You are on page 1of 16

Reproductive Toxicology 49 (2014) 101–116

Contents lists available at ScienceDirect

Reproductive Toxicology
journal homepage: www.elsevier.com/locate/reprotox

Zebrafish embryotoxicity test for developmental (neuro)toxicity:


Demo case of an integrated screening approach system using
anti-epileptic drugs
Anna Beker van Woudenberg a , Cor Snel b , Eke Rijkmans a , Didima de Groot a ,
Marga Bouma a , Sanne Hermsen c , Aldert Piersma c , Aswin Menke b , André Wolterbeek a,∗
a
TNO, Research Group Risk Analysis for Products In Development (RAPID), Zeist, The Netherlands
b
TNO Triskelion B.V., Zeist, The Netherlands
c
Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands

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

Article history: To improve the predictability of the zebrafish embryotoxicity test (ZET) for developmental (neuro)toxicity
Received 16 December 2013 screening, we used a multiple-endpoints strategy, including morphology, motor activity (MA), histopath-
Received in revised form 14 July 2014 ology and kinetics. The model compounds used were antiepileptic drugs (AEDs): valproic acid (VPA),
Accepted 31 July 2014
carbamazepine (CBZ), ethosuximide (ETH) and levetiracetam (LEV). For VPA, histopathology was the
Available online 8 August 2014
most sensitive parameter, showing effects already at 60 ␮M. For CBZ, morphology and MA were the
most sensitive parameters, showing effects at 180 ␮M. For ETH, all endpoints showed similar sensitivity
Keywords:
(6.6 mM), whereas MA was the most sensitive parameter for LEV (40 mM). Inclusion of kinetics did not
Zebrafish embryotoxicity test (ZET)
Developmental (neuro)toxicity
alter the absolute ranking of the compounds, but the relative potency was changed considerably. Taking
Antiepileptic drugs all together, this demo-case study showed that inclusion of multiple-endpoints in ZET may increase the
Integrated test strategy sensitivity of the assay, contribute to the elucidation of the mode of toxic action and to a better definition
Histopathology of the applicability domain of ZET.
Kinetics © 2014 Elsevier Inc. All rights reserved.
Behavior
Gene expression

1. Introduction systems, making it a suitable model organism for medium/high-


throughput screening [11,12].
Developmental toxicity testing in laboratory animals is required Currently, one of the most commonly used endpoints for devel-
for the preclinical safety assessment of new pharmaceuticals [1]. opmental toxicity screening in zebrafish is its morphology, which
Although studies with rodents are still the “gold standard” used to makes it relatively easy to detect various anomalies [13,14] that
study developmental toxicity for regulatory purposes, over the last correspond quite well with higher mammalian species [15,16]. In a
decades efforts have been made to replace these studies with cell- previous study [17], we demonstrated that additional histopatho-
based alternatives (e.g. stem cells) [2–5]. Rodent studies are time logical examination of the larvae contributed to an improved
consuming, expensive and involve large numbers of laboratory ani- understanding of the mode of toxicity action, thereby increasing the
mals. To date, however, the search for alternatives has not been predictability of the zebrafish assay for toxicity screening. The use
very successful, due to the difficulty of investigating the complex- of zebrafish larvae to test compounds for potential developmental
ity of developmental processes in relatively simple cellular assays (neuro)toxicity involves various behavioral tests, such as the motor
[6]. The zebrafish (Danio rerio) is emerging as a candidate lower activity test (MA). Tests of this kind are based on measuring the
vertebrate animal model capable of filling the gap between high effects on factors such as total distance moved and velocity [18–20].
throughput in vitro cellular assays and conventional preclinical ani- Recently, an extension of the MA test analysis was proposed, in
mal testing [7–10]. The embryonic/larval zebrafish model offers an which a wider range of endpoints can be evaluated from the same
intact whole animal model with many of the advantages of in vitro collected data set [17]. Furthermore, other endpoints than morpho-
logical scoring have been used in the embryonic and larval model to
evaluate the potential toxicity of compounds, including proteomics
∗ Corresponding author. Tel.: +31 888665053. [21] and transcriptomics [22]. Although each endpoint evaluation
E-mail address: andre.wolterbeek@tno.nl (A. Wolterbeek). has its own relevance, in general, they are studied separately,

http://dx.doi.org/10.1016/j.reprotox.2014.07.082
0890-6238/© 2014 Elsevier Inc. All rights reserved.
102 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

so the results of different studies need to be integrated. In addition, Table 1


Concentration range from the DRF and main experiments.
the measurement of test compound uptake by zebrafish embryos
and larvae has been shown to be a critical factor, one that can influ- Compound VPA (␮M) CBZ (␮M) ETH (mM) LEV (mM)
ence the predictability of the assay. Various studies have revealed DRF 30–1500 30–720 2–20 0.5–150
an overestimation [23] or an underestimation of exposure, leading Main 30–730 30–480 3–18 20–140
to false positive or false negative assay results. Abbreviations: VPA, valproic acid; CBZ, carbamazepine; ETH, ethosuximide; LEV,
Here we report on an integrated zebrafish screening approach levetiracetam.
based on morphology, histopathology, motor activity and kinetics,
using anti-epileptic drugs (AEDs) as a group of model compounds.
removing the partition. Eggs were collected using an 800 ␮m mesh
Some AEDs can exert their effects via inhibition of the histone
and transferred to a Petri dish containing aquarium water spiked
deacetylases (HDAC) [24,25] by binding to retinoic acid (RA) recep-
with 0.05% methylene blue. Staining indicates that eggs are either
tor [26]. Using in situ hybridization (ISH), we also investigated
non-fertilized or that they have a damaged membrane. Any such
the gene expression profile of two genes (Aldh1a2 and Cyp26a1)
eggs were discarded. Only batches in which more than 80% of the
involved in the retinoic acid (RA) signaling pathway and ways in
eggs were fertilized were used in the experiments.
which these are affected by AEDs. Aldh1a2 is involved in the syn-
thesis of RA, through the oxidation of retinaldehyde, while Cyp26a1
is involved in the degradation of RA, by the conversion of RA into 2.2. Compounds
more polar metabolites [27].
It is known that, when administered during pregnancy, most Valproic acid sodium salt (VPA, CAS No. 1069-66-5), carba-
AEDs are capable of inducing developmental (neuro)toxicity mazepine (CBZ, CAS No. 298-46-4) and ethosuximide (ETH, CAS No.
[28,29]. The effects of AEDs on the offspring may range from major 77-67-8) were purchased by Sigma-Aldrich (St. Louis, MO, USA).
congenital malformations (MCM) to delayed postnatal cognitive Levetiracetam (LEV, CAS No. 102767-28-2) was purchased by TCI
development and growth [30]. The developmental toxic potential Europe N.V. (Zwijndrecht, Belgium). First, a stock solution of each
of first generation AEDs, such as valproic acid (VPA) and carba- compound was prepared in dimethyl sulfoxide (DMSO). The stock
mazepine (CBZ), has been clearly demonstrated in humans and in solution was then further diluted to the desired concentration, in
various animal models [31,32]. Only a limited amount of data has aquarium water. The final concentration of DMSO in the water sup-
been published on the developmental effects of other first genera- plemented with the compound was 0.2%. Therefore, 0.2% DMSO in
tion AEDs such as ethosuximide (ETH) and for newer compounds, aquarium water was used as negative control.
such as levetiracetam (LEV). Despite the limited availability of data,
teratogenicity and cognitive impairment have been described for 2.3. Experimental design
ETH in rodents [33,34]. Levetiracetam was found to have develop-
mental effects only at very high dose levels [35]. In addition, it was 2.3.1. Dose range finding (DRF) experiments
recently reported that LEV might be a safer alternative to VPA, as it In the DRF experiments, a wide range of concentrations (partly
involves a lower risk of major congenital malformations in women based on studies from Berghmans et al. [40] and Weigt et al. [41])
with epilepsy who are of childbearing age [36]. was selected for each test compound. Embryos were exposed to the
The aim of this study was to improve the predictability of selected concentrations to narrow down the relevant concentration
the zebrafish model and to extend its applicability domain by range for further testing in main experiments. In the DRF studies,
using an integrated screening strategy, including a wide-range the embryos were assessed for lethal embryotoxic and/or devel-
of endpoints (modules): morphology, behavior (motor activity opmental toxic effects at 24, 48, 72 and 96 hours post fertilization
parameters), histopathology, kinetics and phenotyping (in situ (hpf) (results not shown). The concentration ranges of the various
hybridization). Four well-known AEDs (valproic acid (VPA), car- compounds used in the DRF and subsequent main experiments are
bamazepine (CBZ), ethosuximide (ETH) and levetiracetam (LEV)) shown in Table 1.
with different developmental (neuro)toxic potencies were chosen
as model compounds. 2.3.2. Main experiments
The main experiments were performed after the concentration
range of interest, which were the concentrations around the EC50
2. Material and methods for morphological endpoints, had been established in the DRF stud-
ies. Selected embryos were at first placed in a Petri dish containing
2.1. Fish husbandry and egg production the test concentrations of interest (see Table 1). Subsequently,
each embryo was transferred to a 12-well plate containing 3 ml of
Adult wild type AB line Zebrafish (Zebrafish International test medium per well, one embryo per well, unless otherwise indi-
Resource Center, Eugene, OR, USA) were kept in a 12 h dark/12 h cated. Embryos were kept in an incubator (Binder Germany, Type
light cycle at 28 ◦ C in self-regulating aquaria (Tecniplast, Tecnilab- BD115) at 26.5 ± 1 ◦ C with a 12 h/12 h dark/light cycle and relative
BMI, The Netherlands) at pH 7.5 and a water conductivity of humidity of 100% (to prevent evaporation). For all experiments,
500 ␮S/cm. The animals were fed twice a day with dry flake food we used intact embryos, the embryos were not dechorionated
(SDS) and once a day with live food (Artemia), as recommended before exposure. Test solutions were not refreshed during the
by Westerfield [37] and in accordance with the OECD draft guide- exposure period. The embryos were subjected to morphological
line on the use of zebrafish for chemical testing [38]. Eggs and evaluations at 24 and 48 hpf, primarily for lethal endpoints (data
larvae used in this study were derived from a zebrafish colony not shown). Evaluations at 72 and 96 hpf were used for detailed
held in the animal facility of TNO. Housing and management of the morphological developmental endpoints (see Section 2.4). Follow-
zebrafish colony fully comply with EU/2010/63 regulations [39]. ing a motor activity assessment (see Section 2.5) the larvae were
For egg production, fish of between 7 and 11 months old were euthanized, and processed for histopathological investigations
used. On the evening prior to the day of breeding, two male fish (see Section 2.6). For morphology and motor activity analysis,
and one female fish were placed in a breeding tank containing three independent experiments were performed per compound,
a partition (Techniplast) and egg traps to prevent egg predation. unless otherwise indicated. In each independent experiment, 12
Next morning, spawning was triggered by turning the light on and embryo/larvae per concentration were used. Histopathology was
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 103

