Anti-Epileptogenesis in Rodent Post-Traumatic Epilepsy Models

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Neuroscience Letters 497 (2011) 163–171

Contents lists available at ScienceDirect

Neuroscience Letters
journal homepage: www.elsevier.com/locate/neulet

Review

Anti-epileptogenesis in rodent post-traumatic epilepsy models


Asla Pitkänen a,b,∗ , Tamuna Bolkvadze a , Riikka Immonen c
a
Department of Neurobiology, Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland
b
Department of Neurology, Kuopio University Hospital, P.O. Box 1777, FIN-70211 Kuopio, Finland
c
Department of Neurobiology, Biomedical NMR Group, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland

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

Article history: Post-traumatic epilepsy (PTE) accounts for 10–20% of symptomatic epilepsies. The urgency to understand
Received 9 August 2010 the process of post-traumatic epileptogenesis and search for antiepileptogenic treatments is emphasized
Received in revised form 25 January 2011 by a recent increase in traumatic brain injury (TBI) related to military combat or accidents in the aging
Accepted 15 February 2011
population. Recent developments in modeling of PTE in rodents have provided tools for identification of
novel drug targets for antiepileptogenesis and biomarkers for predicting the risk of epileptogenesis and
Keywords:
treatment efficacy after TBI. Here we review the available data on endophenotypes of humans and rodents
Antiepileptic drug
with TBI associated with epilepsy. Also, current understanding of the mechanisms and biomarkers for
Biomarker
Controlled cortical impact
PTE as well as factors associated with preclinical study designs are discussed. Finally, we summarize the
Lateral fluid-percussion attempts to prevent PTE in experimental models.
Traumatic brain injury © 2011 Elsevier Ireland Ltd. All rights reserved.
Magnetic resonance imaging

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
2. Risk factors for PTE in human and experimental TBI – how do they match? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164
2.1. Human PTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164
2.2. Experimental PTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165
3. In search of treatment targets – molecular and network reorganization in PTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
4. Biomarkers and surrogate markers for PTE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
5. Challenge of designing antiepileptogenesis trials to prevent PTE in rodents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
6. Disease-modification treatment approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
6.1. Antiepileptogenesis after TBI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
6.2. Co-morbidity modification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170

1. Introduction
[48]. After TBI, the 30-year cumulative incidence of epilepsy is 2.1%
for mild, 4.2% for moderate, and 16.7% for severe injuries [2], and TBI
The rate of hospital admissions for traumatic brain injury (TBI)
is estimated to cause 10–20% of all symptomatic epilepsies [2]. It
across Europe is estimated to be 235/100 000/year [5]. In the
can be estimated that in the USA and the European Union (EU), with
USA, the incidence of emergency department visits for TBI is
a total population of about 800 million, at least 0.5 million surviv-
403/100 000/year and that of hospital admissions 85/100 000/year
ing individuals live with post-traumatic epilepsy (PTE). Data on the
economic burden of PTE are unavailable. Some idea is provided by
the average lifetime cost of TBI, which in the USA is about $200 000
∗ Corresponding author at: A.I. Virtanen Institute for Molecular Sciences, Univer-
per case when scaled to 2004 price levels [5]. Thus, in addition to the
sity of Eastern Finland,
P.O. Box 1627, FIN-70211 Kuopio, Finland. Tel.: +358 50 517 2091;
subjective burden, the economic burden caused by PTE is substan-
fax: +358 17 163025. tial, and means to prevent post-traumatic epileptogenesis would
E-mail address: asla.pitkanen@uef.fi (A. Pitkänen). benefit both the individual and society.

0304-3940/$ – see front matter © 2011 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.neulet.2011.02.033
164 A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171

TBI refers to a brain injury caused by an external mechan- Table 1


Comparison of risk factors for human post-traumatic epilepsy (PTE) and rat PTE in
ical force such as a blow to the head, concussive forces,
lateral fluid percussion (LFP) injury model.
acceleration–deceleration forces, blast injury or a projectile missile
such as a bullet [53]. Epilepsy is a disorder of the brain character- Risk factor for human PTE Presence in LFP injury
ized by an enduring predisposition to generate epileptic seizures Severe Moderate
and by the neurobiological, cognitive, psychological, and social con-
Penetrating injury No No
sequences of this condition. The definition of epilepsy requires the Skull fracture/wounds with Craniectomy, dura left Craniectomy, dura left
occurrence of at least one epileptic seizure [25]. After TBI, the occur- dural penetration intact intact
rence of seizures has been categorized as immediate (<24 h), early Biparietal contusions No No
(1–7 d), or late seizures (>1 wk after TBI) [27]. The immediate and Acute intracerebral Detectable in 15/15a Detectable in 15/15a
hematoma Score 3–4 in 7/15 Score 3–4 in 3/15
early seizures are considered to reflect the severity of the injury Score 4 in 5/15 Score 4 in 1/15
itself, whereas the late seizures result from maturation of epilep- Intracranial hemorrhage Yes (15/15)b Yes (15/15)b
togenic pathology. Thus, TBI associated with one unprovoked late Multiple or bilateral No No
seizure qualifies for diagnosis of PTE. contusions
Multiple intracranial n.d. n.d.
Currently, there is no universally accepted definition for epilep-
procedures
togenesis or anti-epileptogenesis. This complicates the analysis Lesion location (frontal, Yes n.a.
and interpretation of data obtained in preclinical and clinical temporal)
antiepileptogenic trials. Often “epileptogenesis” or “latency period” Evidence of greater than 1.5–3% increase in the 1.5–3% increase in the
are used synonymously as operational terms that refer to a 5-mm midline shift width of ipsilateral width of ipsilateral
hemisphere (11/15) hemisphere (6/15)
time period between the insult and the occurrence of the first
Loss of consciousness for No No
unprovoked seizure. Evidence is accumulating to show that neu- >24 h
robiological changes that occur during the latency period continue Prolonged length of n.d. n.d.
to progress even after epilepsy diagnosis [67,70]. Therefore, we post-traumatic amnesia
Coma duration >24 h n.a. n.a.
recently proposed a revised definition for epileptogenesis. The
Low GCS (<10) n.a. n.a.
major difference from the previous definition is that epileptogen- Old age n.d. n.d.
esis extends from the latency period to include the epilepsy phase Early PTS (within first week) Yes n.d.
[72]. Thus, the term “epileptogenesis” is defined as the development Epileptiform EEG activity 1 n.d. n.d.
and extension of tissue capable of generating spontaneous seizures, month post-TBI

