ArticlePDF Available

Unexpected Effect of IL-1β on the Function of GABAA Receptors in Pediatric Focal Cortical Dysplasia

Authors:

Abstract and Figures

Focal cortical dysplasia (FCD) type II is an epileptogenic malformation of the neocortex, as well as a leading cause of drug-resistant focal epilepsy in children and young adults. The synaptic dysfunctions leading to intractable seizures in this disease appear to have a tight relationship with the immaturity of GABAergic neurotransmission. The likely outcome would include hyperpolarizing responses upon activation of GABAARs. In addition, it is well-established that neuroinflammation plays a relevant role in the pathogenesis of FCD type II. Here, we investigated whether IL-1β, a prototypical pro-inflammatory cytokine, can influence GABAergic neurotransmission in FCD brain tissues. To this purpose, we carried out electrophysiological recordings on Xenopus oocytes transplanted with human tissues and performed a transcriptomics analysis. We found that IL-1β decreases the GABA currents amplitude in tissue samples from adult individuals, while it potentiates GABA responses in samples from pediatric cases. Interestingly, these cases of pediatric FCD were characterized by a more depolarized EGABA and an altered transcriptomics profile, that revealed an up-regulation of chloride cotransporter NKCC1 and IL-1β. Altogether, these results suggest that the neuroinflammatory processes and altered chloride homeostasis can contribute together to increase the brain excitability underlying the occurrence of seizures in these children.
Content may be subject to copyright.
Citation: Alfano, V.; Romagnolo, A.;
Mills, J.D.; Cifelli, P.; Gaeta, A.;
Morano, A.; Mühlebner, A.; Aronica,
E.; Palma, E.; Ruffolo, G. Unexpected
Effect of IL-1βon the Function of
GABAAReceptors in Pediatric Focal
Cortical Dysplasia. Brain Sci. 2022,12,
807. https://doi.org/10.3390/
brainsci12060807
Academic Editors: Enrico Cherubini
and Yehezkel Ben-Ari
Received: 30 May 2022
Accepted: 17 June 2022
Published: 19 June 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
brain
sciences
Article
Unexpected Effect of IL-1βon the Function of GABAA
Receptors in Pediatric Focal Cortical Dysplasia
Veronica Alfano 1,2, , Alessia Romagnolo 3, , James D. Mills 3,4,5, Pierangelo Cifelli 6, Alessandro Gaeta 1,
Alessandra Morano 7, Angelika Mühlebner 3,8, Eleonora Aronica 3, 9, *, Eleonora Palma 1,2 ,*,‡
and Gabriele Ruffolo 1,2,
1Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti,
University of Rome Sapienza, 00185 Rome, Italy; veronica.alfano@uniroma1.it (V.A.);
alessandro.gaeta@uniroma1.it (A.G.); gabriele.ruffolo@uniroma1.it (G.R.)
2IRCCS San Raffaele Roma, 00163 Rome, Italy
3Department of (Neuro) Pathology Amsterdam Neuroscience, Amsterdam UMC Location University of
Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; a.romagnolo@amsterdamumc.nl (A.R.);
j.d.mills@amsterdamumc.nl (J.D.M.); a.muehlebnerfahrngruber@amsterdamumc.nl (A.M.)
4Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology,
London WC1E 6BT, UK
5Chalfont Centre for Epilepsy, Chalfont St Peter SL9 0RJ, UK
6Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
pierangelo.cifelli@univaq.it
7Department of Human Neuroscience, University of Rome Sapienza, 00185 Rome, Italy;
alessandra.morano@uniroma1.it
8Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
9Stichting Epilepsie Instellingen Nederland, 2013 SW Heemstede, The Netherlands
*Correspondence: e.aronica@amsterdamumc.nl (E.A.); eleonora.palma@uniroma1.it (E.P.)
These authors contributed equally to this work.
These authors contributed equally to this work.
Abstract:
Focal cortical dysplasia (FCD) type II is an epileptogenic malformation of the neocortex, as
well as a leading cause of drug-resistant focal epilepsy in children and young adults. The synaptic
dysfunctions leading to intractable seizures in this disease appear to have a tight relationship with
the immaturity of GABAergic neurotransmission. The likely outcome would include hyperpolarizing
responses upon activation of GABA
A
Rs. In addition, it is well-established that neuroinflammation
plays a relevant role in the pathogenesis of FCD type II. Here, we investigated whether IL-1
β
,
a prototypical pro-inflammatory cytokine, can influence GABAergic neurotransmission in FCD
brain tissues. To this purpose, we carried out electrophysiological recordings on Xenopus oocytes
transplanted with human tissues and performed a transcriptomics analysis. We found that IL-1
β
decreases the GABA currents amplitude in tissue samples from adult individuals, while it potentiates
GABA responses in samples from pediatric cases. Interestingly, these cases of pediatric FCD were
characterized by a more depolarized E
GABA
and an altered transcriptomics profile, that revealed an
up-regulation of chloride cotransporter NKCC1 and IL-1
β
. Altogether, these results suggest that the
neuroinflammatory processes and altered chloride homeostasis can contribute together to increase
the brain excitability underlying the occurrence of seizures in these children.
Keywords: human GABAAreceptor; GABAAcurrent; FCD; IL-1β
1. Introduction
Focal cortical dysplasias (FCDs) are a group of malformations of cortical development
(MCD), frequently associated with drug-resistant epilepsy, and are predominant among the
pediatric population [
1
3
]. FCDs type II are the most common MCD in epilepsy surgery
case series [
3
]. From a histopathological point of view, FCDs type II are characterized
Brain Sci. 2022,12, 807. https://doi.org/10.3390/brainsci12060807 https://www.mdpi.com/journal/brainsci
Brain Sci. 2022,12, 807 2 of 10
by alteration of the cortical lamination and cellular abnormalities. Furthermore, these
architectural abnormalities are coupled with a “dysmature” function of the neurons found
in the dysplastic areas [
4
]. Indeed, an anomalous synaptic transmission may be one of
the key pathogenic factors leading to epilepsy in this disease. Specifically, it appears that
hallmark alterations in GABAergic neurotransmission, determining its “immature” activity,
have a pivotal role in this process, since GABA-mediated inhibitory neurotransmission
becomes unable to regulate brain excitability and eventually even drives epileptiform
activity by itself [5,6].
At present, it is a common speculation that an abnormal GABA current reversal poten-
tial (E
GABA
), mainly determined by the dysregulation of the expression of the two main
cation-chloride cotransporters (NKCC1 and KCC2) [
7
,
8
], could represent one of the key
pathophysiological events leading to this “depolarizing GABAergic transmission” [
9
,
10
].
Further support to this hypothesis comes from the observation that these pathophysiologi-
cal mechanisms are a common feature of neurodevelopmental disorders, characterized by
a high incidence of epilepsy, such as FCD [
4
], TSC [
11
], Rett syndrome [
12
,
13
] or Dravet
syndrome [14].
In particular, it is widely known that pediatric FCD shows a peculiar alteration of
GABAergic neurotransmission [
4
]. Indeed, GABA responses can behave as depolariz-
ing, especially in the most severe cases [
15
], and increased GABAergic synaptic activity
can be responsible of the network hyperexcitability, instead of acting as an inhibitory
mechanism [16].
On the other hand, it is known that the generation and recurrence of seizures have
widespread consequences on whole brain homeostasis and the onset of a “vicious cy-
cle” of neuroinflammation is likely one of the most important factors contributing to the
consolidation of the epileptogenic mechanisms [
17
]. In the specific case of FCD type II,
recent evidence supports the hypothesis that factors contributing to the maintenance of
the pro-inflammatory drive in this condition include the up-regulation of inflammatory
mediators in dysmorphic cells [
18
], imbalance of cytokines’ regulatory networks [
19
] and
the pro-inflammatory potential of the seizures themselves [20].
The prototypical pro-inflammatory interleukin-1
β
(IL-1
β
) has a prominent role in
these inflammatory processes in both epileptic patients and animal models of epilepsy [
21
].
Accordingly, we have previously shown that IL-1
β
can decrease GABA currents amplitude
in human drug-resistant temporal lobe epilepsy (TLE) by activation of IL-1
β
signaling [
22
].
Here, we investigated the effect of IL-1
β
on GABAergic neurotransmission in pediatric
brain tissues from patients affected by FCD IIb, which is the most severe form of FCD
considering the high level of neuroinflammation [
18
]. Indeed, even though several reports
supporting the role of this cytokine in neurodevelopmental epilepsies have already been
published [
23
,
24
], there is still scarce evidence regarding its potential role in the “plastic”
neurotransmission that characterizes neurodevelopmental disorders.
2. Materials and Methods
2.1. Patients
The cases included in this study were obtained from the archives of the Departments
of Neuropathology of the Amsterdam UMC (Amsterdam, The Netherlands) and the Uni-
versity Medical Center Utrecht (UMCU, Utrecht, The Netherlands). Cortical brain samples
were obtained from patients undergoing surgery for drug-resistant epilepsy and diagnosed
with FCD type IIb (with brain somatic mutations in the MTOR gene) [
2
]. After resection,
the tissue was immediately snap-frozen in liquid nitrogen and then part of the sample
was used to perform the electrophysiology experiments. All the autopsies were performed
within 16 to <48 h after death, with the acquisition of appropriate written consent for brain
autopsy and subsequent use for research purposes. Control autopsy cases had no known
history of epilepsy, a normal cortical structure for the corresponding age and no signifi-
cant brain pathology. The transcriptional profiles of post-mortem and surgical resected
tissues have previously been compared, showing minimal differences if the tissue is of
Brain Sci. 2022,12, 807 3 of 10
high quality (i.e., handled and stored as in our study) [
25
]. Tissue was obtained and used
in accordance with the Declaration of Helsinki and the Amsterdam UMC Research Code
provided by the Medical Ethics Committee. Please refer to Supplementary Table S1 for
clinical details of the patients. The electrophysiology experiments were performed with
three pediatric samples up to 5 years old (Table 1). However, for the bioinformatics analysis,
we had a larger cohort of patients and only patients below 12 years old were considered
pediatric (
18 patients
). Adult patients diagnosed with FCD IIb, including those 18 years
old, underwent transcriptomic analysis (12 patients), whilst three of these were used for the
electrophysiology experiments (Table 1). A set of electrophysiology experiments were also
performed on two FCD IIa tissues (3 years old, female; 11 years old, male). Throughout
the manuscript, patients are referred to with the symbol “#”, and their clinical features are
summarized in Table 1.
