You are on page 1of 8

Epilepsy & Behavior 51 (2015) 191–198

Contents lists available at ScienceDirect

Epilepsy & Behavior

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

Comparison of short-term effects of midazolam and lorazepam in the


intra-amygdala kainic acid model of status epilepticus in mice
Mairead Diviney, James P. Reynolds, David C. Henshall ⁎
Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland

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

Article history: Benzodiazepines remain as the first-line treatment for status epilepticus (SE), but debate continues as to the
Received 26 June 2015 choice and delivery route of pharmacotherapy. Lorazepam is currently the preferred anticonvulsant for clinical
Revised 27 July 2015 use, but midazolam has become a popular alternative, particularly as it can be given by nonintravenous routes.
Accepted 28 July 2015
Anticonvulsants are also commonly used to terminate SE in animal models. Here, we aimed to compare the effi-
Available online 24 August 2015
cacy of midazolam with that of lorazepam in an experimental model of focal-onset SE. Status epilepticus was in-
Keywords:
duced by intra-amygdala microinjection of kainic acid in 8 week old C57Bl/6 mice. Forty minutes later, mice were
Status epilepticus treated with an intraperitoneal injection of either lorazepam or midazolam (8 mg/kg). Electroencephalogram
Anticonvulsants (EEG) activity, histology, and behavioral tests assessing recovery of function were evaluated and compared be-
Midazolam tween groups. Intraperitoneal injection of either lorazepam or midazolam resulted in similar patterns of reduced
Animal welfare EEG epileptiform activity during 1-hour recordings. Damage to the hippocampus and presentation of postinsult
3Rs (Replacement, Refinement, Reduction) anxiety-related behavior did not significantly differ between treatment groups at 72 h. However, return of nor-
Behavior mal behaviors such as grooming, levels of activity, and the evaluation of overall recovery of SE mice were all su-
perior at 24 h in animals given midazolam compared with lorazepam. Our results indicate that midazolam is as
effective as lorazepam as an anticonvulsant in this model while also providing improved animal recovery after SE.
These data suggest that midazolam might be considered by researchers as an anticonvulsant in animal models of
SE, particularly as it appears to satisfy the requirements of refining procedures involving experimental animals at
early time-points after SE.
© 2015 Elsevier Inc. All rights reserved.

1. Introduction the route of administration, where intravenous delivery is not always


practical.
Status epilepticus (SE) is a life-threatening neurological emergency The cellular and molecular mechanisms by which SE develops are in-
and is regarded as one of the most extreme and severe forms of seizure completely understood but are thought to be from a combination of ex-
activity. It is defined as a prolonged seizure and, operationally, as five cessive excitatory and impaired inhibitory neuronal mechanisms [8].
minutes of continuous seizure activity or as two or more seizures with- Current treatment, therefore, includes administration of benzodiaze-
out complete recovery between seizure episodes [1]. Status epilepticus pines which act by potentiating GABAergic inhibition [9]. The most
is associated with a risk of significant damage involving brain structures widely used agents for first line therapies for SE in the clinical environ-
such as the hippocampus, cognitive impairment, and epileptogenesis ment are. lorazepam and diazepam [10]. Lorazepam remains the phar-
[2,3]. Status epilepticus is also associated with high morbidity and mor- macotherapy of choice as it has fewer contraindications, such as lower
tality [4], and a critical variable in these outcomes is the duration of the incidence of respiratory depression [11]. The mode of delivery of these
SE period [5]. Specifically, the longer that SE persists, the more likely it is drugs is via intravenous (i.v.) administration, and while effective, estab-
to become unresponsive to drug therapy, resulting in poor patient out- lishing i.v. access in a patient having a seizure can be difficult and time-
comes [6]. As such, SE requires prompt treatment using anticonvulsant consuming [12]. These treatments can be administered intramuscularly
drugs [7]. However, there remains a debate regarding the most effective (i.m.); however, they are absorbed more slowly [8], and due to their
prehospital treatment regimen with a particular consideration being vehicle (i.e., propylene glycol), irritation at the injection site can occur.
An alternative benzodiazepine, midazolam, can be safely administered
i.m. enabling quick and accurate delivery. It has also been shown to be
Abbreviations: SE, status epilepticus; EEG, electroencephalogram. highly effective at terminating seizures, both when given as an initial
⁎ Corresponding author at: Department of Physiology and Medical Physics, Royal
College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland. Tel.: +353 1
agent for SE and when given as second line therapy for refractory
402 8629. SE [13,14]. Recent evidence has also emerged to suggest that the anti-
E-mail address: dhenshall@rcsi.ie (D.C. Henshall). seizure efficacy of midazolam for SE is equivalent to lorazepam in

http://dx.doi.org/10.1016/j.yebeh.2015.07.038
1525-5050/© 2015 Elsevier Inc. All rights reserved.
192 M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198

