. GABA
(500 μM) alone elicited inward currents (IGABA) with amplitude ranging
between − 15 and − 170 nA; these currents were blocked by the (2.5-ng/ml; 120 min pre-incubation) induced a 17% decrease of IGABA
GABAA-R antagonist bicuculline (100 μM; not shown). Co-application amplitude (GABA, 58.0 ± 9.7 nA; + IL-1β, 48.2 ± 8.2 nA; n = 10;
of IL-1β (2.5–500 ng/ml) with GABA in specimens from patients with #6,21, Table 1) that was not statistically significant. We found no mod-
TLE did not alter IGABA amplitude (n = 15, not shown; #10–13, ification of current amplitude in oocytes incubated for 10 to 120 min
Table 1). When oocytes were incubated with 25 ng/ml IL-1β for 4 or with oocytes' Ringer only (n = 40, not shown; #1–16, 20–26, Table 1).
10 min before GABA (Fig. 3A), the IGABA amplitude was not modified. These findings suggest that the IL-1β effect on IGABA is time and dose-
However, when oocytes were exposed to the same concentration of dependent. Moreover, the IGABA decrease induced by 25 ng/ml IL-1β
IL-1β from 60 to 250 min before GABA application, then IGABA amplitude was not associated with changes in current decay (T0.5, GABA, 3.4 ±
progressively decreased in 80% of oocytes, reaching a maximal ~30% re- 0.12 s; + IL-1β, 3.1 ± 0.15 s; n = 21; #6–10, Table 1) suggesting that
duction after 120 min incubation (GABA, 73.2 ± 8.3 nA; +IL-1β, 52.5 ± the decrease of IGABA amplitude is not related to a change in the rate of
7.1 nA; n = 273 oocytes from 32 frogs; p b 0.001; #1-16,20,21, Table 1 current inactivation.
and Supplementary Table 2; Fig. 3A–C). This effect was slowly reversible The lack of IL-1β effect in 20% oocytes was unrelated to which pa-
during a 3 to 5-h wash-out period (Fig. 3C). Similar changes in IGABA am- tients were used for membrane injection and it was not dependent on
plitude were obtained using higher IL-1β concentrations (250 and the membrane potential or GABA reversal potential of the oocytes.
400 ng/ml; not shown; #6–9, Table 1), while a lower IL-1β concentration This phenomenon was previously observed in these cell preparations
316 C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320

Fig. 2. IL-1R1 expression increases in temporal cortex. Distribution of IL-1RI immunoreactivity (IR) in the temporal cortex of patients with TLE and controls. Panels A and B: control cortex (A,
white matter, WM; B, gray matter, GM) without detectable IL-1R1 IR. Panels C and D: temporal cortex of patients with TLE (C, white matter, WM; D, gray matter, GM) showing increased IL-
1R1 expression in both glial (arrows in C) and neuronal cells (arrows in D). Scale bar in D: A–D, 40 μm.

(Roseti et al., 2013), and it may be due to a low efficiency in the expres- 120 min pre-treatment with 25 ng/ml IL-1β did not affect IGABA run-
sion of IL-1R1 or related signaling molecules in some microinjected down (GABA, 52.5 ± 9.7%; + IL-1β, 48 ± 6.8%; n = 12, #3–6, Table 1)
oocytes. while still decreasing IGABA amplitude (128 ± 12.5 nA vs 85.3 ±
Using oocytes transplanted with membranes from patients with TLE 14 nA).
without HS, we found that a 120 min pre-incubation with 25 ng/ml IL- Finally, in order to exclude that the IL-1β induced IGABA decrease was
1β induced a significant decrease of IGABA (GABA, 22.4 ± 1.6 nA; +IL- a particular effect related to the oocyte expression system, we per-
1β, 16.0 ± 1.6 nA; n = 48, p b 0.01; #22–26, Table 1), similar to that ob- formed whole-cell patch clamp recordings of IGABA in neurons of ento-
served in the hippocampi of patients with TLE and HS, in accordance rhinal cortex from six control and four epileptic rats. Applications of
with the common increase in IRAK-1 levels. GABA (100 μM) by pressure ejection evoked whole-cell currents of
Likewise, 120 min incubation with 25 ng/ml IL-1β reduced the 632 ± 139 pA (range 166–1165 pA; n = 10) in controls and of 974 ±
IGABA amplitude by about 30% in oocytes injected with temporal cor- 409 pA (range 692–1781 pA; n = 6) in epileptic rat slices. Bath applica-
tex membranes of patients with TLE (GABA, 108.3 ± 6.9 nA; + IL-1β, tion of IL-1β (25 ng/ml) for 15 min, decreased the peak amplitude of
77 nA ± 5.6; n = 240, p = 0.014; #1–16,20,21, Table 1; Fig. 4). The IGABA by ~ 40% (590 ± 360 pA; p b 0.01) in slices from epileptic rats
IGABA decrease, in both the hippocampus and cortex from patients whereas no effect was observed in control slices (653 ± 140 pA)
with TLE, was prevented by 30 min pre-treatment with 10 μM of (Fig. 1S, Supplementary data).