assessed using larvae from one experiment (12 embryo/larvae measured intensity of 0 lx. The movements of the zebrafish larvae
per concentration, per compound, unless otherwise indicated). were tracked using ViewPoint software.
The analysis of total body burden (kinetics) was performed in In each MA test, the tracks of individual larvae were analyzed
a separate experiment (see Section 2.7), with three samples for using the analysis software (Zebrabox) for Distance Moved (DM)
analysis per concentration of each compound. For each sample 18, (mm) as measure of locomotor activity. Tracks were analyzed per
25 or 32 larvae were used. The in situ hybridization experiments unit time (1 min intervals and 15 min test sessions) as described
(WISH) were performed in two independent experiments for each previously [17]. The parameters analyzed were: (i) Distance Moved
transcript (see Section 2.9). In each experiment, embryos and (DM) per unit time, i.e. DM15min , DM1min ; (ii) Slope of Habituation
larvae of different age were exposed to various concentrations (HSL); and (iii) Startle Response (StR). With the exception of the
of the test compounds. For each of these experiments, 15–30 Startle Response, these parameters were measured during each
embryos/larvae per concentration, per age were used. of the three 15-min test sessions, i.e.: Dark I (0–15 min); Light
(16–30 min) and Dark II (31–45 min). The Startle Response was
defined as a response by the larvae to a sudden change in the light-
2.4. Morphology score
ing conditions. For more details see Beker van Woudenberg et al.
[17].
We have investigated the shape and structure (morphology)
of the whole zebrafish embryo. Scoring was performed according
2.6. Histopathology
to the method described by Fraysse and coworkers [42], involv-
ing a single observer and a stereomicroscope (Nikon, Japan), at
After the MA test, all larvae were euthanized using 1 g/L tricaine
20–50 times magnification, in non-dechorionated embryos/larvae.
solution. They were subsequently fixed overnight in Bouin’s fix-
The endpoints were assessed using binary responses (yes/no) as
ative (Klinipath; Duiven, The Netherlands), embedded in paraffin
stipulated in the DIN standard [43]. Any embryos or larvae with
in the same orientation and sectioned completely into serial 4 ␮m
evidence of coagulation and/or no heartbeat were considered to
thick sagittal sections. After staining with hematoxylin and eosin
be dead. Those experiments in which the controls exhibited sur-
(H&E), ≥100 sections of each larva were viewed using an Axioskop
vival rates of >90% were considered to be valid. Per compound, in
2 plus microscope (Zeiss). In general, for each concentration, 12
general, three independent experiments were performed using 12
larvae were used for histopathological analysis. However, since
embryos/larvae per concentration which were evaluated on differ-
occasionally, a larva was lost during processing, the exact number
ent time points for effects on the various morphological endpoints.
of analyzed larvae is listed in Table 3.
For each endpoint, which was assessed on 72 and 96 hpf, an EC50
was calculated using Graph Pad software (see Section 2.9). The EC50
2.7. Kinetics
was defined as the concentration at which there was a 50% increase
(or decrease) of the incidence of a given parameter, in comparison
For the analysis of total body burden (kinetics), a separate exper-
to the control.
iment was performed. First, embryos were placed in a Petri dish
containing the test compound at the concentration to be inves-
2.5. Motor activity (MA) test tigated. They were then transferred to 12-well or 48-well plates
containing as average of 3 embryos/ml of the test solution. The
A motor activity (MA) test was performed to assess develop- embryos were kept in an incubator for 96 h at 26.5 ± 1 ◦ C with a
mental (neuro)toxicity. Just before the start of the MA test, all larvae 12 h/12 h dark/light cycle and relative humidity of 100% (see Sec-
were transferred to 96-well, square well, flat-bottom plates (What- tion 2.3.2). Test solutions were not refreshed during the exposure
man, cat no. 7701-1651) in their own test solution (±500 ␮l), one period.
larvae per well. No selection was made before transfer. However, The concentrations used in these experiments were: VPA 0, 60,
larvae scoring positive on any of the developmental toxicity end- 150, 320 and 730 ␮M; ETH 0, 6, 9 and 12 mM; CBZ 0, 60, 120 and
points were measured in the MA test but excluded from further 180 ␮M; LEV 0, 20, 40 and 100 mM which were in the same range
data analysis in order not to confound MA results. All concentra- as used for the other endpoints. Three samples, each consisting of
tions from the same compound and the respective control were 18, 25 or 32 larvae, per concentration of each compound were used.
tested in the same plate. Before sampling the larvae, they were morphologically evaluated
The MA test was always performed in the early afternoon, after to confirm that the incidences of effects were comparable to the
the last morphological examination (±100 hpf) as described pre- main experiments. Then samples were collected in three 1.5 ml safe
viously [17]. Briefly, MA was assessed using a ViewPoint behavior lock Eppendorf® tubes, euthanized with tricaine (1 g/L) and washed
recording system, together with the associated movement tracking three times with aquarium water. After the final wash, the water
and analysis software (Zebrabox, ViewPoint, France). The system was removed and the larvae were frozen (−20 ◦ C) until analysis.
itself consisted of a light-box platform and an infrared-sensitive The samples were analyzed using an Acquity ultra-performance
camera within a closed box. The larvae were maintained at a liquid chromatography (UPLC) system (Waters, Belgium) coupled
constant environmental temperature by passing a thermostati- to a Waters Xevo TQ MS tandem mass spectrometer (Waters-
cally controlled, continuous water flow (27 ± 1 ◦ C) into the closed Micromass, Manchester, UK). Both of these devices were operated
Zebrabox. The larvae were allowed an average of 15 min to accli- using MassLynx® software.
matize in the Zebrabox, under visible light conditions, before MA The UPLC system was equipped with a 2.1 × 100 mm, 1.7 ␮m
recording commenced. The larvae were then exposed to the fol- ACQUITY BEH C18 column (Waters, Milford, PA, USA) kept at
lowing lighting conditions: 15 min of darkness (i.e. infrared light), 50 ◦ C. A binary gradient mobile phase was used at a flow rate of
15 min of light (i.e. visible light plus infrared light) and a final 15 min 0.4 ml min−1 with solvent A (0.1% FA in MilliQ water) and solvent
period of darkness (i.e. infrared light). These sessions are hereafter B (0.1% FA in MeCN).
referred to as Dark I, Light and Dark II, respectively. A spherical The mass spectrometer was operated in electrospray positive
sensor was placed in the Zebrabox (with a closed lid), to mea- mode, while data acquisition was performed using the multiple
sure the visible light intensity in the locomotion apparatus. At the reactions monitoring mode (MRM). The source settings were as
same light intensity as that used during the experiments, the sen- follows: capillary voltage 2.0 kV, source temperature 150 ◦ C, des-
sor gave a reading of 700 lx, while the infrared light resulted in a olvation temperature 600 ◦ C, cone voltage 32 V, cone gas flow
104 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