prehospital settings [12,15,16]. Patients treated with midazolam also Ireland). Baseline EEG was established over a 10-minute recording
had better short-term outcomes, with a significantly lower number of period. Following this, the animal was lightly restrained while a
patients requiring hospital admittance than those treated with loraze- 31-gauge injection cannula was lowered 3.75 mm below the brain sur-
pam [12]. Lorazepam, however, has often been chosen over midazolam face for injection of KA [0.3 μg in 0.2-microliter vehicle (phosphate-
in clinical treatment as it has a longer duration of action, with one of the buffered saline (PBS)), pH adjusted to 7.4; Sigma-Aldrich] into the
major disadvantages of midazolam being tachyphylaxis, resulting in the basolateral amygdala nucleus to induce SE. After 40 min, mice were
need for increased dosing to maintain seizure control [17]. Furthermore, administered with either lorazepam or midazolam (see Section 2.2).
with prolonged infusion, midazolam begins to accumulate, resulting in Electroencephalogram was recorded for a further 1 h thereafter before
a protracted time to consciousness [18]. Therefore, it can be difficult animals were disconnected and placed in a warmed recovery chamber.
to reconcile the need for immediacy of treatment with longer term out- Mice were sacrificed 72 h after anticonvulsant administration by pento-
comes, adding further complexity to decisions around anticonvulsant barbital overdose and perfused with ice-cold PBS to remove intravascu-
treatment. lar blood components. Whole brains were flash-frozen in 2-
Animal models of SE are widely used to study epilepsy and methylbutane (at −30 °C) and stored at −80 °C for cryostat sectioning
epileptogenesis and are crucial in the development of effective thera- (Fig. 1B).
peutics [19,20]. Antiseizure drugs are often administered to reduce sei-
zures during SE and minimize morbidity and mortality. Indeed, the 2.2. Drug treatments
timing of benzodiazepine administration has time-dependent effects
on attendant brain damage and epilepsy development [21]. The choice For anticonvulsant treatment, animals were randomized to either
of anticonvulsant is, therefore, important for ensuring satisfactory lorazepam (LZ, 8 mg/kg) or midazolam (MDZ, 8 mg/kg; n = 6/group).
interanimal reproducibility in seizure suppression and postinsult recov- Drugs were delivered via a single intraperitoneal injection in 0.25-
ery. Commonly, either lorazepam [22,23] or diazepam [24] is adminis- milliliter volume of either PBS (lorazepam) or sterile H2O (midazolam),
tered via i.v. or intraperitoneal (i.p.) injection in models of chemically 40 min after intra-amygdala KA injection.
induced SE in rodents. Little is known, however, about the performance
of midazolam in rodent SE models. A small number of studies have
reported that midazolam provides protective effects against pentylene- 2.3. Recovery and behavior scoring
tetrazol (PTZ)-induced seizures and also results in a reduction in overall
time spent in seizure [25,26]. Others have focused on the pharmacody- Following SE, mice were housed individually for behavior assess-
namics of midazolam in various epilepsy models [27]. Differing behav- ments. To determine the effect of treatment on recovery, mice were
ioral profiles in parameters such as spontaneous motor activity and observed at 4 h, 24 h, 48 h and 72 h postanticonvulsant treatment.
muscle relaxation have also been noted in naïve mice following treat- Measurements included weight, with baseline weight measured imme-
ment with four benzodiazepines, suggesting that drug type may influ- diately postsurgery — this was to account for additional head-piece
ence recovery [28]. weight (i.e., EEG electrode, cannula, and cement assembly) — and
For many years, our group has used systemic injection of diazepam were monitored at each time-point post-SE. Behaviors observed fell
or lorazepam as anticonvulsants in the intra-amygdala kainic acid into a number of distinct categories including: 1) well-being scoring,
(KA) model in mice [22,29]. Here, we aimed to compare the effect of 2) activity-related behavior, 3) grooming behavior, and 4) attentive
midazolam with that of lorazepam as an alternative anticonvulsant behavior. A modified welfare score sheet which comprised of four cate-
in this model, focusing on EEG, hippocampal injury, and recovery of gories including appearance, locomotion, unprovoked behavior, and
normal behavior patterns in an experimental mouse model of SE. behavior to external stimuli was used (see Table 1). These categories
are modified from Kirsch et al. [31] and Roughan and Flecknell [32].
2. Materials and methods Subscores for each category were determined, and the total, overall
score was calculated by adding all subscores. Scores were interpreted
2.1. Seizure model as: 0 to 3, normal; 4 to 7, animal requires careful monitoring; 8 to 11, an-
imal appears in distress and requires regular observation; and 12 to 15,
All animal experiments were performed in accordance with the severe distress [31]. Activity, grooming, and attentive behavior were
European Union Directive (2010/63/EU) and were reviewed and ap- scored in relation to the number of occurrences within a 10-minute ob-
proved by the Research Ethics Committee of the Royal College of Sur- servation period. Activity-related behaviors included ambulation, head
geons in Ireland, under license from the Health Products Regulatory turns, drinking, eating, and digging, climbing, and biting bars. Grooming
Authority, Ireland (AE19127/P001; AE19127/I089). All experiments ob- included licking fur, washing face, scratching body, and licking paws.
served the principles outlined in the ARRIVE guidelines and the Basel Attentive behavior included sniffing, high rearing, and low rearing.
declaration including adherence to the 3Rs (Replacement, Refinement, The observer scoring all behaviors was blinded to treatment group.
Reduction). Adult (20–25 g) male C57BL/6 mice were purchased from
Harlan (UK). Animals were housed in a vivarium on a 12-hour light/ 2.4. Exploratory and anxiety-related behaviors
dark cycle with access to food and water ad libitum. Mice were anesthe-
tized using isoflurane (3–5%) in oxygen and placed in a mouse- Seventy-two hours after SE, exploratory behavior and anxiety were
adapted stereotaxic frame. Following a midline scalp incision, bregma assessed in mice from both treatment groups using the open-field task
was located and three partial craniectomies were performed for the [33,34]. Each mouse was placed individually in the center of a clear,
placement of skull-mounted recording screws (Bilaney Consultants) plexiglass open-field apparatus (ENV-510; 27.9 × 27.9 cm; Med
to record EEG (see Fig. 1A). A fourth craniectomy was drilled for Associates) and allowed to freely explore for 10 min. Arenas were
the placement of a 26-gauge steel guide cannula (coordinates from wiped with 70% ethanol before each session and between mice. An
bregma; AP = − 0.95 mm, L = − 2.85 mm) based on a stereotaxic overhead camera captured all animal movements, and video recordings
atlas [30]. The cannula and electrode assembly were fixed in place were later analyzed using EthoVision videotracking (Noldus Informa-
using dental cement (Plastics One, Inc.), anesthesia was discontinued, tion Technologies, Wageningen, Netherlands). To assess exploratory be-
and mice were placed in an incubator until they had recovered and havior, distance traveled (cm) and velocity (cm/s) were measured.
were freely moving. For EEG recordings, mice were placed in a clear Anxiety was determined by measuring time spent in either a demarcat-
Perspex recording chamber which allowed free movement. The EEG ed central zone or peripheral zone of the arena. Prior to behavior testing,
was recorded using a Grass Comet digital EEG (Medivent Ltd, Lucan, mice were allowed to acclimatize to the testing environment for 1 h
M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198 193