IL-1 receptor antagonist (GABA, 49.5 ± 3.4 nA; + IL-1Ra, 49.7 ±
4 nA; n = 29; #1, 9–11, 13, Table 1; Fig. 5A,B), thus demonstrating
that the effect of IL-1β was mediated by the activation of IL-1R1/ 3.3. IL-1β effect on GABA currents in TLE is mediated by PKC
IRAK-1 signaling.
To confirm that the effect of IL-1β was mediated by the IL-1R1 sig- In another set of experiments we explored the intracellular pathway
naling machinery transplanted in oocytes by human epileptic tissue, mediating the IL-1β-induced IGABA decrease in oocytes injected with
we intranuclearly injected a separate group of oocytes with the cDNAs hippocampal membranes from patients with TLE. We examined the
encoding α1β2γ2 GABAA-R, the predominant GABAA-R subtype canonical signaling activated down-stream of IL-1R1/IRAK1, namely
expressed in healthy brain (MacDonald et al., 2010). We found that Src and PI3K kinases, by pre-treating oocytes for 60 min with the
the currents evoked by the activation of α1β2γ2 GABAA-Rs were not af- specific inhibitors PP2 (Viviani et al., 2003) (10 μM) and BEZ 235
fected by IL-1β (GABA, 1550 ± 214 nA; +IL-1β, 1670 ± 172 nA; n = 10, (Chi et al., 2014) (1 μM), respectively. Both inhibitors failed to pre-
not shown). Likewise, we found no IL-1β effect on GABA evoked currents vent the IL-1β effect on IGABA (PP2, n = 14; BEZ, n = 16, not shown;
in oocytes co-injected with the cDNAs encoding α1β2γ2 GABAA-R and #9–11, Table 1). We also tested the involvement of TRPV1 channels
IL-1R1 (GABA, 1849 ± 230 nA; + IL-1β, 1877 ± 206 nA; n = 10, not since they mediate the IL-1β decrease of GABAergic synaptic cur-
shown). rents in mouse corticostriatal slices (Musumeci et al., 2011). Sixty
In contrast to the situation in tissue from patients with TLE, IL-1β did minutes pre-treatment with the TRPV1 channel blocker capsazepine
not modify IGABA amplitude in oocytes injected with hippocampal (50 μM) did not prevent the IL-1β effect (n = 14, not shown; #4,9,
(GABA, 65.9 ± 12.5 nA; +IL-1β, 61.8 ± 13.3 nA; n = 81; Fig. 3B,C) or Table 1). We showed, however, that 60 min pre-treatment with
cortical (GABA, 116.9 ± 15.4 nA; + IL-1β, 106.3 ± 13.4 nA; n = 20; 5 μM staurosporine or 1 μM bisindolylmaleimide, two PKC inhibitors
Fig. 4A) membranes from autopsies of controls without epilepsy (#27, (Palma et al., 2005) which per se did not affect I GABA amplitude,
28, 30–34, 38–41, Table 1) . Notably, the lack of IL-1β effect on IGABA am- prevented the IL-1β-induced decrease of IGABA (GABA, 66 ± 8.4 nA;
plitude in control tissue was confirmed using surgical samples from the staurosporine, 64.1 ± 7.5 nA; n = 18; #5,12,13, Table 1; GABA,
temporal cortex of patients with meningioma but without epilepsy 40.9 ± 8.9 nA; bisindolylmaleimide, 39.8 ± 10.7 nA; n = 12; #6
(GABA, 115.1 ± 17.1 nA; GABA + IL-1β, 107.7 ± 16 nA; n = 36; #42– and 8, Table 1; Fig. 5A,B). Accordingly, 120 min pre-treatment of oo-
45, Table 1; Fig. 4A, Inset). These findings confirm the similarity between cytes with 50 nM phorbol 12-myristate 13-acetate (PMA), a PKC ac-
surgical and post-mortem control tissues, as previously reported (Conti tivator (Bright and Smart, 2013), decreased I GABA by about 50%
et al., 2011; Roseti et al., 2013). (GABA, 44.9 ± 8.8 nA; + PMA, 25.1 ± 5.7 nA, n = 15, p = 0.026;
In a different set of experiments, we evoked a repetitive activa- #6,8,15,16, Table 1; Fig. 5A,B). PMA was ineffective in oocytes
tion of GABAA -Rs in microtransplanted oocytes from TLE patients injected with tissue from controls without epilepsy (n = 10, not
with HS in order to induce the IGABA rundown since we previously shown; #27,28, Table 1). These results show that PKC is involved in
showed that this phenomenon is a hallmark of GABAergic impair- the IL-1β-induced decrease of IGABA in tissue from patients with
ment in TLE tissue (Ragozzino et al., 2005; Roseti et al., 2013). A epilepsy.