150 L h−1 , desolvation gas flow 1000 L h−1 , collision gas flow (argon) with PBS, and images were recorded using a Zeiss Stereo Discovery
0.15 ml min−1 , collision cell pressure 3.4 × 10−3 mbar. The results V8 microscope equipped with a Canon powershot G12 camera. The
were quantified using Target Lynx software. For the four com- embryos/larvae were stored in 4% paraformaldehyde (PFA) at 4 ◦ C.
pounds analyzed, the limit of detection was 20 ng/ml and the
recovery was 80–120%. Results of the analysis were expressed as
2.9. Statistical analysis
nanomol test compound per larvae.
For morphological examinations, a dose-response curve was
2.8. Whole mount in situ hybridization (WISH)
fitted for each relevant endpoint and an EC50 concentration was cal-
culated using Graph Pad (GraphPad Prism 5, version 5.03, December
Whole mount in situ hybridization (WISH) was performed to
2009). The following four-parameter logistic equation was used for
investigate the expression patterns of two selected genes (Aldh1a2
curve fitting: y = A + ((B − A)/(1 + ((C/x)D))); where A = minimum y
and Cyp26a1). Being involved in the retinoic acid pathways, these
(constrained to >0), B = maximum y (constrained to <100), C = the
genes also play a part in mechanisms of early development in
EC50 value, and D = slope factor. All of the derived parameters stud-
embryos exposed to anti-epileptic drugs (AEDs) [44–46]. Valproic
ied in the MA test: Distance Moved, Startle Response and Slope of
acid and levetiracetam were selected as model compounds for
Habituation were statistically analyzed using a mixed model using
these WISH experiments since (based on morphology and motor
all data of all experiments. Compound/concentration was used as
activity analyses) these compounds appeared to be the most (VPA)
fixed factor. To correct for plate and row differences and, hence for
and least (LEV) toxic compounds tested in the present study.
instance, to correct for offset differences between controls, loca-
tion (row within a plate) was used as random factor in the mixed
2.8.1. Synthesis of digoxigenin-labeled probes
model. All data were log-transformed before analysis. In all of the
Sense and anti-sense digoxigenin (DIG)-labeled probes for
tests performed, the hypothesis was rejected at the 5% probability
Aldh1a2 (GeneBank: NM 131850.1) and Cyp26a1 (GeneBank:
level (alpha = 0.05). A Dunnett’s post hoc test was performed to cor-
NM 131146.2) were synthesized following the protocol described
rect for multiple testing. All MA analysis were performed using SAS
by Thisse and Thisse [47]. Primer sequences for Cyp26a1: sense:
9.3 (SAS Institute Inc., Cary, NC, USA). For more details on the MA
5 -CATTAACCCTC-ACTAAAGGGAAAGCAGGAGGTGAAGAGCGCC-3
analysis, please see Beker van Woudenberg et al. [17]). The statis-
and antisense: 5 -TAATACGACTCACTATAGGGAGAG-GTTGCAGTAC
tical significance of the histological findings was evaluated by the
TGGCGGTGGT-3 were used. For Aldh1a2 the following primer
Fisher’s exact probability test.
sequences were used; sense: 5 -CATTAACCCTCACTAAAGGGAAGG
CTGCACGTTCGGCCTTCT-3 and antisense: 5 -TAATACGAC-TCACTA
TAGGGAGAGCGGTTGGCCCATAGCCTGG-3 . A digoxigenin(DIG)- 3. Results
labeled probe for the housekeeping gene, fatty acid binding
protein-10 (Fabp-10), was kindly donated by Dr. C. Esguerra (KU 3.1. Morphology
Leuven, Belgium).
The most sensitive effects (based on EC50 ) on morphology of
2.8.2. Embryo exposure and WISH protocol zebrafish embryos exposed to antiepileptic drugs (AEDs) at 72 hpf
Embryos/larvae were exposed to 0, 150, 320 and 730 ␮M VPA and 96 hpf are presented in Table 2.
or to 0, 60, 80, 100 and 140 mM LEV for 24, 48, 72 and 96 hpf. As At 72 hpf, in embryos exposed to valproic acid (VPA), a dose-
a positive control, the expression of Fabp-10 (a gene expressed in dependent increase was found in the incidence of non-hatched
the liver) was tested in larvae at 120 hpf in each experiment. Sense embryos. This endpoint appeared to be the most sensitive param-
probes of Cyp26a1 and Aldh1a2 were used as negative controls. A eter, with an EC50 of 221 ␮M. At this time point, pericardial edema
group without a probe was also used as a negative control. Per tran- was also observed in 46% and 50% of the embryos exposed to
script, two independent experiments were performed (see Section VPA at 320 ␮M and 730 ␮M, respectively (data not shown). More-
2.3.2). The WISH protocol was adapted from Thisse and Thisse [47]. over, at 96 hpf, the percentage of larvae showing pericardial edema
Briefly, after treatment, the embryos were mechanically dechorion- increased to 92% and 100% at 320 ␮M and 730 ␮M VPA, respec-
ated (where applicable) and subsequently euthanized at the desired tively, and it appeared to be the most sensitive parameter for larvae
developmental stages using a 1 g/L tricaine solution. They were of this age, with an EC50 of 165 ␮M. In addition, the group of lar-
then fixed overnight (O/N) in 4% paraformaldehyde. Embryos at vae exposed to 320 ␮M and 730 ␮M VPA had a high incidence of
24 hpf were then immediately dehydrated in 100% methanol and non-hatching (50% and 67%, respectively).
stored at −20 ◦ C until required. Older embryos and larvae were first As with VPA, non-hatching was the most sensitive parameter
bleached by immerging them in a 3% H2 O2 –5% KOH solution until in 72 hpf embryos exposed to carbamazepine (CBZ), with an EC50
all pigmentation disappeared (±1 h). Prior to storage at −20 ◦ C, of 193 ␮M. At 96 hpf, pericardial edema was the most sensitive
the embryos/larvae were gradually dehydrated by being passed parameter, with an EC50 of 220 ␮M. The group of larvae exposed
through a series of methanol solutions (25%, 50%, 75% and 100%) to 240 ␮M and 480 ␮M CBZ also exhibited a high incidence of
in phosphate buffered saline (PBS). non-hatching (50% and 100%, respectively). At relatively high etho-
On the day of the WISH experiments, the embryos/larvae suximide (ETH) concentrations, dose-dependent pericardial edema
were rehydrated through successive baths of 75%, 50% and 25% was observed. This appeared to be the most sensitive parameter at
methanol in PBS. After washing, the embryos/larvae were perme- 72 and 96 hpf, with EC50 values of 9.9 mM and 8.9 mM, respectively.
abilized using 10 ␮g/ml Proteinase K (Roche Diagnostics, Almere, Yolk sac and chorda malformations (defined as larvae showing
The Netherlands). WISH was carried out using DIG-labeled probes a bent spine) were also observed at 96 hpf in larvae exposed to
for Aldh1a2 and Cyp26a, which were synthesized as described 15 mM ETH (42% and 40%, respectively) and 18 mM ETH (75% and
above (see Section 2.8.1). Hybridization was performed O/N at 40%, respectively). Of the 96 hpf larvae exposed to 18 mM ETH, 69%
65 ◦ C in a hybridization mix containing 50% formamide (Sigma- showed an obviously – i.e. clearly visible – decreased heart rate as
Aldrich), 5× SSC and 40 ␮g/ml Salmon sperm DNA (Invitrogen). compared to control larvae.
Probes were detected using alkaline phosphatase conjugated anti- Embryos exposed to levetiracetam (LEV) showed only two
bodies, and colorization was performed O/N with BM purple (Roche adverse developmental effects, and then only at very high concen-
Diagnostics). The embryos/larvae were mounted in 70% glycerol trations. The first of these was tail malformation at 96 hpf, with
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 105

Table 2
Most sensitive effects (based on EC50 ) on morphology of zebrafish embryos/larvae exposed to anti-epileptic drugs (AEDs) at 72 and 96 h post fertilization (hpf).

Abbreviations: VPA, valproic acid; CBZ, carbamazepine; ETH, ethosuximide; LEV, levetiracetam.

an EC50 of 109 mM. This was observed in 58% and 71% of larvae to the control group. In the other hand, the response observed from
exposed to 120 mM or 140 mM LEV, respectively. The second was Dark I to Lightness session (L-StR), was significantly higher than
chorda malformations which had an observed incidence of 12% and the Startle Response observed in the control group. Also swimming
33% (120 mM and 140 mM LEV groups, respectively), not reaching activity (DM15min ) – although fluctuating – remained increased dur-
EC50 levels. ing the Light session, thereby significantly differing from the control
group showing the normal – low – activity in Light. The changes in
3.2. Motor activity (MA) test the 30 and 60 ␮M VPA groups – reduction of a Startle Response at
the start of the Dark I session and time-shift in the Dark II session
The results of the MA test performed (100 hpf ± 2) on lar- – did not reach the level of statistical significance. Since all larvae
vae kept in vehicle or exposed to different concentrations of the exposed to higher concentrations (320 or 730 ␮M) VPA were either
AEDs are shown in the left-hand panels of Fig. 1. The right-hand dead, non-hatched or malformed, they were excluded from the MA
panels of this figure show the results of the statistical evalua- analysis.
tion. Exposure to carbamazepine (CBZ) caused a concentration-
Larvae exposed to valproic acid (VPA) showed a reduction in related reduction (significant at concentrations ≥ ␮M CBZ) in total
total Distance Moved per 15 min session (DM15min ) in Dark I and Distance Moved per 15 min test session (DM15min ) in Dark I and
Dark II sessions at the highest concentration analyzed (150 ␮M Dark II, as well as in the Darkness Startle Response (D-StR) (Fig. 1B).
VPA) compared to the control group. In fact, this 150 ␮M VPA con- Swimming activity in the 240 ␮M CBZ concentration group was
centration group consistently exhibited low levels of activity, and – marginal to even absent and so, the curve slope representing a
consequently – no habituation was observed (Fig. 1A). Moreover, no measure of habituation was absent as well. For all groups of lar-
Startle Response was observed from the Light to Darkness session vae exposed to <180 ␮M CBZ, the slope of the swimming activity
(D-StR) in the 150 ␮M VPA, which was significant lower compared curve was not affected by CBZ. Since all larvae exposed to 480 ␮M
106 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