Table 1. Clinical characteristics of the patients.
Patient Age at the Time of the Surgery (y) Epilepsy Onset (y) Gender Diagnosis ASMs
#1 2 0 M FCD IIb OCZ, C, VPA,
#2 3 0 F FCD IIb LEV, OCZ, C, VPA, CL
#3 5 3 M FCD IIb C, CL
#4 18 2 M FCD IIb C, VPA, LMT, LCM
#5 44 10 F FCD IIb LEV, OCZ
#6 45 12 M FCD IIb LEV, OCZ
Three pediatric patients; three adult patients; Abbreviations: M = male, F = female, ASM = anti-seizure med-
ication, C = Carbamazepine, CL = Clobazam, LCM = Lacosamide, LEV = Levetiracetam, LMT= Lamotrigine,
OCZ = Oxcarbazepine, VPA = Valproic acid.
2.2. Membrane Preparation
All the tissues used for electrophysiology were received in dry ice and processed
immediately or stored at
80
. Preparation of human membranes and their injection in
Xenopus laevis oocytes were performed as previously described [
26
]. Briefly, tissues were
homogenized in a membrane buffer solution (200 mM glycine, 150 mM NaCl, 50 mM EGTA,
50 mM EDTA, and 300 mM sucrose; plus 20
µ
L of protease inhibitors “P2714; Sigma”; pH 9,
adjusted with NaOH). Afterwards, the material was centrifuged for 15 min at
9500×g
.
Then, the supernatant was centrifuged for 2 h at 100,000
×
gwith an ultra-centrifuge
(Beckman-Coulter). Finally, the pellet was washed with sterile water, re-suspended in
assay buffer (glycine 5 mM) and immediately used or stored at
80
until use. The use of
Xenopus laevis frogs and the surgical procedures for oocytes extraction and use conformed
to the Italian Ministry of Health guidelines and were approved by the same institution
(authorization no 427/2020-PR).
2.3. Xenopus Oocytes Electrophysiology
All the experiments with microtransplanted oocytes were carried out 24–48 h after
cytoplasmic injection [
26
]. GABA-evoked currents (I
GABA
) were recorded using the tech-
nique of “two-electrode voltage clamp” [
27
] after the oocytes were placed in a recording
chamber (0.1 mL volume) and continuously perfused with oocyte Ringer solution (OR:
NaCl 82.5 mM; KCl 2.5 mM; CaCl
2
2.5 mM; MgCl
2
1 mM; Hepes 5 mM, adjusted to pH 7.4
with NaOH) at room temperature (20–22
C). GABA application was controlled through
a computer-operated gravity driven multi-valve perfusion system (9–10 mL/min) (Bi-
ologique RSC-200; Claix, France). With this setup, 0.5 to 1 s are enough to completely
replace the entire volume of applied solution in the recording chamber.
In all the experiments, the stability of GABA-evoked currents (I
GABA
) was evaluated
on two consecutive GABA applications, separated by a 4 min washout. Only the cells
that showed a <5% variation in current amplitude were used to test the effect of IL-1
β
.
Variation in current amplitude was calculated comparing the mean current elicited by
the two GABA applications before and after exposure to cytokines and/or inhibitors.
GABA current reversal potential (E
GABA
) was calculated by constructing current-voltage
Brain Sci. 2022,12, 807 4 of 10
(I-V) relationships that were then elaborated by a linear regression curve-fitting software
(Sigmaplot 12, Systat software Inc., Chicago, IL, USA). In a specific set of experiments, we
used IL1-Ra to block IL-1
β
‘s effect. In these experiments, we pre-incubated the cells for
30 min with the blocker alone, and then proceeded to the incubation with the cytokine
plus the inhibitor for two hours. For the incubation, IL-1
β
and IL-1Ra were diluted at the
desired concentration (specified for each experiment) in Barth’s modified saline solution
(88 mM NaCl; 1 mM KCl; 2.4 mM NaHCO
3
; 10 mM HEPES; 0.82 mM MgSO
4
; 0.33 mM
Ca(NO
3
)
2
; 20.41 mM CaCl
2
). IL-1
β
was purchased from Peprotech (London, UK) and
human IL-1Ra was purchased from Invitrogen (Waltham, MA, USA).
2.4. RNA-Seq Library Preparation and Sequencing
All library preparation and sequencing were performed at GenomeScan (Leiden,
The Netherlands
). The NEBNext Ultra II Directional RNA Library Prep Kit for Illumina
(New England Biolabs, Ipswich, MA, USA) was used for sample processing. Sample
preparation was performed according to the protocol “NEBNext Ultra II Directional RNA
Library prep Kit for Illumina” (NEB #E7760S/L). The mRNA isolation was performed
from the total RNA using oligo-dT magnetic beads and cDNA synthesis followed. Next,
sequencing adapters were ligated to the cDNA fragments followed by PCR amplification.
Clustering and DNA-sequencing were performed using the NovaSeq6000 (Illumina, Foster
City, CA, USA) in accordance with manufacturers’ guidelines. All samples underwent
paired-end sequencing of 150 nucleotides in length and the mean read depth per sample
was 47 million reads.
2.5. Bioinformatics Analysis of RNA-Seq Data
The Bestus Bioinformaticus Decontamination Using Kmers (BBDuk) tool from the
BBTools suite was used for adapter removal, quality trimming and removal of contaminant
sequences (ribosomal/bacterial) [
28
]. A phred33 score of 20 was used to assess the quality
of the read, any read shorter than 31 nucleotides in length was excluded from the down-
stream analysis. Reads were aligned directly to the human GRCh38 reference transcriptome
(Gencode version 33) [
29
] using Salmon v0.11.3 [
30
]. Transcript counts were summarized
to the gene level and scaled using library size and average transcript length using the R
package tximport [
31
]. Genes detected in less than 20% of the samples in any diagnosis
and with counts less than 6 across all samples were filtered out. The gene counts were
than normalized using the weighted trimmed mean of M-values (TMM) method using
the R package edgeR [
32
]. The normalized counts were than log2 transformed using the
voom function from the R package limma [
33
]. The subsequent differential expression
was carried out using the R package limma. A linear model was fit for each gene and
moderated t-statistic was calculated after applying an empirical Bayes smoothing to the
standard errors. Differential expression was defined by a Benjamini–Hochberg adjusted
p-value < 0.05. The analysis compared FCD IIb pediatric tissue samples (18 patients) and
matched control cortices (6 patients).
3. Results
3.1. IL-1βAffects the GABA Current Amplitude in Oocytes Injected with FCD IIb Membranes
At first, we recorded GABA-evoked currents from oocytes microinjected with mem-
branes from adult samples of the FCD IIb brain cortex (Table 1). We recorded responses
ranging from 11.8 nA to 250.0 nA (n= 24; #4–6; 250
µ
M GABA). Subsequently, in order
to test the effect of IL-1
β
on FCD, we measured GABA current amplitude (I
GABA
) before
and after a 2 h incubation with this cytokine (25 ng/mL). We observed that this treatment
induces a decrease in I
GABA
(I
GABA
= 98.4
±
16.6 nA and 87.8
±
16.8 nA, before and af-
ter IL-1
β
incubation, n= 14; p< 0.001, Wilcoxon signed rank test; # 3–5, Table 1). This
effect was comparable to that already reported in TLE tissues [
22
] and behaved similarly,
since IL-1
β
induced GABA current decrease in 70% of the cells (14/20), while the rest
did not respond to the cytokine [
22
]. As previously shown for TLE, this I
GABA
decrease
Brain Sci. 2022,12, 807 5 of 10
was blocked by 30 min pre-treatment with 10
µ
M IL-1Ra (I
GABA
= 86.6
±
10.6 nA and
88.7 ±12.07 nA
, before and after incubation with IL-1
β
+ IL-1Ra, n= 6; p> 0.05, paired
t-test; # 5, Table 1). Interestingly, when we repeated the same experiments on pediatric FCD
IIb samples (range: 5.6 nA to 193.0 nA, n= 44; #1–3; 250
µ
M GABA), we did not obtain the
same results, observing an opposite effect of IL-1
β
, which on these tissues and in the same
experimental conditions reported above (2 h incubation, 25 ng/mL) induced a potentiation
of I
GABA
(
IGABA = 45.4 ±10.9 nA
and 52.7
±
12.9 nA, before and after IL-1
β
incubation,
n= 31
;
p< 0.001
, Wilcoxon signed rank test; # 1–3, Table 1, Figure 1). Also in this case, the
effect was blocked by 30 min pre-treatment with 10
µ
M IL-1Ra (
IGABA = 28.8 ±6.2 nA
and
27.6 ±5.0 nA
, before and after incubation with IL-1
β
+ IL-1Ra, n= 8; p> 0.05, Wilcoxon
signed rank test; # 1–3, Table 1). Subsequently, we repeated the IL-1
β
incubation ex-
periments microinjecting the oocytes with membranes from two FCD IIa tissue samples
(3-year-old female and 11-year-old male). The results showed GABA current potentiation
also in these cases (I
GABA
= 40.9
±
8.4 nA and 47.0
±
10.1 nA, before and after IL-1
β
incubation, n= 16; p< 0.01, Wilcoxon signed rank test).
