You are on page 1of 12

Focal Cortical Dysplasia in Childhood

Epilepsy
Tarek Shaker, MSc,1 Anne Bernier, MD,1 and Lionel Carmant, MD, FRCP (C)*

Focal cortical dysplasia is a common cause of medication resistant epilepsy. A better


understanding of its presentation, pathophysiology and consequences have helped us
improved its treatment and outcome. This paper reviews the most recent classification,
pathophysiology and imaging findings in clinical research as well as the knowledge gained
from studying genetic and lesional animal models of focal cortical dysplasia. This review of this
recently gained knowledge will most likely help develop new research models and new
therapeutic targets for patients with epilepsy associated with focal cortical dysplasia.
Semin Pediatr Neurol 23:108-119 C 2016 Elsevier Inc. All rights reserved.

Introduction classification systems for FCD have been proposed. The


currently accepted one was established in 2011 by a task force

M alformations of cortical development comprise a spec-


trum of brain abnormalities caused by defects in brain
development, failing to elaborate a functional laminated cortex.
of the International League Against Epilepsy, and divides FCD
into 3 groups based mainly on histological findings4 (Table).

Focal cortical dysplasias (FCD) are a subgroup of malforma-


tions of cortical development characterized by aberrant cortical Pathological Findings of FCD
architecture in a localized area of the brain, and have been
reported as the most frequent structural brain lesion encoun- FCD Type I
tered in children submitted to surgery for medication resistant FCD type I consist of isolated areas of dyslamination without
epilepsy.1,2 Increasing efforts have been made in the last any cytologic abnormality. Radial dyslamination (FCD type Ia)
decade to better define the different types of FCD and is characterized by organization of the cortex in microcolumns,
understand their pathophysiology, hoping this could help defined by the presence of more than 8 neurons aligned in a
identify new therapeutic targets. In this article, we review the vertical orientation. Tangential dyslamination (FCD type 1b),
classification, pathology, and recent advances in the patho- on the other hand, constitutes a failure to establish the 6-
physiology of FCD derived from human and animal studies. layered horizontal composition of the neocortex. The combi-
nation of abnormal cortical architecture in both radial and
tangential direction characterizes FCD type Ic.

FCD Types and Classification


FCD Type II
The term FCD was first introduced by Taylor et al in 1971, FCD type II consist of areas of cortical dyslamination with
to describe microscopic abnormalities identified in a series specific cytologic abnormalities: dysmorphic neurons are
of 10 patients who underwent epilepsy surgery.3 Different present in both FCD types IIa and IIb, whereas balloon cells
are observed exclusively in type IIb. Dysmorphic neurons can
From the *Division of Neurology, Department of Pediatrics and Neuros- be recognized by their enlarged cell body and nucleus,
ciences, CHU Sainte-Justine, University of Montreal, Montreal, Quebec, abnormally distributed Nissl substance and cytoplasmic accu-
Canada.
1
mulation of neurofilament proteins. Balloon cells have an
These authors contributed equally to the work. enlarged cell body with opalescent glassy eosinophilic cyto-
Address reprint requests to Lionel Carmant, MD, Division of Neurology,
Department of Pediatrics and Neurosciences, CHU Sainte-Justine, Uni-
plasm, no Nissl substance, and can be indistinguishable from
versity of Montreal, Montreal, Quebec, Canada, H3T 1C5. E-mail: lionel. giant cells found in cortical tubers of tuberous sclerosis
carmant@umontreal.ca complex (TSC).

108 http://dx.doi.org/10.1016/j.spen.2016.06.007
1071-9091/16/& 2016 Elsevier Inc. All rights reserved.
FCD in children 109

Table ILAE Classification of Focal Cortical Dysplasias blurring of the gray or white matter and volume loss of
Focal cortical dysplasia type I (isolated dyslamination) the subcortical white matter leading to lobar atrophy or
FCD type Ia: abnormal radial (vertical) cortical lamination. hypoplasia.5 Characteristics suggestive of FCD type II tend to
FCD type Ib: abnormal tangential (horizontal) cortical be more easily identified, with cortical thickness abnormalities,
lamination. hyperintensities of the cortex and subcortical white matter in
FCD type Ic: abnormal radial and tangential cortical T2WI and FLAIR, blurring of the gray or white matter and sulcal
lamination. or gyral abnormalities.5,9-12 The transmantle sign, a T2WI white
matter signal hyperintensity tapering toward the ventricle,
Focal cortical dysplasia type II (dyslamination with dysmorphic
reported to be specific of FCD type II, is found in approximately
neurons)
FCD type IIa: with dysmorphic neurons.
30% of the patients.5,9,13 Although FCD type I and II can be
FCD type IIb: with dysmorphic neurons and balloon cells. present in any lobe, FCD type I has been reported more
frequently in the temporal lobe, whereas FCD type II seems to
Focal cortical dysplasia type III (associated with principal have a predilection for the frontal lobe14,15 (Fig. 1). Finally, no
lesion) specific sign is reported for FCD type III, but the diagnosis is
FCD type IIIa: cortical lamination abnormalities in the usually made in the periphery of the principal lesion.
temporal lobe associated with hippocampal sclerosis. Technological advances have improved the yield of presur-
FCD type IIIb: cortical lamination abnormalities adjacent to a gical imaging. The latest high-field MRIs (7.0 T) has shown
glial or neuroglial tumor. abnormalities in 20% of patients with focal epilepsy and
FCD type IIIc: cortical lamination abnormalities adjacent to a previously normal MRIs.16 Moreover, detection of cortical
vascular malformation.
thickening, gray or white matter blurring and cortical signal
FCD type IIId: cortical lamination abnormalities adjacent to
any lesion acquired during early life (eg, trauma, ischemic
abnormalities is improved by voxel-based analysis of T1WI
injury, and encephalitis). and FLAIR scans.17,18
ILAE, International League Against Epilepsy.
Diffusion Tensor Imaging
Diffusion tensor imaging (DTI), by enabling the measurement
FCD Type III of diffusion of free water molecules, permits the assessment of
FCD type III refer to alterations in cortical lamination or the cerebral microstructure and fiber tracking. Therefore, it can
cytoarchitectural composition associated with hippocampal be used to better evaluate the extent of the lesion, which is
atrophy (type IIIa), glial or glioneuronal tumors (type IIIb), often larger on DTI than on MRI.19
vascular malformations (type IIIc), or other epileptogenic
lesions like encephalitis, ischemic injury, and trauma (type
IIId). To be considered FCD type III rather than “dual Functional Imaging
pathology,” the area of dyslamination must be adjacent to 18-Fluorodeoxyglucose positron emission tomography is
the lesion or affecting the same lobe. highly sensitive (60%-92%) in the detection of FCD.5,20
Single-photon emission computed tomography can also be used
to locate the epileptogenic dysplastic zone. However, the area
Neuroimaging and localized by fluorodeoxyglucose positron emission tomography
and single-photon emission computed tomography can repre-
Electrophysiological Findings in sent, due to technical issues, not only the primary epileptogenic
FCD zone but also the area of propagation of epileptic activity.20,21
The major predictor for a favorable surgical outcome in Correlation with other modalities is therefore required.
children with FCD is the removal of the entire dysplastic
cortex.5-7 Therefore, a complete presurgical evaluation to Electroencephalography and Other
define the extent of FCD with neuroimaging and electro-
physiology is crucial.
Electrophysiological Evaluations
While the electroencephalogram (EEG) of FCD type I is often
normal or nonspecific, the patients with FCD type II have been
Magnetic Resonance Imaging reported to have rhythmic interictal epileptiform discharges
The diagnosis of FCD by magnetic resonance imaging (MRI) correlating spatially with the anatomical extent of their
can be challenging, and requires specific protocols looking at lesion22,23 (Fig. 2).
cortical thickness, gray or white matter signal and junction, and Electrocorticography shows patterns of repetitive spikes,
sulcal or gyral patterns.8 Even with experienced neuroradiolo- spike-and-waves, polyspikes, or bursts of fasts rhythms
gists, 37% of the patients with FCD type I and 15% of those interspaced with quiescient periods.14 These abnormalities
with FCD type II are considered to have a normal MRI.6 tend to increase during drowsiness and during nonrapid eye
Abnormalities are often subtle in FCD type I, with moderate movement sleep.12,22,23 The ictal pattern usually shows
increases in the white matter signal on T2 weighted image intensification of the interictal activity, with the appearance
(T2WI) and fluid attenuation inversion recovery (FLAIR), mild of low-voltage fast-activity.12,14,23 Magnetoencephalography,
110 T. Shaker et al.