Fig. 1. Effect of anti-epileptic (AEDs) compounds on the swimming activity of zebrafish larvae in the motor activity test. Left panel shows the graphics for the Distance
Moved DM-per minute intervals for larvae exposed to (A) valproic acid (VPA); (B) carbamazepine (CBZ); (C) ethosuximide (ETH) and (D) levetiracetam (LEV). Right panel
presents, respectively, the statistical evaluation of the data for motor activity. Conc = concentration; ns = non-significant; *p < 0.05; **p < 0.01; ***p ≤ 0.001; ****p ≤ 0.0001;
#
total number of independent experiments; ## total number of independent experiments for LEV varied from 2 to 4, depending of the concentration group; ‡ number of larvae
analyzed after exclusion of dead, non-hatched and malformed ones out of the total number of larvae.
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 107

CBZ were either not hatched or dead, this group was excluded from and only necrotic tissue was visible (Fig. 2D). The remaining larvae
the MA analysis. did not hatch and showed severe disruption of normal brain struc-
Larvae exposed to ethosuximide (ETH) showed a biphasic ture, characterized by disorganization and apoptosis. Pericardial
response in total Distance Moved per 15 min test session (DM15min ) edema was occasionally detected in the 150 ␮M VPA and 320 ␮M
(Fig. 1C). Low concentrations (3 mM and 6 mM ETH groups) showed VPA group. The heart could not properly be assessed in the 730 ␮M
an increase in (DM15min ) – hyperactivation – compared to the VPA group.
control group (significant for the 6 mM concentration group in Histopathological analysis of the larvae exposed to carba-
Dark I). Conversely, in the groups exposed to higher concentra- mazepine (CBZ) showed a dose-dependent increase of a general
tions (≥9 mM), a concentration-related reduction in total Distance developmental arrest and a disrupted brain structure (Fig. 3).
Moved (DM15min ) was observed in both Dark I and Dark II, signif- At 240 ␮M, 27% of the larvae failed to hatch. In addition 18%
icant for the 12 mM ETH group. However, the swimming activity showed mild disruption of brain structure, characterized by areas
was slightly increased in the Light session in all concentrations of reduced cellularity (Fig. 3B, arrow). At 480 ␮M, 92% of the lar-
groups, significant for the 9 mM ETH group. Larvae exposed to vae failed to hatch (Fig. 3C) and one was necrotic (Fig. 3D). In
12 mM ETH also startled in response to the sudden Light stimulus addition, 25% showed mild disruption of normal brain structure,
(L-StR), whereas a response to the Darkness stimulus (D-StR) was characterized by areas of reduced cellularity. Pericardial edema was
marginal or nearly absent, thereby differing from the control group. occasionally detected in the 240 ␮M CBZ and 480 ␮M CBZ group.
Moreover, in groups of larvae exposed to ≥9 mM, we observed a A dose-dependent increase in the disruption of normal brain
significant reduction of the Darkness Startle Response (D-StR) and structure was observed in larvae exposed to ethosuximide (ETH)
of the Habituation slope of the Dark II period (DII-HSL). Similarly (Fig. 4A–D). In addition, minimal to mild hepatocellular vacuola-
to VPA and CBZ, larvae exposed to high concentrations ETH (15 tion was observed (Fig. 4F–H). At 6 mM ETH (Table 3), one larva
and 18 mM) were all excluded from the MA analysis, due to a high showed hepatocellular vacuolation and mild disruption of brain
incidence of malformations. structure, characterized by areas of reduced cellularity. At 9 mM
Although the total Distance Moved (DM15min ) in all groups of lar- ETH, 50% of the larvae showed mild disruption of brain structure
vae exposed to levetiracetam (LEV) (Fig. 1D) was reduced in Dark I (Fig. 4B). In addition, 58% of the larvae showed minimal to mild
and Dark II sessions when compared to the control group, only in hepatocellular vacuolation. At 12 mM ETH, all larvae showed mild
the 60 mM LEV concentration group this effect reached statistical disruption of brain structure, characterized by areas of reduced cel-
significance in the Dark I session. Activity in the ≥40 mM LEV con- lularity (Fig. 4C). In addition, 90% of the larvae showed minimal to
centration groups appeared consistently low throughout the three mild hepatocellular vacuolation. At 15 mM ETH, 50% of the larvae
sequential 15 min sessions and showed – unlike the control and showed pericardial edema. In addition, all larvae showed minimal
20 mM LEV groups – a relative increase in activity during Light. to mild hepatocellular vacuolation and mild to moderate disrup-
This increased activity (DM15min ) in Light differed significantly from tion of brain structure, characterized by areas of reduced cellularity
the control group in all groups ≥60 mM LEV. The Lightness Startle and/or apoptosis (Fig. 4D). Similar effects were also observed in lar-
Response (L-StR) was significantly increased at the 60 and 100 mM vae exposed to 18 mM ETH (Table 3), however, the percentage of
LEV concentration groups. It was noticed that in all these groups the larvae showing pericardial edema in this group was significantly
activity over time fluctuated more than the activity of the control higher (92%) compared to the 15 mM group.
and 20 mM LEV concentration groups, and fluctuated more than Histopathological analysis showed that, of all the compounds
usual (compare Fig. 1D with Fig. 1A–C). As a consequence of the studied, levetiracetam (LEV) had the lowest potency in terms of
reduced swimming activity, the Habituation Slope in Dark I (DI-HSL) embryotoxicity. Only at LEV concentration of ≥ 80 mM (Fig. 5),
was significantly reduced – or virtually absent – when larvae were minimal to mild hepatocellular vacuolation was observed. At
exposed to ≥40 mM LEV. The Darkness Startle Response (D-StR) was respectively 80 mM, 100 mM, 120 mM and 140 mM LEV, hepato-
significantly reduced even at the 40 mM LEV. cellular vacuolation was observed in 8%, 36%, 33% and 75% of the
larvae.
3.3. Histopathology
3.4. Kinetics
The histopathological findings of larvae exposed to the differ-
ent AEDs used in this study are listed in Table 3 and presented in The total body burden of larval zebrafish (96 hpf), as measured
Figs. 2–5. by UPLC, showed a dose-related increase for all tested compounds.
In agreement with the morphological analysis, the histopath- However, uptake efficiency (based on the slope of the curve) was
ology of the control larvae, revealed no abnormalities (Fig. 2A, different for each of the four compounds studied (Fig. 6). The
Table 3). Larvae exposed to valproic acid (VPA) revealed pericar- analysis revealed that carbamazepine (CBZ) had the most efficient
dial edema (Fig. 2B, arrow), non-hatching (Fig. 2C) and coagulation uptake (y = 0.621x) of all the AEDs studied, followed by valproic
(death) (Fig. 2D). In addition, a dose-dependent disruption of nor- acid (VPA) (y = 0.530x), ethosuximide (ETH) (y = 0.194x) and leve-
mal brain structure was observed (Fig. 2F–H). Already at 60 ␮M tiracetam (LEV) (y = 0.0118x). The internal concentrations for the
VPA, minimal disruption of normal brain structure was observed, established EC50 values for developmental toxicity at 96 hpf (see
characterized by small regions of reduced cellularity (Fig. 2F, Section 3.1 and Table 4) were 0.087 (VPA), 0.137 (CBZ), 1.286 (LEV)
arrows). At 150 ␮M VPA, all larvae showed mild disruption of and 1.726 (ETH) (nanomoles/larvae), respectively.
normal brain structure, characterized by large areas of reduced cel-
lularity, in particular in the developing preoptic region and tectum 3.5. Whole mount in situ hybridization
opticum (Fig. 2G, arrows). In addition, all larvae showed moderate
disruption of the retinal cell layers with signs of apoptosis (Fig. 2K). 3.5.1. Aldh1a2 expression
At 320 ␮M VPA, 75% of the larvae did not hatch. All larvae showed Fig. 7 shows Aldh1a2 expression in the control group (A), the
moderate disruption of normal brain structure (Fig. 2H), character- 320 ␮M valproic acid group (VPA) (B) and the 100 mM levetirac-
ized by large areas of reduced cellularity (arrow), disorganization etam group (LEV) (C) at all of the time points studied (24, 48, 72
and signs of apoptosis (Fig. 2H, insert, arrows head). In addition, the and 96 hpf). In the control groups of VPA and LEV, Aldh1a2 was
retinal layers were completely disrupted, showing severe apoptosis expressed in the retina, branchial arches and somites at 24 hpf.
and necrosis (Fig. 2L). At 730 ␮M VPA 55% of the larvae were dead At 48 hpf it was expressed in the retina, branchial arches, the
108 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

Table 3
Histopathological findings of zebrafish larvae exposed to anti-epileptic drugs (AEDs).