Brain Sci. 2022, 12, x FOR PEER REVIEW 5 of 11
comparable to that already reported in TLE tissues [23] and behaved similarly, since IL-
1β induced GABA current decrease in 70% of the cells (14/20), while the rest did not re-
spond to the cytokine.[23]. As previously shown for TLE, this IGABA decrease was blocked
by 30 min pre-treatment with 10 μM IL-1Ra (IGABA= 86.6 ± 10.6 nA and 88.7 ± 12.07 nA,
before and after incubation with IL-1β + IL-1Ra, n = 6; p > 0.05, paired t-test; # 5, Table 1).
Interestingly, when we repeated the same experiments on pediatric FCD IIb samples
(range: 5.6 nA to 193.0 nA, n = 44; #13; 250 μM GABA), we did not obtain the same results,
observing an opposite effect of IL-1β, which on these tissues and in the same experimental
conditions reported above (2 h incubation, 25 ng/mL) induced a potentiation of IGABA (IGABA
= 45.4 ± 10.9 nA and 52.7 ± 12.9 nA, before and after IL-1β incubation, n = 31; p < 0.001,
Wilcoxon signed rank test; # 13, Table 1, Figure 1). Also in this case, the effect was blocked
by 30 min pre-treatment with 10 μM IL-1Ra (IGABA = 28.8 ± 6.2 nA and 27.6 ± 5.0 nA, before
and after incubation with IL-1β + IL-1Ra, n= 8; p > 0.05, Wilcoxon signed rank test; # 13,
Table 1). Subsequently, we repeated the IL-1β incubation experiments microinjecting the
oocytes with membranes from two FCD IIa tissue samples (3-year-old female and 11-year-
old male). The results showed GABA current potentiation also in these cases (IGABA = 40.9
± 8.4 nA and 47.0 ± 10.1 nA, before and after IL-1β incubation, n = 16; p < 0.01, Wilcoxon
signed rank test).
Figure 1. IL1β potentiates GABA-evoked currents in pediatric FCD IIb tissue samples. The bar
graphs show the average GABA current percent (%) changes ± s.e.m. before and after incubation
with IL-1β (25 ng/mL; 2 h) in oocytes transplanted with adult FCD IIb tissues (blue bar; n = 14, # 4
6, Table 1) and pediatric FCD IIb tissues (red bar; n = 31, # 13, Table 1). Insets represent sample
currents before (left trace) and after (right trace) incubation with IL-1β in adult (lower inset) and
pediatric (upper inset) FCD IIb. White horizontal bars represent GABA application (250 μM). There
is a significantly different effect of IL- between the two experimental groups. * p < 0.05 with Stu-
dent t-test.
3.2. GABA Reversal Potential in Oocytes Injected with FCD IIb Membranes
In order to better characterize the IGABA in our samples, we performed additional ex-
periments to measure EGABA in pediatric FCD IIb samples. Interestingly, we found that
EGABA was more depolarized in pediatric samples (−17.2 ± 1.1 mV, n = 13; #13, Table 1,
Figure 2) compared to adult ones (22.0 ± 0.73 mV, n = 10; p < 0.05, Wilcoxon signed rank
test; #46, Table 1, Figure 2), suggesting that in pediatric patients, GABA currents have
unique characteristics. Indeed, these EGABA values are not comparable with those found in
neurons [8] but are consistent with those previously found using the microtransplantation
technique for the electrophysiological study of human brain tissue. [35] Furthermore, we
Figure 1.
IL
1
β
potentiates GABA-evoked currents in pediatric FCD IIb tissue samples. The bar
graphs show the average GABA current percent (%) changes
±
s.e.m. before and after incubation
with IL-1
β
(25 ng/mL; 2 h) in oocytes transplanted with adult FCD IIb tissues (blue bar; n= 14,
# 4–6, Table 1) and pediatric FCD IIb tissues (red bar; n= 31, # 1–3, Table 1). Insets represent sample
currents before (left trace) and after (right trace) incubation with IL-1
β
in adult (lower inset) and
pediatric (upper inset) FCD IIb. White horizontal bars represent GABA application (250
µ
M). There is
a significantly different effect of IL-1
β
between the two experimental groups. * p< 0.05 with Student
t-test.
3.2. GABA Reversal Potential in Oocytes Injected with FCD IIb Membranes
In order to better characterize the I
GABA
in our samples, we performed additional
experiments to measure E
GABA
in pediatric FCD IIb samples. Interestingly, we found that
E
GABA
was more depolarized in pediatric samples (
17.2
±
1.1 mV, n= 13; #1–3, Table 1,
Figure 2) compared to adult ones (
22.0
±
0.73 mV, n= 10; p< 0.05, Wilcoxon signed rank
test; #4–6, Table 1, Figure 2), suggesting that in pediatric patients, GABA currents have
unique characteristics. Indeed, these E
GABA
values are not comparable with those found in
neurons [
8
] but are consistent with those previously found using the microtransplantation
technique for the electrophysiological study of human brain tissue [
34
]. Furthermore,
we repeated the same measurements in two FCD IIa pediatric samples (see methods for
patients’ details) and we still found a depolarized E
GABA
, which was not statistically
different compared to that recorded in pediatric FCD IIb (16.8 ±0.3 mV, n= 8, p> 0.05).
Brain Sci. 2022,12, 807 6 of 10
Brain Sci. 2022, 12, x FOR PEER REVIEW 6 of 11
repeated the same measurements in two FCD IIa pediatric samples (see methods for pa-
tients details) and we still found a depolarized EGABA, which was not statistically different
compared to that recorded in pediatric FCD IIb (−16.8 ± 0.3 mV, n = 8, p > 0.05).
Figure 2. GABA reversal potential in FCD IIb tissue samples. The two panels represent the mean
EGABA value recorded on adult (A); black circles; n = 10, # 46, Table 1) and pediatric (B); red squares;
n = 13, # 13, Table 1) FCD samples. The dots represent mean ± s.e.m. of GABA currents at corre-
spondent holding potential value (VH) normalized to the maximum currents (Imax = 73.4 for adult
and 42.4 nA for pediatric). Insets represent sample GABAevoked currents (250 μM) at different VH
as shown in the respective panels. Note that the mean EGABA was significantly more depolarized in
pediatric FCD tissues compared to adult FCD (p < 0.05, Wilcoxon signed rank test).
3.3. Transcriptomic Analysis
In order to explain the aforementioned results, we performed transcriptomics analy-
sis on a cohort of pediatric FCD IIb tissue samples. We found that the pro-inflammatory
cytokine IL-and its receptor antagonist, IL-1Ra (IL-1 receptor antagonist) were signifi-
cantly upregulated in FCD IIb pediatric tissue samples (IL- log2FC = 4.535; IL-1Ra log2FC
of 2.042) compared to age-matched control cortices (Figure 3), suggesting IL-1Ra tries to
counteract the overexpression of IL-. However, their common receptor, IL-1R1 did not
show a significant upregulation in these samples (log2FC = 0.753) (Figure 3). Interestingly,
sodium-potassium-chloride cotransporter, NKCC1, was significantly up-regulated
(log2FC = 0.696) (Figure 3). Altogether, these results indicate the presence of an acute neu-
roinflammatory process in these children and altered chloride homeostasis.
Figure 2.
GABA reversal potential in FCD IIb tissue samples. The two panels represent the mean
E
GABA
value recorded on adult (
A
); black circles; n= 10, # 4–6, Table 1) and pediatric (
B
); red
squares; n= 13, # 1–3, Table 1) FCD samples. The dots represent mean
±
s.e.m. of GABA currents
at correspondent holding potential value (V
H
) normalized to the maximum currents (I
max
= 73.4
for adult and 42.4 nA for pediatric). Insets represent sample GABA
evoked currents (250
µ
M) at
different V
H
as shown in the respective panels. Note that the mean E
GABA
was significantly more
depolarized in pediatric FCD tissues compared to adult FCD (p< 0.05, Wilcoxon signed rank test).
3.3. Transcriptomic Analysis
In order to explain the aforementioned results, we performed transcriptomics analysis
on a cohort of pediatric FCD IIb tissue samples. We found that the pro-inflammatory cy-
tokine IL-1
β
and its receptor antagonist, IL-1Ra (IL-1 receptor antagonist) were significantly
upregulated in FCD IIb pediatric tissue samples (IL-1
β
log
2
FC = 4.535; IL-1Ra log
2
FC of
2.042) compared to age-matched control cortices (Figure 3), suggesting IL-1Ra tries to coun-
teract the overexpression of IL-1
β
. However, their common receptor, IL-1R1 did not show a
significant upregulation in these samples (log
2
FC = 0.753) (Figure 3). Interestingly, sodium-
potassium-chloride cotransporter, NKCC1, was significantly up-regulated (log
2
FC = 0.696)
(Figure 3). Altogether, these results indicate the presence of an acute neuroinflammatory
process in these children and altered chloride homeostasis.
Brain Sci. 2022, 12, x FOR PEER REVIEW 7 of 11
Figure 3. Expression levels of the genes of interest in FCD IIb pediatric tissue samples compared to
age-matched controls. IL, IL1Ra and NKCC1 are significantly upregulated in FCD IIb pediatric
tissue samples compared to controls. Expression levels are described in logarithmic scale. * p < 0.05;
**** p< 0.0001. A linear model was fit for each gene and moderated t-statistic was calculated after
applying an empirical Bayes smoothing to the standard errors. Those genes with a Benjamini
Hochberg adjusted p-value < 0.05 were considered significant. Differential expression analysis com-
pared 18 FCD IIb patients and six age-matched control cortices.
4. Discussion
This study focused on the analysis of the effect of IL-1β on GABAergic neurotrans-
mission in FCD IIb samples. This cytokine can decrease GABA currents amplitude in adult
FCD IIb, while it increases these responses in pediatric FCD samples. Interestingly, we
found that these latter tissues were indeed characterized by a more depolarized EGABA
compared to adult FCD IIb, thus suggesting an alteration in chloride homeostasis. [8] Fi-
nally, the transcriptomic analysis revealed an up-regulation of the expression of IL-1β, IL-
1Ra and NKCC1 in a cohort of pediatric FCD IIb samples that fits well with the aforemen-
tioned electrophysiological results.