L R

L R

FLAIR (3 mm slices)

Figure 1 (A) MRI of a patient with medication resistant frontal lobe epilepsy that was read as normal despite a suspected
bottom of the sulcus focal cortical dysplasia (crossing of the lines). Texture analysis showed not only blurring of the affected
sulcus (upper arrow) but also the characteristic transmantle sign (lower arrow). (B) Cortical thickness analysis further
confirmed the suspected lesion (arrow). After removal of the lesion, the patient has remained seizure free for the past 8 years
and is now off medications and attending university. (Color version of figure is available online.)
FCD in children 111

Figure 2 (A) EEG of a patient with focal seizures postinfantile spasms demonstrating continuous focal spikes in right
occipital region with focal slowing (arrows). Review of the MRI demonstrated asymmetrical myelination and blurring of the
gray-white matter in the corresponding area (arrow). This patient also underwent successful epilepsy surgery and has
remained seizure free over the past 16 months but remains on monotherapy antiepileptic drug. (Color version of figure is
available online.)
112 T. Shaker et al.

by allowing measurements of the brain’s magnetic fields, can with increased levels of the cytokine IL-1ß, an important
be used for mapping the epileptogenic zone in MRI negative regulator of neuroinflammation.43 Studies suggest that the
epilepsy and has been shown to improve mapping of the inflammatory response is more prominent in FCD type II
epileptogenic zone.24 than type I,44 and seems to be in direct correlation with
seizure frequency.42
Other causes of FCD are included in the subtypes of FCD
Hypotheses of FCD Formation type III.4 However, whether the FCD is acquired or not in
those situations is a matter of debate, particularly for hippo-
The exact mechanism that leads to the development of FCD campal sclerosis (HS) and Rasmussen encephalitis. It has in fact
remains to be elucidated, but a few hypotheses have been been suggested that FCD could be causal in those conditions,
advanced. Since FCD is most likely a heterogeneous condition, creating a predisposing epileptogenic environment that trig-
both genetic and acquired causes could be involved. gered by certain events (“double-hit hypothesis”), could cause
a cascade of events leading to HS or Rasmussen encephali-
Genetic Causes tis.45,46 However, results of our animal model of the double-hit
Disturbances in the proteins involved in the Wnt/Notch hypothesis for HS cannot be extrapolated for FCD type IIId,
pathway, implicated in cortical development, have been found since our model is one of dual pathology.45
in FCD type IIb.25 However, it is unlikely that mutations in
genes of this pathway could cause FCD, since they are usually
lethal in the embryo.26 Owing to pathological similarities FCD and Epileptogenesis
between the tubers of TSC and FCD type IIb, many experts
The understanding of how a FCD induces epileptogenesis
have advanced that a link could exist between these two
could lead to new therapeutic targets and is therefore an
entities. This hypothesis has been confirmed over the last
important area of research.47 The intrinsic epileptogenicity of
decade, with increasing evidence showing that FCD type IIb is
FCD has clearly been demonstrated,48,49 suggesting impaired
associated with an abnormal activation of the mammalian
balance between excitation and inhibition. One hypothesis is
target of rapamycin (mTOR) pathway.27 However, the cause of
that dysplastic tissue could recapitulate or maintain immature
this aberrant activation is not known. Polymorphisms in TSC1
properties, including a paradoxical excitatory effect of GABA.
and TSC2 genes have been studied with conflicting results:
Indeed, depolarizing GABA activity has been documented in
some found increased polymorphisms in TSC1 gene in surgical
dysplastic tissue resected from patients with FCDs.50 However,
samples of patients with FCD type IIb when compared with
reduced GABAergic inhibition has also been demonstrated,
controls,28 whereas others did not.29
with abnormal organization of GABAergic interneurons.51 On
Recent evidences suggest defects elsewhere in the mTOR
the molecular level, expression of GABAA receptor is decreased
pathway for other subtypes of FCD. Families with focal
in areas of FCD, whereas expression of glutamate receptors
epilepsy and FCD type IIa have been shown to share germline,
(GluR) and N-methyl-D-aspartate [NMDA] receptors is
germline mosaic and brain somatic mutations in DEPDC5.30,31
increased.52,53 Moreover, the increased expression of NMDA
Moreover, upregulation of AKT3 was found in a patient with
receptors is more pronounced in highly epileptogenic dys-
FCD type Ib and infantile spasms,32 and brain somatic mTOR
plastic tissue when compared with less epileptogenic lesions.54
mutations were found in patients with FCD type II.33,34
Nevertheless, the nature of human studies limits the possibility
to know if these changes are causative, adaptative or are only
Acquired Causes markers of epileptic activity.
Viral infections transmitted by transplacental route have been
hypothesized to be responsible for the development of some
FCD. Evidence of human papillomavirus 16 has been found in Models of FCD
FCD type IIb by 2 independent teams,35,36 which is interesting
because human papillomavirus 16 oncoprotein E6 is known to Animal Models
be a potent activator of mTOR signaling.37 One of these teams To better understand the mechanisms of epileptogenicity in
also found evidence of cytomegalovirus and human herpes FCD, a number of animal models have been developed. These
virus type 6 in FCD type IIb.36 However, others did not find models can be generally categorized into either genetic or
any proof of its presence.38,39 acquired. In genetic models, genes involved in neuronal
Evidences of an implication of immune and inflammatory proliferation, differentiation as well as migration in the central
processes in FCD are also increasingly found.40 In fact, the nervous system (CNS) have been targeted to create transgenic
presence of activated microglia, astrocytic proliferation, and animals harboring cortical malformations, whereas acquired
proinflammatory cytokines in the dysplastic cortex of models are based on inducing cortical defects using chemical,
patients with FCD supports the role of the inflammatory like methylazoxymethanol (MAM), or physical injury, like
response in the clinical behavior of FCD.41,42 Interestingly, freeze-lesion and in utero irradiation, within a developmental
increased levels of P2X7R, a purinergic receptor presumed time window, that is, during the late stages of neuronal
to induce microglial activation, have recently been found in migration. To date, none of the models available has succeeded
dysplastic tissue from patients with FCD, and correlated to fully replicate all the defining features of FCD in humans,
FCD in children 113