Valproic acid (␮M) 0 60 150 320 730

Number of larvae analyzed 12 12 12 12 11


Brain Minimal disruption of brain structure 0 1 0 0 0
Mild disruption of brain structure 0 0 12** 0 0
Moderate disruption of brain structure 0 0 0 12** 0
Severe disruption of brain structure 0 0 0 0 5*

Eye Moderate apotosis and disruption of retinal layers 0 0 12** 0 0


Severe apotosis and disruption of retinal layers 0 0 0 12** 1

Heart Pericardial edema 0 0 3 3 ND


**
General Not hatched 0 0 0 9 5*
Dead 0 0 0 0 6*

Carbamazepine (␮M) 0 30 60 120 180 240 480

Number of larvae analyzed 12 12 12 12 12 11 12


Brain Mild disruption of brain structure 0 0 0 0 0 2 3

Heart Pericardial edema 0 0 0 0 0 3 1

General Not hatched 0 0 0 0 0 3 11**


Dead 0 0 0 0 0 0 1

Ethosuximide (mM) 0 3 6 9 12 15 18

Number of larvae analyzed 12 12 12 12 10 10 12


Liver Minimal to mild hepatocellular vacuolation 0 0 1 7* 9** 10** 9**

Brain Mild disruption of brain structure 0 0 1 6* 10** 2 3


Moderate disruption of brain structure 0 0 0 0 0 8** 8**

Heart Pericardial edema 0 0 0 0 0 5* 11**

General Not hatched 0 0 0 0 0 0 1


Dead 0 0 0 0 0 0 1

Levetiracetam (mM) 0 20 40 60 80 100 120 140

Number of larvae analyzed 12 12 12 12 12 11 12 12


Liver Mild to moderate hepatocellular vacuolation 0 0 0 0 1 4# 4# 9**

General Not hatched 0 0 0 0 0 1 0 0

ND = not determined.
*
p < 0.01.
**
p < 0.0001.
#
p < 0.05.

tectum opticum, spinal cord motor neurons and cerebellum. At 72 and 96 hpf). In the control groups of VPA and LEV, Cyp26a1 was
72 hpf Aldh1a2 was weakly expressed in the tectum opticum and generally weakly expressed. The only clear evidence of Cyp26a1
strongly expressed in spinal cord motor neurons and the branchial expression was at 24 hpf, in the tail bud and posterior notochord. At
arches. At 96 hpf, expression was observed in spinal cord motor 96 hpf, no Cyp26a1 expression was observed in the control groups.
neurons, the tectum opticum and the branchial arches. Exposure to 320 ␮M VPA for 24 hpf (Fig. 8B) increased the
Aldh1a2 expression in larvae exposed to 320 ␮M VPA differed expression of Cyp26a1 as compared to control embryos, mainly at
from that seen in control larvae of the same age (Fig. 7B). At the tail bud and (weakly) at the branchial arches. An up-regulated
24 hpf, exposure to 320 ␮M VPA induced increased Aldh1a2 expres- expression of Cyp26a1 was also observed in the tail bud of larvae
sion in the somites and decreased expression in the branchial exposed to 150 ␮M and 730 ␮M VPA (data not shown). At 48 and
arches. At 48 and 72 hpf, compared to the control, a clear decrease 72 hpf, as with the control group, the expression of Cyp26a1 in lar-
in Aldh1a2 expression was observed, especially in the tectum vae exposed to VPA was less pronounced. At 96 hpf, no expression
opticum, branchial arches, spinal cord motor neurons and cere- of Cyp26a1 was observed in larvae exposed to VPA, as in the control
bellum. At 96 hpf, only weak levels of Aldh1a2 expression were larvae.
observed in the spinal cord motor neurons and branchial arches. Resembling the expression pattern of Aldh1a2 (Fig. 7C), expo-
In general, it can be stated that down-regulation of Aldh1a2 was sure to 100 mM of LEV (Fig. 8C) and to the other concentrations of
observed in larvae exposed to VPA, especially in those at 48 hpf or LEV studied (not shown) did not induce any visible alterations in
older. the expression of Cyp26a1.
When exposing larvae to 100 mM LEV (Fig. 7C) or to any of
the other concentrations studied, no clear difference in Aldh1a2 4. Discussion
expression was observed at any of the time points investigated (not
shown). The present study describes an integrated modular approach
to test developmental (neuro)toxicity using the embryonic/larval
3.5.2. Cyp26a1 expression zebrafish model. In general, this screening assay is based on mor-
Fig. 8 shows Cyp26a1 expression in the control group (A), the phological screening for developmental toxicity [13,14] and on a
320 ␮M valproic acid group (VPA) (B) and the 100 mM levetirac- motor activity assay to evaluate potential developmental neurotox-
etam group (LEV) (C) for each of the time points studied (24, 48, icity [18–20], in which the different endpoints are usually studied
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 109

Fig. 2. Histopathology of larvae exposed to valproic acid (VPA). (A) Control. (B) Histopathological analysis confirmed the morphological observations of pericardial edema
(arrow); (C) non-hatching (chorion; arrow) and (D) death. In addition, disruption of normal brain structure was observed, characterized by areas of reduced cell density (F,
G, H, black arrows), and signs of apoptosis (H-insert; black arrow heads). Histopathological analysis of the eye showed VPA-induced disorganization of the retinal layers and
signs of apoptosis (K, L). (0) Pigment epithelium; (1) photoreceptor cell layer; (2) inner nuclear layer; (3) inner plexiform layer; (4) ganglion cell layer; (5) lens. Magnification
A–D: 100×. Magnification E–H: 200×. Magnification I–L: 400×.

separately. We present an extension to – and a further integra- and mode of actions involved in the toxic effects observed. In this
tion of – this approach, applying morphological and motor activity study only a limited number of compounds was tested, therefore, in
screening in combination with histopathology and kinetics. order to be able to draw firm conclusions about the specificity and
The results of this demo-case study with a limited set of sensitivity of each of the assays applied in this study, an extended
compounds, showed the potency of integration of all measured panel of positive and negative compounds covering different mech-
endpoints (each with its own distinct specificity and sensitivity) anisms of action should be studied in future.
to enhance the predictability of the zebrafish assay. Further- By analyzing the larvae treated with valproic acid (VPA)
more, inclusion of in situ hybridization may well contribute histopathologically, we observed that 8% of them had reduced brain
to further elucidate underlying toxicity pathways by linking cellularity (in the optic regions in particular) already at 60 ␮M
structural/functional endpoints on the one hand and molecular whereas no effects were observed on morphology and motor activ-
gene-expression data on the other. Altogether, this may increase ity at this concentration. This indicates that, for VPA, histopathology
the reliability of the model for potency ranking of developmental appeared to be one of the most sensitive endpoint analyzed. Addi-
toxicity, while providing a better understanding of the processes tionally, at 150 ␮M VPA, an analysis of the motor activity (MA)

Fig. 3. Histopathology of larvae exposed to carbamazepine (CBZ). (A) Control. (B–D) Examples of CBZ-induced pathology showing disruption of normal brain structure,
characterized by reduced cell density (B; arrow); non-hatching (C) and death (D). Magnification A–D: 100×.
110 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

Fig. 4. Histopathology of larvae exposed to ethosuximide (ETH). (A) Control. (B and C) Histopathological analysis showed disruption of normal brain structure, characterized
by areas of reduced cell density (arrows), and (D) signs of apoptosis (black arrow heads). (E) Control liver. (F–H) Examples of different degrees of hepatocellular vacuolation
like minimal hepatocellular vacuolation (F), mild hepatocellular vacuolation (G and H). Magnification A1–D1: 100×. Magnification A2–D2: 200×. Magnification E–H: 200×.

test showed a significant reduction in total Distance Moved dur- darkness (D-StR), the observed changes in brain structure have also
ing both of the Darkness sessions and also on the Darkness Startle – at least to some extent – contributed to changes in motor activ-
Response (Table 4). These effects are indicative of neurodevelop- ity behavior, suggesting developmental neurotoxic potency of VPA.
mental toxicity [19]. Indeed, various adverse effects of VPA on The results of histopathology and motor activity, taken together,
neurodevelopment in rodents [48] and in humans (i.e. cognitive, once again, underline the importance of histopathology surveys to
behavior, verbal IQ) have been extensively described in the lit- understanding of the mode of toxic action and the importance of
erature [48–51]. By including the histopathological examination, integrating this technique into the ZET test.
indeed we were able to demonstrate that 100% of the examined In the case of embryos/larvae exposed to carbamazepine
larvae that had been exposed to ≥150 ␮M VPA exhibited reduced (CBZ), unlike VPA, histopathology does not appear to be the
brain cellularity (especially in the optic regions). However, we most sensitive endpoint. This compound only induced observ-
also observed severely damaged retinas. Clearly, in a design that able histopathological effects (reduced cellularity in the brain) at
involves light/darkness sessions (where the integrity of the retina high concentrations (≥240 ␮M). Instead, embryos/larvae exposed
is clearly of pivotal importance) the observed brain (optic region) to 180 ␮M CBZ already showed a delay in the onset of hatching
and retinal damage could well account for the failure of the larvae to at 72 hpf, and also a reduced Darkness Startle Response. This is in
react to changes in light intensity, i.e. no effect in Startle Response. agreement with the literature, in which general growth retardation
However, since there was a response to light (L-StR) – though not to was observed in rodents embryos exposed to CBZ [52]. Our results