The technical approach we used to perform our functional experiments, membrane
microtransplantation in Xenopus oocytes, has the potential of allowing electrophysiologi-
cal recordings from rare human diseases using little amounts of brain tissue, thus making
it easier to test cytokines or other mediators on these pathologies. On the other hand, with
this technique, we record “wholeglial and neuronal currents without discriminating
among cellular subtypes. Nonetheless, it has been demonstrated that neurotransmitter re-
ceptors transplanted in Xenopus oocytes from transfected cells retain their functional char-
acteristics [36].
To our knowledge, this is the first time that IL-1β was reported to affect GABAergic
function in human FCD IIb tissues, even though its ability to reduce the amplitude of
GABA-evoked currents upon activation of its specific receptor was already described in
human TLE [23].
Figure 3.
Expression levels of the genes of interest in FCD IIb pediatric tissue samples compared to
age-matched controls. IL
1
β
,IL
1Ra and NKCC1 are significantly upregulated in FCD IIb pediatric
tissue samples compared to controls. Expression levels are described in logarithmic scale. * p< 0.05;
**** p< 0.0001. A linear model was fit for each gene and moderated t-statistic was calculated
after applying an empirical Bayes smoothing to the standard errors. Those genes with a Benjamini–
Hochberg adjusted p-value < 0.05 were considered significant. Differential expression analysis
compared 18 FCD IIb patients and six age-matched control cortices.
Brain Sci. 2022,12, 807 7 of 10
4. Discussion
This study focused on the analysis of the effect of IL-1
β
on GABAergic neurotransmis-
sion in FCD IIb samples. This cytokine can decrease GABA currents amplitude in adult FCD
IIb, while it increases these responses in pediatric FCD samples. Interestingly, we found
that these latter tissues were indeed characterized by a more depolarized E
GABA
compared
to adult FCD IIb, thus suggesting an alteration in chloride homeostasis [
8
]. Finally, the
transcriptomic analysis revealed an up-regulation of the expression of IL-1
β
,IL-1Ra and
NKCC1 in a cohort of pediatric FCD IIb samples that fits well with the aforementioned
electrophysiological results.
The technical approach we used to perform our functional experiments, membrane
microtransplantation in Xenopus oocytes, has the potential of allowing electrophysiological
recordings from rare human diseases using little amounts of brain tissue, thus making
it easier to test cytokines or other mediators on these pathologies. On the other hand,
with this technique, we record “whole” glial and neuronal currents without discriminating
among cellular subtypes. Nonetheless, it has been demonstrated that neurotransmitter
receptors transplanted in Xenopus oocytes from transfected cells retain their functional
characteristics [35].
To our knowledge, this is the first time that IL-1
β
was reported to affect GABAergic
function in human FCD IIb tissues, even though its ability to reduce the amplitude of
GABA-evoked currents upon activation of its specific receptor was already described in
human TLE [22].
Additionally, we also described the effect of IL-1
β
on two FCD IIa pediatric tissue
samples, where we obtained the same results as the recordings in FCD IIb tissues. Indeed,
FCD IIa and IIb represent two histopathological subtypes of FCD type II, and FCD IIb is
further distinguished from FCD IIa by the additional presence of balloon cells [2,36]. This
broadens the relevance of our findings, since we can hypothesize that the alteration in
synaptic transmission by inflammatory mediators may be a general feature of FCD, at least
of those forms frequently associated with drug-resistant epilepsy.
Here, we can infer that the mechanism underlying IL-1
β
mediated modulation of
GABAergic transmission depends upon the activation of transplanted
IL-1β
receptors
(
IL-1R1
) [
22
], since the blockade with IL-1Ra, the endogenous antagonist [
37
] which specif-
ically prevents the binding of IL-
β
to this receptor, could prevent both GABA current
decrease and increase in adult and pediatric specimens, respectively.
Alongside this mechanism, it is also possible that GABA
A
Rs modulation takes place
as a consequence of the altered expression or dysregulated function of the two main cation-
chloride cotransporters expressed in the brain: NKCC1, which mediates chloride influx, and
KCC2, responsible for chloride extrusion [
8
]. Here, we described an about 5 mV E
GABA
shift
in pediatric FCD, which correlates with the reported NKCC1 upregulation. Furthermore,
this is also supported by previous findings describing a depolarizing shift of about 7–8 mV
in membranes extracted from epileptic subiculum, where both an upregulation of NKCC1
and a downregulation of KCC2 were reported [34].
Interestingly, recent evidence supports the hypothesis that inflammatory stimuli, in
particular IL-1
β
, can alter GABAergic neurotransmission also by modulating the expression
and function of the aforementioned chloride transporters [
38
]. This is particularly relevant,
since chloride homeostasis contributes to the pathogenesis of several neurodevelopmental
disorders such as TSC, autism and epileptic syndromes [
38
]. Any kind of pathogenic factor
acting on this delicate equilibrium, such as neuroinflammation, could serve as a therapeutic
target for conditions that are often characterized by seizures resistant to known ASMs
or medically untreatable cognitive impairment. In pediatric patients, the inflammatory
processes can originate from prenatal stress, infection or traumatic injuries [
38
,
39
] that
contribute to the occurrence of seizures in the first months of life. Therefore, in these
patients, it could be relevant to therapeutically intervene as soon as possible to quench the
neuroinflammatory processes.
Brain Sci. 2022,12, 807 8 of 10
Interestingly, our data indicate that IL-1Ra is also up-regulated, suggesting an attempt
to compensate the IL-1
β
increase in our cohort of FCD IIb samples. However, the need for
epilepsy surgery in these cases suggests that the endogenous anti-inflammatory cytokines
failed to dampen inflammation, which can lead to brain hyperexcitability [40].
The role of GABAergic transmission in this scenario is indeed pivotal. An interesting
hypothesis revolves around the ability of aberrant GABAergic transmission to promote epilep-
togenesis in malformed FCD cerebral cortex, especially in pediatric
cases [4,6,41]
. In fact, the
shift in GABA reversal potential has been associated with a state of brain “dysmaturity that is
a hallmark of several neurodevelopmental diseases such as
TSC [11,42], Dravet [14]
and Rett
syndromes [12,13].
Here, we provided novel evidence in favor of this idea, since pediatric FCD displayed
aberrant, depolarized GABA responses, which were further potentiated by incubation
with IL-1β.
Indeed, inflammatory mediators may be relevant for the pathogenesis of epilepsy
in FCD, as confirmed by the reported up-regulation of inflammatory cytokines and their
receptors and/or downstream effectors (such as IL-1
β
, IL-6, CCL3, CCL4, STAT3, C-JUN
and CCR5) in this disease [
19
,
43
]. Hence, concerning the evident implications on the
sustainment of a pro-inflammatory milieu and the detrimental consequences it possesses on
the progression of the disease, IL-1
β
would also potentiate a kind of GABAergic activity
that does not counteract seizures, but rather contributes to their generation and recurrence.
5. Conclusions
To understand the pathophysiological mechanisms underlying the dysfunction of
synaptic transmission in FCD IIb is a fundamental step towards the development of
new therapeutic strategies in this disease. Here, we highlighted a potential link between
two relevant phenomena, such as neuroinflammation and GABAergic neurotransmission,
and shed light on how the differential effect of IL-1
β
in pediatric versus adult tissues
may depend on a disturbed chloride homeostasis in pediatric FCD IIb samples. Since
endogenous anti-inflammatory response, as demonstrated by our transcriptomic analysis,
may not be enough to compensate for the excessive pro-inflammatory stimuli, we can
infer that an early pharmacological potentiation of anti-inflammatory mechanisms in FCD
IIb may favorably affect the progression of the disease and development of epilepsy in
these patients.
Supplementary Materials:
The following supporting information can be downloaded at: https://
www.mdpi.com/article/10.3390/brainsci12060807/s1, Table S1: Clinical information of the pediatric
study cohort used for transcriptomic analysis.
Author Contributions:
Conceptualization, E.P., E.A. and G.R.; methodology, E.P., E.A. and G.R.;
investigation, V.A., A.R., J.D.M., A.G., P.C. and G.R.; data curation, V.A., A.R., J.D.M., P.C., G.R., A.M.
(Alessandra Morano) and A.M. (Angelika Mühlebner); writing—original draft preparation, A.R., G.R.
and E.P.; writing—review and editing, E.P. and E.A.; supervision, E.P. and E.A.; funding acquisition,
E.P. and E.A. All authors have read and agreed to the published version of the manuscript.
Funding:
The work was supported by grants from Ateneo Project (Sapienza University)”, grant n
RM11916B84D24429 (EP). This research was supported by intramural “DISCAB” GRANT 2022 code
07_DG_2022_05 to P.C. This project has received funding from the European Union’s Horizon 2020
Research and Innovation Program under grant agreement No. 952455; EpiEpiNet (EP, EA).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Acknowledgments:
GR was supported by Italian Ministry of Health “Ricerca corrente”. The authors
wish to thank Anwesha Ghosh for English proofreading.
Conflicts of Interest: The authors declare no conflict of interest.
Brain Sci. 2022,12, 807 9 of 10
References
1.
Iffland, P.H.; Crino, P.B. Focal Cortical Dysplasia: Gene Mutations, Cell Signaling, and Therapeutic Implications. Annu. Rev.
Pathol. 2017,12, 547–571. [CrossRef] [PubMed]
2.
Najm, I.; Lal, D.; Alonso Vanegas, M.; Cendes, F.; Lopes-Cendes, I.; Palmini, A.; Paglioli, E.; Sarnat, H.B.; Walsh, C.A.;
Wiebe, S.; et al
. The ILAE Consensus Classification of Focal Cortical Dysplasia: An Update Proposed by an Ad Hoc Task
Force of the ILAE Diagnostic Methods Commission. Epilepsia 2022.Online ahead of print. [CrossRef]
3.
