You are on page 1of 8

Epilepsy Research 138 (2017) 124–131

Contents lists available at ScienceDirect

Epilepsy Research
journal homepage: www.elsevier.com/locate/epilepsyres

Phenylalanine derivatives with modulating effects on human α1-glycine T


receptors and anticonvulsant activity in strychnine-induced seizure model in
male adult rats

Bassem Sadeka, , Murat Oza,b, Syed M. Nurulaina,c, Petrilla Jayaprakasha, Gniewomir Lataczd,
Katarzyna Kieć-Kononowiczd, Ewa Szymańskad
a
Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, P.O. Box 17666, United Arab
Emirates
b
Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
c
Department of Bioscience, COMSATS Institute of Information Technology, Islamabad 45550, Pakistan
d
Department of Technology and Biotechnology of Drugs Jagiellonian University Medical College, Medyczna 9, PL 30-688 Krakow, Poland

A R T I C L E I N F O A B S T R A C T

Keywords: The critical role of α1-glycine receptor (α1-GLYRs) in pathological conditions such as epilepsy is well known. In
Human α1-glucine receptor the present study, structure-activity relations for a series of phenylalanine derivatives carrying selected hydrogen
Phenyl alanine derivatives bond acceptors were investigated on the functional properties of human α1-GLYR expressed in Xenopus oocytes.
Inhibitor The results indicate that one particular substitution position appeared to be of special importance for control of
Potentiator
ligand activity. Among tested ligands (1-8), the biphenyl derivative (2) provided the most promising
Anticonvulsant
Strychnine
antagonistic effect on α1-GLYRs, while its phenylbenzyl analogue (5) exhibited the highest potentiation effect.
Moreover, ligand 5 with most promising potentiating effect showed in-vivo moderate protection when tested in
strychnine (STR)-induced seizure model in male adult rats, whereas ligand 2 with highest antagonistic effect
failed to provide appreciable anti(pro)convulsant effect. Furthermore, ligands 2 and 5 with the most promising
effects on human α1-GLYRs were examined for their toxicity and potential neuroprotective effect against
neurotoxin 6-hydroxydopamine (6-OHDA). The results show that ligands 2 and 5 possessed neither significant
antiproliferative effects, nor necrotic and mitochondrial toxicity (up to concentration of 50 μM). Moreover,
ligand 2 showed weak neuroprotective effect at the 50 μM against 100 μM toxic dose of 6-OHDA. Our results
indicate that modulatory effects of ligands 2 and 5 on human α1-GLYRs as well as on STR-induced convulsion
can provide further insights for the design of therapeutic agents in treatment of epilepsy and other pathological
conditions requiring enhanced activity of inhibitory glycine receptors.

1. Introduction motor-control, and pain sensation (Callister and Graham, 2010; Camp
et al., 2010). In recent studies, the critical role of GLYRs in modulating
In central, as well as peripheral, nervous system, GLY functions as a temporal lobe epilepsy has been proposed based on clinical findings
neurotransmitter at inhibitory synapses, where it activates strychnine- that patients suffering from temporal lobe epilepsy disease would
sensitive glycine receptors (GLYRs) which are composed of five possibly be advantaged most from facilitated glycinergic inhibition,
subunits (α1-4 and β) (Lynch, 2004) and belong to pentameric Cys- e.g. through GLYR modulators. Accordingly, mounting evidences have
loop ligand-gated ion channel superfamily (Cys-loop LGICs)(Castro accentuated the contribution of GLYRs to tonic hippocampal inhibition
et al., 2012; Lester et al., 2004). in response to glycine, and glycine and taurine as well as glycine uptake
It has been well established that GLYRs play important roles in inhibitors were found to be of anticonvulsive role in epilepsy models
various pathological conditions such as epilepsy, hyperexcitability, (Bedet et al., 2006; Eichler et al., 2008; Meier and Grantyn, 2004; Song

Abbreviations: ANOVA, analysis of variance; BAPTA, 1,2-bis(o-aminophenoxy)ethane-N,N,N9,N9-tetraacetic acid; DA, dopamine; DMSO, dimethylsulfoxide; MBS, Barth’s solution;
nAChR, nicotinic acetylcholine receptor; α1-GLYRs, α1-glycine receptor; ROS, radical oxygen species; 6-OHDA, 6-hydroxydopamine; ATP, adenosine triphosphate; HEK-293, human
embryonic kidney cell line; DMEM, Dulbecco’s Modified Eagle’s Medium; STR, strychnine; VPA, valproic acid; AED, antiepileptic drug; AMPA, (RS)-2-amino-3-(3-hydroxy-5-methyl-4-
isoxazolyl)propionic acid; KA, kainite; NMDA, N-methyl-D-aspartic acid; GluK1, homomeric kainate receptor

Corresponding author.
E-mail address: bassem.sadek@uaeu.ac.ae (B. Sadek).

http://dx.doi.org/10.1016/j.eplepsyres.2017.05.008
Received 22 February 2017; Received in revised form 1 May 2017; Accepted 19 May 2017
Available online 26 May 2017
0920-1211/ © 2017 Elsevier B.V. All rights reserved.
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