Fig. 5. Histopathology of larvae exposed to levetiracetam (LEV). (A) Control liver. (B and C) Examples of the different degrees of hepatocellular vacuolation found in all larvae
exposed to ≥80 mM LEV, like mild hepatocellular vacuolation (B) and moderate hepatocellular vacuolation (C). Magnification A–C: 200×.
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 111

Fig. 6. Total body burden of zebrafish larvae exposed to anti-epileptic (AEDs) measured by ultra-performance liquid chromatography (UPLC). Abbreviations: VPA, valproic
acid; CBZ, carbamazepine; ETH, ethosuximide; LEV, levetiracetam.

are also in agreement with human data, as children exposed to hepatocellular vacuolation in 8% of the larvae at a concentration
CBZ in utero presented increased frequency of neurodevelopmental of just 6 mM ETH. At functional levels (MA test), larvae exposed to
disorders, compared to non-exposed children [50]. ETH showed a clear biphasic dose-response effect. At low concen-
The morphological examination of embryos/larvae exposed to trations (≤ 6 mM) an increase in the MA parameters (i.e. Distance
ethosuximide (ETH) showed developmental toxicity (pericardial Moved, Darkness Startle Response, etc.) was observed, while at
edema) at 72 hpf and 96 hpf, with respective EC50 levels of 9.9 mM higher concentrations (≥9 mM) the value of these parameters
and 8.9 mM. No developmental delay was observed at any of the decreased.
concentrations studied (the incidence of larvae that were hatched Observations of larvae exposed to levetiracetam (LEV) also
at 72 hpf and 96 hpf was not affected). However, histopatho- showed that multiple-endpoints are complementary to each
logy showed neurodegeneration (reduced brain cellularity) and other and that they increase the model’s potential for pinpointing

Table 4
Summarizing results.

Compound Morphologya Motor activity Histopathology Kineticsb (total body burden at EC50 at 96 hpf)

72 hpf 96 hpf

VPA Non-hatching Pericardial edema 150 ␮M 60 ␮M (8% larvae showing reduced (0.530 × 0.165) = 0.087 nmol/larvae
EC10 : 101 ␮M EC10 : 76 ␮M cellularity in brain)
EC20 : 135 ␮M EC20 : 101 ␮M 150 ␮M (100% larvae showing reduced
EC50 : 221 ␮M EC50 : 165 ␮M cellularity in brain and 100% showing
disruption of retinal cell layers)
CBZ Non-hatching Pericardial edema ≥180 ␮M 240 ␮M (18% larvae showing reduced (0.621 × 0.22c ) = 0.137 nmol/larvae
EC10 : 148 ␮M EC10 : 167 ␮M cellularity in brain)
EC20 : 163 ␮M EC20 : 185 ␮M 480 ␮M (25% larvae showing reduced
EC50 : 193 ␮M EC50 : 220 ␮M cellularity in brain)
ETH Pericardial Pericardial ≥6 mM 6 mM (8% larvae showing reduced cellularity (0.194 × 8.9) = 1.726 nmol/larvae
edema edema in brain and hepatocellular vacuolation)
EC10 : 6.6 mM EC10 : 6.6 mM 9 mM (50% larvae showing reduced
EC20 : 7.7 mM EC20 : 7.3 mM cellularity in brain and 58% showing
EC50 : 9.9 mM EC50 : 8.9 mM hepatocellular vacuolation)
12 mM (100% larvae showing reduced
cellularity in brain and 90% showing
hepatocellular vacuolation)
LEV Malformation Malformation ≥40 mM 80 mM (8% larvae showing hepatocellular (0.0118 × 109c ) = 1.286 nmol/larvae
tail tail vacuolation)
EC10 : 69 mM EC10 : 53 mM 120 mM (33% larvae showing hepatocellular
EC20 : 84 mM EC20 : 69 mM vacuolation)
EC50 : 118 mM EC50 : 109 mM 140 mM (75% larvae showing hepatocellular
vacuolation)

Abbreviations: VPA, valproic acid; CBZ, carbamazepine; ETH, ethosuximide; LEV, levetiracetam.
a
Here we also present the morphology EC10 and EC20 for a better comparison with the histopathological findings.
b
The internal concentrations (total body burden) were calculated, for each compound, by multiplying the uptake efficiency (slope of curve, Fig. 6) with the respective
morphology EC50 value at 96 hpf.
c
Observed EC50 concentration for CBZ (220 ␮M) and LEV (109 mM) were outside the range of concentrations used for total body burden measurements as shown in Fig. 6.
Consequently, the values shown in this table were based on extrapolation of the results shown in Fig. 6.
112 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

Fig. 7. Representative illustrations of Aldh1a2 expression in zebrafish embryos/larvae at 24, 48, 72 and 96 hpf. (A) Control; (B) embryos/larvae exposed to 320 ␮M VPA;
(C) embryos exposed to 100 mM LEV. (1) Retina; (2) branchial arches; (3) somites; (4) tectum opticum; (5) spinal cord motorneurons; (6) cerebellum; *a-specific staining;
Abbreviations: VPA, valproic acid; LEV, levetiracetam; n.d., not done.

the most sensitive parameter for toxicity. The most sensitive damage (histopathology) was observed at any of the concentra-
parameter, by far, for this compound was neither morphology nor tions studied. However, significant functional effects on motor
histopathology, but motor activity. In embryos/larvae that were activity (i.e. Darkness Startle Response) were observed already
exposed to LEV, effects on morphology (malformation tail) and at 40 mM. The remarkable fluctuations observed in swimming
histopathology (hepatocellular vacuolation) were only observed activity at concentrations of ≥60 mM LEV might represent early
at high concentrations (≥80 mM). Moreover, no structural brain alterations in tail morphology that go unrecognized during
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 113

Fig. 8. Representative illustrations of Cyp26a1 expression in zebrafish embryos/larvae at 24, 48, 72 and 96 hpf. (A) Control; (B) embryos/larvae exposed to 320 ␮M VPA; (C)
embryos exposed to 100 mM LEV. (1) Notochord; (2) tail bud; (3) branchial arches. Abbreviations: VPA, valproic acid; LEV, levetiracetam; n.d., not done.
114 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