but all do mimic certain aspects of the disorder. On the one discharges within 2-9 days of gene deletion, despite lack of
hand, dysmorphic neurons and giant cells reminiscent of discernible histological abnormalities.81 This suggests that the
balloon cells are observed in genetic models, however, molecular mechanisms downstream of TSC1/2, which con-
dyslamination and neuronal disorientation extend beyond tribute to epileptogenesis are potentially disjoint from the ones
the cortex into other brain regions.55-59 On the other hand, inducing lesion formation.
despite the ability of injury-inducing models to produce In order to restrict Cre recombination to the cortex,
aberrant architecture and neuronal positioning more specific Feliciano et al82 elegantly developed a localized TSC1 inacti-
to the cortical structures, these models lack, for the most part, vation model by taking advantage of the in utero electro-
the presence of cells with pronounced morphological abnor- poration technique.82 They stereotaxically delivered Cre-
malities.60-62 containing plasmids to cortical cells of E15/16 transgenic
Nonetheless, a high incidence of spontaneous seizures has mouse embryos that have the Tsc1 allele flanked by LoxP
been reported in a number of transgenic mouse lines where sites, leading to discrete focal lesions confined to the cortex.82
genes suppressing the mTOR pathway, primarily Tsc1/2, have The lesions showed some tuber hallmarks, including dyslami-
been inactivated.55,56,58,59,63 Furthermore, animals with nation and ectopic hypertrophic neurons, but were devoid of
acquired FCDs exhibit a heightened susceptibility to seizure balloon cells or increased GFAP immunoreactivity, which is a
development,64-68 and in some models resulted in sponta- marker of enhanced gliosis. However, the model failed to
neous epileptiform activity in vivo.69-73 In agreement with induce spontaneous seizures.
the molecular changes noticed in human epilepsy, upregula-
tion or potentiation of NMDA and GABA receptors were also
identified in multiple animal FCD models.71,74-76 Thus, FCD Injury-Induced Models
models are valuable tools to study how putative structural,
molecular and functional changes in the cortex contribute to Chemical Injury Model
epileptogenesis. Administration of antiproliferative compounds, such as MAM
and 1,3-bis (2-chloroethyl)-N-nitrosourea (or Carmustine), to
rodents during embryogenesis interferes with CNS develop-
Genetic Models ment by inhibiting cell division of neural progenitors in the
brain, thus ultimately disrupting layer formation.60,83 When
TSC1/2 Models compared with other antimitotic agents, MAM effects are
Different types of transgenic animals have been developed in highly selective to the CNS,60 and therefore are considered to
attempt to model FCD. Given the strong association between be a suitable neurotoxin to induce cortical lesions. Although
mutations in the mTOR pathway and cortical malformations, the MAM model has been extensively examined, MAM-
inactivation of the upstream mTOR signaling regulators TSC1/ induced in vivo spontaneous seizures were only reported in
2 has been the main experimental approach in recent years to one study.73 Yet, hyperexcitability and seizure susceptibility
explore FCD genetic etiologies. were common observations in the bulk of the literature on this
Conventional Tsc1/2 homozygous knockout was embryonic FCD model.84
lethal,77,78 and heterozygous Tsc1/2 mutation did not manifest The model is generated in rats by in utero injection of MAM
any pathological abnormalities or spontaneous seizures.79,80 at E15/E16, resulting in selective ablation of layers II-IV, while
More recently, using the Cre-lox recombination system in mice other cortical layers are spared.85 In addition, MAM treatment
to create Tsc1 conditional knockout (cKo) in neurons, that is, causes ectopic neuronal migration ultimately leading to
Cre-mediated excision of the Tsc1 gene driven by the neuron- heterotopic neuronal clusters. However, the effect of MAM is
specific promoter Synapsin, resulted in severe developmental widespread to other areas beside the cortex, mainly the
neuropathologies, namely enlarged dysplastic neurons.55 Sim- hippocampus and to a lesser extent the striatum, thalamus,
ilarly, abnormal cortical lamination and neuronal dispersion and hypothalamus.85 The most consistent characteristics of the
were respectively detected in astrocyte-specific (Gfap pro- MAM model are cortical thinning, along with heterotopia
moter-driven)56,59 and neural progenitor-specific (Emx1 and within the neocortex, in the periventricular white matter and
Gfap2 promoter-driven)57,58,63 Tsc1/2 cKo mice. Furthermore, CA1-CA2 regions of the hippocampus.60,83,85,86 Of note, there
adult mice in all the earlier-mentioned cKo models displayed is evidence suggesting that hippocampal heterotopic neurons
epileptogenesis, which was dependent on mTOR activation, are dysplastic cells of cortical lineage.86,87
demonstrated by the ability of the mTOR inhibitors, mainly Imaging and tracing experiments demonstrated ectopic
rapamycin, to suppress seizures.56-59,63 Although the pathol- mossy fiber innervations outside of heterotopia in the hippo-
ogy of these mice lacked FCD key features, including (1) the campus88 as well as excessive synaptic connectivity between
phenotype was diffuse as opposed to focal lesions in tubers, heterotopic cortex and other brain regions.89 Electrophysio-
and (2) lesions were not confined to the cortex. Also, in the logical recordings and c-fos activity marker assay confirmed
case of neuron- and neural progenitor-specific cKo, hypomye- functional activity between these synapses.90,91 Interestingly,
lination was reported,55,57,58,63 hence, marking mutation- when the long-term effect of prenatal MAM treatment was
induced molecular mechanisms that are not specific to FCD. investigated in adult rats (P70 and P160) via immunohisto-
Surprisingly, inducible neuron-specific Tsc1Nestin cKo, where chemistry, neuronal density in the CA1-CA3 and dentate
Tsc1 was timely deleted in adult mice, developed epileptogenic gyrus, and mossy fiber synaptogenesis were similar to control
114 T. Shaker et al.