including (a) development of an epileptic condition and (b) pro- Abbreviations: EEG, electroencephalogram; GCS, Glasgow Coma Scale; n.a., not appli-
gression after the condition is established. To describe the effect of cable; n.d., no data; PTS, post-traumatic seizure. For references, see the text. Some
of the changes in LFP injury listed in the table are demonstrated in Fig. 1.
treatments, we suggest the term “disease or syndrome modification” a
Number and size of intracerebral hematomas were scored from 0 (not present)
which refers to a process that alters the development or progres- to 4 (several large hematomas).
sion of a “disease”, in this case epilepsy (either epileptic disease b
Includes both microbleed in white and/or grey matter as well as hematomas.
or epilepsy syndrome). Disease or syndrome modifying interven-
tions may be “antiepileptogenic”, which refers to a process that
counteracts the effects of epileptogenesis, including (a) prevention relevant ways, and optimize the selection of animals for pre-
(lower percentage of subjects develop epilepsy), (b) seizure modi- clinical antiepileptogenesis studies. Identification of epileptogenic
fication (e.g., shorter and less frequent seizures), and (c) cure. These endophenotypes is also relevant for the identification of epilepto-
interventions could also modify comorbidities by reducing or pre- genic mechanisms and presentation of biomarkers (or surrogate
venting deleterious nonepileptic functional changes in the brain markers) at different phases of the post-traumatic epileptogenic
(e.g., memory, emotional behavior). process. We will next compare the human and experimental data.
The number of reports demonstrating the disease modifying
effects of various treatments on the TBI aftermath in experimen- 2.1. Human PTE
tal models is abundant [55,68]. In particular, these studies show
favorable PTE-related co-morbidity modifying effects, including After the first late seizure, 86% of patients were reported
enhanced recovery of memory, motor performance, or emotional to develop a second seizure within 2 years, suggesting the
behavior, whereas evidence for antiepileptogenesis is meager. establishment of an epileptogenic network [31]. Further, recent
Surprisingly, more information is available from humans than magnetoencephalography (MEG) studies revealed that some
experimental models, which could relate to the relatively recent patients without reported seizures after mild TBI have epileptiform
recognition of the occurrence of epilepsy in injured animals. To take spikes when assessed at 12–140 months post-TBI [49]. Whether
the field forward and conduct a study that focuses on prevention some of these individuals would later develop epilepsy remains to
of PTE after TBI, we need valid models that are reproducible and be assessed. Some studies have also investigated the persistence of
easy to use, treatments that are designed to counteract the mech- seizures after TBI. For example, Eftekhar et al. [21] reported that not
anisms of post-TBI epileptogenesis, biomarkers and/or surrogate all, but 75% of patients with penetrating TBI during Iraq–Iran war
markers for predicting treatment efficacy, and optimized preclini- continue to have seizures after a 21-year follow-up, even when data
cal study designs that reveal true positive data. We will next discuss were adjusted for AED treatment. The risk was associated with >3
each one of these aspects and outline challenges that we are facing pieces of shrapnel and with sphincter disturbances. Interestingly,
when trying to tackle post-traumatic epileptogenesis. no association was found between the persistence of epilepsy and
lesion size. Some important questions arising from these observa-
tions are: what are the mechanisms of post-TBI epileptogenesis,
2. Risk factors for PTE in human and experimental TBI – and what mechanisms can counteract the persistence of epilepsy
how do they match? in a subpopulation of patients with TBI?
Table 1 summarizes the risk factors for PTE obtained from epi-
TBI is a heterogeneous disorder with different forms of clinical demiological studies conducted in civilian or military populations.
presentation. Identification of endophenotypes in injured subjects Many of the identified risk factors reflect the severity of brain
with a high risk for PTE can guide attempts to model PTE in clinically injury, and show that the risk for PTE increases with the severity
A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171 165

of injury. However, data about the association of the type of injury

Reference
with epileptogenesis are meager. A recent study investigating the

[14,15]
[43,44]

[36,37]
civilian population with TBI that included MRI analysis performed

[29]

[81]

[82]

[10]
[7]

[8]
1–360 months (median 12 months) post-TBI reported an associ-
ation of PTE with contusions but not with an isolated diagnosis
of diffuse axonal injury (DAI) [74]. This study found no association
with microbleeds and PTE, which challenges the data from previous

Seizure susceptibility ↑ to ECS-induced sz at


epidemiologic studies (see Table 1).

Sz threshold as adult ±0; Sz threshold for


80% had epileptiform spiking; Increased
Increased susceptibility to PTZ-induced

Increased susceptibility to PTZ-induced

Increased susceptibility to PTZ-induced


One possible epileptogenic factor occurring during the post-TBI
susceptibility to PTZ-induced seizures aftermath is acute symptomatic status epilepticus (SE), which has
been reported to be associated with epilepsy in 41% of cases [33].
88% had epileptiform spiking
Vespa et al. [88] monitored 20 patients with severe non-penetrating
minimal clonic sz as adult ↓

TBI using cortical EEG while they received therapeutic levels of


phenytoin. Ten patients had seizures and seven of these patients
had nonconvulsive SE lasting on average 41 min during the first
week post-injury.

7 d post-TBI
Other recently reported clinical risk factors for PTE in civilian

Abbreviations: CCI, controlled cortical impact; ECS, electroconvulsive shock; FPI, flud-percussio injury; n.d., no data; sz, seizure; P, postnatal day; PTZ, pentylenetetrazol.
seizures

seizures

seizures
populations are a history of depression and three or more chronic
Other

medical conditions at the time of injury [24]. Data on genetic risk


factors for outcome from TBI is emerging, but very few studies have
assessed the linkage of genes to PTE [16]. One study found that
ApoE4 allele is associated with a 2.4-fold increased risk of late post-
traumatic seizures after moderate to severe TBI [19]. However, this
Ictal episode ≤10

∼90 (behavioral

was not confirmed by another study [1] which also reported no


Sz duration (s)

assessment)

association with haptoglobin phenotype.


Epileptogenesis occurs in parallel to post-injury recovery [71].
45–60

Recent data suggests that epilepsy has a negative effect on gen-


104
n.d.

n.d.

n.d.
91
50

eral functional outcome when tested using the Glasgow Outcome


Scale (GOSE) [74]. Whether this implies that the severely injured
brain recovers poorly and consequently, is prone to epileptoge-
Sz frequency

nesis remains to be studied. Other testable hypotheses are that


epileptogenic mechanisms counteract those resulting in favor-
0.23/d
0.3/d

0.1/d

able functional recovery, or that epilepsy-related psychological


n.d.

n.d.

n.d.

n.d.

n.d.
n.d

and work-related consequences compromise the outcome when


assessed using GOSE.
spontaneous sz
Latency to

2.2. Experimental PTE


6 months

6 months
4–11 wk

42–71 d
∼2 wk

260 d
n.d.

n.d.

n.d.

A large number of animal models have been developed and used


to investigate the molecular and cellular mechanisms of post-TBI
injury in rodents and pigs, and to test the efficacy of novel treat-
ments on functional recovery [60]. However, it has only recently
20%(mild) to 36%

been pointed out that a significant percentage of injured rodents


(severe injury)

develop epileptiform activity and epilepsy in long-term follow-up


Animals with
epilepsy (%)

1 of 8 (13%)

1 of 17 (6%)

(Table 2). Whether the risk factors for epileptogenesis in a sub-


Epileptogenesis after experimental traumatic brain injury in rodents.

group of animals that develop PTE after TBI correspond to that


∼92%
50%
n.d.

n.d.

n.d.

in humans is little understood. Experiments in the lateral fluid-


9%

percussion (LFP) injury model provide some data. First, as evidence


so far suggests, the development of PTE requires a severe external
impact, that is, about 3 atm when “severity” is calculated based on
Mouse (CD-1) Adult

Mouse (CD-1) Adult


Species, age (strain)

Mouse (B6) Adult

Mouse (B6) Adult

the mortality (>30%) within the injury group. This correlates with
the observations in humans showing that the risk of PTE increases
Rat P32–35

Rat P16–18

with the severity of TBI. Second, the location of the injury in the
Rat Adult

Rat Adult

Rat P17

posterior aspects of the cortex including the perirhinal, postrhinal


and entorhinal cortices is associated with rat PTE [46]. Patient data
suggests that parietal or temporal location of injury is associated
with PTE [35]. However, comparison of the injury location between
rodents and humans is difficult because of the different brain ori-
Closed skull traumatic

entation relative to the skull as well as a lissencephalic appearance


of the rodent brain. Third, as LFP injury causes both grey and white
Parasagittal FPI

brain injury

matter damage, it is of interest that also CCI, in which the damage


Weight-drop

is more confined to the grey matter, can trigger epileptogenesis


Lateral FPI

Lateral FPI

[8,36]. Whether this suggests that, as seen in patients, white mat-


Model
Table 2

ter damage in rodents is not required for epileptogenesis remains


CCI

CCI
CCI

CCI

a testable hypothesis.
166 A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171

Fig. 1. Examples of T2 maps (A and B) and T2*-weighted MR images (C and D) of rats with severe (impact force >3.0 atm; left column) or moderate (1.9–2.1 atm; right
column) lateral fluid-percussion (LFP) injury. (A) At 3 d after severe TBI, several hematomas (yellow arrows), a series of microhemorrhages in white matter structures (orange
arrows), and ipsilateral cortical edema (white arrowheads) were observed. In a subgroup of animals with severe TBI, edema was associated with an increased (about 2%)
width of the ipsilateral hemisphere (indicated with white dashed line). (B) After moderate injury, MRI findings were milder than after severe TBI, particularly, the extent of
intracortical hematomas. However, there was significant overlap between the rats with severe or mild injury. For example, 5 of 15 rats with severe and 1 of 15 rats with
moderate injury has the score 4+ (most extensive) hematoma (p = 0.068, Chi-Square test). Swollen ipsilateral hemisphere was found in 11 of 15 rats with severe and 6 of 15
rats with moderate LFP injury (p = 0.065). This resulted in a 1.5–3% increase in the width of the ipsilateral hemisphere when compared to that of the contralateral hemisphere
in the slice taken from the injury epicenter. Also, as we noted in the original description, cortical lesion volume assessed by MRI was 23.9 ± 16.8 mm3 (range 0–69.5 mm3 )
in the moderate group and 31.8 ± 10.9 mm3 (range 12.9–48.4 mm3 ) in the severe group [45]. (C) A T2*-weighted MR images from a rat with severe and (D) with a moderate
injury. The arrow points to the microbleeds in the external capsule that is associated with microglial and astrocytic accumulation (see panel E and also [96]). Panels (E and F)
show the histology at 18 d post-TBI from the same rats. The black arrowhead indicates the cortical lesion. Note the remarkable neurodegeneration that happened in 2–3 wk
post-TBI. The orange arrow indicates gliosis at the site of white matter microhemorrhage detected in MRI Images 2 wk earlier (panels A–D). Scale bar in A equals 1 mm and
in E 1 mm.