morphology testing. It should be borne in mind that larvae with more toxic than LEV (EC50 VPA: 165 ␮M; LEV: 109 mM). However,
recognized tail aberrations were excluded from motor activity when taking the kinetics (TBB at EC50 at 96 hpf) into consideration,
analysis. This is also an example how integration of different the difference in potency falls from a factor of 660 to a factor of
parameters may help to elucidate the mode of actions underlying just 15 (TBB VPA: 0.087 nM/larvae; LEV: 1.286 nM/larvae) (Table 4
functional toxic effects. In this case, most probably, the effects on and Fig. 6). Most likely this is due to the fact that the uptake of
motor activity were related to effects on tail morphology whereas, LEV (y = 0.0118x) is 45 times lower than that of VPA (y = 0.530x).
for example, in case of VPA the effects on motor activity were The observation that, in the present study, the results of kinetic
related to histopathological effects on brain and eye. analysis did not lead to a change in the absolute ranking of AEDs,
Taking the results of all endpoints together (Table 4), we note was probably related to the wide range of EC50 values involved (see
that valproic acid (VPA) showed significant effects at concentra- Table 4).
tions ≥150 ␮M (yet histopathology reveals that 8% of the larvae In situ hybridization (ISH) was included in the present study in
studied showed a reduction in brain cellularity even at concentra- an attempt to phenotypically relate effects on gene expression pro-
tions as low as 60 ␮M VPA). With regard to carbamazepine (CBZ), files to histopathological findings. For this purpose, we first studied
clear effects were observed at concentrations ≥180 ␮M, while the spatial–temporal expression of Aldh1a2 and Cyp26a1, two
zebrafish embryos/larvae exposed to ethosuximide (ETH) showed important genes involved in the retinoic acid signaling pathway
significant effects only at millimolar concentrations (≥6 mM). Lev- and therefore involved in the embryo/fetal development. Further
etiracetam (LEV) showed the lowest developmental toxic potency we studied the possible involvement of those genes in the develop-
of the AEDs studied. In the MA test, significant effects were detected mental and neurodevelopmental effects of the most potent (VPA)
at concentrations ≥40 mM LEV, whereas significant effects on and least potent (LEV) AEDs (as ranked in this study). Therefore,
morphology and histopathology were only observed at higher con- we initially performed ISH on Formalin-Fixed Paraffin-Embedded
centrations (≥80 mM). No lethal effects whatsoever were observed (FFPE) serial sections. Unfortunately, we did not obtain consistent
at any of the concentrations studied (up to 140 mM). results, as the gene expression patterns detected by ISH in these
Therefore, based on the external dose exposure, our results sections appeared to be very localized. For instance, the expression
show that the AEDs can be ranked in terms of their developmental of Aldh1a2 was detected in only one or two slides out of an entire
toxicity/neurotoxicity potency as follows. The most potent com- series. The adjacent sections showed only mild expression, or none
pound is valproic acid (VPA), followed by carbamazepine (CBZ), at all. Accordingly, far too many sections would have to be ana-
then ethosuximide (ETH). Finally, levetiracetam (LEV) is the least lyzed to obtain absolute certainty about the expression of genes in
potent compound. The above AED developmental (neuro)toxicity histopathological sections and about how this is affected by test
ranking shows good agreement with results from in vivo studies compounds. The follow-up experiments were, therefore, carried
in animals (see review by Finnel and Dansky [31]) and humans. In out using whole mount in situ hybridization (WISH) (Fig. 7).
a recent human review study, Tomson and Battino [28] described One way in which VPA exerts its teratogenic effect is by inhib-
that exposure to monotherapy with valproate produced a higher iting histone deacetylases, by binding to the RA receptor [26]. We
rate of major congenital malformations (MCM) than was the case therefore investigated the effects of VPA and LEV (respectively, the
with carbamazepine. This is also in agreement with the results most and least potent AEDs studied) on the expression of the two
of a prospective study by Morrow et al. [53], who found that major genes involved in RA signaling. In control larvae, at 24 hpf, a
women receiving VPA monotherapy during pregnancy were at strong expression of Aldh1a2 was observed in the retina, branchial
greater risk of MCM than those given CBZ. Moreover, fetal expo- arches and somites. Concurrently with embryo development (in
sure to antiepileptic drugs at doses lower than those that result time), no further Aldh1a2 expression in the somites was observed
in structural malformations can produce behavioral and cogni- from 48 hpf onwards, while it was still occurring in neuronal tis-
tive deficits. Indeed, Meador and collaborators [54] reported that sue up to 96 hpf (Fig. 7). In contrast, control larvae only exhibited a
children exposed (in utero) to valproate had a reduced cognitive weak expression of Cyp26a1 at 24 hpf, and in the tail bud and pos-
outcome (low IQ score) compared those exposed to carbamazepine. terior notochord. In addition, unlike the pattern seen in Aldh1a,
Regarding ETH, our results are in agreement with Sullivan and col- no expression of Cyp26a1 was observed in any neuronal tissue
laborators [55], who performed a study on the ranking potency of from 48 hpf onwards. This is in agreement with Dobbs-McAuliffe
AEDs in mice. These workers found that ETH was less potent than et al. [58], who reported that, in early development, Cyp26a1
CBZ in inducing developmental effects. In the present study, LEV expression occurred either adjacent to, or opposite, tissues express-
was the least potent compound for developmental (neuro)toxicity. ing Aldh1a2. Interestingly, when zebrafish embryos/larvae were
This is in agreement with a recent study showing that LEV had lower exposed to VPA, they showed an up-regulation of Aldh1a2 during
MCM incidence than valproate when used as monotherapy dur- the first 24 hpf, especially in the somites. However, a general down-
ing pregnancy [36]. Moreover, children exposed (in utero) to LEV regulation was observed after this period (from 48 hpf onwards).
achieved significantly higher scores on a mental development scale Similarly to the Aldh1a2 pattern, the expression pattern of Cyp26a1
than those exposed to valproate [56]. in embryos exposed to VPA at 24 hpf also showed evidence of
Although uptake is generally dictated by a compound’s phys- up-regulation. In older embryos/larvae (≥48 hpf), the expression
icochemical properties [57], the potency of a compound in the pattern of Cyp 26a1 in the group exposed to VPA was similar to
zebrafish screening assay cannot be predicted from its aqueous con- that in the control group (i.e. down-regulated). Regarding the expo-
centration or log P value alone. For this reason, additional kinetic sure of zebrafish embryos/larvae to LEV, we did not observe any
analysis is required. In this study, the total body burden (TBB) of significant effects on the expression of Aldh1a2 and Cyp26a1 at
larvae was analyzed (Fig. 6). It was shown that whereas supplemen- any of the time-points studied. These results showed a relation
tation of the present approach with a TBB analysis (kinetics) left between effects on the expression of RA-related enzymes and apical
the absolute ranking (based on EC50 of morphological endpoints) endpoints (morphology, motor activity and histopathology). This
unchanged, it altered the relative potency significantly (Table 4). indicates that these molecular markers are very useful for pheno-
Accordingly, the addition of kinetics analysis provides the most type prediction.
relevant and important information on compound uptake, and on In conclusion, in this demo-case study the developmental toxic
the impact of this uptake in terms of the actual potency of the potential of a series of AEDs was studied at different levels,
compound. For instance, we showed that, based on EC50 of mor- i.e. the structural level (whole animal level by morphology and
phological endpoints (at 96 hpf), VPA was found to be 660 times organ/tissue/cellular level by histopathology), the functional level
A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116 115