Fig. 1. Electroencephalogram and status epilepticus in mice treated with LZ or MDZ. (A) Schematic showing cannula and electrode implantation site. (B) Experimental design illustrating
time of intra-amygdala KA injection followed by 40-minute recording post-KA and a further 60-minute recording after anticonvulsant treatment. (C) Representative EEG traces depicting
electrographic seizure development following KA and also after treatment with either LZ or MDZ. Below, an EEG heat map depicting typical frequency–amplitude data during status
epilepticus (SE) and after anticonvulsant administration. Electroencephalogram recordings show no differences between mice in LZ (n = 6) or MDZ (n = 6) treated groups during SE
in (D) total power or (E) amplitude during SE. Antiseizure treatment type also does not result in significant alterations in (F) total power or (G) amplitude during the 60 min after admin-
istration of either LZ or MDZ.

prior to experimentation. The observer was blind to the treatment of followed by a rinse, and then stained with 0.001% w/v FJB (Millipore
the mice. Ireland B.V., Tullagreen, Ireland). Staining was examined using an
epifluorescence microscope (Nikon 2000s) under Ex/Em wavelengths
of 472/520 nm (green), and positive cells within the hippocampus
2.5. Histopathology
were counted in a blinded fashion under 20× magnification. Represen-
tative images were obtained using an Orca 285 camera and processed
Brains (n = 6 per group) were sectioned at 12 μm on a cryostat
using Wasabi software (Hamamatsu Photonics Germany GmbH,
(Leica) in the coronal plane and stored at −80 °C until further use. For
Herrsching, Germany). Images were converted to grayscale and
detection of neurodegeneration, sections at the level of the dorsal hip-
inverted such that degenerated neurons appeared dark on a light back-
pocampus (−1.7 mm) [30] were processed for Fluorojade B (FJB) [35]
ground. Hippocampal FJB counts were carried out on areas CA3, CA1,
(Millipore, Cork, Ireland). Flash-frozen tissue sections were postfixed,
and the hilus and taken as the mean of two adjacent sections. The ob-
dehydrated, immersed in 0.06% potassium permanganate solution
server quantifying FJB positive cells was blinded to treatment group.

2.6. Quantification of EEG


Table 1
Well-being recovery evaluation scoring.
Electroencephalogram data were exported to Labchart 7 (AD Instru-
Category Score Operational Definition ments Ltd, Oxford, UK) for analysis. Baseline EEG was recorded for
Appearance 0 ○ Normal, haircoat smooth, flat with sheen, eyes clear 10 min for each animal. The effects of drug treatments on EEG were
1 ○ Ruffled fur, lack of grooming, lubricant in eyes determined for the 60-minute recording period after injection. Mean
2 ○ Rough hair, pointy, hunched look, eye and nose discharge
amplitude and total power were calculated, and measurements were
3 ○ Ungroomed, abnormal posture, eyes glazed, animal
appears severely depressed normalized to baseline for each animal. The total power was automati-
Locomotion 0 ○ Walking normally cally measured by the software with the default frequency range of
1 ○ Limping, stiffness 0–500 Hz. Quantification of EEG was performed by an observer blinded
2 ○ Swollen limbs to treatment. Treatment group was revealed after all analyses were
3 ○ Severely restricted mobility
Unprovoked 0 ○ Normal — exploring cage, grooming, feeding, bright, alert
complete and only revealed at the time when statistical comparisons
behavior & responsive were conducted.
1 ○ Altered slightly, huddled
2 ○ Abnormal behavior, reduced mobility, alertness or 2.7. Data analysis
responsiveness, inactive, lame, aggressive or huddled
3 ○ Severe distress, vocalizations, restless/no movement,
unresponsive, twitching, lameness All data are presented as mean ± standard error of the mean (SEM).
Behavior to 0 ○ Normal response to reaching into and tapping cage Two group comparisons were made using independent t-tests, while
external 1 ○ Shows minor exaggerated responses repeated measures analyses were made using analysis of variance
stimuli 2 ○ Moderate abnormalities, more aggressive or docile (ANOVA) followed by appropriate post hoc testing. Significance was
3 ○ Overreaction to stimuli or nonresponsive
accepted at p b 0.05.
194 M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198