rats. In addition, no signs of gliosis were detected in adult rats routing of migrating neurons98 as well as invasion of reactive
of either group,92 which suggests that some of MAM cellular glia99 from surrounding cortical regions to the lesion site,
and histological effects on the hippocampus are short-term. culminating in layering abnormalities. Characterization of
Ex vivo electrophysiological recordings in the CA1 of P25- axonal connectivity in P60 lesioned rats revealed disorganized
P35 MAM-treated rats indicated that extracellular potassium thalamocortical, corticothalamic, and callosal projections in the
concentrations ([Kþ]o) above physiological levels, led to microgyrus and surrounding cortical tissue.100,101
spontaneous epileptiform-like bursts upon fiber stimulation, Structural dyslamination is restricted to the hemisphere
a phenomenon that was not detected in control slices under ipsilateral to the lesion, and the hippocampal and subcortical
the same conditions.93 In a separate study, patch-clamp structures have normal histological appearance.102 Thus,
recordings revealed augmented hyperexcitability caused by unlike other FCD models, histological disruptions in the
loss of potassium channel-mediated A-type current, specifi- freeze-lesion model are localized rather than diffuse. Never-
cally Kv4.2 channel subunit, in heterotopic pyramidal-like cells theless, neuropathological alterations are widespread beyond
in the CA1, whereas other intrinsic electrophysiological the lesion. For example, when compared with sham-operated
properties were not changed.87 These findings were corrobo- rats, AMPA, and kainic acid receptor binding was significantly
rated with a notable reduction of Kv4.2 subunit detected via enhanced not only in dysplastic neurons within the micro-
immunohistochemistry and in situ hybridization,87 thus, gyrus but in remote cortical regions as well.75 Also, potentia-
implicating heterotopic neurons as a plausible source of high tion of NMDA current was reported in cortical dysplastic
[Kþ]o-induced spontaneous epileptiform activity. However, neurons and CA1 hippocampal pyramidal neurons, where
recent data showed that epileptiform activity is triggered upregulation of NR2B subunit was suggested to drive this
independent of heterotopic neurons94 (discussed “hyperexcit- enhanced hyperexcitability.75,103-105
able belt” in the next section). Furthermore, there was a global downregulation in GABAA
CA1 hippocampal recordings in slices obtained from MAM- receptor expression and binding throughout the ipsilateral
treated rats also demonstrated that the NMDA component of cortex, along with reduced GABAA expression in the ipsilateral
evoked glutamate-mediated excitatory postsynaptic currents hippocampus and restricted areas of the contralateral
has amplified amplitude and slower decay time as compared cortex.75,106 Electrophysiological data confirmed functional
with slices derived from saline injected control animals.74 decrease in GABAergic currents. In contrast, GABAB down-
Further support for NMDA receptor impairment comes from regulation was restricted to dysplastic neurons.75
immunoprecipitation and Western blot analysis where cortical So far no spontaneous seizures have been reported in freeze-
and hippocampal tissue of MAM-treated rats exhibited a lesioned newborn rats (i.e., without a second insult), however,
significant reduction in NR2A/B subunit assembly into NMDA many studies described propagating epileptiform activity in
receptor complex as well as NR2A/B phosphorylation, specif- response to presynaptic stimulation during ex vivo electro-
ically in postsynaptic membranes.95 Similar alterations in physiological recordings.107-109 Strikingly, epileptogenicity
postsynaptic NMDA composition and phosphorylation have was not generated within the dysplastic tissue, but rather by
been reported in epilepsy patients diagnosed with periven- cortical neurons that are in immediate vicinity of the micro-
tricular heterotopia.96 Therefore, the MAM model bears some gyrus,107-109 thus suggesting that the epileptic network lies
similarities to human cortical malformations. outside the site of the lesion. This corresponds to clinical data
where in epileptic FCD patients, ictal activity extends beyond
the anatomical lesion visible via imaging approaches.110 The
paramicrogyral zone (also referred to as the hyperexcitable
Cortical Freeze-Lesion Model belt) exhibits normal architecture, yet electophysiological data
In this model, physical injury is induced in newborn pups by indicated pronounced imbalance between excitatory and
local cooling of the neocortical surface, usually by bringing a inhibitory networks,75 likely to be caused by ectopic excitatory
liquid nitrogen-cooled probe in contact with the cranium input to these neurons.111 Moreover, paramicrogyral neurons
overlying the cortex, thus, leading to a focal necrotic lesion.61 displayed aberrant intrinsic properties, including increased
In adult rodents, cortical freeze-lesion mimics traumatic brain input resistance and a hyperpolarized resting membrane
injury,97 whereas in prenatal and neonatal rats or mice (up to potential.112
P4), and while cortical cells are still in the migratory state, the Recently, Takase et al113 found that cortical dysplasias
pathophysiology produced by the same experimental protocol elicited by freeze-lesions in prenatal rats at E18 were more
resembles human neuronal migration disorders,61 including severe than those in neonatal rats, as indicated by complete loss
3-4 layered cortex, that is, microgyrus or cortical cleft, that is, of local laminar organization.113 As postmitotic astrocytes do
schizencephaly. not complete populating the cortex until P0,114 the pro-
The model is well studied in rats, but comparable results nounced defects were attributed to a reduced repair capacity
have been reported in mice. Focal cortical freeze-lesions in of the embryonic cortex, caused by diminished reactive
neonatal (P0/P1) rodents, produced by 8-10 seconds contact of astrocytic gliosis at that stage.113 A follow-up study by the
probes 1-2 mm in size, have consistently proven to lead to a same group demonstrated that multiple, bilateral freeze-lesions
localized four layered microgyrus by P10 as opposed to the during embryogenesis are sufficient to provoke in vivo sponta-
normal 6-layered cortex.62,69 Following the freezing proce- neous seizures in adult rats, where 12 of 16 lesioned animals
dure, necrosis within the lesion is believed to be repaired by re- exhibited spikes confined to the bilateral hippocampi.71