et al., 2006; Zhang et al., 2008). Also, STR-sensitive glycine receptors described procedures (Szymanska et al., 2011, 2009). Stock solutions
were previously found to depress hyperexcitability in rat dentate gyrus of the respective test compound used in this study were prepared in
(Eichler et al., 2008; Kirchner et al., 2003; Mori et al., 2002). Based on saline at a concentration of 10 mM. The cDNA plasmids for human α1-
the aforementioned role of GLYRs in central and peripheral nervous glycine receptor expression were kindly provided by Prof. Joe Lynch
system pathologies, and as a continuation of our research (Szymanska (The University of Queensland, Brisbane, Australia). Capped cRNA
et al., 2011, 2009), in the present study effects of a series of unnatural transcripts were synthesized in vitro using a mMESSAGE mMACHINE kit
racemic phenylalanine derivatives (1–8) on the functional properties of from Ambion (Austin, TX, USA) and analyzed on a 1.2% formaldehyde
the cloned human α1-GLYR expressed in Xenopus oocytes were agarose gel to check the size and quality of the transcripts.
investigated. Moreover, selected compounds have been examined for
their anti(pro)convulsant activity in STR-induced seizure model in 2.2. Data analysis
adult rats.
In addition, the most interesting phenylalanine derivatives were Average values were calculated as the mean of 6–8
tested for their toxicity using antiproliferative EZ4U test as well as for experiments ± SEM. Throughout this study, n defines the number of
their potential neuroprotective properties in cell-death in vitro model oocytes or number of samples tested in each experiment. Statistical
using neurotoxin 6-OHDA. Cytotoxicity of 6-OHDA is believed to result significance was analyzed using paired t-test. The criterion for statis-
from radical oxygen species (ROS) derived by 6-OHDA intraneuronal tical significance was set at a P value of less than 0.05.
autooxidation as well as a possible direct effect of 6-OHDA on the
mitochondrial respiratory chain (Rodriguez-Pallares et al., 2007), and 2.3. Cell line studies
recent studies demonstrated the direct association of mitochondrial
dysfunction and oxidative stress with epileptogenesis and acquired Human embryonic kidney HEK-293 cell line (ATCC CRL1573) was
chronic epilepsy (Blum-Degen et al., 1998; Waldbaum and Patel, 2010). kindly donated by Prof. Dr. Christa Müller (Pharmaceutical Institute,
In this context, the new multi-target acting anti-epileptic compounds Pharmaceutical Chemistry I, University of Bonn, Germany).
which would act not only via epilepsy-relevant receptors but also Neuroblastoma IMR-32 cell line was provided by Department of
possessing neuroprotective activity preventing mitochondria from Oncogenomics, Academisch Medisch Centrum, Amsterdam, Holland
ROS may contribute towards development of the therapy of epilepsy. (Cheng et al., 1995).

2. Materials and methods 2.3.1. Antiproliferative assay against HEK-293


The HEK-293 cells were seeded in 96-well flat-bottomed microtiter
2.1. Human α1-glycine receptor recordings from oocytes plate at a concentration of 1 × 104 cells/well and cultured in 200 μl/
well of Dulbecco’s Modified Eagle’s Medium − DMEM (Gibco,
Mature female Xenopus laevis frogs were purchased from Xenopus Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS),
Express (Haute-Loire, France), housed in dechlorinated tap water at 100 mg mL−1 streptomycin and 100 U mL−1 penicillin in a humidified
19–21 °C with a 12/12-h light/dark cycle, and fed food pellets supplied atmosphere (5% CO2, 95% air) at 37 °C for 24 h to reach 60%
by Xenopus Express. The procedures followed in this study were in confluence. The stock solutions of 2 and 5 in DMSO (25 mM) were
accordance with the Guide for the Care and Use of Laboratory Animals diluted into the fresh growth medium and added to the wells in the
(8th edition) of the National Institutes of Health (Bethesda, MD) and concentration range 0.1–100 μM. Final DMSO concentration did not
approved by the Institutional Animal Care and Use Committee at the exceed 1%. After 48 h of incubation, the EZ4U (Biomedica, Vienna,
UAEU. Austria) labeling mixture (20 μl) was added to each well and the cells
As described previously by Sadek et al., 2014 (Ashoor et al., were incubated under the same conditions for 5 h. The absorbance of
2013a,b; Mahgoub et al., 2013; Sadek et al., 2012, 2014a, 2015a), the samples was measured using a microplate reader (EnSpire,
clusters of oocytes were removed surgically under benzocaine (Sigma, PerkinElmer, Waltham, MA USA) at 492 nm. The reference compound
St. Louis, MO) local anesthesia (0.15% w/V), and individual oocytes doxorubicin (DX) was used during this study as a cytotoxic standard
were dissected manually in a solution containing (in mM): NaCl, 88; against HEK-293 cells.
KCl, 1; NaHCO3, 2.4; MgSO4, 0.8; HEPES, 10 (pH 7.5). Dissected
oocytes were then stored 2–7 days in modified Barth’s solution (MBS) 2.3.2. Antiproliferative assay against IMR-32
containing (in mM): NaCl, 88; KCl, 1; NaHCO3, 2.4; CaCl2, 2; MgSO4, The IMR-32 cell line was seeded in 96-well flat-bottomed microtiter
0.8; HEPES, 10 (pH 7.5), supplemented with sodium pyruvate, 2 mM, plate at a concentration of 1 × 104 cells/well and incubated under the
penicillin, 10,000 IU/L, streptomycin, 10 mg/L, gentamicin, 50 mg/L, same conditions as described above. The stock solutions of 2, 5 and 6-
and theophylline, 0.5 mM. Briefly, oocytes were placed in a 0.2 ml OHDA in DMSO (25 mM) were diluted into the fresh growth medium
recording chamber and superfused at a rate of 2–3 ml/min. The bathing and added to the cells in concentration range 1–100 μM and incubated
solution consisted of (in mM): NaCl, 95; KCl, 2; CaCl2, 2; and HEPES 5 for 48 h. Final DMSO concentration did not exceed 1%. Next, the EZ4U
(pH 7.5). The cells were impaled with two glass microelectrodes filled assay was performed in the same way as described above.
with a 3 M KCl. The oocytes were routinely voltage clamped at a
holding potential of −70 mV using a GeneClamp-500 amplifier (Axon 2.3.3. Neuroprotection test
Instruments Inc., Burlingame, CA). Glycine-induced currents were The Mitochondrial ToxGlo™ Assay and protocols were provided by
evoked every 5 min by 3–4 s application of 30 μM glycine. During Promega (Madison, WI, USA). The neuroblastoma IMR-32 cell line was
10 min application time, strychnine or series of phenylalanine deriva- seeded in 96-well white microtiter Plate 24 h before experiments at a
tives were applied externally by addition to the superfusate. These concentration 100 000 cells/ml. The cells were incubated in 200 μl of
compounds (strychnine and series of phenylalanine derivatives) were DMEM (Gibco, Invitrogen Ltd, Paisley, UK) supplemented with 10%
also included in glycine containing agonist solution.All chemicals used FBS in a humidified atmosphere (5% CO2) at 37 °C. Next, the old media
in preparing the solutions were from Sigma-Aldrich (St. Louis, MO, were removed and the 2, 5 at the concentrations 50 μM, and 6-OHDA at
USA) whereas test compounds 1–8 were synthesized as racemates in the the concentration 100 μM dissolved in DMEM supplemented with 10%
Department of Drug Design and Pharmacology, Faculty of Health and FBS and 4.5 g/L glucose were added to the plate. The cells were
Medical Sciences, University of Copenhagen/Denmark or in the Depart- incubated at 37 °C in a humidified and CO2-supplemented incubator for
ment of Technology and Biotechnology of Drugs, Jagiellonian Uni- 90 min. After addition of 20 μl fluorescence-based Cytotoxicity Reagent
versity Medical College, Kraków/Poland, according to previously (Promega) the plate was mixed (500 rpm) and incubated at 37 °C (5%