Blumcke, I.; Spreafico, R.; Haaker, G.; Coras, R.; Kobow, K.; Bien, C.G.; Pfäfflin, M.; Elger, C.; Widman, G.; Schramm, J.; et al.
Histopathological Findings in Brain Tissue Obtained during Epilepsy Surgery. N. Engl. J. Med. 2017,377, 1648–1656. [CrossRef]
[PubMed]
4.
Cepeda, C.; André, V.M.; Levine, M.S.; Salamon, N.; Miyata, H.; Vinters, H.V.; Mathern, G.W. Epileptogenesis in Pediatric Cortical
Dysplasia: The Dysmature Cerebral Developmental Hypothesis. Epilepsy Behav. 2006,9, 219–235. [CrossRef]
5.
Blauwblomme, T.; Dossi, E.; Pellegrino, C.; Goubert, E.; Iglesias, B.G.; Sainte-Rose, C.; Rouach, N.; Nabbout, R.; Huberfeld, G.
Gamma-Aminobutyric Acidergic Transmission Underlies Interictal Epileptogenicity in Pediatric Focal Cortical Dysplasia. Ann.
Neurol. 2019,85, 204–217. [CrossRef]
6.
Cepeda, C.; Chen, J.Y.; Wu, J.Y.; Fisher, R.S.; Vinters, H.V.; Mathern, G.W.; Levine, M.S. Pacemaker GABA Synaptic Activity May
Contribute to Network Synchronization in Pediatric Cortical Dysplasia. Neurobiol. Dis. 2014,62, 208–217. [CrossRef]
7.
Kaila, K.; Price, T.J.; Payne, J.A.; Puskarjov, M.; Voipio, J. Cation-Chloride Cotransporters in Neuronal Development, Plasticity
and Disease. Nat. Rev. Neurosci. 2014,15, 637–654. [CrossRef]
8.
Ben-Ari, Y.; Khalilov, I.; Kahle, K.T.; Cherubini, E. The GABA Excitatory/Inhibitory Shift in Brain Maturation and Neurological
Disorders. Neuroscientist 2012,18, 467–486. [CrossRef]
9.
Ben-Ari, Y. Excitatory Actions of Gaba during Development: The Nature of the Nurture. Nat. Rev. Neurosci.
2002
,3, 728–739.
[CrossRef]
10.
Cherubini, E.; Griguoli, M.; Safiulina, V.; Lagostena, L. The Depolarizing Action of GABA Controls Early Network Activity in the
Developing Hippocampus. Mol. Neurobiol. 2011,43, 97–106. [CrossRef]
11.
Talos, D.M.; Sun, H.; Kosaras, B.; Joseph, A.; Folkerth, R.D.; Poduri, A.; Madsen, J.R.; Black, P.M.; Jensen, F.E. Altered Inhibition in
Tuberous Sclerosis and Type IIb Cortical Dysplasia. Ann. Neurol. 2012,71, 539–551. [CrossRef] [PubMed]
12.
Ruffolo, G.; Cifelli, P.; Miranda-Lourenço, C.; De Felice, E.; Limatola, C.; Sebastião, A.M.; Diógenes, M.J.; Aronica, E.; Palma, E.
Rare Diseases of Neurodevelopment: Maintain the Mystery or Use a Dazzling Tool for Investigation? The Case of Rett Syndrome.
Neuroscience 2019,439, 146–152. [CrossRef] [PubMed]
13.
Tang, X.; Kim, J.; Zhou, L.; Wengert, E.; Zhang, L.; Wu, Z.; Carromeu, C.; Muotri, A.R.; Marchetto, M.C.N.; Gage, F.H.; et al. KCC2
Rescues Functional Deficits in Human Neurons Derived from Patients with Rett Syndrome. Proc. Natl. Acad. Sci. USA
2016
,
113, 751–756. [CrossRef] [PubMed]
14.
Ruffolo, G.; Cifelli, P.; Roseti, C.; Thom, M.; van Vliet, E.A.; Limatola, C.; Aronica, E.; Palma, E. A Novel GABAergic Dysfunction
in Human Dravet Syndrome. Epilepsia 2018,59, 2106–2117. [CrossRef]
15.
Cepeda, C.; André, V.M.; Wu, N.; Yamazaki, I.; Uzgil, B.; Vinters, H.V.; Levine, M.S.; Mathern, G.W. Immature Neurons and GABA
Networks May Contribute to Epileptogenesis in Pediatric Cortical Dysplasia. Epilepsia 2007,48 (Suppl. 5), 79–85. [CrossRef]
16.
Cepeda, C.; Levinson, S.; Nariai, H.; Yazon, V.-W.; Tran, C.; Barry, J.; Oikonomou, K.D.; Vinters, H.V.; Fallah, A.;
Mathern, G.W.; et al
. Pathological High Frequency Oscillations Associate with Increased GABA Synaptic Activity in Pe-
diatric Epilepsy Surgery Patients. Neurobiol. Dis. 2020,134, 104618. [CrossRef]
17.
Vezzani, A.; Viviani, B. Neuromodulatory Properties of Inflammatory Cytokines and Their Impact on Neuronal Excitability.
Neuropharmacology 2015,96 Pt A, 70–82. [CrossRef]
18.
Zimmer, T.S.; Broekaart, D.W.M.; Luinenburg, M.; Mijnsbergen, C.; Anink, J.J.; Sim, N.S.; Michailidou, I.; Jansen, F.E.; van Rijen,
P.C.; Lee, J.H.; et al. Balloon Cells Promote Immune System Activation in Focal Cortical Dysplasia Type 2b. Neuropathol. Appl.
Neurobiol. 2021,47, 826–839. [CrossRef]
19.
Srivastava, A.; Dixit, A.B.; Paul, D.; Tripathi, M.; Sarkar, C.; Chandra, P.S.; Banerjee, J. Comparative Analysis of Cy-
tokine/Chemokine Regulatory Networks in Patients with Hippocampal Sclerosis (HS) and Focal Cortical Dysplasia (FCD). Sci.
Rep. 2017,7, 15904. [CrossRef]
20.
Butler, T.; Li, Y.; Tsui, W.; Friedman, D.; Maoz, A.; Wang, X.; Harvey, P.; Tanzi, E.; Morim, S.; Kang, Y.; et al. Transient and Chronic
Seizure-Induced Inflammation in Human Focal Epilepsy. Epilepsia 2016,57, e191–e194. [CrossRef] [PubMed]
21.
Vezzani, A.; Balosso, S.; Ravizza, T. Neuroinflammatory Pathways as Treatment Targets and Biomarkers in Epilepsy. Nat. Rev.
Neurol. 2019,15, 459–472. [CrossRef] [PubMed]
22.
Roseti, C.; van Vliet, E.A.; Cifelli, P.; Ruffolo, G.; Baayen, J.C.; Di Castro, M.A.; Bertollini, C.; Limatola, C.; Aronica, E.;
Vezzani, A.; et al
. GABAA Currents Are Decreased by IL-1
β
in Epileptogenic Tissue of Patients with Temporal Lobe Epilepsy:
Implications for Ictogenesis. Neurobiol. Dis. 2015,82, 311–320. [CrossRef] [PubMed]
23.
Fuso, A.; Iyer, A.M.; van Scheppingen, J.; Maccarrone, M.; Scholl, T.; Hainfellner, J.A.; Feucht, M.; Jansen, F.E.; Spliet, W.G.;
Krsek, P.; et al
. Promoter-Specific Hypomethylation Correlates with IL-1
β
Overexpression in Tuberous Sclerosis Complex (TSC).
J. Mol. Neurosci. 2016,59, 464–470. [CrossRef]
Brain Sci. 2022,12, 807 10 of 10
24.
Rossini, L.; Villani, F.; Granata, T.; Tassi, L.; Tringali, G.; Cardinale, F.; Aronica, E.; Spreafico, R.; Garbelli, R. FCD Type II and
MTOR Pathway: Evidence for Different Mechanisms Involved in the Pathogenesis of Dysmorphic Neurons. Epilepsy Res.
2017
,
129, 146–156. [CrossRef]
25.
Mills, J.D.; van Vliet, E.A.; Chen, B.J.; Janitz, M.; Anink, J.J.; Baayen, J.C.; Idema, S.; Devore, S.; Friedman, D.; Diehl, B.; et al.
Coding and Non-Coding Transcriptome of Mesial Temporal Lobe Epilepsy: Critical Role of Small Non-Coding RNAs. Neurobiol.
Dis. 2020,134, 104612. [CrossRef]
26.
Palma, E.; Esposito, V.; Mileo, A.M.; Di Gennaro, G.; Quarato, P.; Giangaspero, F.; Scoppetta, C.; Onorati, P.; Trettel, F.;
Miledi, R.; et al
. Expression of Human Epileptic Temporal Lobe Neurotransmitter Receptors in Xenopus Oocytes: An Innovative
Approach to Study Epilepsy. Proc. Natl. Acad. Sci. USA 2002,99, 15078–15083. [CrossRef]
27.
Miledi, R.; Eusebi, F.; Martínez-Torres, A.; Palma, E.; Trettel, F. Expression of Functional Neurotransmitter Receptors in Xenopus
Oocytes after Injection of Human Brain Membranes. Proc. Natl. Acad. Sci. USA 2002,99, 13238–13242. [CrossRef]
28.
Bushnell, B.; Rood, J.; Singer, E. BBMerge—Accurate Paired Shotgun Read Merging via Overlap. PLoS ONE
2017
,12, e0185056.
[CrossRef]
29.
Harrow, J.; Frankish, A.; Gonzalez, J.M.; Tapanari, E.; Diekhans, M.; Kokocinski, F.; Aken, B.L.; Barrell, D.; Zadissa, A.;
Searle, S.; et al
. GENCODE: The Reference Human Genome Annotation for The ENCODE Project. Genome Res.