FCD in children 115

However, the mortality rate was relatively high (20%-30%), as Electrophysiological data also indicated that excitation in
opposed to neonatal lesions (5%). the dysplastic cortex is notably enhanced. Whole-cell patch-
Similar to neonatal lesions,75,105 there was a notable increase clamp recordings of dysplastic and control neurons demon-
in the expression of NMDA glutamatergic subunits NR2A/2B strated a significant increase in spontaneous excitatory post-
in the cortex and the hippocampus of epileptic animals vs synaptic current amplitude and frequency, decrease in
shams at P28, that is, before seizure manifestation.71 The same spontaneous IPSC amplitude, and decrease in miniature
NMDA upregulation was detected at P78, that is, after seizure Inhibitory Post-Synaptic Currents (IPSC) amplitude and
development, only in lesioned rats. Moreover, the glutamate frequency in dysplastic neurons.123 In addition, patch-
transporters Glutamate Aspartate Transporter and Glutamate clamp recordings combined with paired-pulse stimulation
Transporter 1 along with GABAergic markers glutamic acid revealed augmented presynaptic release probability in pyr-
carboxylase 65/67 (GAD65/67) were markedly upregulated in amidal neurons.124 Extracellular field recordings in dysplastic
the cortex at P28.71 Further investigations are necessary to slices showed that hyperexcitability induced by the GABAA
elucidate the epileptogenic mechanism in this novel model. receptor antagonist, bicuculline, was significantly higher in
pyramidal neurons of irradiated animals than in their control
counterparts.76 Hence, in utero irradiation creates an imbal-
ance between the excitatory and inhibitory networks in
In Utero Irradiation Model dysplastic tissue, possibly leading to epileptogenicity. Indeed,
Initially used as a tool to investigate the effect of radiation on 2 studies have reported spontaneous seizure activity in adult
brain development during ontogenesis,115 in utero gamma- animals, albeit the number of animals used in the experiments
irradiation was found to elicit cortical malformations in was relatively small. The first study identified ictal activity in
rodents, primarily rats, pertinent to human cortical dyspla- 4 of 7 irradiated rats via continuous in vivo EEG monitoring,
sias.62 Depending on the timing and dose of radiation, yet seizures originated from outside the frontal cortex or the
pathological structural changes vary in severity. Migratory hippocampus.125 A subsequent study used EEG accompanied
progenitors and precursors appear to be specifically vulnerable by video recording, where seizures localized to the cortical or
to gamma rays, therefore, irradiation of rat embryos (usually hippocampal electrodes were detected in 3 of 5 animals.72
with 145-225 cGy dosage) during late developmental stages However, these results warrant further investigation.
( E17) mostly targets migrating cells destined to the last
forming layers during corticogenesis, that is, intermediate
layers II-IV, resulting in a 4-layered microgyrus after birth.116
Other cytoarchitectural defects include subcortical and
The Two-Hit Model
periventricular heterotopic gray matter, thinning of the cor- Because most of the available FCD models are not sufficient to
tical mantle, clusters of disoriented neurons, and the pre- trigger in vivo recurrent spontaneous seizures, it is hypothesized
sence of scattered giant pyramidal neurons.116 However, the that cortical lesions rather facilitate seizure development by
irradiation-induced phenotype is diffuse and not uniform predisposing the brain to hyperexcitability. The findings that
across the cortex. Also, neither dysmorphic nor balloon cells cortical lesions in different models can lower seizure threshold
were described in this model. and enhance seizure activity support this notion. For example,
Postnatally, the total number of neurons in irradiated MAM-treated rats postnatally exposed to anesthetic agents,
animals (rP6) was comparable with control animals, how- such as flurothyl, or proconvulsant agents, like pilocarpine,
ever, irradiated rats displayed over 50% reduction of cortical kainic acid (KA), and bicuculline, showed significantly lower
inhibitory interneurons, where the parvalbumin (PVþ)- and threshold for seizures than nontreated controls.64,65,67 Also, in
calbindin (CBþ)-expressing interneuronal subtypes were sig- 2 different studies, subjecting neonatal freeze-lesioned and
nificantly decreased compared to controls, whereas the cell MAM-treated rats to prolonged hyperthermia, which is a
number of calretinin (CRþ)-expressing subtype was not model for febrile seizures, led to reduced seizure latency.66,68
changed.117 Notably, this robust reduction of CBþ and PVþ Therefore, if the perinatal FCD is followed by a seizure-
interneurons, measured by cell density, continues throughout provoking insult, additive pathological contributions of both
adulthood.118 Furthermore, Zhou and Roper119 demonstrated insults (or two hits) can potentiate ictal discharges.
a functional impairment in surviving CBþ and somatostatin- So far, seizures were reported in 2 distinct two-hit models
expressing interneurons within cortical dysplasias, mainly that combined perinatal cortical injury with an epileptogenic
marked decline in excitatory input and lower spontaneous stressor. The first model used in utero irradiation at E17,
firing rate as opposed to controls, while CR interneuron firing followed by a single sub-convulsive dose of pentylenetetrazole
rate was not affected by radiation. More recently, dysplastic fast at P45, where only the rats exposed to both insults (8 of 11)
spiking PVþ interneurons were found to have reduced output displayed chronic frequent epileptiform spikes (up to 50 days
synaptic efficiency onto other neurons as well as impaired local postpentylenetetrazole injection) that arose from the bilateral
gap junction coupling within these neuronal populations.120 hippocampi.69 In the second model, cortical freeze-lesion at P1
Previous clinical studies described interneuron reduction was followed by hyperthermia-induced seizures at P10.70 After
in tissue derived from epilepsy patients diagnosed with a latent seizure-free period, spontaneous recurrent seizures
FCD,61,121,122 thus implicating a potentially similar patho- were recorded in adult rats around P80, whereas no seizure
physiology between FCDs and the in utero irradiation model. activity was observed throughout adulthood in rats with the
116 T. Shaker et al.