3. Results groups in the ipsilateral CA1 (t(10) = 0.036, p = 0.94) and the hilus
(t(10) = 0.08, p = 0.93; Fig. 2B, C). There were also no differences
All mice used in this study (n = 12; n = 6 LZ; n = 6 MDZ) survived observed in any subregion of the contralateral hippocampus (see repre-
SE induced by intra-amygdala KA, and all reached the planned end- sentative image Fig. 2D).
point at 72 h post-SE.
3.3. Midazolam improves speed of recovery after SE
3.1. Effect of anticonvulsant treatment on EEG after SE
We next examined a number of behavioral parameters to determine
Electroencephalogram was recorded for all mice for 10 min prior any changes in the recovery and well-being of mice following the termi-
to injection of KA and referenced as a baseline (100%) to determine nation of SE. Mice were assessed for weight, alongside a number of
changes in EEG during and after SE. Following intra-amygdala injection other parameters which would indicate the occurrence of any alter-
of KA, all mice entered SE with no differences in EEG total power be- ations as a result of anticonvulsant treatment on recovery from SE.
tween the mice later treated with lorazepam (LZ) (587.7 ± 149.0%) or The postsurgical weight of mice was recorded at baseline and change
midazolam (MDZ) (648.1 ± 129.4%; t(10) = 0.31, p = 0.76) or in am- in weight was measured at 4 h, 24 h, 48 h, and 72 h after SE. A repeated
plitude between the groups (LZ: 186.6 ± 18.97; MDZ: 214.5 ± 26.66%; measures ANOVA revealed an overall reduction in weight for both
t(10) = 0.85, p = 0.41; Fig. 1C–E). This would indicate that all groups across time (F(4, 40) = 12.76, p b 0.001), with a significant re-
mice, prior to random assignment to treatment group, experienced duction from baseline to 4 h (p b 0.001), 24 h (p b 0.001), and 48 h
similar SE. To assess the effect of drug treatment on the reduction of (p b 0.002). However, there was no difference in weight between treat-
EEG activity after SE, total power and mean amplitude were analyzed ment groups overall (F(1, 10) = 0.038, p = 0.85; Fig. 3A).
for a 60-minute period following anticonvulsant drug administra- We next sought to determine if there were changes in recovery
tion. No differences were observed in EEG total power between LZ evaluation scores (see Table 1). Mice were observed over a 10-minute
(398.0 ± 122.9%) and MDZ (328.5 ± 127.2%) treated mice (t(10) = period in their home cages at each of the four time points. A repeated
0.54, p = 0.59). Similarly, analysis of mean amplitude also indicates measures ANOVA indicates an overall improvement in general recovery
that treatment with MDZ (163.9 ± 6.38%) did not alter the pattern scores over time (F(3, 30) = 96.75, p b 0.001). There was no difference
of seizure termination when compared with LZ treatment (173.2 ± in recovery between groups (F(1, 10) = 2.73, p = 0.13); however, a sig-
27.46%; t(10) = 0.33, p = 0.75; Fig. 1F, G). nificant interaction effect was found for time × group (F(3, 30) = 5.57,
p b 0.01). Further analysis revealed that MDZ-treated mice displayed
3.2. Effect of anticonvulsant treatment on neurodegeneration after SE significant improvements at 24 h in comparison to LZ-treated mice
(p b 0.05; Fig. 3B). Grooming, a good indicator of normal behavior
To determine whether there were differences in hippocampal dam- in mice, also improved over time for both groups (F(3, 30) = 14.03,
age between the MDZ and LZ groups, we next examined hippocampal p b 0.001), and while there was no overall effect for group (F(1,
damage in tissue sections from mice from each treatment group, 72 h 10) = 3.09, p = 0.109), there was a significant time × group interaction
after SE. Semiquantitative counts of FJB positive cells in ipsilateral CA3 effect (F(3, 30) = 6.31, p b 0.01). Crucially, further analysis suggests that
indicated no significant difference between LZ-treated (62.75 ± 6.9) MDZ had a significant impact on initial recovery after intra-amygdala
and MDZ-treated mice (76.17 ± 15.92; t(10) = 0.77, p = 0.45; injection of KA, with a significantly higher level of grooming behavior
Fig. 2A). Similarly, no differences in FJB counts were noted between in MDZ than LZ mice 24 h after SE (p b 0.05; see Fig. 3C). Similarly, the

Fig. 2. Hippocampal damage 72 h after status epilepticus in LZ-treated and MDZ-treated mice. FJB-positive cell counts in the subfields of the ipsilateral hippocampus 72 h after SE
(n = 6/group). Treatment type did not significantly alter the level of neuronal death in ipsilateral (A) CA3, (B) CA1, or in the (C) hilus. (D) Representative photomicrographs of FJB-stained
neurons in the ipsilateral (right) and contralateral (left) hippocampi at 72 h for each group. Note the little damage evident in the contralateral hippocampus with no differences between
groups in the contralateral area CA3 (p = 0.41), CA1 (p = 0.27) and the hilus (p = 0.50). Scale bar: 500 μm.
M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198 195

Fig. 3. Welfare and typical behavior assessment of mice treated with LZ and MDZ. (A) There was an initial reduction in weight 4 h and 24 h post-SE; however, treatment had no effect
on weight change. (B) Improvements in welfare were observed across days in all mice. Midazolam-treated mice display improved typical behaviors and appearance at 24 h in comparison
to LZ-treated mice. (C) Significant improvements in levels of activity across time in all mice were noted. Again, significantly higher active behavior was seen in mice treated with
MDZ. (D) Patterns of grooming behavior indicate increased activity at 24 h for MDZ mice when compared with LZ mice. (E) The ability of mice to attend to environmental cues also sig-
nificantly improved over time. Again, treatment with MDZ resulted in a higher level of occurrence of this behavior when compared with LZ treatment at 24 h (p b 0.001). *p b 0.05,
**p b 0.01, ***p b 0.001.