2012
,
22, 1760–1774. [CrossRef]
30.
Patro, R.; Duggal, G.; Love, M.I.; Irizarry, R.A.; Kingsford, C. Salmon Provides Fast and Bias-Aware Quantification of Transcript
Expression. Nat. Methods 2017,14, 417–419. [CrossRef] [PubMed]
31.
Soneson, C.; Love, M.I.; Robinson, M.D. Differential Analyses for RNA-Seq: Transcript-Level Estimates Improve Gene-Level
Inferences. F1000Research 2015,4, 1521. [CrossRef] [PubMed]
32.
Robinson, M.D.; McCarthy, D.J.; Smyth, G.K. EdgeR: A Bioconductor Package for Differential Expression Analysis of Digital
Gene Expression Data. Bioinformatics 2010,26, 139–140. [CrossRef] [PubMed]
33.
Ritchie, M.E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C.W.; Shi, W.; Smyth, G.K. Limma Powers Differential Expression Analyses for
RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015,43, e47. [CrossRef]
34.
Palma, E.; Amici, M.; Sobrero, F.; Spinelli, G.; Di Angelantonio, S.; Ragozzino, D.; Mascia, A.; Scoppetta, C.; Esposito, V.;
Miledi, R.; et al
. Anomalous Levels of Cl- Transporters in the Hippocampal Subiculum from Temporal Lobe Epilepsy Patients
Make GABA Excitatory. Proc. Natl. Acad. Sci. USA 2006,103, 8465–8468. [CrossRef]
35.
Palma, E.; Trettel, F.; Fucile, S.; Renzi, M.; Miledi, R.; Eusebi, F. Microtransplantation of Membranes from Cultured Cells to
Xenopus Oocytes: A Method to Study Neurotransmitter Receptors Embedded in Native Lipids. Proc. Natl. Acad. Sci. USA
2003
,
100, 2896–2900. [CrossRef]
36.
Blümcke, I.; Thom, M.; Aronica, E.; Armstrong, D.D.; Vinters, H.V.; Palmini, A.; Jacques, T.S.; Avanzini, G.; Barkovich, A.J.;
Battaglia, G.; et al. The Clinico-Pathological Spectrum of Focal Cortical Dysplasias: A Consensus Classification Proposed by an
Ad Hoc Task Force of the ILAE Diagnostic Methods Commission. Epilepsia 2011,52, 158–174. [CrossRef]
37.
Koh, S.; Wirrell, E.; Vezzani, A.; Nabbout, R.; Muscal, E.; Kaliakatsos, M.; Wickström, R.; Riviello, J.J.; Brunklaus, A.;
Payne, E.; et al.
Proposal to Optimize Evaluation and Treatment of Febrile Infection-Related Epilepsy Syndrome (FIRES): A Report from FIRES
Workshop. Epilepsia Open 2021,6, 62–72. [CrossRef]
38.
Pozzi, D.; Rasile, M.; Corradini, I.; Matteoli, M. Environmental Regulation of the Chloride Transporter KCC2: Switching
Inflammation off to Switch the GABA On? Transl. Psychiatry 2020,10, 349. [CrossRef]
39.
Semple, B.D.; O’Brien, T.J.; Gimlin, K.; Wright, D.K.; Kim, S.E.; Casillas-Espinosa, P.M.; Webster, K.M.; Petrou, S.; Noble-
Haeusslein, L.J. Interleukin-1 Receptor in Seizure Susceptibility after Traumatic Injury to the Pediatric Brain. J. Neurosci.
2017
,
37, 7864–7877. [CrossRef]
40.
Iori, V.; Frigerio, F.; Vezzani, A. Modulation of Neuronal Excitability by Immune Mediators in Epilepsy. Curr. Opin. Pharmacol.
2016,26, 118–123. [CrossRef] [PubMed]
41.
Shao, Y.; Ge, Q.; Yang, J.; Wang, M.; Zhou, Y.; Guo, J.-X.; Zhu, M.; Shi, J.; Hu, Y.; Shen, L.; et al. Pathological Networks Involving
Dysmorphic Neurons in Type II Focal Cortical Dysplasia. Neurosci. Bull. 2022.Online ahead of print. [CrossRef] [PubMed]
42.
Ruffolo, G.; Iyer, A.; Cifelli, P.; Roseti, C.; Mühlebner, A.; van Scheppingen, J.; Scholl, T.; Hainfellner, J.A.; Feucht, M.;
Krsek, P.; et al
.
Functional Aspects of Early Brain Development Are Preserved in Tuberous Sclerosis Complex (TSC) Epileptogenic Lesions.
Neurobiol. Dis. 2016,95, 93–101. [CrossRef] [PubMed]
43.
Zhang, Z.; Liu, Q.; Liu, M.; Wang, H.; Dong, Y.; Ji, T.; Liu, X.; Jiang, Y.; Cai, L.; Wu, Y. Upregulation of HMGB1-TLR4 Inflammatory
Pathway in Focal Cortical Dysplasia Type II. J. Neuroinflammation 2018,15, 27. [CrossRef] [PubMed]
... The preparation of human membranes, the cytoplasmic injection in Xenopus laevis oocytes and the electrophysiological recordings of GABA currents were executed as previously described [1,44]. These membranes are extracted from control and pathological tissues and injected in the cytoplasm of the oocytes. ...
Article
Full-text available
GABAergic interneurons play a critical role in maintaining neural circuit balance, excitation–inhibition regulation, and cognitive function modulation. In tuberous sclerosis complex (TSC), GABAergic neuron dysfunction contributes to disrupted network activity and associated neurological symptoms, assumingly in a cell type-specific manner. This GABAergic centric study focuses on identifying specific interneuron subpopulations within TSC, emphasizing the unique characteristics of medial ganglionic eminence (MGE)- and caudal ganglionic eminence (CGE)-derived interneurons. Using single-nuclei RNA sequencing in TSC patient material, we identify somatostatin-expressing (SST+) interneurons as a unique and immature subpopulation in TSC. The disrupted maturation of SST+ interneurons may undergo an incomplete switch from excitatory to inhibitory GABAergic signaling during development, resulting in reduced inhibitory properties. Notably, this study reveals markers of immaturity specifically in SST+ interneurons, including an abnormal NKCC1/KCC2 ratio, indicating an imbalance in chloride homeostasis crucial for the postsynaptic consequences of GABAergic signaling as well as the downregulation of GABAA receptor subunits, GABRA1, and upregulation of GABRA2. Further exploration of SST+ interneurons revealed altered localization patterns of SST+ interneurons in TSC brain tissue, concentrated in deeper cortical layers, possibly linked to cortical dyslamination. In the epilepsy context, our research underscores the diverse cell type-specific roles of GABAergic interneurons in shaping seizures, advocating for precise therapeutic considerations. Moreover, this study illuminates the potential contribution of SST+ interneurons to TSC pathophysiology, offering insights for targeted therapeutic interventions.
... In addition, recent evidence supports the hypothesis that inflammatory stimuli, in particular IL-1β can alter GABAergic neurotransmission by modulating the functional expression of the aforementioned CCCs (Pozzi et al. 2020). Alfano et al. 2022 indicated that IL-1Ra is also upregulated, suggesting an attempt to compensate for the IL-1β increase in a cohort of FCDIIb samples. According to this information, endogenous anti-inflammatory cytokines fail to reduce neuroinflammation and can lead to brain hyperexcitability (Iori et al. 2016). ...
Chapter
Despite the fact that there are several drugs available for the treatment of epilepsy, pharmacoresistance remains a major challenge in seizure control. Therefore, a significant part of epilepsy research has focused on revealing the mechanisms underlying drug resistance in order to develop new rationally designed pharmaceutical therapies for refractory epilepsies. Based on experimental and clinical studies, epilepsy-induced structural and functional alterations in brain targets have been postulated to lead to decreased sensitivity to antiepileptic drugs, more recently antiseizure medications (ASM). Also, evidence shows that GABA neurotransmission system plays a leading role in the pathophysiology of epilepsy. Canonically, GABA (gamma-aminobutyric acid) is considered the main inhibitory neurotransmitter in the central nervous system, but due to the variability in the location and composition of its receptors by different types of subunits, as well as neural physiological immaturity, GABA may have excitatory effects. Abnormalities in the GABAergic system identified in animal models of epilepsy and in samples of brain tissue samples resected surgically from patients with drug-resistant epilepsy, mainly at level of GABAA receptors (GABAARs), whose changes lead to an altered response to some ASMs, among other neuroleptic drugs.Here we review the current evidence on changes in the GABAergic system related to seizure generation, epilepsy, and pharmacoresistance, with particular emphasis on GABAARs and genetic polymorphisms of its subunits associated with refractory human epilepsy.KeywordsPharmacoresistant epilepsyGABA neurotransmissionGABAReceptorsHuman dataAnimal modelsGABA geneticsAntiseizure medication
... The research article by Alfano et al. focuses on the function of GABA A receptors in type II focal cortical dysplasia (FCD), one of the most frequent drug-resistant forms of epilepsy in pediatric patients [2]. Using Xenopus oocytes transplanted with human tissues from patients affected by FCD, the authors found that while the prototypical proinflammatory cytokine IL-1β reduced the amplitude of GABA responses via its signaling pathway in tissues obtained from adults, it potentiated those obtained from children. ...
Article
Full-text available
This Special Issue, “GABA Signaling: Therapeutic Targets for Neurodegenerative and Neurodevelopmental Disorders”, focuses on a fundamental property of the neurotransmitter γ-aminobutyric acid (GABA), namely its capacity to shift, in particular conditions, from the hyperpolarizing to the depolarizing direction [...]