C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320 317

4. Discussion 2000; Ravizza et al., 2008; Vezzani et al., 2011b). Accordingly, IL-1R1
immunoreactivity was enhanced in temporal cortex from patients
We report the novel finding that IL-1β decreases IGABA amplitude in with TLE, as previously shown in hippocampi from patients with TLE
tissue from patients with TLE, specifically in hippocampal and temporal (Ravizza et al., 2008).
cortex specimens, but not in control specimens. This effect was shown To study the effect of IL-1β on IGABA, we took advantage of the
by recording GABAA evoked currents in oocytes transplanted with microtransplantation technique in Xenopus oocytes (Eusebi et al.,
membranes from either surgically resected brain tissue from patients 2009). The exact glial or neuronal origin of the membrane patches
with TLE, or from autoptic and surgical control tissues. In the same transplanted onto the oocytes surface is not known, but this approach
TLE specimens, we found an increase in the level of IRAK-1, a key prox- has a two-fold advantage by allowing: 1. investigation of “whole”, glial
imal kinase associated with IL-1R1, thus supporting previous evidence and neuronal, GABA currents in oocytes expressing human GABAA-Rs
of the activation of IL-1β/IL-1R1 axis in human TLE (Henshall et al., that retain their native properties, (Eusebi et al., 2009; Palma et al.,
2003); 2. use of human tissues from patients without epilepsy as suit-
able controls since these individuals did not have neurological diseases
or ongoing inflammatory processes. Due to the limited quantity of tissue
available, control specimens are usually unsuitable for obtaining viable
brain slices or isolated neurons for physiological investigations with al-
ternative techniques.
We found that pathophysiological concentrations (≥25 ng/ml) of IL-
1β decreased the IGABA amplitude by up to 30% in specimens from pa-
tients with TLE; this corresponds to a relatively high concentration of
IL-1β compared to the normal range reported in blood or brain (e.g.,
pg/ml or pg/mg), thereby arguing in favor of an IL-1β effect compatible
with a pathologic rather than physiological scenario. Similar high con-
centrations were previously shown to affect GABAergic neurotransmis-
sion in in vitro rodent preparations (Wang et al., 2000).
The effect on IGABA by IL-1β in oocytes transplanted with human tis-
sue was replicated in a limited number of neurons in cortical slices from
epileptic rats, and, as in human specimens, was absent in control rat
slices. This evidence suggests that the IL-1β induced decrease in IGABA
was not a particular effect related to the oocyte expression system.
These results suggest the need for more in-depth investigations in ex-
perimental models of epilepsy.