levels of activity-related behavior increased significantly over the 72 h 3.4. Exploratory and anxiety-related behaviors
after treatment for both groups (F(3, 30) = 14.43, p b 0.01) with a sig-
nificant difference in the level of activity between treatment groups Seventy-two hours after SE, exploratory behavior and anxiety were
(F(1, 10) = 5.11, p b 0.05). Further analysis indicates that MDZ mice assessed in mice using the open-field task to determine if the treat-
were, again, significantly more active than LZ mice at 24 h (p b 0.001; ment had any gross effects on animal behavior. To assess exploratory
Fig. 3D), displaying higher occurrences of ambulatory behavior, head- behavior, distance traveled (cm) was measured with no differences
turns, climbing, eating, and drinking. The occurrence of attentive behav- noted between LZ (2582 ± 545.1 cm) and MDZ (2845 ± 588.6 cm)
iors, such as high and low rearing and sniffing also increased for both treated mice (t(10) = 0.87, p = 0.74; Fig. 4B). Similarly, assessment
groups at each time point subsequent to KA injection (F(3, 30) = of velocity (cm/s) in the arena identified no significant differences
16.01, p b 0.001). Treatment did not appear to have an overall effect between groups (LZ: 4.23 ± 0.95 cm/s; MDZ: 4.72 ± 0.99 cm/s;
on the level of attentive behaviors (F(1, 10) = 2.01, p = 0.187); how- t(10) = 0.36, p = 0.72, Fig. 4C) suggesting that anticonvulsant treat-
ever, a significant interaction effect was noted between group and time ment type did not differentially affect exploratory behaviors in
after SE (F(3, 30) = 3.03, p b 0.05), with MDZ-treated mice showing sig- the open-field task. Anxiety was assessed by measuring time spent in
nificant improvements over LZ mice at 24 h after treatment (p b 0.001; either a demarcated central zone or peripheral zone of the open-field
Fig. 3E). On all behavior measures, differences between treatment arena. Animals normally spend greater time exploring the periphery,
groups were no longer present after the 24-hour time-point as mice and this was found for both groups (Fig. 4D). There was no significant
continued to recover over the subsequent 48 h. difference in total time spent in the peripheral zone between LZ

Fig. 4. Exploratory and anxiety-related behaviors in mice treated with lorazepam and midazolam. (A) Representative image of the open-field and demarcated peripheral and central zones.
The open-field, carried out 72 h after SE, revealed no differences between treatment groups (n = 6/group) in (B) distance traveled (p = 0.74) or (C) velocity (p = 0.72). Assessment of
anxiety-related behavior revealed no differences in percentage of time spent in a (D) peripheral zone (p = 0.40) and (E) central zone (p = 0.43) between LZ or MDZ mice (p N 0.05).
196 M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198

(68.46 ± 14.04%) and MDZ (81.21 ± 3.75%) treated mice (t(10) = 0.87, in seizure activity, supporting the utility of midazolam as a treatment
p = 0.40; Fig. 4D). Similarly, there was no difference between the LZ in experimental SE.
(32.96 ± 13.99%) and MDZ (21.01 ± 4.22%) groups in time spent in While the electrographic and histologic features of the i.a. KA model
the central area of the arena (t(10) = 0.81, p = 0.43; Fig. 4E). are well documented, relatively less is known about post-SE recovery
and behavioral deficits. Work to date has focused on hippocampus-
4. Discussion dependent tasks [40], but here, we show that a number of behaviors
are altered post-SE in mice. Examination of these other outcome param-
The main findings of the present study are that midazolam was eters revealed significant differences between treatment groups, with
noninferior to lorazepam in the treatment of SE in an experimental midazolam-treated animals displaying a better recovery of function at
model. Our data indicate that midazolam was as effective as lorazepam 24 h after SE. Specifically, midazolam-treated mice displayed higher
in curtailing seizures during SE and did not alter the level of acute neu- levels of overall activity including eating, drinking, and ambulation, as
ronal loss as a result of prolonged seizures, indicating that it was neither well as a higher frequency of grooming behavior and ability to attend
significantly more protective nor deleterious. There was also no impact to environmental alterations. While no differences in EEG between
of treatment on later-measured anxiety-related behavior or in locomo- groups during SE would indicate that any alterations in behavior are
tor activity in an open-field task. Surprisingly, we found that midazolam due to anticonvulsant effects and treatment, there may be, albeit unlike-
was superior to lorazepam in initial animal recovery 24 h after SE. Be- ly, alternative targets beyond seizure management resulting in im-
havioral measures including welfare monitoring, grooming, and activity proved outcomes. Although there are many similarities among the
levels all indicated that midazolam-treated mice displayed better out- various benzodiazepine compounds, the pharmacokinetics vary and
comes than lorazepam-treated mice at the early stages of recovery. may account for clinically relevant differences between drugs. The supe-
Thus, midazolam offered equivalent seizure-stopping performance riority of midazolam at 24 h after administration is potentially a result of
and reliable histopathology while also improving early recovery. elimination half-life differences between therapies. The elimination
Time is one of the key factors in treating SE, with the overall dura- half-life in humans for lorazepam is 12–15 h, significantly longer than
tion of seizure activity affecting later outcomes [5,36]; as such, early midazolam at approximately 3 h [37]. These are, however, more rapidly
treatment is essential. There is disagreement, however, on the best eliminated by rodents with an elimination half-life plasma of 1.5 h for
treatment regimen in clinical settings [17,37]. The factors to be consid- lorazepam [45] and 0.5 h for midazolam [46] and, as such, may not
ered include ease of delivery, access routes, onset, and duration of ac- fully explain the improvements seen. Midazolam's shorter duration of
tion, as well as recovery time. In the present study, we explored the action is due to its rapid metabolism by the liver via the cytochrome
efficacy of midazolam in a model of partial-onset SE, which is the P450 enzyme system to active and inactive metabolites [47]. Whether
most common form in patients with epilepsy [38]. The intra- this is beneficial or not remains contentious. Some argue that due to a
amygdala (i.a.) kainic acid model in mice has been in use since 2001 short duration of action, seizure relapse can occur in patients in the ab-
and is becoming a popular model due to its consistent and reliable sence of repeated dosing [17]. However, in the current study, the supe-
SE response, reproducible hippocampal pathology, being relatively rior animal recovery over lorazepam treatment at 24 h suggests the
high-throughput, with minimal mortality and versatility to translate utility of midazolam in treating SE in this experimental model. These
to other strains and transgenic lines [22,39–41]. Here, we find evidence data could also assist researchers in meeting obligations under the
to support the use of midazolam as an anticonvulsant treatment in this 3Rs (replacement, refinement, and reduction), now a mandatory re-
experimental model of SE. Our first major finding was that while both quirement under EU legislation.
lorazepam- and midazolam-treated animals experienced similar epi- Our findings are also in agreement with work comparing the clinical
sodes of SE as measured by EEG, no differences were found between efficacy of various benzodiazepine therapies. Crucially, our findings sup-
groups during a 1-hour recording after anticonvulsant administration. port a randomized control trial that specifically compared the efficacy of
This would indicate that midazolam is as effective as the frontline i.m midazolam treatment with i.v. lorazepam in the treatment of SE in a
treatment lorazepam in terminating SE. nonhospital setting [12]. Patients treated with midazolam displayed at
Furthermore, the level of neuronal loss was also similar between least comparable outcomes as those administered lorazepam, with ad-
groups. The i.a. KA model is associated with a highly reproducible injury ditional parameters, such as percentage requiring admittance to hospi-
to the ipsilateral hippocampus, and the damage seen here was very tal, indicating better outcomes for midazolam-treated patients [12].
similar to that reported previously in this model when performed by Midazolam was also found to be as effective as lorazepam in treating
another researcher [23]. Here, we found again that damage was mainly SE in a pediatric population [15]. When compared with other benzodi-
confined to area CA3, with some lesser and more variable injury to CA1. azepine treatments, midazolam has also shown effective results, with
There was also no difference in damage between lorazepam-treated a recent meta-analysis of 19 studies concluding that non-i.v. midazolam
and midazolam-treated mice at 72 h. This fits with the EEG data and is as effective or superior to diazepam in treating SE [48,49]. In experi-
with previous studies having found a close association between seizure mental models of SE, midazolam has also shown its effectiveness, with
duration and resulting damage [22,42]. Since hippocampal pathology is tonic–clonic seizures being terminated within 1.5–2 min of i.m. midazo-
a necessary feature of the i.a. KA model in order to assess potential lam administration [26,50].
disease-modifying treatments and in longer-term studies of epilepsy, While the majority of studies report i.m. administration of mid-
midazolam could be used in place of lorazepam while displaying the azolam, the current study is the first to determine its effectiveness
advantage of improved animal recovery at early time-points. administered i.p. in an experimental animal model of SE. Electroen-
When looking at the mechanism of action of these drugs, it is cephalogram recordings showed an ~ 50% reduction of EEG power
perhaps not surprising that we see similar effects of midazolam and over a 1-hour recording after administration of midazolam. This is
lorazepam treatment in these parameters. Similar to lorazepam and potentially an underestimation of its effect as it is compared with EEG
other benzodiazepines, midazolam acts primarily through potentiating recordings from the entire 40-minute period prior to midazolam treat-
the benzodiazepine–GABAA receptor complex [43]. This enhances the ment and which takes in a 10- to 15-minute period after KA injection
inhibitory action of GABAA by increasing the number of chloride channel before seizures begin in the model. Previous observations from our
openings resulting in the hyperpolarization of neurons. Status epilepti- group indicated that lorazepam administered i.p. results in similar anti-
cus is thought to result from a failure of the normal mechanisms for sei- convulsant properties as i.v. administration [22]. Intramuscular admin-
zure termination, in particular, a decrease in GABAA receptor function istration is also not advised in smaller rodents such as mice [51]. Here,
[44] and an increase in glutamate receptor density [1]. In line with this we show that midazolam is as effective in treating SE as lorazepam
view, both benzodiazepine treatments resulted in similar reductions when administered i.p. This is likely due to its solubility in water,
M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198 197