125
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

Fig. 1. Chemical structures of phenylalanine derivatives tested in the current study.

CO2) for next 30 min. The fluorescence was measured at 485 nmEx/ et al., 2014b, 2016a,b,c, 2015b, 2014c, 2013; Sadek and Stark, 2015).
530 nmEm by microplate reader (EnSpire, PerkinElmer, Waltham, MA First, different doses of the proconvulsant compound STR have been
USA) The 65 μM of digitonin (DGT) was used as a positive necrosis examined (in six to eight rats per dose) to define the minimum dose that
control. Then, the assay plate was adjusted to room temperature induces convulsions in all treated animals (Branco Cdos et al., 2012;
(5–10 min) and 100 μl of Adenosine triphosphate (ATP) Detection Coppola et al., 2010; Loscher, 2011; Sancheti et al., 2013; Sowemimo
Reagent (Promega) was added to each well. After orbital shaking et al., 2011). This minimal proconvulsant dose was then applied to
(500–700 rpm) for 1–5 min, the bioluminescence was measured by screen potential activities of the test compounds 2 and 5. Consequently,
microplate reader (EnSpire, PerkinElmer, Waltham, MA USA). STR (3.5 mg/kg, i.p.) was administered to all experimental test groups
(six to eight rats per group), ie, saline and treated rats. Saline, VPA
2.4. Data analysis (300 mg/kg, i.p.) (Loscher, 2011) or tested ligands 2 and 5 (5, 10, and
15 mg/kg, i.p.) were injected 30 min prior to STR (3.5 mg/kg, i.p.)
Statistical significance was analyzed by GraphPad Prism™ software administration, and rats were immediately observed for 30 min (ex-
(version 5.01, San Diego, CA, USA) using One-way ANOVA and periment period) for any convulsive signs. The convulsion signs were
Bonferroni's Multiple Comparison Post Test. The experiments were observed, and graded scores have been used to assess the severity of
performed in triplicate wells. Statistical significance was set at a P value convulsions according to the following scale: score 0 = no seizures,
of less than 0.05. score 1 = eye or facial twitches, score 2 = convulsive waves across the
body, score 3 = myo-clonic jerks or rearing, score 4 = turn over on to
one side position, and score 5 = turn over on to back position,
2.5. In-vivo STR-induced seizure generalized tonic-clonic seizures, or die during the experiment period.
The animals were divided into groups of seven rats. Animals in the
2.5.1. Animals positive control group were injected with VPA at a dose of 300 mg/kg,
Inbred male Wistar rats (Central Animal Facility of the UAE this being the minimal dose of VPA that protected animals against STR-
University) of body weight 180–220 g were used for this study. The induced seizures without mortality in rats. Animals in the experimental
animals were retained in an air-conditioned animal facility room with groups were administered test compounds at doses of 5, 10, or 15 mg/
controlled temperature (24 °C ± 2 °C) and humidity (55% ± 15%) kg, i.p., 30 min before the STR treatment. The dose of STR used in this
under a 12-h light/dark cycle. study formed convulsions (score 4–5) in 100% of animals with
The animals were allowed free access to food and water. The mortality, and this dose was used according to previously described
experiments of this study were carried out between 9 and 12 AM, and experimental protocols (Sadek et al., 2016a,c, 2015b; Serdiuk et al.,
all procedures were performed according to the guidelines of the 2014). In further two control experiments, two groups of seven animals
European Communities Council Directive of November 24, 1986 (86/ were injected with test ligands 2 and 5 (5, 10, and 20 mg/kg, i.p.)
609/EEC) and were previously approved for epilepsy study by the 30 min prior to saline administration, and rats were immediately
College of Medicine and Health Sciences, United Arab Emirates observed for 30 min (experiment period) for any convulsive signs.
University (Institutional Animal Ethics Committee, approval number;
A9-14). In the present study and according to previously used experi-
mental procedures, STR was utilized to induce convulsions (Sadek

126
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

Fig. 2. Effect of phenyl alanine derivatives (1–8) on human α1-glycine receptor-mediated


ion currents. GLY, glycine; STR, strychnine. Comparison of the extent of inhibition caused
by 30 μM glycine (GLY), 10 μM of strychnine (STR), and 10 μM of each test compound
after 10 min application time. Each bar represents the normalized mean ± SEM from 6 to
8 cells. **P < 0.005.

2.5.2. Data analysis


For statistical comparisons, the software package SPSS 20.0 (IBM
Middle East, Dubai, UAE) was used. All data were expressed as the
mean ± standard error of the mean. The effects of test compounds on
epilepsy were analyzed using One-way analysis of variance (ANOVA)
with Treatment (vehicle or test compound) and Dose (test compound)
as the between-subjects factor. In case of a significant main effect, post
hoc comparisons were performed with Bonferroni’s test. The criterion
for statistical significance was set at a P-value of < 0.05.