We re-analyzed our recent dataset from rats with LFP-induced gest that the occurrence of epilepsy is lower than in CD-1 mice,
TBI, which were imaged with quantitative MRI at 3 d post-injury implying that genetic background affects post-TBI epileptogene-
[44]. The goal was to assess whether some of the parameters known sis (Table 1) [8,36,37]. Genetic background has also been shown
to associate with a high risk of PTE in humans could also be found to the affect the development of co-morbidities, including motor
in rats with severe but not with moderate injury. The parameters and cognitive decline after experimental TBI when C57BL/6, FVB/N,
assessed included the occurrence of hematoma(s), hemorrhage, and 129/SvEMS mice were compared [26]. The effects on post-
and early hemispheric swelling on MRI. Our data show that a sub- injury neurodegeneration have been negligible when C57BL/10J
group of rats with severe impact, and thus more likely to develop and C57BL/6J mice with subtle differences in genetic background
epilepsy, had a tendency to a larger number of hematomas and were compared [94]. So far, no data about single gene mutations
more apparent brain swelling as compared to rats with moderate and the risk of PTE in rodents have been published. As some data
injury (Table 1; Fig. 1). from patients suggest, there is an inverse association between the
The effect of age and genetic background on experimental occurrence of epilepsy and behavioral recovery. It is, therefore, of
post-TBI epileptogenesis has been assessed recently. CCI injury interest to note that in the CCI model rats receiving two electrocon-
was induced at P16–18 and rats were followed-up for up to 2.5 vulsive seizures (ECSs) within the first 24 h post-contusion showed
months. No spontaneous seizures were detected, but CCI injury accelerated recovery of beam-walking ability, reduced volume of
resulted in permanent lowering of the threshold for clonic seizures necrosis and less spontaneous activity compared to animals receiv-
[81]. Interestingly preliminary data from CCI-injured B6 mice sug- ing only contusions. The effect was even more pronounced after
A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171 167

7 ECSs [22]. Moreover, daily injection of PTZ (25 mg/kg) started at recently shown that hilar cell loss includes over a 50% reduction
24 h post-TBI induced by central FPI in rats and continued for about in the total number of parvalbumin positive cells which associates
4 wk improved the recovery of water-maze performance despite with a decrease in phasic inhibition in granule cells [38,66]. Other
of strengthening of the severity of behavioral seizures during the cell types lost in the hilus include mossy cells (35% remaining),
follow-up, that is, induction of kindling [32]. Finally, we have not cholecystokinin immunoreactive (IR) cells (44%), and somatostatin-
observed SE in any of our injured animals which could have com- IR cells [87]. In addition, neurogenesis, gliosis, dendritic plasticity,
promised the interpretation of the cause of epileptogenesis after and axonal injury have been demonstrated in several models of
TBI. TBI associated with PTE but their contribution to epileptogenesis is
unexplored (see [69]). The cortex and different nuclei of the thala-
mus also undergo remarkable neurodegeneration, but there are no
3. In search of treatment targets – molecular and network data detailing the cell types involved [80].
reorganization in PTE One of the major challenges for future work is to find means to
localize the ictogenic focus/network in post-traumatic brain. This
After TBI, the primary injury occurring within minutes to hours would help to specify which one of the observed molecular and
is related to the impact itself, and includes shearing of white mat- cellular changes are relevant for post-traumatic epileptogenesis,
ter tracts, focal contusions, hematomas, (micro)hemorrhages, and and consequently, guide targeting of therapies to the right locations
diffuse swelling. At the cellular level this is seen as microporation in antiepileptogenesis trials.
of membranes, leaky ion channels, and conformational changes
in proteins. Secondary damage develops over hours to days,
4. Biomarkers and surrogate markers for PTE
and includes neurotransmitter release, free-radical generation,
calcium-mediated damage, gene activation, mitochondrial dys-
The search for biomarkers that predict post-TBI recovery is
function, and inflammatory responses [53]. Many of the secondary
active and includes both serum and CSF analyses as well as the
changes have previously been associated with epileptogenesis
use of various imaging modalities [18]. Again, however, PTE has
caused by other etiologies like SE [70]. However, causality and the
not been included as an outcome measure in these studies. We
details of the overlap between molecular pathways underlying each
recently performed quantitative MRI in the hippocampus ipsilat-
change in various etiologies need to be explored.
eral to LFP injury. We found that the diffusion trace (Dav ) measured
Several large scale molecular profiling studies are available from
at both early (3 h) and chronic (23 d, 2, 3, 6, 7 and 11 months)
mice and rats [11–13,23,39,40,47,50,51,57,73,90,93,95] and one in
post-injury time points correlated with the number of spikes or
humans with TBI [59]. In most of the studies the timing of sampling
epileptiform discharges quantified in the EEG recorded during a
has been within 1 wk post-injury, and therefore, involves only the
pentylenetetrazol (PTZ) seizure susceptibility test at 12 months
early post-injury period. Post-traumatic epileptogenicity of identi-
post-TBI. Correlations were also found between the Dav and the
fied genes has not been further analyzed.
density of mossy fiber sprouting that is a typical plastic response of
One of the prevailing hypotheses for epileptogenesis is acquired
granule cell axons found in the hippocampus both in experimental
channelopathy which appears as alterations in the expression of
and human temporal lobe epilepsy [44]. These results suggest that
ion channel subunits resulting in altered receptor composition
quantitative diffusion MRI could serve as a tool to facilitate predic-
and increased excitability (for review, see [70]). Interestingly, we
tion of increased seizure susceptibility in experimental models of
recently showed that in the LFP injury model there are changes in
PTE.
GABAA receptor subunits in the dentate granule cell layer which
last for several months [66]. Also, Gibson et al. [28] demonstrated
changes in the ␣1, ␣2, ␣3, and ␥2 subunits of the GABAA recep- 5. Challenge of designing antiepileptogenesis trials to
tor when assessed within 24 h after central FPI. Changes in GABAA prevent PTE in rodents
receptor subunit expression have a temporal association with
decreased phasic and/or tonic inhibition in the dentate granule cells Table 2 summarizes the models of TBI in which epileptogene-
[54,61,66]. Mao et al. [54] recently showed a 3-fold increase in the sis has been described. We focus on those models that are used in
expression of the Nav 1.6 subunit in the hippocampal neurons dur- the trauma field [60]. A common feature for most of the models is
ing the 72 h post-LFP injury in rats, but its association with post-TBI that ≤50% of animals develop epilepsy, latency is weeks to months
hyperexcitability needs to be explored. rather than days, and seizure frequency is low. Further, the occur-
Even though the data on pathologic changes after TBI in humans rence of seizures can vary substantially within the study population
is abundant, little data are available that indicate a connection (Fig. 2). One study reported the occurrence of epileptiform spiking
between the cellular changes and PTE in patients. The data available activity in 80% of rats after LFP injury, occurring also in rats with no
show that hippocampal pathology includes mossy fiber sprouting detected seizures [43].
as well as a 50–70% neuronal loss in pyramidal cell layers, which One caveat in the data obtained is that even the longest video-
have been described in TLE with other etiologies [56,83]. How- EEG monitoring studies cover only a small fragment of the entire
ever, there is no available comparison of whether these changes epileptogenesis period, and in fact, the duration of the delay
would differ between TBI patients with and without PTE. Recently between the insult and the occurrence of first late seizures is not
Tomkins et al. [86] compared the extent of blood–brain bar- known. In some models such as weight-drop or closed skull trau-
rier (BBB) disruption with MRI in TBI patients with and without matic brain injury, only hyperexcitability has been demonstrated,
epilepsy. They found larger perilesional BBB disruption in PTE and the occurrence of spontaneous seizures remains to be shown
patients as compared to TBI patients without epilepsy. Interest- [29]. These data on the phenotype of experimental PTE makes us
ingly, there was no difference in lesion size between the groups. consider the value of PTE models in antiepileptogenesis studies rel-
In animal models, mossy fiber sprouting has been described ative to that of, for example, status epilepticus (SE) models. As well
both in rats and mice after CCI and/or LFP injury [36,43]. In the described, after chemically or electrically induced SE typically ≥80%
LFP model, there is also hilar cell loss that varies from 20% to 75% of rats develop epilepsy within 1 month, and the seizure frequency
depending on the time point investigated as well as on the count- can be up to 50/d (for review, see [41]), making the SE models sub-
ing method used [30,43,52,80]. In addition, there is a 10–20% loss stantially less labor-intensive than PTE models. However, there is
of principal cells, particularly in the CA3a region [30]. We have no data that would qualify SE models to replace the PTE models
168 A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171