resulting in rapid absorbance from the injection site [52]. A potential [8] Towne AR, DeLorenzo RJ. Use of intramuscular midazolam for status epilepticus.
J Emerg Med 1999;17:323–8.
limitation of our findings is that this effect could be unique to mice [9] Prasad M, Krishnan PR, Sequeira R, Al-Roomi K. Anticonvulsant therapy for status
or to the i.a. KA model of SE. Furthermore, due to the drug solubil- epilepticus. Cochrane Database Syst Rev 2014;9:Cd003723.
ity, there were different vehicles used for each drug, i.e., phosphate- [10] Alldredge BK, Gelb AM, Isaacs SM, Corry MD, Allen F, Ulrich S, et al. A comparison of
lorazepam, diazepam, and placebo for the treatment of out-of-hospital status epilep-
buffered saline and sterile water. This is potentially a confounding factor ticus. N Engl J Med 2001;345:631–7.
that should be considered and may have indirect effects on resultant [11] Chiulli DA, Terndrup TE, Kanter RK. The influence of diazepam or lorazepam on the
recovery. frequency of endotracheal intubation in childhood status epilepticus. J Emerg Med
1991;9:13–7.
A potential limitation of our study is the short duration of EEG re- [12] Silbergleit R, Durkalski V, Lowenstein D, Conwit R, Pancioli A, Palesch Y, et al. Intra-
cording and relatively short-term behavioral observations. While we muscular versus intravenous therapy for prehospital status epilepticus. N Engl J Med
saw similar EEG patterns during the 60-minute posttreatment between 2012;366:591–600.
[13] Mandrioli R, Mercolini L, Raggi MA. Benzodiazepine metabolism: an analytical
groups, there is the possibility we missed recurrence of seizures over
perspective. Curr Drug Metab 2008;9:827–44.
the subsequent 24 h. In addition, although there is a return to a similar [14] Portela JL, Garcia PC, Piva JP, Barcelos A, Bruno F, Branco R, et al. Intramuscular
level of recovery of function by 72 h in both treatment groups and no midazolam versus intravenous diazepam for treatment of seizures in the pediat-
differences in the levels of neuronal loss at these early time-points, ric emergency department: a randomized clinical trial. Med Intensiva 2015;39:
160–6.
it is possible that differences between treatments emerge later, for ex- [15] Welch RD, Nicholas K, Durkalski-Mauldin VL, Lowenstein DH, Conwit R, Mahajan PV,
ample during epilepsy development, or affect delayed neurodegenera- et al. Intramuscular midazolam versus intravenous lorazepam for the prehospital
tion. Future studies could incorporate prolonged EEG recording, for treatment of status epilepticus in the pediatric population. Epilepsia 2015;56:254–62.
[16] Lowenstein DH, Alldredge BK, Allen F, Neuhaus J, Corry M, Gottwald M, et al. The
example using telemetry, and explore longer-term outcomes between prehospital treatment of status epilepticus (PHTSE) study: design and methodology.
midazolam-treated and lorazepam-treated animals. Also, our study Control Clin Trials 2001;22:290–309.
did not include a saline-treated group of SE mice, and it may be valuable [17] Costello DJ, Cole AJ. Treatment of acute seizures and status epilepticus. J Intensive
Care Med 2007;22:319–47.
to have data on the duration of SE and recovery pattern in animals in the [18] Naritoku DK, Sinha S. Prolongation of midazolam half-life after sustained infusion for
absence of treatment. status epilepticus. Neurology 2000;54:1366–8.
In summary, the present study provides important insights into the [19] Bialer M, White HS. Key factors in the discovery and development of new antiepilep-
tic drugs. Nat Rev Drug Discov 2010;9:68–82.
efficacy of midazolam as a pharmacotherapy for SE in an experimental [20] Loscher W, Schmidt D. Modern antiepileptic drug development has failed to deliver:
model of epilepsy. We found evidence supporting the utility of midazo- ways out of the current dilemma. Epilepsia 2011;52:657–78.
lam as an anticonvulsant treatment as compared with the more widely [21] Löscher W, Brandt C. Prevention or modification of epileptogenesis after brain
insults: experimental approaches and translational research. Pharmacol Rev 2010;
used lorazepam. Importantly, our data indicated significant benefits of
62:668–700.
midazolam at early time-points after drug administration on animal re- [22] Tanaka K, Jimenez-Mateos EM, Matsushima S, Taki W, Henshall DC. Hippocampal
covery without altering baseline EEG and histological features. Midazo- damage after intra-amygdala kainic acid-induced status epilepticus and seizure
lam is quickly becoming considered as the drug of choice for persistent preconditioning-mediated neuroprotection in SJL mice. Epilepsy Res 2010;88:
151–61.
acute seizures and SE [53], and the results reported here support the ef- [23] Engel T, Gomez-Villafuertes R, Tanaka K, Mesuret G, Sanz-Rodriguez A, Garcia-
ficacy of midazolam administered i.p. in treating acute, early SE in a Huerta P, et al. Seizure suppression and neuroprotection by targeting the purinergic