Fig. 3. α1-GLYR-mediated ion currents is blocked by ligand 2 and potentiated by ligand


3. Results 5. (A) Records of currents activated by glycine (GLY, 30 μM) in control conditions (left),
during co application of 10 μM STR and GLY (30 μM) after10 min pretreatment
3.1. Effects of ligands 1-8 on α1-GLYR-mediated ion currents with10 μM STR (middle), and15 min following STR washout (right). (B) Records of
currents activated by glycine (GLY, 30 μM) in control conditions (left), during co
In un-injected oocytes (n = 5), glycine (GLY) at the highest application of 10 μM (2) and GLY after10 min pretreatment with 10 μM (2) (middle),
and15 min following (2) washout (right). (C) Records of currents activated by glycine
concentration used in this study (30 μM) did not cause detectable
(GLY, 30 μM) in control conditions (left), during co application of 10 μM (5) and GLY
currents (data not shown). On the other hand, application of 30 μM GLY after10 min pretreatment with 10 μM (5) (middle), and 15 min following (5) washout
for 3–4 s induced rapidly activated inward-currents in oocytes injected (right).
with cRNA encoding the human α1-GLYR. These α1-GLYR-mediated
inward-currents were completely inhibited by 10 min bath application
of 10 μM STR (n = 7) (Figs. 2 and 3 A ), indicating that currents were
mediated by the activation of α1-GLYR. Effects of phenylalanine
derivatives 1–8 at a concentration of 10 μM on the function of α1-
GLYR are shown in Fig. 2. The results show that ligands 1–8 did not
induce any detectable current, when they were applied alone for 10 min
at the concentration of 10 μM (n = 7–9). Ligands 1, 3, 4, 6–8 in the
concentration of 10 μM did not cause a significant change in the
maximum amplitudes of α1-GLYR-mediated currents (Fig. 2). However,
10 min bath application of ligand 2 significantly inhibited these
currents (Figs. 2 and 3 B). Interestingly, ligand 5 when tested in the
same concentration (10 μM) for 10 min significantly potentiated the
function of GLYR (Figs. 2 and 3 C).

Fig. 4. Antiproliferative test against normal human HEK-293 cell line. Values represent
3.2. Antiproliferative assay against HEK-293 the mean of n = 4 experiments. ***P < 0.001.

To evaluate in vitro toxicity of ligands 2 and 5 the normal human 3.3. Antiproliferative assay against IMR-32
embryonic kidney HEK-293 cell line and formazan based EZ4U anti-
proliferative assay were used. The cells were incubated with the To determine the neurotoxicity and most suitable concentrations for
presence of ligand 2 and ligand 5 in the concentration range further neuroprotection studies, the antiproliferative effect of neuro-
0.1–100 μM for 48 h. The cytostatic compound DX was used as a toxin 6-OHDA and selected ligands 2, 5 against neuroblastoma IMR-32
reference. As a result, no antiproliferative activity of compound 5 was cells was also studied using EZ4U assay. The 6-OHDA, and phenylala-
observed. For compound 2, very weak antiproliferative effect in nine derivatives were examined in three concentrations: 1, 10 and
compare to DX was observed only at the highest concentration 100 μM. As was shown in Fig. 5, compounds 2 and 5 did not show any
(100 μM) (Fig. 4). antiproliferative activity after 48 h of incubation with IMR-32, even in

127
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

Fig. 5. Antiproliferative test against neuroblastoma cell line IMR-32. Values represent the
mean of n = 4 experiments. ***P < 0.001.

the highest used doses 100 μM, whereas 6-OHDA significantly inhibited Fig. 7. ATP level assessment after 130 min of IMR-32 cells incubation with 6-OHDA
IMR-32 proliferation at 100 μM. Thus, for the neuroprotection studies (100 μM), 2 (50 μM), 5 (50 μM) and after simultaneously addition of 50 μM of
phenylalanine derivatives with 100 μM of 6-OHDA. Values represent the mean of n = 3
the 50 μM concentrations of ligands 2 and 5 and 100 μM of 6-OHDA
experiments. ***P < 0.0001, *P < 0.05.
were selected.