The decrease of IGABA provoked by IL-1β was receptor-mediated, as-
sociated with increased IL-1R1 and IRAK-1 in epileptogenic tissue from
humans, and concentration-dependent, all features with pathophysio-
logical relevance (Ravizza et al., 2008; Vezzani et al., 2011a). These find-
ings also imply that the effect of IL-1β on IGABA occurs in the presence of
an up-regulated signaling machinery, in line with the evidence that
neuroinflammation can lead to brain dysfunction when prolonged or
excessive (Vezzani et al., 2011a; Vezzani and Viviani, 2015). Persistent
up-regulation of the IL-1β/IL-1R1 signaling has been reported in
human TLE and in related experimental models (Henshall et al., 2000;
Ravizza et al., 2006; Ravizza et al., 2008; Maroso et al., 2011; Tan et al.,
2015), and is confirmed by increased IL-1R1 and IRAK-1 in our TLE spec-
imens. When IL-1R1 is activated by IL-1β, ΙRAK-1 associates to the cell
membrane-bound receptor, therefore the IL-1R1/IRAK-1 complex is
transplanted into the oocytes. Interestingly, the induction of IRAK-1
was similar in the hippocampi of patients with TLE with or without
HS, thus suggesting that the persistent activation of the IL-1R1 signaling
in TLE may be driven by recurrent seizures rather than the underlying
neuropathology, as also suggested by findings in experimental models

Fig. 3. IL-1β decreases IGABA amplitude in oocytes transplanted with TLE hippocampus.(A) Time-
course of IL-1β-induced decrease of IGABA amplitude in oocytes microtransplanted with hip-
pocampal membranes from specimens from patients with TLE and HS. GABA alone was ap-
plied at 500 μM at the beginning of the experiment (time 0), and at various time points after
treatment with 25 ng/ml IL-1β. Data (mean ± s.e.m. of 15 oocytes/time point; patients #1–7,
Table 1) represent the percentage decrease of the peak amplitude induced by IL-1β. (B). His-
tograms show the average % change in IGABA amplitude after the 120 min application of 25
ng/ml IL-1β. The holding potential was −60 mV. Tissue from patients with TLE (n = 273 oo-
cytes; #1–16,20,21, Table 1) and patients without epilepsy (n = 81 oocytes, * p b 0.001 vs
GABA, Kruskal–Wallis test; patients #27,28,30–34,38–41, Table 1).(C) top traces are record-
ings in one representative oocyte transplanted with TLE membranes: 120 min pre-incuba-
tion with IL-1β reduced IGABA amplitude (middle trace) compared with GABA alone (first
trace). The effect was reverted by a three hour wash-out (third trace). Bottom traces show lack
of IL-1β effect on IGABA amplitude in one representative oocyte transplanted with hippo-
campal membranes from an individual without epilepsy. Holding potential and concentra-
tions are as in (A). Black bars indicate GABA application.
318 C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320

Fig. 4. IL-1 β decreases IGABA amplitude in oocytes transplanted with the cortex of patients with
TLE. (A) Histograms show the average % change in IGABA amplitude after a 120 min appli-
cation of 25 ng/ml IL-1β in specimens from patients with TLE specimens (patients #1–
16,20,21, Table 1) and autoptic control tissue from patients without epilepsy (patients
#27,28,30–34,38–41, Table 1). The holding potential was −60 mV. Black bars indicate
GABA application, *p = 0.014 vs GABA alone, Kruskal–Wallis test. (Inset) Histograms
show the lack of IL-1β effect in oocytes injected with membranes from surgical control tis-
sue from patients without epilepsy undergoing surgery for meningioma (WHO grade III).
(n = 36; patients #42–45, Table 1). Sample traces in inset are from one representative oo-
cyte transplanted with cortical membranes from a patient with meningioma but without
epilepsy. (B) Sample traces in one representative oocyte transplanted with membranes
from a patient with TLE before (first trace) and after 120 min pre-treatment with IL-1β
(middle trace). The effect reverted by 3 h wash-out (third trace). Holding potential and
black bars as in (A).

Fig. 5. IL-1β effect on IGABA amplitude in TLE is mediated by IL-1R1 and PKC. (A) Histograms
(Dube' et al., 2005, 2010; Maroso et al., 2010; Akin et al., 2011). Thus, the
show IGABA amplitude (mean ± s.e.m., expressed as percentage of control peak amplitude)
decrease of IGABA amplitude mediated by IL-1β was similar in patients after the indicated treatments in oocytes injected with hippocampal membranes from pa-
with TLE with or without HS. tients with TLE and HS. (10 μM IL-1Ra, n = 29; patients #1, 9–11, 13, Table 1; 5 μM
We found that IL-1β does not affect IGABA rundown caused by repet- staurosporine, n = 18; patients #5, 12, 13, Table 1; 1 μM bisindolylmaleimide, n = 12; pa-
itive GABAA receptor stimulation which is increased in TLE hippocam- tients #6 and 8, Table 1; 50 nM PMA, n = 15,*p = 0.026 vs GABA alone, Kruskal–Wallis
test; patients #6,8,15,16, Table 1). The holding potential was −60 mV. GABA was applied
pus vs control tissue, and reduced by fractalkine (Roseti et al., 2013).