chemically induced experimental model of epilepsy. P2X7 receptor during status epilepticus in mice. FASEB J 2012;26:1616–28.
[24] Kretschmann A, Danis B, Andonovic L, Abnaof K, van Rikxoort M, Siegel F, et al. Dif-
ferent microRNA profiles in chronic epilepsy versus acute seizure mouse models.
J Mol Neurosci 2015;55:466–79.
Ethical publication statement [25] Dhir A, Zolkowska D, Rogawski MA. Seizure protection by intrapulmonary delivery
of midazolam in mice. Neuropharmacology 2013;73:425–31.
We confirm that we have read the Journal's position on issues in- [26] Raines A, Henderson TR, Swinyard EA, Dretchen KL. Comparison of midazolam and
diazepam by the intramuscular route for the control of seizures in a mouse model
volved in ethical publication and affirm that this report is consistent
of status epilepticus. Epilepsia 1990;31:313–7.
with those guidelines. [27] Cleton A, Voskuyl RA, Danhof M. Adaptive changes in the pharmacodynamics of
midazolam in different experimental models of epilepsy: kindling, cortical stimula-
tion and genetic absence epilepsy. Br J Pharmacol 1998;125:615–20.
Acknowledgments [28] Bourin M, Hascoet M, Mansouri B, Colombel MC, Bradwejn J. Comparison of behav-
ioral effects after single and repeated administrations of four benzodiazepines in
three mice behavioral models. J Psychiatry Neurosci 1992;17:72–7.
We would like to thank Dr. Edward Mealy for his assistance and ad- [29] Jimenez-Mateos EM, Bray I, Sanz-Rodriguez A, Engel T, McKiernan RC, Mouri G, et al.
vice in the care and welfare of experimental animals. This research was miRNA expression profile after status epilepticus and hippocampal neuroprotection
funded in part by Science Foundation Ireland award 13/IA/1891 and the by targeting miR-132. Am J Pathol 2011;179:2519–32.
[30] Paxinos G, Franklin KBJ. The mouse brain in stereotaxic coordinates. 2nd ed. San
Health Research Board PHD/2007/11. Diego, California: Academic Press; 2001.
[31] Kirsch JH, Klaus JA, Blizzard KK, Hurn PD, Murphy SJ. Pain evaluation and response to
Conflict of interest buprenorphine in rats subjected to sham middle cerebral artery occlusion. Contemp
Top Lab Anim Sci 2002;41:9–14.
[32] Roughan JV, Flecknell PA. Effects of surgery and analgesic administration on sponta-
None of the authors has any conflict of interest to disclose. neous behaviour in singly housed rats. Res Vet Sci 2000;69:283–8.
[33] Prut L, Belzung C. The open field as a paradigm to measure the effects of drugs on
anxiety-like behaviors: a review. Eur J Pharmacol 2003;463:3–33.
References [34] Engel T, Sanz-Rodgriguez A, Jimenez-Mateos EM, Concannon CG, Jimenez-Pacheco
A, Moran C, et al. CHOP regulates the p53-MDM2 axis and is required for neuronal
[1] Wasterlain CG, Liu H, Naylor DE, Thompson KW, Suchomelova L, Niquet J, et al. survival after seizures. Brain 2013;136:577–92.
Molecular basis of self-sustaining seizures and pharmacoresistance during status ep- [35] Jimenez-Mateos EM, Hatazaki S, Johnson MB, Bellver-Estelles C, Mouri G, Bonner C,
ilepticus: the receptor trafficking hypothesis revisited. Epilepsia 2009;50(Suppl. 12): et al. Hippocampal transcriptome after status epilepticus in mice rendered seizure
16–8. damage-tolerant by epileptic preconditioning features suppressed calcium and neu-
[2] Neligan A, Shorvon SD. Prognostic factors, morbidity and mortality in tonic–clonic ronal excitability pathways. Neurobiol Dis 2008;32:442–53.
status epilepticus: a review. Epilepsy Res 2011;93:1–10. [36] Kapur J, Macdonald RL. Rapid seizure-induced reduction of benzodiazepine and
[3] Lowenstein DH, Alldredge BK. Status epilepticus. N Engl J Med 1998;338:970–6. Zn2+ sensitivity of hippocampal dentate granule cell GABAA receptors. J Neurosci
[4] DeLorenzo RJ, Pellock JM, Towne AR, Boggs JG. Epidemiology of status epilepticus. 1997;17:7532–40.
J Clin Neurophysiol 1995;12:316–25. [37] Loscher W. Single versus combinatorial therapies in status epilepticus: novel data
[5] Towne AR, Pellock JM, Ko D, DeLorenzo RJ. Determinants of mortality in status from preclinical models. Epilepsy Behav 2015. http://dx.doi.org/10.1016/j.yebeh.
epilepticus. Epilepsia 1994;35:27–34. 2015.02.027 (pii: S1525-5050(15)00083-9, in press).
[6] Loscher W. Molecular mechanisms of drug resistance in status epilepticus. Epilepsia [38] Wasterlain CG, Treiman DM. Status epilepticus: mechanisms and management.
2009;50(Suppl. 12):19–21. Cambridge, Mass.: MIT; 2006
[7] Rossetti AO, Lowenstein DH. Management of refractory status epilepticus in adults: [39] Henshall DC, Skradski SL, Bonislawski DP, Lan JQ, Simon RP. Caspase-2 activation is
still more questions than answers. Lancet Neurol 2011;10:922–30. redundant during seizure-induced neuronal death. J Neurochem 2001;77:886–95.
198 M. Diviney et al. / Epilepsy & Behavior 51 (2015) 191–198