Fig. 2. Examples of the occurrence of spontaneous late seizures in 3 rats (#73, #2, #46) during a 52-wk follow-up (unpublished data from [43]). The grey bars show the
timing of continuous video-EEG monitoring (1 or 2 wk at a time). Seizures were detected relatively infrequently, and each rat had a variable appearance of seizures.

when assessing the molecular and cellular consequences leading 6. Disease-modification treatment approaches
to PTE after TBI. Also, there is no evidence that any treatment mod-
ifying the epileptogenic process after SE would have a similar effect As discussed earlier (see Section 1), we divide the disease
after TBI. modification after TBI into antiepileptogenesis and co-morbidity
In all models of TBI analyzed so far, the susceptibility to induced modification.
seizures has been shown to be increased (Table 2). These include
LFP injury, CCI, and weight-drop models in rats as well as closed 6.1. Antiepileptogenesis after TBI
skull TBI, LFP injury, and CCI models in mice. Seizure susceptibil-
ity has been reported for kainate, PTZ or ECS induced seizures. Table 3 summarizes the treatment approaches to suppress
Whether increased seizure susceptibility to induced seizures can epileptogenesis after experimental TBI. The Soltez group investi-
be used as a surrogate marker for ongoing epileptogenesis, and as gated the effect of a cannabinoid 1 receptor antagonist, SR141716A
an indicator for pinpointing individual animals that will eventually (Rimonabant® ) on epileptogenesis [20]. Interestingly, SR141716A
develop epilepsy, is under dispute. The little available literature is a proconvulsant that has been demonstrated to lower the
suggests that perhaps it can. Rats with spontaneous seizures trig- threshold for kainate-induced seizures [9]. They induced epilepto-
gered by kainic acid or pilocarpine-induced SE at 30–45 d before genesis by lateral fluid-percussion induced TBI, and administered
a PTZ test showed enhanced susceptibility to PTZ-induced gener- SR141716A (1 mg/kg or 10 mg/kg, i.p.) as a single injection at
alized convulsions and SE [6,89]. Further, the latency to the first 2 min post-injury. The threshold for kainate-induced seizures was
generalized convulsion was reduced from 1280s (controls) to 389 s assessed at 6 wk post-TBI. The reduction in the latency to kainate-
(post-SE animals)[6]. Interestingly, also after pilocarpine-induced induced seizures was prevented by SR141716A. Also, the total
SE the rats that did not develop SE acutely, and consequently time spent in seizures after kainate administration was reduced
had no spontaneous seizures during a 45 d follow-up, still showed in the SR141716A group as compared to the vehicle group. Impor-
increased seizure susceptibility to PTZ even though it was less tantly, no positive effect was found if SR141716A was administered
than that of rats with spontaneous seizures [6]. It was recently 20 min post-TBI. Another study by Chrzaszcz et al. [10] used
shown that the non-SE rats after pilocarpine injection also actu- the closed-skull midline impact model in mice, and adminis-
ally develop infrequent spontaneous electrographic seizures (0.1–1 tered minozac 3 or 6 h after injury. Minozac is a suppressor of
seizures/wk) within 8 ± 2 months post-pilocarpine [64]. These data pro-inflammatory cytokine up-regulation. Its discovery and devel-
have some similarity to PTE models, in which injured rats do not opment was made using a de novo compound discovery platform
show spontaneous seizures or have very infrequent seizures, but interfaced with hierarchal biological screens for oral bioavailabil-
have enhanced response to chemoconvulsants or electroshock (ES), ity, toxicity, brain penetrance, and stability, and it has been tested
and show enhanced in vitro excitability [20,29,36,37,61]. Finally, for efficacy in animal models of brain disorders [62]. After 1 wk
we recently demonstrated that animals with TBI and spontaneous post-TBI, minozac-treated mice showed less susceptibility to ES-
seizures show the highest seizure susceptibility in the PTZ test (see induced seizures than sham-operated animals. Whether minozac
Fig. 7C in [44]). treatment prevented the long-term increase in seizure susceptibil-
Currently, the PTE rodent models are laborious to use. One way ity and occurrence of late seizures remains to be explored. The effect
to facilitate epileptogenesis and increase the proportion of animals of ketogenic diet on post-TBI epileptogenesis was investigated by
that develop epilepsy within a reasonable time frame would be to Schwartzkroin et al. [75]. They triggered LFP injury to 8-wk-old rats
induce TBI in animals with an epileptogenesis-prone genetic back- and administered ketogenic diet for 3 wk post-injury. Seizure sus-
ground, or select animals with the highest risk based on post-TBI ceptibility to fluorothyl was not any different from that in rats on
imaging. Another issue to be further explored is to assess whether standard diet when assessed at 3 and 6 wk after discontinuation of
a decrease in seizure susceptibility is a valid outcome measure in ketogenic diet. It should be noted, however, that TBI had no effect
antiepileptogenesis studies. on seizure susceptibility to fluorothyl (seizure threshold, seizure
A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171 169

Table 3
Treatment approaches to suppress epileptogenesis after TBI in experimental models.

Treatment Mechanism of Action Model Effect Reference

Rimonabant ®
CB1 receptor antagonist Lateral FPI (rat) Seizure susceptibility to kainate-induced seizures at 6 wk post-TBI ↓ [20]
(SR141716A)
Minozac® Reduction of proinflammatory Closed skull TBI Seizure susceptibility to ECS-induced seizures at 7 d post-TBI ↓ [10]
cytokine production by activated glia (CD-1 mouse)
Ketogenic diet Unknown (likely multiple) Lateral FPI (rat) No effect on fluorothyl-induced seizure susceptibility at 3 or 6 wk [75]
post-TBI
Hypothermia Reduction of brain temperature Parasagittal FPI (rat) Seizure susceptibility to PTZ-induced seizures at 12 wk post-TBI ↓ [3]

Abbreviations: CB1, cannabinoid receptor 1; ECS, electroconvulsive shock; FPI, fluid percussion injury; PTZ, pentylenetetrazol; TBI, traumatic brain injury.