at 500 μM alone and after 120 min exposure to 25 ng/ml IL-1β. Inhibitors were given be-
This evidence suggests that inflammatory cytokines and chemokines, fore IL-1β as indicated in the text. (B) Recordings of GABA currents in 3 representative oo-
both of which contribute to the neuroinflammatory milieu (Fabene cytes before and after drugs treatment as indicated. Holding potential and concentrations
et al., 2010; Vezzani et al., 2011b), have a different effect on the IGABA as in (A). Black bars indicate GABA application.
in epileptogenic tissue.
The relevance of our in vitro findings to the clinical condition is seizure-prone areas, thereby reducing the threshold for seizure
based on the compelling evidence that the activation of the IL-1β/IL- generation.
1R1 axis in experimental models promotes hyperexcitability, seizures We show that PKC is a key down-stream component mediating the IL-
and cell death (Vezzani et al., 2011a,b). Our data, therefore, suggest 1β/IL-1R1/IRAK1-dependent decrease in IGABA. Supporting evidence
that IL-1β, by decreasing GABA currents in brain tissue of patients shows that activation of PKC leads to down-regulation of GABAA-R medi-
with TLE, mediates a reduction of the GABAergic inhibitory tone in ated inhibition through β3 subunit phosphorylation (Brandon et al., 2000;
C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320 319

Bright and Smart, 2013). Although in our experiments we do not distin- Balosso, S., Maroso, M., Sanchez-Alavez, M., Ravizza, T., Frasca, A., Bartfai, T., et al., 2008. A
novel non-transcriptional pathway mediates the proconvulsive effects of interleukin-
guish between synaptic and extrasynaptic inhibition, one possibility is 1beta. Brain 131, 3256–3265.
that GABAA β subunits, that are over-expressed in the hippocampi of pa- Bernard, G., Shevell, M.I., 2008. Channelopathies: a review. Pediatr. Neurol. 38, 73–85.
tients with TLE (Sperk et al., 2009), may be targeted by the activation of Blümcke, I., Thom, M., Aronica, E., Armstrong, D.D., Bartolomei, F., Bernasconi, A., et al.,
2013. International consensus classification of hippocampal sclerosis in temporal
IL-1β/IL-R1/IRAK1 signaling. Indeed, this signaling activates neuronal lobe epilepsy: a task force report from the ILAE commission on diagnostic methods.
PKC in various in vitro and in vivo systems, thereby affecting neuronal Epilepsia 54, 1315–1329.
function and excitability (Vezzani and Viviani, 2015). Therefore, the pos- Brandon, N.J., Delmas, P., Kittler, J.T., McDonald, B.J., Sieghart, W., Brown, D.A., et al., 2000.
GABAA receptor phosphorylation and functional modulation in cortical neurons by a
sibility that IL-1R1 coupling to PKC is an oocyte-specific feature is unlikely. protein kinase C-dependent pathway. J. Biol. Chem. 275, 38856–38862.
Notably, IL-1β does not affect IGABA in control tissue. This lack of ef- Bright, D.P., Smart, T.G., 2013. Protein kinase C regulates tonic GABA(A) receptor-mediated
fect may be due either to the relatively low expression of IL-1R1 and inhibition in the hippocampus and thalamus. Eur. J. Neurosci. 38, 3408–3423.
Brooks-Kayal, A.R., Shumate, M.D., Jin, H., Rikhter, T.Y., Coulter, D.A., 1998. Selective
IRAK-1 in control compared with epileptic tissue, thus resulting in a
changes in single cell GABA(A) receptor subunit expression and function in temporal
less efficient activation of down-stream molecular signaling affecting lobe epilepsy. Nat. Med. 4, 1166–1172.
GABAA-R, or to up-regulation in epileptic tissue of a specific GABAA-R Chi, M., Ye, Y., Zhang, X.D., Chen, J., 2014. Insulin induces drug resistance in melanoma
subunit composition (Brooks-Kayal et al., 1998; Pirker et al., 2003; through activation of the PI3K/Akt pathway. Drug Des. Dev. Ther. 8, 255–262.