lesion alone or hyperthermia alone.70 Seizures occurred in 11. Mellerio C, Labeyrie MA, Chassoux F, et al: Optimizing MR imaging
detection of type 2 focal cortical dysplasia: Best criteria for clinical
86%-100% of male rats arising in the temporal lobe ipsilateral
practice. Am J Neuroradiol 33:1932-1938, 2012
to the lesion.45,70,126 Interestingly, male rats appear to be 12. Tassi L, Garbelli R, Colombo N, et al: Electroclinical, MRI and surgical
particularly vulnerable in this model, as females that received outcomes in 100 epileptic patients with type II FCD. Epileptic Disord
the same insults did not develop epilepsy.127 Analysis of 14:257-266, 2012
epileptic rats revealed hippocampal atrophy, neuronal loss, 13. Barkovich AJ, Kuzniecky RI, Bollen AW, et al: Focal transmantle
and synaptic reorganization in the ipsilateral hippocampus as dysplasia: A specific malformation of cortical development. Neurology
49:1148-1152, 1997
compared to rats that underwent a single insult and naive 14. Tassi L, Colombo N, Garbelli R, et al: Focal cortical dysplasia: neuro-
pups.45 Although the lesion enhanced the function of gluta- pathological subtypes, EEG, neuroimaging and surgical outcome. Brain
matergic networks (as previously indicated in the Cortical 125:1719-1732, 2002
Freeze-Lesion Model Section), hyperthermia potentiated 15. Urbach H, Scheffler B, Heinrichsmeier T, et al: Focal cortical dysplasia of
Taylor’s balloon cell type: A clinicopathological entity with characteristic
GABAergic networks in hippocampal CA1 pyramidal cells,
neuroimaging and histopathological features, and favorable postsurgical
thus, producing additive excitatory and inhibitory effects on outcome. Epilepsia 43:33-40, 2002
hippocampal circuits.105 For an overview on the lesion þ 16. De Ciantis A, Barba C, Tassi L, et al: 7 T MRI in focal epilepsy with
hyperthermia two-hit model, see review.128 unrevealing conventional field strength imaging. Epilepsia 57:445-454,
2016
17. Bonilha L, Montenegro MA, Rorden C, et al: Voxel-based morphometry
reveals excess gray matter concentration in patients with focal cortical
Conclusions dysplasia. Epilepsia 47:908-915, 2006
18. Focke NK, Symms MR, Burdett JL, et al: Voxel-based analysis of whole
FCD represent a common cause of medication resistant brain FLAIR at 3 T detects focal cortical dysplasia. Epilepsia 49:786-793,
epilepsy in children. Their recognition has been improved by 2008
modern imaging techniques and their complete removal leads 19. Eriksson SH, Rugg-Gunn FJ, Symms MR, et al: Diffusion tensor imaging
to a successful surgical outcome. However, both electro- in patients with epilepsy and malformations of cortical development.
Brain 124:617-626, 2001
physiological and functional data in animal and human studies 20. Kim SK, Na DG, Byun HS, et al: Focal cortical dysplasia:
suggest that in a number of these lesions, the epileptogenic Comparison of MRI and FDG-PET. J Comput Assist Tomogr 24:
zone extends beyond the anatomical margins because of 296-302, 2000
modifications of both excitatory and synaptic networks. 21. Sood S, Chugani HT: Functional neuroimaging in the preoperative
Finally, surgical tissue studies over the next few years would evaluation of children with drug-resistant epilepsy. Childs Nerv Syst
22:810-820, 2006
further improve our knowledge on the mechanisms leading to 22. Chassoux F, Devaux B, Landré E, et al: Stereoelectroencephalography in
FCD as somatic mutations or mosaicisms would be identified focal cortical dysplasia: A 3D approach to delineating the dysplastic
in critical pathways involved in cortical development. cortex. Brain 123:1733-1751, 2000
23. Gambardella A, Palmini A, Andermann F, et al: Usefulness of focal
rhythmic discharges on scalp EEG of patients with focal cortical dysplasia
References and intractable epilepsy. Electroencephalogr Clin Neurophysiol
1. Harvey AS, Cross JH, Shinnar S, et al: Defining the spectrum of 98:243-249, 1996
international practice in pediatric epilepsy surgery patients. Epilepsia 24. Bast T, Oezkan O, Rona S, et al: EEG and MEG source analysis of single
49:146-155, 2008 and averaged interictal spikes reveals intrinsic epileptogenicity in focal
2. Palmini A, Holthausen H: Focal malformations of cortical development: cortical dysplasia. Epilepsia 45:621-631, 2004
A most relevant etiology of epilepsy in children. Handb Clin Neurol 25. Cotter D, Honavar M, Lovestone S, et al: Disturbance of Notch-1 and
111:549-565, 2013 Wnt signalling proteins in neuroglial balloon cells and abnormal large
3. Taylor DC, Falconer MA, Bruton CJ, et al: Focal dysplasia of the cerebral neurons in focal cortical dysplasia in human cortex. Acta Neuropathol
cortex in epilepsy. J Neurol Neurosurg Psychiatry 34:369-387, 1971 98:465-472, 1999
4. Blumcke I, Thom M, Aronica E, et al: The clinicopathologic spectrum of 26. Wang VY, Chang EF, Barbaro NM: Focal cortical dysplasia: a review of
focal cortical dysplasias: A consensus classification proposed by an ad hoc pathological features, genetics, and surgical outcome. Neurosurg Focus
Task Force of the ILAE Diagnostic Methods Commission. Epilepsia 20:E7, 2006
52:158-174, 2011 27. Crino PB: mTOR: A pathogenic signaling pathway in developmental
5. Lerner JT, Salamon N, Hauptman JS, et al: Assessment and surgical brain malformations. Trends Mol Med 17:734-742, 2011
outcomes for mild type I and severe type II cortical dysplasia: A critical 28. Becker AJ, Urbach H, Scheffler B, et al: Focal cortical dysplasia of Taylor’s
review and the UCLA experience. Epilepsia 50:1310-1335, 2009 balloon cell type: Mutational analysis of the TSC1 gene indicates a
6. Kim DW, Lee SK, Chu K, et al: Predictors of surgical outcome and pathogenic relationship to tuberous sclerosis. Ann Neurol 52:29-37,
pathologic considerations in focal cortical dysplasia. Neurology 2002
72:211-216, 2009 29. Gumbinger C, Rohsbach CB, Schulze-Bonhage A, et al: Focal cortical
7. Krsek P, Maton B, Jayakar P, et al: Incomplete resection of focal cortical dysplasia: A genotype-phenotype analysis of polymorphisms and
dysplasia is the main predictor of poor postsurgical outcome. Neurology mutations in the TSC genes. Epilepsia 50:1396-1408, 2009
72:217-223, 2009 30. Baulac S, Ishida S, Marsan E, et al: Familial focal epilepsy with focal
8. Duncan JS: Imaging in the surgical treatment of epilepsy. Nat Rev Neurol cortical dysplasia due to DEPDC5 mutations. Ann Neurol 77:675-683,
6:537-550, 2010 2015
9. Krsek P, Pieper T, Karlmeier A, et al: Different presurgical characteristics 31. Scheffer IE, Heron SE, Regan BM, et al: Mutations in mammalian target
and seizure outcomes in children with focal cortical dysplasia type I or II. of rapamycin regulator DEPDC5 cause focal epilepsy with brain
Epilepsia 50:125-137, 2009 malformations. Ann Neurol 75:782-787, 2014
10. Colombo N, Tassi L, Deleo F, et al: Focal cortical dysplasia type IIa and 32. Conti V, Pantaleo M, Barba C, et al: Focal dysplasia of the cerebral cortex
IIb: MRI aspects in 118 cases proven by histopathology. Neuroradiology and infantile spasms associated with somatic 1q21.1-q44 duplication
54:1065-1077, 2012 including the AKT3 gene. Clin Genet 88:241-247, 2015
FCD in children 117