duration) when injured and sham-operated animals on standard a structurally-novel broad-spectrum anticonvulsant that modu-
diet were compared. Finally, Atkins et al. [3] induced moderate lates the activity of several ligand- and voltage-gated ion channels
parasagittal FP injury in adult rats. Animals were kept under nor- in neurons. These include an inhibitory effect on the AMPA and
mothermic or moderate hypothermic temperatures for 4 h starting kainate subtypes of glutamate receptors, mixed modulatory effects
at 30 min post-injury. Seizure susceptibility was tested at 12-wk (usually positive) on some types of GABAA receptors, negative
post-TBI by using PTZ. Behavioral analysis of data indicated reduced modulatory effects on some types of voltage-gated Na+ and Ca2+
number of induced seizures during the 60 min post-PTZ injection. channels, and a positive modulatory effect on at least one type
The behavioral severity of seizures was not affected of K+ channel [76]. Hoover and colleagues [34] reported that the
The list of antiepileptogenic treatment attempts in animal mod- administration of topiramate 30 min after lateral FPI improved
els is substantially shorter than that in human PTE which includes sensorimotor behavior when assessed 4 wk postinjury, but had
several antiepileptic drugs as monotherapy or polytherapy (car- no favorable effects on neuronal survival or learning. Talampanel,
bamazepine, phenytoin, valproate, phenobarbital), glucocorticoids, a non-competitive AMPA antagonist, administered 30 min after
and magnesium sulphate [84,85,92]. Clinical studies have been parasagittal FPI reduced neurodegeneration in the contusion area
conducted with relatively little or no evidence for antiepileptoge- as well as the hippocampal CA1 subfield when assessed 7 d after
nesis in animal models. Many of these compounds were, however, TBI [4]. We recently reported that lacosamide, when administra-
shown to have neuroprotective effects after experimental TBI. In tion was started at 30 min post-LFP injury and continued for 3 d,
particular, MgSO4 was shown to be one of the most efficient neuro- had no effect on lesion size or motor impairment when assessed
protectants both in acute and chronic experiments after LFP injury 2 wk postinjury [65]. Also carisbamate, when administered at
[58,78], and still when given to patients, no effect was seen, rather 15 min post LFP injury for 1 d, had no effect on brain edema (48 h
they worsened [84]. As a note, we use doses of MgSO4 in our anes- post-injury), lesion size, motor function or learning when ana-
thesia cocktail when inducing LFP injury that are comparable to lyzed 4 wk post-injury [42]. Recently, Dash et al. [17] reported
that used in neuroprotection studies, and in our hands 50% of the that administration of 400 mg/kg valproate starting at 30 min or
rats developed epilepsy in 1 year follow-up [43]. It should be noted 3 h post-CCI induced injury and continued for 5 d improved BBB
that even though there are reports on the disappearance of seizures integrity, reduced TBI-associated hippocampal dendritic damage,
in 25–45% of patients after established PTE (see [21] and refer- lessened cortical contusion volume, and improved motor function
ences therein), there is no evidence that the AEDs used to treat and spatial memory. This was associated with inhibition of glyco-
the condition in these patients had any contribution to remission. gen synthase kinase 3 (GSK-3) and histone deacetylase (HDAC).
Interestingly, these behavioral improvements were not observed
6.2. Co-morbidity modification when SAHA (suberoylanilide hydroxamic acid), a selective HDAC
inhibitor, was administered. Taken together, there is meager evi-
The list of compounds that have been tested to enhance post- dence pointing to significant neuroprotective effects of any AEDs
TBI recovery in experimental models is extensive and includes after TBI in animal models which could associate with co-morbidity
AEDs, excitatory amino acid modulators, Ca-channel modulators, modification. However, AEDs like valproate that have mechanisms
ROS scavengers, anti-inflammatory drugs, neurotrophic factors, of action other than just those on receptor or ligand gated ion
caspase inhibitors, calpain inhibitors, endocrinological modulators, channels, including epigenetic modulatory effects on histone acety-
and modulators of neurotransmission; furthermore, administra- lation and DNA methylation, may warrant further studies.
tion routes range from systemic pharmacotherapy to cell therapy,
gene therapy, and vagal nerve stimulation [55,68,77]. Even though 7. Conclusions
many of these therapies have shown favorable effects on co-
morbidities (motor impairment, cognitive and emotional decline) The first article published on TBI included in PubMed is from
which associate with PTE in these models, a change in seizure sus- 1894 [63] and that on PTE from 1947, more than 50 years later
ceptibility or occurrence of seizures have never been included as [91]. Thereafter, about 430 articles have been listed in PubMed on
outcome measures. “posttraumatic epilepsy”. In animal models of TBI that have been
To control immediate and early seizures the use of AEDs is indi- commonly used in the trauma field, epileptogenesis was detected
cated. During the past years several laboratories have assessed less than 10 years ago [16,36,43]. Not surprisingly, comparisons of
whether AEDs are neuroprotective, and whether they affect post- endophenotypes between patients and animals with PTE are still
TBI recovery in experimental models. Even though epileptogenesis in their infancy. Similarly, few attempts have been made to test the
has not been assessed, these data give information on the effects effects of candidate antiepileptogenic treatments in models of PTE
of AEDs on other components of disease or syndrome modifi- are few. To move further in the search for a cure for PTE, more work
cation, that is, on co-morbidity modification. It has been shown is needed to understand the specific clinical, molecular, and net-
that the administration of remacemide, which has an affinity for work characteristics of the injured brain in humans and rodents,
the NMDA receptor ionophore and Na+ channels, 15 min postin- which lead to PTE at different ages. Application of novel imaging
jury reduced cortical lesion volume when assessed at 48 h. There techniques can be expected to help in localization of the developing
were no favorable effects on spatial memory [79]. Topiramate is epileptogenic focus/network in post-traumatic brain, and conse-
170 A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171