Conti, L., Palma, E., Roseti, C., Lauro, C., Cipriani, R., de Groot, M., et al., 2011. Anomalous
Sperk, 2007,2009) that is particularly sensitive to IL-1β; both scenarios levels of Cl-transporters cause a decrease of GABAergic inhibition in human
are conceivable and not mutually exclusive. peritumoral epileptic cortex. Epilepsia 52, 1635–1644.
In summary, since the IL-1β/IL1R1/IRAK1 signaling is activated in Dubé, C., Vezzani, A., Behrens, M., Bartfai, T., Baram, T.Z., 2005. Interleukin-1beta contributes
to the generation of experimental febrile seizures. Ann. Neurol. 57, 152–155.
human and experimental TLE, it may cause a protracted reduction of Dubé, C.M., Ravizza, T., Hamamura, M., Zha, Q., Keebaugh, A., Fok, K., et al., 2010.
GABAA-R current, thus contributing to hyperexcitability and resulting Epileptogenesis provoked by prolonged experimental febrile seizures: mechanisms
in seizure generation and recurrence. Since drugs specifically blocking and biomarkers. J. Neurosci. 30, 7484–7494.
Eusebi, F., Palma, E., Amici, M., Miledi, R., 2009. Microtransplantation of ligand-gated
the IL-1β system are clinically available (Vezzani and Viviani, 2015), receptor-channels from fresh or frozen nervous tissue into Xenopus oocytes: a potent
we suggest that this mechanism may be therapeutically targeted in tool for expanding functional information. Prog. Neurobiol. 88, 32–40.
drug-resistant epilepsies associated with neuroinflammation (Vezzani Fabene, P.F., Bramanti, P., Constantin, G., 2010. The emerging role for chemokines in
epilepsy. J. Neuroimmunol. 224, 22–27.
et al., 2011a,b).
González, M.I., Grabenstatter, H.I., Cea-Del, Christian A., Rio, C.A., 2015. Seizure-related
regulation of GABAA receptors in spontaneously epileptic rats. Neurobiol. Dis. 77,
5. Conclusions and outlook 246–256.
Gottipati, S., Rao, N.L., Fung-Leung, W.P., 2008. IRAK1: a critical signaling mediator of
innate immunity. Cell. Signal. 20, 269–276.
We report the novel finding that pathophysiological concentrations Henshall, D.C., Clark, R.S., Adelson, P.D., Chen, M., Watkins, S.C., Simon, R.P., et al., 2000.
of IL-1β significantly decrease GABAA-evoked currents in hippocampal Alterations in bcl-2 and caspase gene family protein expression in human temporal
lobe epilepsy. Neurology 55, 250–257.
and cortical specimens of patients with TLE characterized by the induc- Iyer, A., Zurolo, E., Spliet, W.G., van Rijen, P.C., Baayen, J.C., Gorter, J.A., et al., 2010.
tion of IL-1R1/IRAK1 signaling. This effect was not observed in control Evaluation of the innate and adaptive immunity in type I and type II focal cortical
specimens from patients without epilepsy and ongoing inflammatory dysplasias. Epilepsia 51, 1763–1773.
Iyer, A., Zurolo, E., Prabowo, A., Fluiter, K., Spliet, W.G., van Rijen, P.C., et al., 2012.
processes. Likewise, IL-1β reduced GABAA currents in cortical slices
MicroRNA-146a: a key regulator of astrocyte-mediated inflammatory response.
from epileptic rats, but not in control rats. These findings should prompt PLoS ONE 7 (9), e44789. http://dx.doi.org/10.1371/journal.pone.0044789.
additional investigations into the GABAA-R subunits that are specifically Jung, R., 1958. Excitation, inhibition and coordination of cortical neurones. Exp. Cell Res.
14, 262–271.
targeted by IL-1β, to see whether IL-1β affects synaptic or extrasynaptic
Kwan, P., Arzimanoglou, A., Berg, A.T., Brodie, M.J., Allen Hauser, W., Mathern, G., et
GABAergic transmission, or both. The IL-1β effect on GABA-mediated al., 2010. Definition of drug resistant epilepsy: consensus proposal by the ad hoc
neurotransmission may contribute to the well-established ictogenic task force of the ILAE commission on therapeutic strategies. Epilepsia 51,
actions of this cytokine, thus supporting the use of anti-inflammatory 1069–1077.