... We found increased glutamatergic and GABAergic synaptic density in the peri-soma and primary dendrites of NPNFP-expressing interstitial neurons in the white matter of drug-resistant epilepsy patients when compared with control brain tissue. The imbalance of synaptic excitation and the inhibition of dysmorphic neurons in the grey matter could contribute to seizure genesis in FCD [21,42,43]. Therefore, the increased glutamatergic and GABAergic synaptic density of interstitial neurons in the white matter could be integrated into the epileptic neuronal networks and contribute to seizure genesis in drug-resistant epilepsy. ...
Article
Full-text available
The interstitial neurons in the white matter of the human and non-human primate cortex share a similar developmental origin with subplate neurons and deep-layer cortical neurons. A subset of interstitial neurons expresses the molecular markers of subplate neurons, but whether interstitial neurons express cortical layer markers in the adult human brain remains unexplored. Here we report the expression of cortical layer markers in interstitial neurons in the white matter of the adult human brain, supporting the hypothesis that interstitial neurons could be derived from cortical progenitor cells. Furthermore, we found increased non-phosphorylated neurofilament protein (NPNFP) expression in interstitial neurons in the white matter of drug-resistant epilepsy patients. We also identified the expression of glutamatergic and g-aminobutyric acid (GABAergic) synaptic puncta that were distributed in the perikarya and dendrites of interstitial neurons. The density of glutamatergic and GABAergic synaptic puncta was increased in interstitial neurons in the white matter of drug-resistant epilepsy patients compared with control brain tissues with no history of epilepsy. Together, our results provide important insights of the molecular identity of interstitial neurons in the adult human white matter. Increased synaptic density of interstitial neurons could result in an imbalanced synaptic network in the white matter and participate as part of the epileptic network in drug-resistant epilepsy.
Article
Epidemiological research has shown that a variety of circulating bioactive factors are associated with epilepsy, including macrophage colony-stimulating factor, interleukin-1β, and tumor necrosis factor-α. To further investigate the associations between epilepsy and 41 inflammatory cytokines, this Mendelian randomization was performed. This study presents genome-wide association study summary data on 41 inflammatory cytokines and epilepsy. Epilepsy incorporates generalized and focal epilepsy. A two-sample Mendelian randomization method was used. In order to analyze causal relationships between exposures and outcomes, the inverse variance-weighted method was mainly used. The findings suggested that increased levels of interleukin-1 receptor antagonists and interleukin-5 may be significantly associated with increased risks of focal epilepsy (beta: 0.080, P = .043; beta: 0.083, P = .015). In addition, regulated upon activation normal T cell expressed and secreted factor and Macrophage colony-stimulating factor may be significantly associated with generalized epilepsy (beta: 0.110, P = .042; beta: –0.114, P = .024). Furthermore, inflammatory cytokines such as interleukin-10, interleukin-1β, interleukin-1Ra, interleukin-7, tumor necrosis factor-α, and interferon-γ may be identified as the result of focal epilepsy (beta: 0.152, P = .031; beta: 0.214, P = .037; beta: 0.214, P = .047; beta: 0.222, P = .031; beta: 0.224, P = .025; beta: 0.161, P = .018). This study suggests that interleukin-5 and interleukin-1 receptor antagonists are potentially correlated factors with focal epilepsy etiology, macrophage colony-stimulating factor and regulated upon activation normal T cell expressed and secreted factor are potentially correlated factors with generalized epilepsy etiology, while several inflammatory cytokines possibly contribute to focal epilepsy development downstream.
Article
Tuberous sclerosis complex (TSC) is a genetic disorder caused by inactivating mutations in the TSC1 and TSC2 genes, causing overactivation of the mechanistic (previously referred to as mammalian) target of rapamycin (mTOR) signaling pathway in fetal life. The mTOR pathway plays a crucial role in several brain processes leading to TSC-related epilepsy, intellectual disability, and autism spectrum disorder (ASD). Pre-natal or early post-natal diagnosis of TSC is now possible in a growing number of pre-symptomatic infants. We searched PubMed for peer-reviewed publications published between January 2010 and April 2023 with the terms “tuberous sclerosis”, “autism”, or “autism spectrum disorder”,” animal models”, “preclinical studies”, “neurobiology”, and “treatment”. Prospective studies have highlighted that developmental trajectories in TSC infants who were later diagnosed with ASD already show motor, visual and social communication skills in the first year of life delays. Reliable genetic, cellular, electroencephalography and magnetic resonance imaging biomarkers can identify pre-symptomatic TSC infants at high risk for having autism and epilepsy. Preventing epilepsy or improving therapy for seizures associated with prompt and tailored treatment strategies for autism in a sensitive developmental time window could have the potential to mitigate autistic symptoms in infants with TSC.
Article
Focal cortical dysplasia (FCD) represents a group of malformations of cortical development, which are speculated to be related to early developmental defects in the cerebral cortex. According to dysmature cerebral development hypothesis of FCD altered GABAA receptor function is known to contribute to abnormal neuronal network. Here, we studied the possible association between age at seizure onset in FCD with the subunit configuration of GABAA receptors in resected brain specimens obtained from patients with FCD. We observed a significantly higher ratio of α4/α1 subunit-containing GABAA receptors in patients with early onset (EO) FCD as compared to those with late onset (LO) FCD as is seen during the course of development where α4-containing GABAA receptors expression is high as compared to α1-containing GABAA receptors expression. Likewise, the influx to efflux chloride co-transporter expression of NKCC1/KCC2 was also increased in patients with EO FCD as seen during brain development. In addition, we observed that the ratio of GABA/Glutamate neurotransmitters was lower in patients with EO FCD as compared to that in patients with LO FCD. Our findings suggest altered configuration of GABAA receptors in FCD which could be contributing to aberrant depolarizing GABAergic activity. In particular, we observed a correlation of age at seizure onset in FCD with subunit configuration of GABAA receptors, levels of NKCC1/KCC2 and the ratio of GABA/Glutamate neurotransmitters such that the patients with EO FCD exhibited a more critically modulated GABAergic network.
Article
Full-text available
Ongoing challenges in diagnosing focal cortical dysplasia (FCD) mandate continuous research and consensus agreement to improve disease definition and classification. An International League Against Epilepsy (ILAE) Task Force (TF) reviewed the FCD classification of 2011 to identify existing gaps and provide a timely update. The following methodology was applied to achieve this goal: a survey of published literature indexed with ((Focal Cortical Dysplasia) AND (epilepsy)) between 01/01/2012 and 06/30/2021 (n = 1349) in PubMed identified the knowledge gained since 2012 and new developments in the field. An online survey consulted the ILAE community about the current use of the FCD classification scheme with 367 people answering. The TF performed an iterative clinico‐pathological and genetic agreement study to objectively measure the diagnostic gap in blood/brain samples from 22 patients suspicious for FCD and submitted to epilepsy surgery. The literature confirmed new molecular‐genetic characterizations involving the mechanistic Target Of Rapamycin (mTOR) pathway in FCD type II (FCDII), and SLC35A2 in mild malformations of cortical development (mMCDs) with oligodendroglial hyperplasia (MOGHE). The electro‐clinical‐imaging phenotypes and surgical outcomes were better defined and validated for FCDII. Little new information was acquired on clinical, histopathological, or genetic characteristics of FCD type I (FCDI) and FCD type III (FCDIII). The survey identified mMCDs, FCDI, and genetic characterization as fields for improvement in an updated classification. Our iterative clinico‐pathological and genetic agreement study confirmed the importance of immunohistochemical staining, neuroimaging, and genetic tests to improve the diagnostic yield. The TF proposes to include mMCDs, MOGHE, and “no definite FCD on histopathology” as new categories in the updated FCD classification. The histopathological classification can be further augmented by advanced neuroimaging and genetic studies to comprehensively diagnose FCD subtypes; these different levels should then be integrated into a multi‐layered diagnostic scheme. This update may help to foster multidisciplinary efforts toward a better understanding of FCD and the development of novel targeted treatment options.
Article
Full-text available
Focal cortical dysplasia (FCD) is one of the most common causes of drug-resistant epilepsy. Dysmorphic neurons are the major histopathological feature of type II FCD, but their role in seizure genesis in FCD is unclear. Here we performed whole-cell patch-clamp recording and morphological reconstruction of cortical principal neurons in postsurgical brain tissue from drug-resistant epilepsy patients. Quantitative analyses revealed distinct morphological and electrophysiological characteristics of the upper layer dysmorphic neurons in type II FCD, including an enlarged soma, aberrant dendritic arbors, increased current injection for rheobase action potential firing, and reduced action potential firing frequency. Intriguingly, the upper layer dysmorphic neurons received decreased glutamatergic and increased GABAergic synaptic inputs that were coupled with upregulation of the Na+-K+-Cl- cotransporter. In addition, we found a depolarizing shift of the GABA reversal potential in the CamKII-cre::PTENflox/flox mouse model of drug-resistant epilepsy, suggesting that enhanced GABAergic inputs might depolarize dysmorphic neurons. Thus, imbalance of synaptic excitation and inhibition of dysmorphic neurons may contribute to seizure genesis in type II FCD.
Article
Full-text available
Aims Focal cortical dysplasia (FCD) type 2 is an epileptogenic malformation of the neocortex associated with somatic mutations in the mammalian target of rapamycin (mTOR) pathway. Histopathologically, FCD 2 is subdivided into FCD 2a and FCD 2b, the only discriminator being the presence of balloon cells in FCD 2b. While pro-epileptogenic immune system activation and inflammatory responses are commonly detected in both subtypes, it is unknown what contextual role balloon cells play. Methods The present study employed RNA sequencing of surgically resected brain tissue from FCD 2a (n=11) and FCD 2b (n=20) patients compared to autopsy control (n=9) focusing on three immune system processes: adaptive immunity, innate immunity and cytokine-production. This analysis was followed by immunohistochemistry on a clinically well-characterized FCD 2 cohort. Results Differential expression analysis revealed stronger expression of components of innate immunity, adaptive immunity and cytokine-production in FCD 2b than in FCD 2a, particularly complement activation and antigen presentation. Immunohistochemical analysis confirmed these findings, with strong expression of leukocyte antigen I and II in FCD 2b as compared to FCD 2a. Moreover, T-lymphocyte tissue infiltration was elevated in FCD 2b. Expression of markers of immune system activation in FCD 2b was concentrated in subcortical white matter. Lastly, antigen presentation was strongly correlated with balloon cell load in FCD 2b lesions. Conclusion We conclude that, next to mutation-driven mTOR activation and seizure activity, balloon cells are crucial drivers of inflammation in FCD 2b. Our findings indicate that therapies targeting inflammation may be beneficial in FCD 2b.