quently, guide the investigations to the right anatomic target(s). [21] B. Eftekhar, M.A. Sahraian, B. Nouralishahi, A. Khaji, Z. Vahabi, M. Ghodsi, H.
Such data would have implications for the model development as Araghizadeh, M.R. Soroush, S.K. Esmaeili, M. Masoumi, Prognostic factors in the
persistence of posttraumatic epilepsy after penetrating head injuries sustained
well as understanding of the mechanisms of post-traumatic epilep- in war, J. Neurosurg. 110 (2) (2009) 319–326.
togenesis, identification of biomarkers and surrogate markers, and [22] D.M. Feeney, B.Y. Bailey, M.G. Boyeson, D.A. Hovda, R.L. Sutton, The effect of
optimization of preclinical trial designs. seizures on recovery of function following cortical contusion in the rat, Brain
Inj. 1 (1) (1987) 27–32.
[23] S. Ferguson, B. Mouzon, G. Kayihan, M. Wood, F. Poon, S. Doore, V. Mathura, J.
Humphrey, B. O’Steen, R. Hayes, A. Roses, M. Mullan, F. Crawford, Apolipopro-
Acknowledgements tein E genotype and oxidative stress response to traumatic brain injury,
Neuroscience 168 (3) (2010) 811–819.
This study was supported by the Academy of Finland, the Sigrid [24] P.L. Ferguson, G.M. Smith, B.B. Wannamaker, D.J. Thurman, E.E. Pickelsimer,
A.W. Selassie, A population-based study of risk of epilepsy after hospitalization
Juselius Foundation, and CURE. We thank Nick Hayward, M.Sc., for
for traumatic brain injury, Epilepsia 51 (5) (2010) 891–898.
revising the language of the manuscript. [25] R.S. Fisher, W. van Emde Boas, W. Blume, C. Elger, P. Genton, P. Lee, J. Engel
Jr., Epileptic seizures and epilepsy: definitions proposed by the International
League Against Epilepsy (ILAE) and the International Bureau for Epilepsy (IBE),
References Epilepsia 46 (4) (2005) 470–472.
[26] G.B. Fox, R.A. LeVasseur, A.I. Faden, Behavioral responses of C57BL/6, FVB/N,
[1] G.D. Anderson, N.R. Temkin, S.S. Dikmen, R. Diaz-Arrastia, J.E. Machamer, and 129/SvEMS mouse strains to traumatic brain injury: implications for gene
C. Farhrenbruch, J.W. Miller, S.M. Sadrzadeh, Haptoglobin phenotype and targeting approaches to neurotrauma, J. Neurotrauma 16 (5) (1999) 377–389.
apolipoprotein E polymorphism: relationship to posttraumatic seizures and [27] L.C. Frey, Epidemiology of posttraumatic epilepsy: a critical review, Epilepsia
neuropsychological functioning after traumatic brain injury, Epilepsy Behav. 44 (Suppl. 10) (2003) 11–17.
16 (3) (2009) 501–506. [28] C.J. Gibson, R.C. Meyer, R.J. Hamm, Traumatic brain injury and the effects of
[2] J.F. Annegers, W.A. Hauser, S.P. Coan, W.A. Rocca, A population-based study of diazepam, diltiazem, and MK-801 on GABA-A receptor subunit expression in
seizures after traumatic brain injuries, N. Engl. J. Med. 338 (1) (1998) 20–24. rat hippocampus, J. Biomed. Sci. 17 (2010) 38.
[3] C.M. Atkins, J.S. Truettner, G. Lotocki, J. Sanchez-Molano, Y. Kang, O.F. Alonso, [29] G. Golarai, A.C. Greenwood, D.M. Feeney, J.A. Connor, Physiological and struc-
T.J. Sick, W.D. Dietrich, H.M. Bramlett, Post-traumatic seizure susceptibility is tural evidence for hippocampal involvement in persistent seizure susceptibility
attenuated by hypothermia therapy, Eur. J. Neurosci. 32 (11) (2010) 1912–1920. after traumatic brain injury, J. Neurosci. 21 (21) (2001) 8523–8537.
[4] L. Belayev, O.F. Alonso, Y. Liu, A.S. Chappell, W. Zhao, M.D. Ginsberg, R. Busto, [30] M.S. Grady, J.S. Charleston, D. Maris, B.M. Witgen, J. Lifshitz, Neuronal and glial
Talampanel, a novel noncompetitive AMPA antagonist, is neuroprotective after cell number in the hippocampus after experimental traumatic brain injury:
traumatic brain injury in rats, J. Neurotrauma 18 (10) (2001) 1031–1038. analysis by stereological estimation, J. Neurotrauma 20 (10) (2003) 929–941.
[5] J. Berg, F. Tagliaferri, F. Servadei, Cost of trauma in Europe, Eur. J. Neurol. 1 [31] A.M. Haltiner, N.R. Temkin, S.S. Dikmen, Risk of seizure recurrence after the first
(2005) 85–90. late posttraumatic seizure, Arch. Phys. Med. Rehabil. 78 (8) (1997) 835–840.
[6] M.M. Blanco, J.G. dos Santos Jr., P. Perez-Mendes, S.R. Kohek, C.F. Cavarsan, [32] R.J. Hamm, B.R. Pike, M.D. Temple, D.M. O’Dell, B.G. Lyeth, The effect of postin-
M. Hummel, C. Albuquerque, L.E. Mello, Assessment of seizure susceptibility jury kindled seizures on cognitive performance of traumatically brain-injured
in pilocarpine epileptic and nonepileptic Wistar rats and of seizure reinduc- rats, Exp. Neurol. 136 (2) (1995) 143–148.
tion with pentylenetetrazole and electroshock models, Epilepsia 50 (4) (2009) [33] D.C. Hesdorffer, G. Logroscino, G. Cascino, J.F. Annegers, W.A. Hauser, Risk of
824–831. unprovoked seizure after acute symptomatic seizure: effect of status epilepti-
[7] T. Bolkvadze, J. Nissinen, I. Kharatishvili, A. Pitkänen, Development of lowered cus, Ann. Neurol. 44 (6) (1998) 908–912.
seizure threshold after lateral fluid-percussion brain injury in mouse, Epilepsia [34] R.C. Hoover, M. Motta, J. Davis, K.E. Saatman, S.T. Fujimoto, H.J. Thompson,
49 (Suppl. 7) (2008) 462. J.F. Stover, M.A. Dichter, R. Twyman, H.S. White, T.K. McIntosh, Differential
[8] T. Bolkvadze, J. Nissinen, I. Kharatishvili, A. Pitkänen, Development of post- effects of the anticonvulsant topiramate on neurobehavioral and histological
traumatic epilepsy in C57BL/6 mice after controlled cortical impact injury, J. outcomes following traumatic brain injury in rats, J. Neurotrauma 21 (5) (2004)
Neurotrauma 26 (8) (2009), A-75. 501–512.
[9] K. Chen, A. Neu, A.L. Howard, C. Földy, J. Echegoyen, L. Hilgenberg, M. Smith, K. [35] A.M. Hudak, K. Trivedi, C.R. Harper, K. Booker, R.R. Caesar, M. Agostini, P.C.