Macdonald, R.L., Kang, J.Q., Gallagher, M.J., 2010. Mutations in GABAA receptor subunits
drugs as adjuvant treatment in pharmacoresistant forms of epilepsy. associated with genetic epilepsies. J. Physiol. 588, 1861–1869.
Maroso, M., Balosso, S., Ravizza, T., Liu, J., Aronica, E., Iyer, A.M., et al., 2010. Toll-like receptor
4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to
Conflict of interest reduce seizures. Nat. Med. 16, 413–419.
Maroso, M., Balosso, S., Ravizza, T., Iori, V., Wright, C.I., French, J., et al., 2011. Interleukin-
None of the authors declares conflict of interest. 1β biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic
activity in mice. Neurotherapeutics 8, 304–315.
Mazzuferi, M., Palma, E., Martinello, K., Maiolino, F., Roseti, C., Fucile, S., et al., 2010. En-
Acknowledgments hancement of GABA(A)-current run-down in the hippocampus occurs at the first
spontaneous seizure in a model of temporal lobe epilepsy. Proc. Natl. Acad. Sci. U. S.
A. 107, 3180–3185.
This work was supported by AICE-FIRE 2014 (E.P.), PRIN project Miledi, R., Palma, E., Eusebi, F., 2006. Microtransplantation of neurotransmitter receptors
2010 (C.R.), Ri.MED Foundation (P. C.), EPITARGET (FP7/2007–2013), from cells to Xenopus oocyte membranes: new procedure for ion channel studies.
grant agreement no. 602102 (A.V. and E.A.), and Fondazione Monzino Methods Mol. Biol. 322, 347–355.
Musumeci, G., Grasselli, G., Rossi, S., De Chiara, V., Musella, A., Motta, C., et al., 2011. Tran-
grant no. 5600 (A.V.). We thank Gail Bell (NIHR University College
sient receptor potential vanilloid 1 channels modulate the synaptic effects of TNF-α
London, UK) for language editing and Aldo Giovannelli (University of and of IL-1β in experimental autoimmune encephalomyelitis. Neurobiol. Dis. 43,
Aquila, Italy) and Sergio Fucile (University of Rome Sapienza) for helping 669–677.
Palma, E., Trettel, F., Fucile, S., Renzi, M., Miledi, R., Eusebi, F., 2003. Microtransplantation
with statistics.
of membranes from cultured cells to Xenopus oocytes: a method to study neurotrans-
mitter receptors embedded in native lipids. Proc. Natl. Acad. Sci. U. S. A. 100,
Appendix A. Supplementary data 2896–2900.
Palma, E., Torchia, G., Limatola, C., Trettel, F., Arcella, A., Cantore, G., et al., 2005. BDNF
modulates GABAA receptors microtransplanted from the human epileptic brain to
Supplementary data to this article can be found online at http://dx. Xenopus oocytes. Proc. Natl. Acad. Sci. U. S. A. 102, 1667–1672.
doi.org/10.1016/j.nbd.2015.07.003. Palma, E., Roseti, C., Maiolino, F., Fucile, S., Martinello, K., Mazzuferi, M., et al., 2007. The
GABAA-current run-down of temporal lobe epilepsy is associated with repetitive ac-
tivation of GABAA receptors with low sensititiviy ot GABA and Zn2+. Proc. Natl. Acad.
References Sci. 104, 20944–20948.
Pirker, S., Schwarzer, C., Czech, T., Baumgartner, C., Pockberger, H., Maier, H., et al., 2003.
Akin, D., Ravizza, T., Maroso, M., Carcak, N., Eryigit, T., Vanzulli, I., et al., 2011. IL-1β is Increased expression of GABA(A) receptor beta-subunits in the hippocampus of
induced in reactive astrocytes in the somatosensory cortex of rats with genetic patients with temporal lobe epilepsy. J. Neuropathol. Exp. Neurol. 62, 820–834.
absence epilepsy at the onset of spike-and-wave discharges, and contributes to Racine, R.J., 1972. Modification of seizure activity by electrical stimulation. II. Motor
their occurrence. Neurobiol. Dis. 44, 59–69. seizure. Electroencephalogr. Clin. Neurophysiol. 32, 281–294.