ated next by measuring of ATP level in cells using bioluminescent


method. The generated bioluminescent signal was proportional to the
3.4. Neuroprotective effects of compounds 2 and 5
amount of ATP present in cells. The incubation of 6-OHDA and 6-OHDA
together with ligand 5 decreased significantly (P < 0.05) the ATP level
For the neuroprotection study we used Mitochondrial ToxGlo™
in the IMR-32 cell line, in compare to the control cells. These results
Assay. This test employs a sequential-addition, multiplexed chemistry
showed that compound 5 was not able to inhibit 6-OHDA toxic effect on
for predicting necrosis and potential mitochondrial dysfunction in cells
mitochondria. However, slight but not statistically significant decrease
as a result of the short-term xenobiotic exposure. The IMR-32 cell line
of ATP level in cells treated by 2 with 6-OHDA was observed indicating
was incubated with ligands 2 or 5 (50 μM) together with neurotoxin 6-
that ligand 2 appeared to inhibit moderately the 6-OHDA activity on
OHDA (100 μM) for 90 min to determine their potential neuroprotec-
mitochondria (Fig. 7). Moreover, both tested ligands 2 and 5 did not
tive effects. The tested ligands were also incubated separately without
decrease significantly the ATP level in IMR-32 cells, confirming
6-OHDA for assessing their potential influence on cell membrane
previously obtained data indicating no toxicity of ligands 2 and 5
integrity and mitochondria activity. The necrosis effect was assessed
(Figs. 4, 5 and 7).
first by fluorometric quantification of dead cell protease activity
associated with necrosis. DGT was used as a positive necrosis control.
As depicted in Fig. 6, in comparison with the control cells containing
3.5. Effects of ligands 2 and 5 pretreatment on STR-induced convulsions
media only and positive control with DGT, none of tested phenylalanine
derivatives at 50 μM and 6-OHDA at 100 μM concentration caused
In STR-induced seizure model, ligand 5 (5 mg/kg, i.p.) provided
necrosis after short-term 90 min incubation. Similarly, no necrosis
moderate protective activity only after 30 min of observation
effect was observed, if tested compounds were co-applied with
[F(1,12) = 6.12; P < 0.05], whereas acute systemic administration of
100 μM of 6-OHDA.Thus, these results indicated different from necrosis
ligand 5 in a dose of 10 and 15 mg/kg, i.p. exhibited moderate
cytotoxic mechanism of 6-OHDA on IMR-32 cells and confirmed no
protection against STR-induced convulsion when compared to the
toxic effect of 2 and 5.
saline-treated group after 10, 20, and 30 min of observation [all P-
The effect of tested ligands on the mitochondrial activity and their
values > 0.5] (Fig. 8). Notably, the reference drug VPA (300 mg/kg,
potential protection from 6-OHDA mitochondrial toxicity were evalu-
i.p.) provided full protection when compared with saline-treated group
after 10, 20, and 30 min with [F(1,12) = 148.41; P < 0.05],
[F(1,12) = 1156; P < 0.05], and [F(1,12) = 653.42; P < 0.05], respec-
tively. Moreover, the results show that ligand 2 (5, 10, and 15 mg/kg,
i.p.) neither provided proconvulsant nor anticonvulsant activity when
compared to saline-treated group (all P-values > 0.5) (Fig. 8). Notably,
neither Saline + Saline vs. 2 (5 and 10 mg/kg, i.p.) + Saline, nor
Saline + Saline vs. 5 (5, 10, and 15 mg/kg, i.p.) + Saline differences
were significant (all P-values > 0.05) (Fig. 9). However, acute systemic
administration of ligand 2 (15 mg/kg, i.p.) exhibited slight proconvul-
sant effect when compared with saline- treated group after 10, 20, and
30 min with [F(2,11)(2.11) = 6.12; P < 0.05], [F(2,11) = 10.57;
P < 0.05], and [F(2,11) = 4.24; P < 0.05], respectively (Fig. 9). How-
ever, the convulsions induced after acute systemic administration of
ligand 2 (15 mg/kg, i.p.) is significantly lower than that provided by
STR (3.5 mg/kg, i.p.) after 10, 20, and 30 min with [F(2,11) = 26.27;
P < 0.001], [F(2,11) = 87.63; P < 0.001], and [F(2,11) = 38.46;
Fig. 6. Necrosis assessment after 90 min of IMR-32 cells incubation with 6-OHDA
P < 0.001], respectively (Fig. 9).
(100 μM), 2 (50 μM), 5 (50 μM) and after simultaneously addition of 50 μM of
phenylalanine derivatives with 100 μM of 6-OHDA. Values represent the mean of n = 3
experiments. ***P < 0.001.

128
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

cantly potentiated the α1-GLYR-mediated effects (Figs. 2 and 3 B).


Interestingly, none of compounds 2 and 5, previously designed as
ligands of ionotropic glutamate receptors, showed significant affinity
for native AMPA, KA, NMDA or homomeric GluK1 kainate receptors
(Szymanska et al., 2009).
From a structural point of view, two particular differences in the
structure of both compounds appear to be of special importance for the
control of ligand activity on human α1-GLYRs. Accordingly, the amino
acid 2 containing biphenyl moiety provided the most promising
antagonistic effect on α1-GLYRs, while separation two phenyl rings
by methylene linker in the analogue 5 resulted in the highest potentia-
tion effect (Figs. 2 and 3 C). The second difference lies in moving away
of the carboxyl function in the ligand 2 compared to 5. Both modifica-
tions significantly influence the spatial shape of the whole molecule and
place the carboxyl group in various positions regarding the amino acid
moiety. The location of the carboxyl substituent seems to be crucial for
Fig. 8. Effect of acute systemic pretreatment of 2 and 5 on STR-induced convulsions in high affinity for α1-GLYRs of the ligand 2 and is not reproduced in any
male adult rats. VPA (300 mg/kg, i.p.) and ligands 2 and 5 (A–C in a dose of 5, 10, and other compound within the investigated series; the similar situation can
15 mg/kg, i.p., respectively) were injected 30 min before STR (3.5 mg/kg, i.p.) treat-
be observed in case of the compound 5. As it is shown in our studies,
ments. Values are expressed as the mean ± SEM (n = 7). *P < 0.05 and **P < 0.001
vs (STR-saline)-treated group. #Full protection. Abbreviations: STR, strychnine; VPA,
none of the positional isomers of 2 and 5 showed any significant affinity
valproic acid; SEM, standard error of the mean. for α1-GLYRs (Figs. 1 and 2, ligands 1 and 8). Furthermore, replace-
ment of the acidic substituent with a nitro group (6, 7), or removal of
an additional aromatic ring (3, 4) resulted in a failure to provide
affinity at α1-GLYRs. Accordingly, a role could also be played by other
additional substituents in the respective test compound, e.g. −Cl, for
compound 3; −Cl and −NO2, for compound 4; or two −Cl substitu-
ents, for compounds 6 and 7. These results are in agreement with a
previous study in which the pharmacological properties of the respec-
tive α-amino acid at the α1-GLYRs are critically determined by the size
and polarity of a certain substitution position (Schmieden and Betz,
1995).
In further experiments, compared to the reference compound
doxorubicin (DX), ligand 5 did not show any antiproliferative activity
after 48 h of incubation with HEK-293 cells whereas for 2 very weak
effect, but only at the 100 μM was observed (Fig. 4). Moreover, ligands
2 and 5 did not act on neuroblastoma IMR-32 cell line viability in the
concentration range 1–100 μM, whereas 6-OHDA significantly inhibited
its proliferation −when applied at 100 μM (Fig. 5). Consequently,
50 μM concentration of the respective ligand and 100 μM toxic
concentration of 6-OHDA were selected for neuroprotection studies.
Fig. 9. Effect of acute systemic pretreatment of 2 and 5 on saline-treated male adult rats. As depicted in Fig. 6 none of tested phenylalanine derivatives and 6-
STR (3.5 mg/kg, i.p.) and ligands 2 and 5 (A, B, and C in a dose of 5, 10, and 15 mg/kg, OHDA produced necrosis. Next, the effect of ligands 2 and 5 on the
i.p., respectively) were injected 30 min before observation time. Values are expressed as mitochondrial activity and 6-OHDA induced mitochondrial toxicity
the mean ± SEM (n = 7). *P < 0.05 vs saline-saline-treated groups. #P < 0.001 vs were assessed by measuring of ATP level in cells. Results confirmed that
STR-saline)-treated groups. Abbreviations;: STRstrychnine; SEMstandard error of the
selected phenylalanine derivatives are safe at the 50 μM and do not
mean.
have significant effect on mitochondrial functions (Fig. 7). Regarding
the neuroprotection activity, the comparison of the ATP level in cells
4. Discussion
treated with 6-OHDA and 6-OHDA together with 2 or 5 showed that
compound 5 did not possess neuroprotective properties. However,
GLY receptors are known to play important roles in epilepsy, pain
ligand 2 appeared to inhibit moderately toxic 6-OHDA activity on
transmission, and motor coordination in central as well as peripheral
mitochondria according to the measured ATP levels (Fig. 7).
nervous systems (Bedet et al., 2006; Eichler et al., 2008; Kirchner et al.,
In a further in vivo experiment and among promising ligands 2 and
2003; Meier and Grantyn, 2004; Mori et al., 2002; Song et al., 2006;
5, the results showed that acute systemic administration of ligand 5 (10
Zhang et al., 2008). Based on several previous clinical findings that
and 15 mg/kg, i.p.) provided some appreciable protection against STR-
patients would benefit from a facilitated glycinergic inhibition, the
induced convulsions in male adult rats, whereas the standard AED VPA
central role of GLYRs in modifying temporal lobe epilepsy has been
(300 mg/kg, i.p.) exhibited full protection following 10–30 min time
proposed,
observation (Fig. 8). Moreover, ligand 2 (5, 10 and 15 mg/kg, i.p.) and
In the current study, a series of unnatural racemic phenylalanine
the lower dose of ligand 5 (5 mg/kg, i.p.) failed to provide protection
derivatives (1–8) bearing a carboxyl or nitro group −has been explored
against STR-induced convulsions throughout observation time (Fig. 8).
on the functional properties of the human α1-GLYR expressed in
Interestingly, the results observed for ligand 5 (10 and 15 mg/kg, i.p.)
Xenopus oocytes. When tested in a concentration of 10 μM, compounds
are in agreement with previous studies in which anticonvulsant effects
1, 3, 4, 6–8 −failed to cause a significant change in the amplitude of
were obtained at the same dose range for several α-amino acid
α1-GLYR-mediated currents (Fig. 2). However, ligands 2 and 5
derivatives (De Caro et al., 2014; Paruszewski et al., 2001; Torregrosa
functionally interacted with human α1-GLYRs. Interestingly, ligand 2
et al., 2015). Moreover, a previous study showed that glycine potenti-
(10 μM) significantly inhibited the GLY-induced currents (Figs. 2 and 3
ates the anticonvulsant effect of diazepam and concluded that the
B), whereas ligand 5 when tested in the same concentration signifi-
potentiating effect involves a direct pharmacodynamic interaction