Article
Full-text available
Febrile infection‐related epilepsy syndrome (FIRES) is a rare catastrophic epileptic encephalopathy that presents suddenly in otherwise normal children and young adults causing significant neurological disability, chronic epilepsy, and high rates of mortality. To suggest a therapy protocol to improve outcome of FIRES, workshops were held in conjunction with American Epilepsy Society annual meeting between 2017 and 2019. An international group of pediatric epileptologists, pediatric neurointensivists, rheumatologists and basic scientists with interest and expertise in FIRES convened to propose an algorithm for a standardized approach to the diagnosis and treatment of FIRES. The broad differential for refractory status epilepticus (RSE) should include FIRES, to allow empiric therapies to be started early in the clinical course. FIRES should be considered in all previously healthy patients older than two years of age who present with explosive onset of seizures rapidly progressing to RSE, following a febrile illness in the preceding two weeks. Once FIRES is suspected, early administrations of ketogenic diet and anakinra (the IL‐1 receptor antagonist that blocks biologic activity of IL‐1β) are recommended.
Article
Full-text available
Chloride homeostasis, the main determinant factor for the dynamic tuning of GABAergic inhibition during development, has emerged as a key element altered in a wide variety of brain disorders. Accordingly, developmental disorders such as schizophrenia, Autism Spectrum Disorder, Down syndrome, epilepsy, and tuberous sclerosis complex (TSC) have been associated with alterations in the expression of genes codifying for either of the two cotransporters involved in the excitatory-to-inhibitory GABA switch, KCC2 and NKCC1. These alterations can result from environmental insults, including prenatal stress and maternal separation which share, as common molecular denominator, the elevation of pro-inflammatory cytokines. In this review we report and systemize recent research articles indicating that different perinatal environmental perturbations affect the expression of chloride transporters, delaying the developmental switch of GABA signaling, and that inflammatory cytokines, in particular interleukin 1β, may represent a key causal factor for this phenomenon. Based on literature data, we provide therefore a unifying conceptual framework, linking environmental hits with the excitatory-to-inhibitory GABA switch in the context of brain developmental disorders.
Article
Full-text available
Our understanding of mesial temporal lobe epilepsy (MTLE), one of the most common form of drug-resistant epilepsy in humans, is derived mainly from clinical, imaging, and physiological data from humans and animal models. High-throughput gene expression studies of human MTLE have the potential to uncover molecular changes underlying disease pathogenesis along with novel therapeutic targets. Using RNA- and small RNA-sequencing in parrallel, we explored differentially expressed genes in the hippocampus and cortex of MTLE patients who had undergone surgical resection and non-epileptic controls. We identified differentially expressed genes in the hippocampus of MTLE patients and differentially expressed small RNAs across both the cortex and hippocampus. We found significant enrichment for astrocytic and microglial genes among up-regulated genes, and down regulation of neuron specific genes in the hippocampus of MTLE patients. The transcriptome profile of the small RNAs reflected disease state more robustly than mRNAs, even across brain regions which show very little pathology. While mRNAs segregated predominately by brain region for MTLE and controls, small RNAs segregated by disease state. In particular, our data suggest that specific miRNAs (e.g., let-7b-3p and let-7c-3p) may be key regulators of multiple pathways related to MTLE pathology. Further, we report a strong association of other small RNA species with MTLE pathology. As such we have uncovered novel elements that may contribute to the establishment and progression of MTLE pathogenesis and that could be leveraged as therapeutic targets.
Article
Pathological high-frequency oscillations (HFOs), specifically fast ripples (FRs, >250 Hz), are pathognomonic of an active epileptogenic zone. However, the origin of FRs remains unknown. Here we explored the correlation between FRs recorded with intraoperative pre-resection electrocorticography (ECoG) and spontaneous synaptic activity recorded ex vivo from cortical tissue samples resected for the treatment of pharmacoresistant epilepsy. The cohort included 47 children (ages 0.22-9.99 yr) with focal cortical dysplasias (CD types I and II), tuberous sclerosis complex (TSC) and non-CD pathologies. Whole-cell patch clamp recordings were obtained from pyramidal neurons and interneurons in cortical regions that were positive or negative for pathological HFOs, defined as FR band oscillations (250-500 Hz) at ECoG. The frequency of spontaneous excitatory and inhibitory postsynaptic currents (sEPSCs and IPSCs, respectively) was compared between HFO+ and HFO- regions. Regardless of pathological substrate, regions positive for FRs displayed significantly increased frequencies of sIPSCs compared with regions negative for FRs. In contrast, the frequency of sEPSCs was similar in both regions. In about one third of cases (n = 17), pacemaker GABA synaptic activity (PGA) was observed. In the vast majority (n = 15), PGA occurred in HFO+ areas. Further, fast-spiking interneurons displayed signs of hyperexcitability exclusively in HFO+ areas. These results indicate that, in pediatric epilepsy patients, increased GABA synaptic activity is associated with interictal FRs in the epileptogenic zone and suggest an active role of GABAergic interneurons in the generation of pathological HFOs. Increased GABA synaptic activity could serve to dampen excessive excitability of cortical pyramidal neurons in the epileptogenic zone, but it could also promote neuronal network synchrony.
Article
Epilepsy is a chronic neurological disease characterized by an enduring propensity for generation of seizures. The pathogenic processes of seizure generation and recurrence are the subject of intensive preclinical and clinical investigations as their identification would enable development of novel treatments that prevent epileptic seizures and reduce seizure burden. Such treatments are particularly needed for pharmacoresistant epilepsies, which affect ~30% of patients. Neuroinflammation is commonly activated in epileptogenic brain regions in humans and is clearly involved in animal models of epilepsy. An increased understanding of neuroinflammatory mechanisms in epilepsy has identified cellular and molecular targets for new mechanistic therapies or existing anti-inflammatory drugs that could overcome the limitations of current medications, which provide only symptomatic control of seizures. Moreover, inflammatory mediators in the blood and molecular imaging of neuroinflammation could provide diagnostic, prognostic and predictive biomarkers for epilepsy, which will be instrumental for patient stratification in future clinical studies. In this Review, we focus on our understanding of the IL-1 receptor-Toll-like receptor 4 axis, the arachidonic acid-prostaglandin cascade, oxidative stress and transforming growth factor-β signalling associated with blood-brain barrier dysfunction, all of which are pathways that are activated in pharmacoresistant epilepsy in humans and that can be modulated in animal models to produce therapeutic effects on seizures, neuronal cell loss and neurological comorbidities.
Article
The investigation on neurotransmission function during normal and pathologic development is a pivotal component needed to understand the basic mechanisms underlying neurodevelopmental pathologies. To study these diseases, many animal models have been generated which allowed to face the limited availability of human tissues and, as a consequence, most of the electrophysiology has been performed on these models of diseases. On the other hand, the technique of membrane microtransplantation in Xenopus oocytes allows the study of human functional neurotransmitter receptors thanks to the use of tissues from autopsies or surgeries, even in quantities that would not permit other kinds of functional studies. In this short article, we intend to underline how this technique is well-fit for the study of rare diseases by characterizing the electrophysiological properties of GABAA and AMPA receptors in Rett syndrome. For our purposes, we used both tissues from Rett syndrome patients and Mecp2-null mice, a well validated murine model of the same disease, in order to strengthen the solidity of our results through the comparison of the two. Our findings retrace previous results and, in the light of this, further argue in favor of Prof. Miledi's technique of membrane microtransplantation that proves itself a very useful tool of investigation in the field of neurophysiology. This article is part of a Special Issue entitled: SI: Miledi’s contributions.
Article
Objective Dysregulation of GABAergic transmission has been reported in lesional acquired epilepsies (gliomas, hippocampal sclerosis). We investigated its involvement in a developmental disorder, human Focal Cortical Dysplasia, focusing on chloride regulation driving GABAergic signals. Methods In vitro recordings of 47 human cortical acute slices from 11 pediatric patients operated from a Focal Cortical Dysplasia were performed on Multi Electrode Arrays. GABAergic receptors and chloride regulators were pharmacologically modulated. Immunostaining for chloride co‐transporter KCC2 and interneurons were performed on recorded slices to correlate electrophysiology and expression patterns. Results Focal Cortical Dysplasia slices retain intrinsic epileptogenicity. 36/47 slices displayed spontaneous interictal discharges, along with a pattern specific to the histological subtypes. Ictal discharges were induced in pro‐epileptic conditions in 6/8 slices in the areas generating spontaneous interictal discharges, with a transition to seizure involving the emergence of preictal discharges. Interictal discharges were sustained by GABAergic signaling as a GABAA receptors blocker stopped them in 2/3 slices. Blockade of NKCC1 Cl‐ co‐transporters further controlled interictal discharges in 9/12 cases, revealing a Cl‐ dysregulation affecting actions of GABA. Immunohistochemistry highlighted decreased expression and changes in KCC2 sub‐cellular localization and a decrease in the number of GAD67‐positive interneurons in regions generating interictal discharges. Interpretation Altered chloride cotransporters expression and changes in interneuron density in FCD may lead to paradoxical depolarization of pyramidal cells. Spontaneous interictal discharges are consequently mediated by GABAergic signals and targeting chloride regulation in neurons may be considered for the development of new antiepileptic drugs. This article is protected by copyright. All rights reserved.