33. Lim JS, Lee JH: Brain somatic mutations in MTOR leading to focal 57. Magri L, Cambiaghi M, Cominelli M, et al: Sustained activation of mTOR
cortical dysplasia. BMB Rep 49:71-72, 2016 pathway in embryonic neural stem cells leads to development of
34. Nakashima M, Saitsu H, Takei N, et al: Somatic mutations in the MTOR tuberous sclerosis complex-associated lesions. Cell Stem Cell
gene cause focal cortical dysplasia type IIb. Ann Neurol 78:375-386, 9:447-462, 2011
2015 58. Magri L, Cominelli M, Cambiaghi M, et al: Timing of mTOR activation
35. Chen J, Tsai V, Parker WE, et al: Detection of human papillomavirus in affects tuberous sclerosis complex neuropathology in mouse models. Dis
human focal cortical dysplasia type IIB. Ann Neurol 72:881-892, 2012 Model Mech 6:1185-1197, 2013
36. Liu S, Lu L, Cheng X, et al: Viral infection and focal cortical dysplasia. 59. Zeng LH, Rensing NR, Zhang B, et al: Tsc2 gene inactivation causes a
Ann Neurol 75:614-616, 2014 more severe epilepsy phenotype than Tsc1 inactivation in a mouse
37. Lu Z, Hu X, Li Y, et al: Human papillomavirus 16 E6 oncoprotein model of tuberous sclerosis complex. Hum Mol Genet 20:445-454,
interferences with insulin signaling pathway by binding to tuberin. J Biol 2011
Chem 279:35664-35670, 2004 60. Benardete EA, Kriegstein AR: Increased excitability and decreased
38. Coras R, Korn K, Bien CG, et al: No evidence for human papillomavirus sensitivity to GABA in an animal model of dysplastic cortex. Epilepsia
infection in focal cortical dysplasia IIb. Ann Neurol 77:312-319, 43:970-982, 2002
2015 61. Dvorak K, Feit J: Migration of neuroblasts through partial necrosis of the
39. Shapiro KA, McGuone D, Deshpande V, et al: Failure to detect human cerebral cortex in newborn rats-contribution to the problems of
papillomavirus in focal cortical dysplasia type IIb. Ann Neurol 78:63-67, morphological development and developmental period of cerebral
2015 microgyria. Histological and autoradiographical study. Acta Neuro-
40. Khurana DS: Focal epilepsies: immunologic and inflammatory mecha- pathol 38:203-212, 1977
nisms. Semin Pediatr Neurol 21:207-213, 2014 62. Roper SN: In utero irradiation of rats as a model of human cerebrocort-
41. Boer K, Spliet WG, van Rijen PC, et al: Evidence of activated microglia in ical dysgenesis: A review. Epilepsy Res 32:63-74, 1998
focal cortical dysplasia. J Neuroimmunol 173:188-195, 2006 63. Way SW, McKenna 3rd J, Mietzsch U, et al: Loss of Tsc2 in radial glia
42. Choi J, Nordli Jr DR, Alden TD, et al: Cellular injury and neuro- models the brain pathology of tuberous sclerosis complex in the mouse.
inflammation in children with chronic intractable epilepsy. J Neuro- Hum Mol Genet 18:1252-1265, 2009
inflammation 6:38, 2009 64. Baraban SC, Schwartzkroin PA: Flurothyl seizure susceptibility in rats
43. Wei YJ, Guo W, Sun FJ, et al: Increased expression and cellular following prenatal methylazoxymethanol treatment. Epilepsy Res
localization of P2X7R in cortical lesions of patients with focal cortical 23:189-194, 1996
dysplasia. J Neuropathol Exp Neurol 71:61-68, 2015 65. de Feo MR, Mecarelli O, Ricci GF: Seizure susceptibility in immature rats
44. Iyer A, Zurolo E, Spliet WG, et al: Evaluation of the innate and adaptive with micrencephaly induced by prenatal exposure to methylazoxyme-
immunity in type I and type II focal cortical dysplasias. Epilepsia thanol acetate. Pharmacol Res 31:109-114, 1995
51:1763-1773, 2010 66. Germano IM, Zhang YF, Sperber EF, et al: Neuronal migration disorders
45. Gibbs S, Chattopadhyaya B, Desgent S, et al: Long-term consequences of increase susceptibility to hyperthermia-induced seizures in developing
a prolonged febrile seizure in a dual pathology model. Neurobiol Dis rats. Epilepsia 37:902-910, 1996
43:312-321, 2011 67. Germano IM, Sperber EF: Increased seizure susceptibility in adult rats
46. Wang D, Blümcke I, Gui Q, et al: Clinico-pathological investigations of with neuronal migration disorders. Brain Res 777:219-222, 1997
Rasmussen encephalitis suggest multifocal disease progression and 68. Scantlebury MH, Ouellet PL, Psarropoulou C, et al: Freeze lesion-
associated focal cortical dysplasia. Epileptic Disord 15:32-43, 2013 induced focal cortical dysplasia predisposes to atypical hyperthermic
47. Wong M: Mechanisms of epileptogenesis in tuberous sclerosis complex seizures in the immature rat. Epilepsia 45:592-600, 2004
and related malformations of cortical development with abnormal 69. Oghlakian RO, Tilelli CQ, Hiremath GK, et al: Single injection of a low
glioneuronal proliferation. Epilepsia 49:8-21, 2008 dose of pentylenetetrazole leads to epileptogenesis in an animal model of
48. Otsubo H, Iida K, Oishi M, et al: Neurophysiologic findings of neuronal cortical dysplasia. Epilepsia 50:801-810, 2009
migration disorders: Intrinsic epileptogenicity of focal cortical dysplasia 70. Scantlebury MH, Gibbs SA, Foadjo B, et al: Febrile seizures in the
on electroencephalography, electrocorticography, and magnetoencepha- predisposed brain: A new model of temporal lobe epilepsy. Ann Neurol
lography. J Child Neurol 20:357-363, 2005 58:41-49, 2005
49. Palmini A, Gambardella A, Andermann F, et al: Intrinsic epileptogenicity 71. Kamada T, Sun W, Takase K, et al: Spontaneous seizures in a rat model
of human dysplastic cortex as suggested by corticography and surgical of multiple prenatal freeze lesioning. Epilepsy Res 105:280-291, 2013
results. Ann Neurol 37:476-487, 1995 72. Kellinghaus C, Kunieda T, Ying Z, et al: Severity of histopathologic
50. D’Antuono M, Louvel J, Köhling R, et al: GABAA receptor-dependent abnormalities and in vivo epileptogenicity in the in utero radiation model
synchronization leads to ictogenesis in the human dysplastic cortex. of rats is dose dependent. Epilepsia 45:583-591, 2004
Brain 127:1626-1640, 2004 73. Harrington EP, Möddel G, Najm IM, et al: Altered glutamate receptor -
51. Calcagnotto ME, Paredes MF, Tihan T, et al: Dysfunction of synaptic transporter expression and spontaneous seizures in rats exposed to
inhibition in epilepsy associated with focal cortical dysplasia. J Neurosci methylazoxymethanol in utero. Epilepsia 48:158-168, 2007
25:9649-9657, 2005 74. Calcagnotto ME, Baraban SC: Prolonged NMDA-mediated responses,
52. Crino PB, Duhaime AC, Baltuch G, et al: Differential expression of altered ifenprodil sensitivity, and epileptiform-like events in the mal-
glutamate and GABA-A receptor subunit mRNA in cortical dysplasia. formed hippocampus of methylazoxymethanol exposed rats. J Neuro-
Neurology 56:906-913, 2001 physiol 94:153-162, 2005
53. Mikuni N, Babb TL, Ying Z, et al: NMDA-receptors 1 and 2A/B 75. Zilles K, Qü M, Schleicher A, et al: Characterization of neuronal
coassembly increased in human epileptic focal cortical dysplasia. migration disorders in neocortical structures: quantitative receptor
Epilepsia 40:1683-1687, 1999 autoradiography of ionotropic glutamate, GABA(A) and GABA
54. Najm IM, Ying Z, Babb T, et al: Epileptogenicity correlated with (B) receptors. Eur J Neurosci 10:3095-3106, 1998
increased N-methyl-D-aspartate receptor subunit NR2A/B in human 76. Roper SN, King MA, Abraham LA, et al: Disinhibited in vitro
focal cortical dysplasia. Epilepsia 41:971-976, 2000 neocortical slices containing experimentally induced cortical dys-
55. Meikle L, Talos DM, Onda H, et al: A mouse model of tuberous sclerosis: plasia demonstrate hyperexcitability. Epilepsy Res 26:443-449,
neuronal loss of Tsc1 causes dysplastic and ectopic neurons, reduced 1997
myelination, seizure activity, and limited survival. J Neurosci 77. Onda H, Lueck A, Marks PW, et al: Tsc2(þ/) mice develop tumors in
27:5546-5558, 2007 multiple sites that express gelsolin and are influenced by genetic
56. Uhlmann EJ, Wong M, Baldwin RL, et al: Astrocyte-specific TSC1 background. J Clin Invest 104:687-695, 1999
conditional knockout mice exhibit abnormal neuronal organization and 78. Kobayashi T, Minowa O, Sugitani Y, et al: A germ-line Tsc1 mutation
seizures. Ann Neurol 52:285-296, 2002 causes tumor development and embryonic lethality that are similar, but
118 T. Shaker et al.