129
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

between these two compounds (Peterson and Frye, 1987). Furthermore, Wistar rats. Brain Res. Bull. 92, 60–68.
Callister, R.J., Graham, B.A., 2010. Early history of glycine receptor biology in
the latter in vivo findings for ligand 5 further confirmed our in vitro Mammalian spinal cord circuits. Front. Mol. Neurosci. 3, 13.
results for its observed significant potentiating effects on the α1-GLYR- Camp, A.J., Lim, R., Anderson, W.B., Schofield, P.R., Callister, R.J., Brichta, A.M., 2010.
induced currents (Figs. 1–3), since the mechanism underlying STR- Attenuated glycine receptor function reduces excitability of mouse medial vestibular
nucleus neurons. Neuroscience 170, 348–360.
induced convulsions is thought to be credited mainly to its blocking Castro, P.A., Figueroa, M., Yevenes, G.E., San Martin, L.S., Aguayo, L.G., 2012. The basic
effect on glycine receptors in the brain and in the spinal cord (Sadek property of Lys385 is important for potentiation of the human alpha1 glycine
et al., 2016a,b; Sowemimo et al., 2011). Importantly, the results for receptor by ethanol. J. Pharmacol. Exp. Ther. 340, 339–349.
Cheng, N.C., Van Roy, N., Chan, A., Beitsma, M., Westerveld, A., Speleman, F., Versteeg,
ligands 2 and 5 were further confirmed as a moderate proconvulsant R., 1995. Deletion mapping in neuroblastoma cell lines suggests two distinct tumor
effect was observed for ligand 2 (15 mg/kg, i.p.) in saline-treated male suppressor genes in the 1p 35–36 region, only one of which is associated with N-myc
adult animals whereas no appreciable effect was obtained for ligand 5 amplification. Oncogene 10, 291–297.
Coppola, G., Arcieri, S., D'Aniello, A., Messana, T., Verrotti, A., Signoriello, G., Pascotto,
(5, 10, and 15 mg/kg, i.p.) (Fig. 9).
A., 2010. Levetiracetam in submaximal subcutaneous pentylentetrazol-induced
seizures in rats. Seizure 19, 296–299.
5. Conclusion De Caro, V., Scaturro, A.L., Sutera, F.M., Avellone, G., Schiera, G., Ferrantelli, E., Carafa,
M., Rizzo, V., Carletti, F., Sardo, P., Giannola, L.I., 2014. N-valproyl-L-phenylalanine
as new potential antiepileptic drug: synthesis, characterization and in vitro studies on
The results indicate that phenyl alanine derivatives carrying stability, toxicity and anticonvulsant efficacy. Med. Chem. 11, 30–40.
selected hydrogen bond acceptors can constitute a new group of Eichler, S.A., Kirischuk, S., Juttner, R., Schaefermeier, P.K., Legendre, P., Lehmann, T.N.,
molecules with micromolar potencies for human α1-GLYRs. The Gloveli, T., Grantyn, R., Meier, J.C., 2008. Glycinergic tonic inhibition of
hippocampal neurons with depolarizing GABAergic transmission elicits
structure-activity relationships revealed that one particular substituent histopathological signs of temporal lobe epilepsy. J. Cell. Mol. Med. 12, 2848–2866.
position seemed to be crucial for control of ligand activity. The Kirchner, A., Breustedt, J., Rosche, B., Heinemann, U.F., Schmieden, V., 2003. Effects of
biphenyl derivative 2 provided the most promising antagonistic effect taurine and glycine on epileptiform activity induced by removal of Mg2+ in
combined rat entorhinal cortex-hippocampal slices. Epilepsia 44, 1145–1152.
on α1-GLYRs, while its structural analogue 5 displayed the highest Lester, H.A., Dibas, M.I., Dahan, D.S., Leite, J.F., Dougherty, D.A., 2004. Cys-loop
potentiation effect. Moreover, ligand 5 (potentiator of human α1- receptors: new twists and turns. Trends Neurosci. 27, 329–336.
GLYRs) provided moderate in vivo protection, whereas ligand 2 Loscher, W., 2011. Critical review of current animal models of seizures and epilepsy used
in the discovery and development of new antiepileptic drugs. Seizure 20, 359–368.
(inhibitor of human α1-GLYRs) failed to show anti(pro)convulsant
Lynch, J.W., 2004. Molecular structure and function of the glycine receptor chloride
effect when tested in STR-induced convulsion model in male adult rats. channel. Physiol. Rev. 84, 1051–1095.
Furthermore, the safety studies showed ligands 2 and 5 as non-toxic. A Mahgoub, M., Keun-Hang, S.Y., Sydorenko, V., Ashoor, A., Kabbani, N., Al Kury, L.,
Sadek, B., Howarth, C.F., Isaev, D., Galadari, S., Oz, M., 2013. Effects of cannabidiol
very weak antiproliferative effect in comparison to DX was only
on the function of alpha7-nicotinic acetylcholine receptors. Eur. J. Pharmacol. 720,
observed for ligand 2 against HEK-293 cell line at the highest 310–319.
concentration used. Additionally, neither necrosis, nor modulated Meier, J., Grantyn, R., 2004. A gephyrin-related mechanism restraining glycine receptor
mitochondrial toxicity in IMR-32 were observed for 2 and 5 at anchoring at GABAergic synapses. J. Neurosci. 24, 1398–1405.
Mori, M., Gahwiler, B.H., Gerber, U., 2002. Beta-alanine and taurine as endogenous
50 μM, while ligand 2 showed weak neuroprotective properties. In agonists at glycine receptors in rat hippocampus in vitro. J. Physiol. 539, 191–200.
light of the present findings, we conclude that the novel ligands 2 and 5 Paruszewski, R., Strupinska, M., Stables, J.P., Swiader, M., Czuczwar, S., Kleinrok, Z.,