320 C. Roseti et al. / Neurobiology of Disease 82 (2015) 311–320

Ragozzino, D., Palma, E., Di Angelantonio, S., Amici, M., Mascia, A., Arcella, A., et al., 2005. Sperk, G., Drexel, M., Pirker, S., 2009. Neuronal plasticity in animal models and the epileptic
Rundown of GABA type A receptors is a dysfunction associated with human drug- human hippocampus. Epilepsia 50 (Suppl. 12), 29–31.
resistant mesial temporal lobe epilepsy. Proc. Natl. Acad. Sci. U. S. A. 102, Tan, C.C., Zhang, J.G., Tan, M.S., Chen, H., Meng, D.W., Jiang, T., et al., 2015. NLRP1
15219–15223. inflammasome is activated in patients with medial temporal lobe epilepsy and con-
Ravizza, T., Lucas, S.M., Balosso, S., Bernardino, L., Ku, G., Noé, F., et al., 2006. Inactivation of tributes to neuronal pyroptosis in amygdala kindling-induced rat model.
caspase-1 in rodent brain: a novel anticonvulsive strategy. Epilepsia 47, 1160–1168. J. Neuroinflammation 12, 18. http://dx.doi.org/10.1186/s12974-014-0233-0.
Ravizza, T., Gagliardi, B., Noé, F., Vaghi, V., Sifringer, M., Vezzani, A., 2008. Innate and adap- Vezzani, A., Viviani, B., 2015. Neuromodulatory properties of inflammatory cytokines and
tive immunity during epileptogenesis and spontaneous seizures: evidence from ex- their impact on neuronal excitability. Neuropharmacology 96, 70–82.
perimental models and human temporal lobe epilepsy. Neurobiol. Dis. 29, 142–160. Vezzani, A., French, J., Bartfai, T., Baram, T.Z., 2011a. The role of inflammation in epilepsy.
Röder, C., Schwarzer, C., Vezzani, A., Gobbi, M., Mennini, T., Sperk, G., 1996. Autoradio- Nat. Rev. Neurol. 7, 31–40.
graphic analysis of neuropeptide Y receptor binding sites in the rat hippocampus Vezzani, A., Maroso, M., Balosso, S., Sanchez, M.A., Bartfai, T., 2011b. IL-1 receptor/Toll-like
after kainic acid-induced limbic seizures. Neuroscience 70, 47–55. receptor signaling in infection, inflammation, stress and neurodegeneration couples
Roseti, C., Fucile, S., Lauro, C., Martinello, K., Bertollini, C., Esposito, V., et al., 2013. hyperexcitability and seizures. Brain Behav. Immun. 25, 1281–1289.
Fractalkine/CX3CL1 modulates GABAA currents in human temporal lobe epilepsy. Viviani, B., Bartesaghi, S., Gardoni, F., Vezzani, A., Behrens, M.M., Bartfai, T., et al., 2003.
Epilepsia 54, 1834–1844. Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase
Scharfman, H.E., Brooks-Kayal, A.R., 2014. Is plasticity of GABAergic mechanisms relevant through activation of the Src family of kinases. J. Neurosci. 23, 8692–8700.
to epileptogenesis? Adv. Exp. Med. Biol. 813, 133–150. Wang, S., Cheng, Q., Malik, S., Yang, J., 2000. Interleukin-1beta inhibits gamma-
Schmidt, D., Löscher, W., 2005. Drug resistance in epilepsy: putative neurobiological and aminobutyric acid type A (GABA(A) receptor) current in cultured hippocampal
clinical mechanisms. Epilepsia 46, 858–877. neurons. J. Pharmacol. Exp. Ther. 292, 497–504.
Smith, P.K., Krohn, R.I., Hermanson, G.T., Mallia, A.K., Gartner, F.H., Provenzano, M.D., et al., Zeise, M.L., Espinoza, J., Morales, P., Nalli, A., 1997. Interleukin-1beta does not increase
1985. Measurement of protein using bicinchoninic acid. Annal. Biochem. 150, 76–85. synaptic inhibition in hippocampal CA3 pyramidal and dentate gyrus granule cells
Sperk, G., 2007. Changes in GABAA receptors in status epilepticus. Epilepsia 48 (Suppl. 8), of the rat in vitro. Brain Res. 768, 341–344.
11–13.

You might also like