Mackie, I. Soltesz, Prevention of plasticity of endocannabinoid signaling inhibits Van Ness, R. Diaz-Arrastia, Evaluation of seizure-like episodes in survivors of
persistent limbic hyperexcitability caused by developmental seizures, J. Neu- moderate and severe traumatic brain injury, J. Head Trauma Rehabil. 19 (4)
rosci. 27 (1) (2007) 46–58. (2004) 290–295.
[10] M. Chrzaszcz, C. Venkatesan, T. Dragisic, D.M. Watterson, M.S. Wainwright, [36] R.F. Hunt, S.W. Scheff, B.N. Smith, Posttraumatic epilepsy after controlled cor-
Minozac treatment prevents increased seizure susceptibility in a mouse “two- tical impact injury in mice, Exp. Neurol. 215 (2) (2009) 243–252.
hit” model of closed skull traumatic brain injury and electroconvulsive shock- [37] R.F. Hunt, S.W. Scheff, B.N. Smith, Regionally localized recurrent excitation in
induced seizures, J. Neurotrauma 27 (7) (2010) 1283–1295. the dentate gyrus of a cortical contusion model of posttraumatic epilepsy, J.
[11] P.J. Crack, J. Gould, N. Bye, S. Ross, U. Ali, M.D. Habgood, C. Morganti- Neurophysiol. 103 (3) (2010) 1490–1500.
Kossman, N.R. Saunders, P.J. Hertzog, Victorian Neurotrauma Research Group, [38] N. Huusko, C. Roemer, A. Pitkänen, Parvalbumin immunoreactivity in epileptic
The genomic profile of the cerebral cortex after closed head injury in mice: brain: Comparison of SE and TBI as epileptogenic etiology. American Society
effects of minocycline, J. Neural. Transm. 116 (1) (2009) 1–12. for Neuroscience Abstracts, number 241.4.
[12] F.C. Crawford, M. Wood, S. Ferguson, V.S. Mathura, B. Faza, S. Wilson, T. Fan, B. [39] C. Israelsson, H. Bengtsson, A. Kylberg, K. Kullander, A. Lewén, L. Hillered,
O’Steen, G. Ait-Ghezala, R. Hayes, M.J. Mullan, Genomic analysis of response to T. Ebendal, Distinct cellular patterns of upregulated chemokine expression
traumatic brain injury in a mouse model of Alzheimer’s disease (APPsw), Brain supporting a prominent inflammatory role in traumatic brain injury, J. Neu-
Res. 1185 (2007) 45–58. rotrauma 25 (8) (2008) 959–974.
[13] F. Crawford, M. Wood, S. Ferguson, V. Mathura, P. Gupta, J. Humphrey, B. [40] C. Israelsson, Y. Wang, A. Kylberg, C.G. Pick, B.J. Hoffer, T. Ebendal, Closed head
Mouzon, V. Laporte, E. Margenthaler, B. O’Steen, R. Hayes, A. Roses, M. Mullan, injury in a mouse model results in molecular changes indicating inflammatory
Apolipoprotein E-genotype dependent hippocampal and cortical responses to responses, J. Neurotrauma 26 (8) (2009) 1307–1314.
traumatic brain injury, Neuroscience 159 (4) (2009) 1349–1362. [41] H. Karhunen, J. Jolkkonen, J. Sivenius, A. Pitkänen, Epileptogenesis after exper-
[14] R. D’Ambrosio, J.P. Fairbanks, J.S. Fender, D.E. Born, D.L. Doyle, J.W. Miller, Post- imental focal cerebral ischemia, Neurochem. Res. 30 (12) (2005) 1529–1542.
traumatic epilepsy following fluid percussion injury in the rat, Brain 127 (Pt. [42] C.A. Keck, H.J. Thompson, A. Pitkänen, D.G. LeBold, D.M. Morales, J.B. Plevy, R.
2) (2004) 304–314. Puri, B. Zhao, M. Dichter, T.K. McIntosh, The novel antiepileptic agent RWJ-
[15] R. D’Ambrosio, J.S. Fender, J.P. Fairbanks, E.A. Simon, D.E. Born, D.L. Doyle, J.W. 333369-A, but not its analog RWJ-333369, reduces regional cerebral edema
Miller, Progression from frontal-parietal to mesial-temporal epilepsy after fluid without affecting neurobehavioral outcome or cell death following experimen-
percussion injury in the rat, Brain 128 (Pt. 1) (2005) 174–188. tal traumatic brain injury, Restor. Neurol. Neurosci. 25 (2) (2007) 77–90.
[16] E. Dardiotis, K.N. Fountas, M. Dardioti, G. Xiromerisiou, E. Kapsalaki, A. Tasiou, [43] I. Kharatishvili, J. Nissinen, T.K. McIntosh, A. Pitkänen, A model of post-
G.M. Hadjigeorgiou, Genetic association studies in patients with traumatic traumatic epilepsy induced by lateral fluid-percussion brain injury in rats,
brain injury, Neurosurg. Focus. 28 (1) (2010) E9. Neuroscience 140 (2) (2006) 685–697.
[17] P.K. Dash, S.A. Orsi, M. Zhang, R.J. Grill, S. Pati, J. Zhao, A.N. Moore, Val- [44] I. Kharatishvili, R. Immonen, O. Gröhn, A. Pitkänen, Quantitative diffusion MRI
proate administered after traumatic brain injury provides neuroprotection and of the hippocampus as a surrogate marker for posttraumatic epileptogenesis,
improves cognitive function in rats, PLoS One 5 (6) (2010) e11383. Brain 130 (Pt. 12) (2007) 3155–3168.
[18] P.K. Dash, J. Zha, G. Hergenroede, A.N. Moore, Biomarkers for the diagnosis, [45] I. Kharatishvili, A. Sierra, R.J. Immonen, O.H.J. Gröhn, A. Pitkänen, Quantitative
prognosis, and evaluation of treatment efficacy for traumatic brain injury, Neu- T2 mapping as a potential marker for the initial assessment of the severity of
rotherapeutics 7 (1) (2010) 100–114. damage after traumatic brain injury in rat, Exp. Neurol. 217 (2009) 154–164.
[19] R. Diaz-Arrastia, Y. Gong, S. Fair, K.D. Scott, M.C. Garcia, M.C. Carlile, M.A. Agos- [46] I. Kharatishvili, A. Pitkänen, Association of the severity of cortical damage with
tini, P.C. Van Ness, Increased risk of late posttraumatic seizures associated with the occurrence of spontaneous seizures and hyperexcitability in an animal
inheritance of APOE epsilon4 allele, Arch. Neurol. 60 (6) (2003) 818–822. model of posttraumatic epilepsy, Epilepsy Res. 90 (2010) 47–59.
[20] J. Echegoyen, C. Armstrong, R.J. Morgan, I. Soltesz, Single application of a CB1 [47] N. Kobori, G.L. Clifton, P. Dash, Altered expression of novel genes in the cere-
receptor antagonist rapidly following head injury prevents long-term hyper- bral cortex following experimental brain injury, Brain Res. Mol. Brain Res. 104
excitability in a rat model, Epilepsy Res. 85 (1) (2009) 123–127. (2002) 148–158.
A. Pitkänen et al. / Neuroscience Letters 497 (2011) 163–171 171