with modulatory effects on human α1-GLYRs can be used as a potential Turski, W., 2001. Amino acid derivatives with anticonvulsant activity. Chem. Pharm.
Bull. (Tokyo) 49, 629–631.
template for further drug design. However, further investigations Peterson, S.L., Frye, G.D., 1987. Glycine potentiates diazepam anticonvulsant activity in
addressing additional in vitro as well as in vivo pro(anti)convulsant electroshock seizures of rats: possible sites of interaction in the brainstem. Brain Res.
effects of ligands 2 and 5 are warranted to exclude potentials of off- Bull. 18, 715–721.
Rodriguez-Pallares, J., Parga, J.A., Munoz, A., Rey, P., Guerra, M.J., Labandeira-Garcia,
target effects and to confirm the possible utility of α1-GLYR modulators J.L., 2007. Mechanism of 6-hydroxydopamine neurotoxicity: the role of NADPH
as AEDs. oxidase and microglial activation in 6-hydroxydopamine-induced degeneration of
dopaminergic neurons. J. Neurochem. 103, 145–156.
Sadek, B., Stark, H., 2015. Cherry-picked ligands at histamine receptor subtypes.
Acknowledgements Neuropharmacology 106.
Sadek, B., Ashoor, A., Al Mansouri, A., Lorke, D.E., Nurulain, S.M., Petroianu, G.,
The authors cordially thank Prof. Joe Lynch of the University of Wainwright, M., Oz, M., 2012. N3,N7-diaminophenothiazinium derivatives as
antagonists of alpha7-nicotinic acetylcholine receptors expressed in Xenopus oocytes.
Queensland, Australia for providing the cDNA for human α1-GLYR.
Pharmacol. Res. 66, 213–218.
Bassem Sadek and Murat Oz were supported by intermural funds from Sadek, B., Shehab, S., Wiecek, M., Subramanian, D., Shafiullah, M., Kiec-Kononowicz, K.,
the College of Medicine and Health Sciences and the Office of Graduate Adem, A., 2013. Anticonvulsant properties of histamine H3 receptor ligands
Studies and Research, UAE University. The financial support of this belonging to N-substituted carbamates of imidazopropanol. Bioorg. Med. Chem. Lett.
23, 4886–4891.
research by the Ministry of Science and Higher Education in Poland Sadek, B., Khanian, S.S., Ashoor, A., Prytkova, T., Ghattas, M.A., Atatreh, N., Nurulain,
(Jagiellonian University statutory project K/ZDS/005594) as well as the S.M., Yang, K.H., Howarth, F.C., Oz, M., 2014a. Effects of antihistamines on the
National Science Centre Poland (DEC-2014/15/B/NZ7/00908) is grate- function of human alpha7-nicotinic acetylcholine receptors. Eur. J. Pharmacol. 746,
308–316.
fully acknowledged. Sadek, B., Kuder, K., Subramanian, D., Shafiullah, M., Stark, H., Lazewska, D., Adem, A.,
Kiec-Kononowicz, K., 2014b. Anticonvulsive effect of nonimidazole histamine H3
References receptor antagonists. Behav. Pharmacol. 25, 245–252.
Sadek, B., Schwed, J.S., Subramanian, D., Weizel, L., Walter, M., Adem, A., Stark, H.,
2014c. Non-imidazole histamine H3 receptor ligands incorporating antiepileptic
Ashoor, A., Nordman, J.C., Veltri, D., Yang, K.H., Al Kury, L., Shuba, Y., Mahgoub, M., moieties. Eur. J. Med. Chem. 77, 269–279.
Howarth, F.C., Sadek, B., Shehu, A., Kabbani, N., Oz, M., 2013a. Menthol binding and Sadek, B., Khanian, S.S., Ashoor, A., Prytkova, T., Ghattas, M.A., Atatreh, N., Nurulain,
inhibition of alpha7-nicotinic acetylcholine receptors. PLoS One 8, e67674. S.M., Yang, K.H., Howarth, F.C., Oz, M., 2015a. Effects of antihistamines on the
Ashoor, A., Nordman, J.C., Veltri, D., Yang, K.H., Shuba, Y., Al Kury, L., Sadek, B., function of human alpha7-nicotinic acetylcholine receptors. Eur. J. Pharmacol. 746,
Howarth, F.C., Shehu, A., Kabbani, N., Oz, M., 2013b. Menthol inhibits 5-HT3 308–316.
receptor-mediated currents. J. Pharmacol. Exp. Ther. 347, 398–409. Sadek, B., Saad, A., Subramanian, D., Shafiullah, M., Łażewska, D., Kieć-Kononowiczc, K.,
Bedet, C., Bruusgaard, J.C., Vergo, S., Groth-Pedersen, L., Eimer, S., Triller, A., Vannier, 2015b. Anticonvulsant and procognitive properties of the non-imidazole histamine
C., 2006. Regulation of gephyrin assembly and glycine receptor synaptic stability. J. H3 receptor antagonist DL77 in male adult rats. Neuropharmacology 106, 46–55.
Biol. Chem. 281, 30046–30056. Sadek, B., Saad, A., Latacz, G., Kuder, K., Olejarz, A., Karcz, T., Stark, H., Kiec-
Blum-Degen, D., Haas, M., Pohli, S., Harth, R., Romer, W., Oettel, M., Riederer, P., Gotz, Kononowicz, K., 2016a. Non-imidazole-based histamine H3 receptor antagonists with
M.E., 1998. Scavestrogens protect IMR 32 cells from oxidative stress-induced cell anticonvulsant activity in different seizure models in male adult rats. Drug Des. Dev.
death. Toxicol. Appl. Pharmacol. 152, 49–55. Ther. 10, 3879–3898.
Branco Cdos, S., Scola, G., Rodrigues, A.D., Cesio, V., Laprovitera, M., Heinzen, H., Dos Sadek, B., Saad, A., Sadeq, A., Jalal, F., Stark, H., 2016b. Histamine H3 receptor as a
Santos, M.T., Fank, B., de Freitas, S.C., Coitinho, A.S., Salvador, M., 2012. potential target for cognitive symptoms in neuropsychiatric diseases. Behav. Brain
Anticonvulsant, neuroprotective and behavioral effects of organic and conventional Res. 312, 415–430.
yerba mate (Ilex paraguariensis St. Hil.) on pentylenetetrazol-induced seizures in Sadek, B., Saad, A., Schwed, J.S., Weizel, L., Walter, M., Stark, H., 2016c. Anticonvulsant