(motor activity test) and molecular level (ISH). The results of the [13] Hermsen SAB, van den Brandhof E, van der Ven LTM, Piersma AH. Relative
study showed the potential of adding multiple endpoints in the embryotoxicity of two classes of chemicals in a modified zebrafish embryo-
toxicity test and comparison with their in vivo potencies. Toxicol In Vitro
zebrafish embryotoxicity test (ZET) that may increase the sensi- 2011;25:745–53.
tivity and predictability of the assay and may contribute to the [14] Padilla S, Corum D, Padnos B, Hunter DL, Beam A, Houck KA, et al. Zebrafish
elucidation of the mode of toxic action. Most probably, the inclu- developmental screening of the ToxCastTM Phase I chemical library. Reprod
Toxicol 2012;33:174–87.
sion of – omics – technology will further elucidate the mechanisms [15] Brannen KC, Panzica-Kelly JM, Danberry TL, Augustine-Rauch KA. Development
underlying developmental toxicity and deliver relevant biomark- of a zebrafish embryo teratogenicity assay and quantitative prediction model.
ers for developmental toxicity. In potency, this will further define Birth Defects Res B: Dev Reprod 2010;89:66–77.
[16] Selderslaghs IWT, Blust R, Witters HE. Feasibility study of the zebrafish assay
the applicability domain and increase the reliability of effects in
as an alternative method to screen for developmental toxicity and embryo-
the ZET. The inclusion of analysis of the uptake of test compounds toxicity using a training set of 27 compounds. Reprod Toxicol 2012;33:
to measure total body burden will refine the potency ranking of 142–54.
[17] Beker van Woudenberg A, Wolterbeek A, te Brake L, Snel C, Menke A, Rubingh
test compounds. In addition, especially where compounds induce
C, et al. A category approach to predicting the developmental (neuro)toxicity of
no effects, it will further increase the predictability of the assay by organotin compounds: the value of the zebrafish (Danio rerio) embryotoxicity
ruling out false negative results (and also false positive results, in test (ZET). Reprod Toxicol 2013;41:35–44.
cases of overexposure) in the zebrafish embryotoxicity test. [18] Irons TD, MacPhail RC, Hunter DL, Padilla S. Acute neuroactive drug expo-
sures alter locomotor activity in larval zebrafish. Neurotoxicol Teratol
2010;32:84–90.
Conflict of interest [19] de Esch C, van der Linde H, Slieker R, Willemsen R, Wolterbeek A, Woutersen R,
et al. Locomotor activity assay in zebrafish larvae: influence of age, strain and
ethanol. Neurotoxicol Teratol 2012;34:425–33.
None declared. [20] Selderslaghs IWT, Hooyberghs J, Blust R, Witters HE. Assessment of the devel-
opmental neurotoxicity of compounds by measuring locomotor activity in
zebrafish embryos and larvae. Neurotoxicol Teratol 2013;37:44–56.
Transparency document
[21] Shrader EA, Henry TR, Greeley Jr MS, Bradley BP. Proteomics in zebrafish
exposed to endocrine disrupting chemicals. Ecotoxicology 2003;12:
The Transparency document associated with this article can be 485–8.
found in the online version. [22] Hermsen SAB, Pronk TE, van den Brandhof E-, van der Ven LTM, Piersma AH.
Transcriptomic analysis in the developing zebrafish embryo after compound
exposure: individual gene expression and pathway regulation. Toxicol Appl
Acknowledgements Pharmacol 2013;272:161–71.
[23] Van den Bulck K, Hill A, Mesens N, Diekman H, De Schaepdrijver L, Lammens L.
Zebrafish developmental toxicity assay: a fishy solution to reproductive toxic-
Authors would like to thank Mieke Havekes and Hakim Rahaoui ity screening, or just a red herring. Reprod Toxicol 2011;32:213–9.
(TNO, Zeist, The Netherlands) for assembling the RNA probes [24] Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Histone deacety-
for ISH, Carina Rubingh and Ruud Boessen (TNO, Zeist, The lase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer,
and teratogen. J Biol Chem 2001;276:36734–41.
Netherlands) for the statistics support and graphic layout. We [25] Lloyd KA. A scientific review: mechanisms of valproate-mediated teratogene-
would also like to thank Gerard van Beek and Dick Veldhuysen sis. Biosci Horizons 2013;6.
(Animal Facilities TNO-Triskelion, Zeist, The Netherlands) for tak- [26] Menegola E, Di Renzo F, Broccia ML, Giavini E. Inhibition of histone deacetylase
as a new mechanism of teratogenesis. Birth Defects Res C: Embryo Today Rev
ing care of the zebrafish. This work was partially financed by the 2006;78:345–53.
Netherlands Toxicogenomics Center (NTC). [27] Rhinn M, Dollé P. Retinoic acid signalling during development. Development
2012;139:843–58.
[28] Tomson T, Battino D. Teratogenic effects of antiepileptic drugs. Lancet Neurol
References 2012;11:803–13.
[29] Ornoy A. Neuroteratogens in man: an overview with special emphasis
[1] Kultima K, Jergil M, Salter H, Gustafson A-L, Dencker L, Stigson M. Early on the teratogenicity of antiepileptic drugs in pregnancy. Reprod Toxicol
transcriptional responses in mouse embryos as a basis for selection of 2006;22:214–26.
molecular markers predictive of valproic acid teratogenicity. Reprod Toxicol [30] Samrén EB, Van Duijn CM, Koch S, Hiilesmaa VK, Klepel H, Bardy AH, et al.
2010;30:457–68. Maternal use of antiepileptic drugs and the risk of major congenital malforma-
[2] Marx-Stoelting P, Adriaens E, Ahr H-J, Bremer S, Garthoff B, Gelbke H-P, et al. A tions: a joint European prospective study of human teratogenesis associated
review of the implementation of the embryonic stem cell test (EST). Altern Lab with maternal epilepsy. Epilepsia 1997;38:981–90.
Anim 2009;37:313–28. [31] Finnell RH, Dansky LV. Parental epilepsy, anticonvulsant drugs, and repro-
[3] West PR, Weir AM, Smith AM, Donley ELR, Cezar GG. Predicting human devel- ductive outcome: epidemiologic and experimental findings spanning three
opmental toxicity of pharmaceuticals using human embryonic stem cells and decades; 1: Animal studies. Reprod Toxicol 1991;5:281–99.
metabolomics. Toxicol Appl Pharmacol 2010;247:18–27. [32] Pennell PB, Klein AM, Browning N, Baker GA, Clayton-Smith J, Kalayjian LA,
[4] Piersma AH, Bosgra S, van Duursen MBM, Hermsen SAB, Jonker LRA, Kroese ED, et al. Differential effects of antiepileptic drugs on neonatal outcomes. Epilepsy
et al. Evaluation of an alternative in vitro test battery for detecting reproductive Behav 2012;24:449–56.
toxicants. Reprod Toxicol 2013;38:53–64. [33] Fabro S, Shull G, Brown NA. The relative teratogenic index and teratogenic
[5] Breier JM, Gassmann K, Kayser R, Stegeman H, De Groot D, Fritsche E, et al. Neu- potency: proposed components of developmental toxicity risk assessment.
ral progenitor cells as models for high-throughput screens of developmental Teratog Carcinog Mutagen 1982;2:61–76.
neurotoxicity: state of the science. Neurotoxicol Teratol 2010;32:4–15. [34] Ponnusamy R, Pradhan N. The effects of chronic administration of ethosuximide
[6] Schenk B, Weimer M, Bremer S, van der Burg B, Cortvrindt R, Freyberger A, on learning and memory: a behavioral and biochemical study on nonepileptic
et al. The ReProTect Feasibility Study, a novel comprehensive in vitro approach rats. Behav Pharmacol 2006;17:573–9.
to detect reproductive toxicants. Reprod Toxicol 2010;30:200–18. [35] Long L. Levetiracetam monotherapy during pregnancy: a case series. Epilepsy
[7] Lieschke GJ, Currie PD. Animal models of human disease: zebrafish swim into Behav 2003;4:447–8.
view. Nat Rev Genet 2007;8:353–67. [36] Mawhinney E, Craig J, Morrow J, Russell A, Smithson WH, Parsons L, et al.
[8] Barros TP, Alderton WK, Reynolds HM, Roach AG, Berghmans S. Zebrafish: an Levetiracetam in pregnancy: results from the UK and Ireland epilepsy and
emerging technology for in vivo pharmacological assessment to identify poten- pregnancy registers. Neurology 2013;80:400–5.
tial safety liabilities in early drug discovery. Br J Pharmacol 2008;154:1400–13. [37] Westerfield M. The zebrafish book: a guide for the laboratory use of
[9] Sukardi H, Chng HT, Chan ECY, Gong Z, Lam SH. Zebrafish for drug toxicity zebrafish (Danio rerio). 5th ed. Eugene, OR, USA: University of Oregon Press;
screening: bridging the in vitro cell-based models and in vivo mammalian 2007.
models. Expert Opin Drug Metabol Toxicol 2011;7:579–89. [38] OECD. Guideline for the testing of chemicals. Draft proposal for a new guideline
[10] Hill A. Zebrafish in drug discovery: safety assessment. In: Vogel HG, Fish Embryo Toxicity (FET) test; 2006.
Maas J, Hock FJ, Mayer D, editors. Drug discovery and evaluation: safety [39] European Commission (EC). EU. Directive 2010/63/EU of the European parlia-
and pharmacokinetic assays. Heidelberg: Springer Berlin; 2013. p. 605–29, ment and of the council of 22 September 2010 on the protection of animals
http://dx.doi.org/10.1007/978-3-642-25240-2 22. used for scientific purposes.; 2010. p. 33–79.
[11] Hill AJ, Teraoka H, Heideman W, Peterson RE. Zebrafish as a model vertebrate [40] Berghmans S, Hunt J, Roach A, Goldsmith P. Zebrafish offer the potential for a
for investigating chemical toxicity. Toxicol Sci 2005;86:6–19. primary screen to identify a wide variety of potential anticonvulsants. Epilepsy
[12] Padilla S. Zebrafish development: high-throughput test systems to assess Res 2007;75:18–28.
developmental toxicity. In: Steinberg P, editor. High-throughput screening [41] Weigt S, Huebler N, Strecker R, Braunbeck T, Broschard T. Zebrafish (Danio rerio)
methods in toxicity testing. USA: John Wiley & Sons, Inc.; 2013. p. 371–83. embryos as a model for testing proteratogens. Toxicology 2011;281:25–36.
116 A. Beker van Woudenberg et al. / Reproductive Toxicology 49 (2014) 101–116

[42] Fraysse B, Mons R, Garric J. Development of a zebrafish 4-day embryo-larval [51] Rihtman T, Parush S, Ornoy A. Developmental outcomes at preschool age after
bioassay to assess toxicity of chemicals. Ecotoxicol Environ Saf fetal exposure to valproic acid and lamotrigine: cognitive, motor, sensory and
2006;63:253–67. behavioral function. Reprod Toxicol 2013;41:115–25.
[43] DIN. Basivalidierung genormter verfahre zur wasser-,abwasser-,und schlam- [52] Piersma AH, Verhoef A, Opperhuizen A, Klaassen R, Van Eijkeren J, Olling
muntersuchung validierungsdokument (38415-T6); 2002. p. 27. M. Embryotoxicity of carbamazepine in rat postimplantation embryo culture
[44] Nau H, Tzimas G, Mondry M, Plum C, Spohr H-L. Antiepileptic drugs alter after in vitro exposure via three different routes. Reprod Toxicol 1998;12:
endogenous retinoid concentrations: a possible mechanism of teratogenesis 161–8.
of anticonvulsant therapy. Life Sci 1995;57:53–60. [53] Morrow J, Russell A, Guthrie E, Parsons L, Robertson I, Waddell R, et al.
[45] Duester G. Retinoic acid synthesis and signaling during early organogenesis. Malformation risks of antiepileptic drugs in pregnancy: a prospective study
Cell 2008;134:921–31. from the UK Epilepsy and Pregnancy Register. J Neurol Neurosurg Psychiatry
[46] Chuang C-M, Chang C-H, Wang H-E, Chen K-C, Peng C-C, Hsieh C-L, et al. Val- 2006;77:193–8.
proic acid downregulates RBP4 and elicits hypervitaminosis a-teratogenesis—a [54] Meador KJ, Baker GA, Browning N, Clayton-Smith J, Combs-Cantrell DT, Cohen
kinetic analysis on retinol/retinoic acid homeostatic system. PLoS ONE 2012: M, et al. Cognitive function at 3 years of age after fetal exposure to antiepileptic
7. drugs. N Engl J Med 2009;360:1597–605.
[47] Thisse C, Thisse B. High-resolution in situ hybridization to whole-mount [55] Sullivan FM, McElhatton PR. A comparison of the teratogenic activity of the
zebrafish embryos. Nat Protoc 2008;3:59–69. antiepileptic drugs carbamazepine, clonazepam, ethosuximide, phenobarbital,
[48] Kaindl AM, Asimiadou S, Manthey D, Hagen MVD, Turski L, Ikonomi- phenytoin, and primidone in mice. Toxicol Appl Pharmacol 1977;40:365–78.
dou C. Antiepileptic drugs and the developing brain. Cell Mol Life Sci [56] Shallcross R, Bromley RL, Irwin B, Bonnett LJ, Morrow J, Baker GA. Child
2006;63:399–413. development following in utero exposure: levetiracetam vs sodium valproate.
[49] Roullet FI, Lai JKY, Foster JA. In utero exposure to valproic acid and Neurology 2011;76:383–9.
autism—a current review of clinical and animal studies. Neurotoxicol Teratol [57] Diekmann H, Hill A. ADMETox in zebrafish. Drug Discov Today: Dis Models
2013;36:47–56. 2013;10:e31–5.
[50] Dean JCS, Hailey H, Moore SJ, Lloyd DJ, Turnpenny PD, Little J. Long term health [58] Dobbs-McAuliffe B, Zhao Q, Linney E. Feedback mechanisms regulate retinoic
and neurodevelopment in children exposed to antiepileptic drugs before birth. acid production and degradation in the zebrafish embryo. Mech Dev
J Med Genet 2002;39:251–9. 2004;121:339–50.

You might also like