[48] J.A. Langlois, W. Rutland-Brown, K.E. Thomas, Traumatic brain injury in the [72] A. Pitkänen, Therapeutic approaches to epileptogenesis – hope on the horizon,
United States: emergency department visits, hospitalizations, and deaths, Cen- Epilepsia 51 (Suppl. 3) (2010) 2–17.
terd for Disease Control and Prevention, National Center for Injury Prevention [73] J.M. Rall, D.A. Matzilevich, P.K. Dash, Comparative analysis of mRNA levels in the
and Control, Atlanta, 2006. frontal cortex and the hippocampus in the basal state and in response to exper-
[49] J.D. Levine, J.T. Davis, E.D. Bigler, R. Thoma, D. Hill, M. Funke, J.H. Sloan, S. Hall, imental brain injury, Neuropathol. Appl. Neurobiol. 29 (2) (2003) 118–131.
W.W. Orrison, Objective documentation of traumayic brain injury subsequent [74] R. Scheid, D.Y. von Cramon, Clinical findings in the chronic phase of traumatic
to mild head trauma: multimodal brain imaging with MEG, SPECT, and MRI, J. brain injury: data from 12 years’ experience in the Cognitive Neurology Out-
Head. Trauma Rehabil. 22 (3) (2007) 141–155. patient Clinic at the University of Leipzig, Dtsch. Arztebl. Int. 107 (12) (2010)
[50] H.H. Li, S.M. Lee, Y. Cai, R.L. Sutton, D.A. Hovda, Differential gene expres- 199–205.
sion in hippocampus following experimental brain trauma reveals distinct [75] P.A. Schwartzkroin, H.J. Wenzel, B.G. Lyeth, C.C. Poon, A. Delance, K.C. Van, L.
features of moderate and severe injuries, J. Neurotrauma 21 (9) (2007) Campos, D.V. Nguyen, Does ketogenic diet alter seizure sensitivity and cell loss
1141–1153. following fluid percussion injury? Epilepsy Res. 92 (1) (2010) 74–84.
[51] Y. Long, L. Zou, H. Liu, H. Lu, X. Yuan, C.S. Robertson, K. Yang, Altered expression [76] R.P. Shank, J.F. Gardocki, A.J. Streeter, B.E. Maryanoff, An overview of the preclin-
of randomly selected genes in mouse hippocampus after traumatic brain injury, ical aspects of topiramate: pharmacology, pharmacokinetics, and mechanism
J. Neurosci. Res. 71 (5) (2003) 710–720. of action, Epilepsia 41 (Suppl. 1) (2000) S3–9.
[52] D.H. Lowenstein, M.J. Thomas, D.H. Smith, T.K. McIntosh, Selective vulnerability [77] D.C. Smith, A.A. Tan, A. Duke, S.L. Neese, R.W. Clough, R.A. Browning, R.A. Jensen,
of dentate hilar neurons following traumatic brain injury: a potential mecha- Recovery of function after vagus nerve stimulation initiated 24 hours after fluid
nistic link between head trauma and disorders of the hippocampus, J. Neurosci. percussion brain injury, J. Neurotrauma 23 (10) (2006) 1549–1560.
12 (12) (1992) 4846–4853. [78] D.H. Smith, K. Okiyama, T.A. Gennarelli, T.K. McIntosh, Magnesium and
[53] A.I.R. Maas, N. Stocchetti, R. Bullock, Moderate and severe traumatic brain injury ketamine attenuate cognitive dysfunction following experimental brain injury,
in adults, Lancet Neurol. 7 (2008) 728–741. Neurosci. Lett. 157 (2) (1993) 211–214.
[54] Q. Mao, F. Jia, X.H. Zhang, Y.M. Qiu, J.W. Ge, W.J. Bao, Q.Z. Luo, J.Y. Jiang, The [79] D.H. Smith, B.R. Perri, R. Raghupathi, K.E. Saatman, T.K. McIntosh, Remacemide
up-regulation of voltage-gated sodium channel NaV1.6 expression following hydrochloride reduces cortical lesion volume following brain trauma in the rat,
fluid percussion traumatic brain injury in rats, Neurosurgery 66 (6) (2010) Neurosci. Lett. 231 (3) (1997) 135–138.
1134–1139. [80] D.H. Smith, X.H. Chen, J.E. Pierce, J.A. Wolf, J.Q. Trojanowski, D.I. Graham, T.K.
[55] N. Marklund, A. Bakshi, D.J. Castelbuono, V. Conte, T.K. McIntosh, Evaluation of McIntosh, Progressive atrophy and neuron death for one year following brain
pharmacological treatment strategies in traumatic brain injury, Curr. Pharm. trauma in the rat, J. Neurotrauma 14 (10) (1997) 715–727.
Des. 12 (13) (2006) 1645–1680. [81] K.D. Statler, S. Swank, T. Abildskov, E.D. Bigler, H.S. White, Traumatic brain
[56] G.W. Mathern, J.K. Pretorius, T.L. Babb, Influence of the type of initial precipi- injury during development reduces minimal clonic seizure thresholds at matu-
tating injury and at what age it occurs on course and outcome in patients with rity, Epilepsy Res. 80 (2–3) (2008) 163–170.
temporal lobe seizures, J. Neurosurg. 82 (2) (1995) 220–227. [82] K.D. Statler, P. Scheerlinck, W. Pouliot, M. Hamilton, H.S. White, F.E. Dudek,
[57] D.A. Matzilevich, J.M. Rall, A.N. Moore, R.J. Grill, P.K. Dash, High-density A potential model of pediatric posttraumatic epilepsy, Epilepsy Res. 86 (2–3)
microarray analysis of hippocampal gene expression following experimental (2009) 221–223.
brain injury, J. Neurosci. Res. 67 (5) (2002) 646–663. [83] B.E. Swartz, C.R. Houser, U. Tomiyasu, G.O. Walsh, A. DeSalles, J.R. Rich, A.
[58] T.K. McIntosh, R. Vink, I. Yamakami, A.I. Faden, Magnesium protects against Delgado-Escueta, Hippocampal cell loss in posttraumatic human epilepsy,
neurological deficit after brain injury, Brain Res. 482 (2) (1989) 252–260. Epilepsia 47 (8) (2006) 1373–1382.
[59] D.B. Michael, D.M. Byers, L.N. Irwin, Gene expression following traumatic brain [84] N.R. Temkin, G.D. Anderson, H.R. Winn, R.G. Ellenbogen, G.W. Britz, J. Schuster,
injury in humans: analysis by microarray, J. Clin. Neurosci. 12 (3) (2005) T. Lucas, D.W. Newell, P.N. Mansfield, J.E. Machamer, J. Barber, S.S. Dikmen,
284–290. Magnesium sulfate for neuroprotection after traumatic brain injury: a ran-
[60] D.M. Morales, N. Marklund, D. Lebold, H.J. Thompson, A. Pitkänen, W.L. domised controlled trial, Lancet Neurol. 6 (1) (2007) 29–38.
Maxwell, L. Longhi, H. Laurer, M. Maegele, D.I. Graham, N. Stocchetti, T.H. McIn- [85] N.R. Temkin, Preventing and treating posttraumatic seizures: the human expe-
tosh, Experimental models of traumatic brain injury: do we really need to build rience, Epilepsia 50 (Suppl. 2) (2009) 10–13.
a better mousetrap? Neuroscience 136 (4) (2005) 971–989. [86] O. Tomkins, I. Shelef, I. Kaizerman, A. Eliushin, Z. Afawi, A. Misk, M. Gidon,
[61] Z. Mtchedlishvili, E. Lepsveridze, H. Xu, E.A. Kharlamov, B. Lu, K.M. Kelly, A. Cohen, D. Zumsteg, A. Friedman, Blood-brain barrier disruption in post-
Increase of GABAA receptor-mediated tonic inhibition in dentate granule cells traumatic epilepsy, J. Neurol. Neurosurg. Psychiatry 79 (7) (2008) 774–777.
after traumatic brain injury, Neurobiol. Dis. 38 (3) (2010) 464–475. [87] Z. Toth, G.S. Hollrigel, T. Gorcs, I. Soltesz, Instantaneous perturbation of den-
[62] L. Munoz, H.R. Ranaivo, S.M. Roy, W. Hu, J.M. Craft, L.K. McNamara, L.W. Chico, tate interneuronal networks by a pressure wave-transient delivered to the
L.J. Van Eldik, D.M. Watterson, A novel p38 alpha MAPK inhibitor suppresses neocortex, J. Neurosci. 17 (21) (1997) 8106–8117.
brain proinflammatory cytokine up-regulation and attenuates synaptic dys- [88] P.M. Vespa, C. Miller, D. McArthur, M. Eliseo, M. Etchepare, D. Hirt, T.C. Glenn,
function and behavioral deficits in an Alzheimer’s disease mouse model, J. N. Martin, D. Hovda, Nonconvulsive electrographic seizures after traumatic
Neuroinflammation 4 (2007) 21. brain injury result in a delayed, prolonged increase in intracranial pressure
[63] H. Mynter III, Contribution to the study of head injuries, and of the results of and metabolic crisis, Crit. Care Med. 35 (12) (2007) 2830–2836.
trephining for subdural haemorrhage, abscess of brain, and epilepsy, Ann. Surg. [89] A. Vezzani, G. Civenni, M. Rizzi, A. Monno, S. Messali, R. Samanin, Enhanced
19 (5) (1894) 539–545. neuropeptide Y release in the hippocampus is associated with chronic seizure
[64] G. Navarro Mora, P. Bramanti, F. Osculati, A. Chakir, E. Nicolato, P. Marzola, A. susceptibility in kainic acid treated rats, Brain Res. 660 (1) (1994) 138–143.
Sbarbati, P.F. Fabene, Does pilocarpine-induced epilepsy in adult rats require [90] A. Von Gertten, A. Flores Morales, S. Holmin, T. Mathiesen, A.C. Nordqvist,
status epilepticus? PLoS One 4 (6) (2009) e5759. Genomic responses in rat cerebral cortex after traumatic brain injury, BMC
[65] J. Nissinen, R. Immonen, T. Stöhr, O. Gröhn, A. Pitkänen, Effect of lacosamide Neurosci. 6 (2005) 69.
on structural and functional recovery after traumatic brain injury in rats, in: R. [91] A.E. Walker, F.A. Quadfasel, C. Marshall, M.G. Netsky, R.G. Fisher, I.C. Kaufman,
Kälviäinen (Ed.), Proceedings from the 7th European Congress on Epileptology, J.W. Gerhardt, E.N. Beresford, Problems in posttraumatic epilepsy, J. Nerv. Ment.
Blackwell Publishing, Oxford, 2006, p. 87. Dis. 105 (6) (1947) 673–678.
[66] I. Pavlov, N. Huusko, J. Nissinen, G. Sperk, A. Pitkänen, M.C. Walker, Phasic [92] N.F. Watson, J.K. Barber, M.J. Doherty, J.W. Miller, N.R. Temkin, Does glucocor-
but not tonic inhibition is decreased in dentate gyrus granule cells in fluid- ticoid administration prevent late seizures after head injury? Epilepsia 45 (6)
percussion brain injury model of posttraumatic epilepsy in rats. American (2004) 690–694.
Society for Neuroscience Abstracts, number 241.3/I10. [93] H.H. Wei, X.C. Lu, D.A. Shear, A. Waghray, C. Yao, F.C. Tortella, J.R. Dave, NNZ-
[67] A. Pitkänen, T.P. Sutula, Is epilepsy a progressive disorder? Prospects for new 2566 treatment inhibits neuroinflammation and pro-inflammatory cytokine
therapeutic approaches in temporal-lobe epilepsy, Lancet Neurol. 1 (3) (2002) expression induced by experimental penetrating ballistic-like brain injury in
173–181. rats, J. Neuroinflammation 6 (2009) 19.
[68] A. Pitkänen, L. Longhi, N. Marklund, D. Morales, T.K. McIntosh, Mechanisms [94] B.M. Witgen, J. Lifshitz, M.S. Grady, Inbred mouse strains as a tool to analyze hip-
of neuronal death and neuroprotective strategies after traumatic brain injury, pocampal neuronal loss after brain injury: a stereological study, J. Neurotrauma
Drug Discov. Today: Dis. Mech. 2 (4) (2005) 409–418. 23 (9) (2006) 1320–1329.
[69] A. Pitkänen, T.K. McIntosh, Animal models of post-traumatic epilepsy, J. Neu- [95] K. Yoshiya, H. Tanaka, K. Kasai, T. Irisawa, T. Shiozaki, H. Sugimoto, Profile
rotrauma 23 (2) (2006) 241–261. of gene expression in the subventricular zone after traumatic brain injury, J.
[70] A. Pitkänen, K. Lukasiuk, Molecular and cellular basis of epileptogenesis in Neurotrauma 20 (11) (2003) 1147–1162.
symptomatic epilepsy, Epilepsy Behav. 14 (Suppl. 1) (2009) 16–25. [96] I. Yu, M. Inaji, J. Maeda, T. Okauchi, T. Nariai, K. Ohno, M. Higuchi, T. Suhara, Glial
[71] A. Pitkänen, R.J. Immonen, O.H. Gröhn, I. Kharatishvili, From traumatic brain cell-mediated deterioration and repair of the nervous system after traumatic
injury to posttraumatic epilepsy: what animal models tell us about the process brain injury in a rat model as assessed by positron emission tomography, J.
and treatment options, Epilepsia 50 (Suppl. 2) (2009) 21–29. Neurotrauma (2010) [Epub ahead of print, May 26].

You might also like