130
B. Sadek et al. Epilepsy Research 138 (2017) 124–131

effects of isomeric nonimidazole histamine H3 receptor antagonists. Drug Des. Dev. phenylalanines as selective AMPA- and kainate receptor ligands. Bioorg. Med. Chem.
Ther. 10, 3633–3651. 17, 6390–6401.
Sancheti, J., Shaikh, M.F., Chaudhari, R., Somani, G., Patil, S., Jain, P., Sathaye, S., 2013. Szymanska, E., Frydenvang, K., Contreras-Sanz, A., Pickering, D.S., Frola, E., Serafimoska,
Characterization of anticonvulsant and antiepileptogenic potential of thymol in Z., Nielsen, B., Kastrup, J.S., Johansen, T.N., 2011. A new phenylalanine derivative
various experimental models. Naunyn. Schmiedebergs Arch. Pharmacol. 387, 59–66. acts as an antagonist at the AMPA receptor GluA2 and introduces partial domain
Schmieden, V., Betz, H., 1995. Pharmacology of the inhibitory glycine receptor: agonist closure: synthesis, resolution, pharmacology, and crystal structure. J. Med. Chem. 54,
and antagonist actions of amino acids and piperidine carboxylic acid compounds. 7289–7298.
Mol. Pharmacol. 48, 919–927. Torregrosa, R., Yang, X.F., Dustrude, E.T., Cummins, T.R., Khanna, R., Kohn, H., 2015.
Serdiuk, S.E., Gmiro, V.E., Veselkina, O.S., 2014. Stimulation of gastric mucosa afferents Chimeric derivatives of functionalized amino acids and alpha-aminoamides:
by phenylephrine potentiates anticonvulsive and eliminates sedative action of compounds with anticonvulsant activity in seizure models and inhibitory actions on
sodium valproate in the pentylenetetrazol kindling model in rats. Ross. Fiziol. Zh. Im. central, peripheral, and cardiac isoforms of voltage-gated sodium channels. Bioorg.
I. M. Sechenova 100, 96–104. Med. Chem. 23, 3655–3666.
Song, W., Chattipakorn, S.C., McMahon, L.L., 2006. Glycine-gated chloride channels Waldbaum, S., Patel, M., 2010. Mitochondrial dysfunction and oxidative stress: a
depress synaptic transmission in rat hippocampus. J. Neurophysiol. 95, 2366–2379. contributing link to acquired epilepsy. J. Bioenerg. Biomembr. 42, 449–455.
Sowemimo, A.A., Adio, O., Fageyinbo, S., 2011. Anticonvulsant activity of the methanolic Zhang, L.H., Gong, N., Fei, D., Xu, L., Xu, T.L., 2008. Glycine uptake regulates
extract of Justicia extensa T. Anders. J. Ethnopharmacol. 138, 697–699. hippocampal network activity via glycine receptor-mediated tonic inhibition.
Szymanska, E., Pickering, D.S., Nielsen, B., Johansen, T.N., 2009. 3-Substituted Neuropsychopharmacology 33, 701–711.

131

You might also like