not identical to, those caused by Tsc2 mutation in mice. Proc Natl Acad 100. Rosen GD, Burstein D, Galaburda AM: Changes in efferent and afferent
Sci U S A 98:8762-8767, 2001 connectivity in rats with induced cerebrocortical microgyria. J Comp
79. Goorden SM, van Woerden GM, van der Weerd L, et al: Cognitive Neurol 418:423-440, 2000
deficits in Tsc1þ/ mice in the absence of cerebral lesions and seizures. 101. Giannetti S, Gaglini P, Granato A, et al: Organization of callosal
Ann Neurol 62:648-655, 2007 connections in rats with experimentally induced microgyria. Childs
80. Ehninger D, Han S, Shilyansky C, et al: Reversal of learning deficits in a Nerv Syst 15:444-448, 1999. [discussion 449-50]
Tsc2þ/ mouse model of tuberous sclerosis. Nat Med 14:843-848, 102. Schwindt W, Burke M, Pillekamp F, et al: Functional magnetic
2008 resonance imaging and somatosensory evoked potentials in rats with a
81. Abs E, Goorden SM, Schreiber J: TORC1-dependent epilepsy caused by neonatally induced freeze lesion of the somatosensory cortex. J Cereb
acute biallelic Tsc1 deletion in adult mice. Ann Neurol 74:569-579, 2013 Blood Flow Metab 24:1409-1418, 2004
82. Feliciano DM, Su T, Lopez J, et al: Single-cell Tsc1 knockout during 103. Ying Z, Babb TL, Comair YG, et al: Induced expression of NMDAR2
corticogenesis generates tuber-like lesions and reduces seizure threshold proteins and differential expression of NMDAR1 splice variants in
in mice. J Clin Invest 121:1596-1607, 2011 dysplastic neurons of human epileptic neocortex. J Neuropathol Exp
83. Singh SC: Ectopic neurones in the hippocampus of the postnatal rat Neurol 57:47-62, 1998
exposed to methylazoxymethanol during foetal development. Acta 104. DeFazio RA, Hablitz JJ: Alterations in NMDA receptors in a rat model of
Neuropathol 40:111-116, 1977 cortical dysplasia. J Neurophysiol 83:315-321, 2000
84. Battaglia G, Colciaghi F, Finardi A, et al: Intrinsic epileptogenicity of 105. Ouardouz M, Lema P, Awad PN, et al: N-methyl-D-aspartate,
dysplastic cortex: converging data from experimental models and hyperpolarization-activated cation current (Ih) and gamma-
human patients. Epilepsia 54:33-36, 2013 aminobutyric acid conductances govern the risk of epileptogenesis
85. Battaglia G, Pagliardini S, Saglietti L, et al: Neurogenesis in cerebral following febrile seizures in rat hippocampus. Eur J Neurosci
heterotopia induced in rats by prenatal methylazoxymethanol treatment. 31:1252-1260, 2010
Cereb Cortex 13:736-748, 2003 106. Redecker C, Luhmann HJ, Hagemann G, et al: Differential down-
86. Paredes M, Pleasure SJ, Baraban SC: Embryonic and early postnatal regulation of GABAA receptor subunits in widespread brain regions in
abnormalities contributing to the development of hippocampal malfor- the freeze-lesion model of focal cortical malformations. J Neurosci
mations in a rodent model of dysplasia. J Comp Neurol 495:133-148, 20:5045-5053, 2000
2006 107. Luhmann HJ, Raabe K, Qü M, et al: Characterization of neuronal
87. Castro PA, Cooper EC, Lowenstein DH, et al: Hippocampal heterotopia migration disorders in neocortical structures: extracellular in vitro
lack functional Kv4.2 potassium channels in the methylazoxymethanol recordings. Eur J Neurosci 10:3085-3094, 1998
model of cortical malformations and epilepsy. J Neurosci 21:6626-6634, 108. Jacobs KM, Gutnick MJ, Prince DA: Hyperexcitability in a model of
2001 cortical maldevelopment. Cereb Cortex 6:514-523, 1996
88. Chevassus-Au-Louis N, Jorquera I, Ben-Ari Y, et al: Abnormal con- 109. Jacobs KM, Hwang BJ, Prince DA: Focal epileptogenesis in a rat model of
nections in the malformed cortex of rats with prenatal treatment with polymicrogyria. J Neurophysiol 81:159-173, 1999
methylazoxymethanol may support hyperexcitability. Dev Neurosci 110. Palmini A, Gambardella A, Andermann F, et al: Operative strategies for
21:385-392, 1999 patients with cortical dysplastic lesions and intractable epilepsy. Epilepsia
89. Colacitti C, Sancini G, Franceschetti S, et al: Altered connections between 35:S57-S71, 1994
neocortical and heterotopic areas in methylazoxymethanol-treated rat. 111. Zsombok A, Jacobs KM: Postsynaptic currents prior to onset
Epilepsy Res 32:49-62, 1998 of epileptiform activity in rat microgyria. J Neurophysiol 98:178-186,
90. Chevassus-Au-Louis N, Congar P, Represa A, et al: Neuronal migration 2007
disorders: Heterotopic neocortical neurons in CA1 provide a bridge 112. Albertson AJ, Yang J, Hablitz JJ: Decreased hyperpolarization-activated
between the hippocampus and the neocortex. Proc Natl Acad Sci U S A currents in layer 5 pyramidal neurons enhances excitability in focal
95:10263-10268, 1998 cortical dysplasia. J Neurophysiol 106:2189-2200, 2011
91. Chevassus-Au-Louis N, Rafiki A, Jorquera I, et al: Neocortex in the 113. Takase K, Shigeto H, Suzuki SO, et al: Prenatal freeze lesioning produces
hippocampus: an anatomical and functional study of CA1 heterotopias epileptogenic focal cortical dysplasia. Epilepsia 49:997-1010, 2008
after prenatal treatment with methylazoxymethanol in rats. J Comp 114. Parnavelas JG: Glial cell lineages in the rat cerebral cortex. Exp Neurol
Neurol 394:520-536, 1998 156:418-429, 1999
92. Park KI, Chu K, Jung KH, et al: Role of cortical dysplasia in epilepto- 115. McGrath JJ, Riggs HE, Schwarz HP: Malformation of the adult brain
genesis following prolonged febrile seizure. Epilepsia 51:1809-1819, (albino rat) resulting from prenatal irradiation. J Neuropathol Exp
2010 Neurol 15:432-446, 1956. [discussion, 446-7]
93. Baraban SC, Schwartzkroin PA: Electrophysiology of CA1 pyramidal 116. Ferrer I: Experimentally induced cortical malformations in rats. Childs
neurons in an animal model of neuronal migration disorders: prenatal Nerv Syst 9:403-407, 1993
methylazoxymethanol treatment. Epilepsy Res 22:145-156, 1995 117. Deukmedjian AJ, King MA, Cuda C, et al: The GABAergic system of the
94. Doisy ET, Wenzel HJ, Mu Y, et al: Nodule excitability in an animal model developing neocortex has a reduced capacity to recover from in utero
of periventricular nodular heterotopia: c-fos activation in organotypic injury in experimental cortical dysplasia. J Neuropathol Exp Neurol
hippocampal slices. Epilepsia 56:626-635, 2015 63:1265-1273, 2004
95. Gardoni F, Pagliardini S, Setola V, et al: The NMDA receptor complex is 118. Roper SN, Eisenschenk S, King MA: Reduced density of parvalbumin-
altered in an animal model of human cerebral heterotopia. J Neuropathol and calbindin D28-immunoreactive neurons in experimental cortical
Exp Neurol 62:662-675, 2003 dysplasia. Epilepsy Res 37:63-71, 1999
96. Battaglia G, Pagliardini S, Ferrario A, et al: AlphaCaMKII and NMDA- 119. Zhou FW, Roper SN: Altered firing rates and patterns in interneurons in
receptor subunit expression in epileptogenic cortex from human experimental cortical dysplasia. Cereb Cortex 21:1645-1658, 2011
periventricular nodular heterotopia. Epilepsia 43:209-216, 2002 120. Zhou FW, Roper SN: Reduced chemical and electrical connections of
97. Hogg S, Moser PC, Sanger DJ: Mild traumatic lesion of the right parietal fast-spiking interneurons in experimental cortical dysplasia. J Neuro-
cortex of the rat: selective behavioural deficits in the absence of physiol 112:1277-1290, 2014
neurological impairment. Behav Brain Res 93:143-155, 1998 121. Ferrer I, Oliver B, Russi A, et al: Parvalbumin and calbindin-D28k
98. Kharazia VN, Jacobs KM, Prince DA: Light microscopic study of GluR1 immunocytochemistry in human neocortical epileptic foci. J Neurol Sci
and calbindin expression in interneurons of neocortical microgyral 123:18-25, 1994
malformations. Neuroscience 120:207-218, 2003 122. Spreafico R, Tassi L, Colombo N, et al: Inhibitory circuits in human
99. Bordey A, Lyons SA, Hablitz JJ, et al: Electrophysiological characteristics dysplastic tissue. Epilepsia 41:S168-S173, 2000
of reactive astrocytes in experimental cortical dysplasia. J Neurophysiol 123. Zhu WJ, Roper SN: Reduced inhibition in an animal model of cortical
85:1719-1731, 2001 dysplasia. J Neurosci 20:8925-8931, 2000
FCD in children 119

124. Chen HX, Xiang H, Roper SN: Impaired developmental switch of short- seizure in the immature rat with a focal neocortical lesion. Neurobiol
term plasticity in pyramidal cells of dysplastic cortex. Epilepsia Dis 32:176-182, 2008
48:141-148, 2007 127. Desgent S, Duss S, Sanon NT, et al: Early-life stress is associated with
125. Kondo S, Najm I, Kunieda T, et al: Electroencephalographic character- gender-based vulnerability to epileptogenesis in rat pups. PLoS One 7:
ization of an adult rat model of radiation-induced cortical dysplasia. e42622, 2012
Epilepsia 42:1221-1227, 2001 128. Sanon NT, Desgent S, Carmant L: Atypical febrile seizures, mesial
126. Gibbs SA, Scantlebury MH, Awad P, et al: Hippocampal atrophy and temporal lobe epilepsy, and dual pathology. Epilepsy Res Treat
abnormal brain development following a prolonged hyperthermic 2012:342928, 2012

You might also like