[40] Liu G, Gu B, He XP, Joshi RB, Wackerle HD, Rodriguiz RM, et al. Transient inhibition of [47] Thummel KE, Shen DD, Podoll TD, Kunze KL, Trager WF, Hartwell PS, et al. Use of
TrkB kinase after status epilepticus prevents development of temporal lobe epilepsy. midazolam as a human cytochrome P450 3A probe: I. In vitro–in vivo correlations
Neuron 2013;79:31–8. in liver transplant patients. J Pharmacol Exp Ther 1994;271:549–56.
[41] Li T, Ren G, Lusardi T, Wilz A, Lan JQ, Iwasato T, et al. Adenosine kinase is a target for [48] McMullan J, Sasson C, Pancioli A, Silbergleit R. Midazolam versus diazepam for the
the prediction and prevention of epileptogenesis in mice. J Clin Invest 2008;118: treatment of status epilepticus in children and young adults: a meta-analysis. Acad
571–82. Emerg Med 2010;17:575–82.
[42] Engel T, Hatazaki S, Tanaka K, Prehn JH, Henshall DC. Deletion of Puma protects hip- [49] Brigo F, Nardone R, Tezzon F, Trinka E. Nonintravenous midazolam versus intrave-
pocampal neurons in a model of severe status epilepticus. Neuroscience 2010;168: nous or rectal diazepam for the treatment of early status epilepticus: a systematic
443–50. review with meta-analysis. Epilepsy Behav 2015. http://dx.doi.org/10.1016/j.
[43] Amrein R, Hetzel W. Pharmacology of drugs frequently used in ICUs: midazolam and yebeh.2015.02.030 (pii: S1525-5050(15)00090-6, in press).
flumazenil. Intensive Care Med 1991;17(Suppl. 1):S1–S10. [50] Domino EF. Comparative seizure inducing properties of various cholinesterase
[44] Naylor DE, Liu H, Wasterlain CG. Trafficking of GABA(A) receptors, loss of inhibition, inhibitors: antagonism by diazepam and midazolam. Neurotoxicology 1987;8:
and a mechanism for pharmacoresistance in status epilepticus. J Neurosci 2005;25: 113–22.
7724–33. [51] Shimizu S. Routes of administration. The laboratory mouse. New York: Elsevier
[45] Granvil CP, Yu AM, Elizondo G, Akiyama TE, Cheung C, Feigenbaum L, et al. Academic; 2004 527–42.
Expression of the human CYP3A4 gene in the small intestine of transgenic [52] Barr J, Zomorodi K, Bertaccini EJ, Shafer SL, Geller E. A double-blind, randomized
mice: in vitro metabolism and pharmacokinetics of midazolam. Drug Metab comparison of i.v. lorazepam versus midazolam for sedation of ICU patients via a
Dispos 2003;31:548–58. pharmacologic model. Anesthesiology 2001;95:286–98.
[46] Mandema JW, Tukker E, Danhof M. Pharmacokinetic–pharmacodynamic modelling [53] McDonough JH, Van Shura KE, LaMont JC, McMonagle JD, Shih TM. Comparison of the
of the EEG effects of midazolam in individual rats: influence of rate and route of intramuscular, intranasal or sublingual routes of midazolam administration for the
administration. Br J Pharmacol 1991;102:663–8. control of soman-induced seizures. Basic Clin Pharmacol Toxicol 2009;104:27–34.

You might also like