• Nie Znaleziono Wyników

Effects of N-(morpholinomethyl)-p-isopropoxyphenylsuccinimide on the protectiveaction of different classical antiepileptic drugsagainst maximal electroshock-induced tonicseizures in mice

N/A
N/A
Protected

Academic year: 2022

Share "Effects of N-(morpholinomethyl)-p-isopropoxyphenylsuccinimide on the protectiveaction of different classical antiepileptic drugsagainst maximal electroshock-induced tonicseizures in mice"

Copied!
10
0
0

Pełen tekst

(1)

Effects of N-(morpholinomethyl)-

p-isopropoxyphenylsuccinimide on the protective action of different classical antiepileptic drugs against maximal electroshock-induced tonic seizures in mice

Dorota ¯ó³kowska1, Mateusz Kominek2,3, Magdalena Florek-£uszczki4, Sergey L. Kocharov5, Jarogniew J. £uszczki2,3,

1Department of Neurology, UC Davis School of Medicine, 4635 2nd Avenue, Sacramento, CA 95817, USA

2Isobolographic Analysis Laboratory, Institute of Rural Health, Jaczewskiego 2, PL 20-950 Lublin, Poland

3Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8, PL 20-090 Lublin, Poland

4Department of Public Health, Institute of Rural Health, Jaczewskiego 2, PL 20-950 Lublin, Poland

5Mndjoyan’s Institute of Fine Organic Chemistry, National Academy of Sciences, Azatutyan Avenue 26, RA 375014 Yerevan, Republic of Armenia

Correspondence: Jarogniew J. £uszczki, e-mail: jarogniew.luszczki@gmail.com or jluszczki@yahoo.com

Abstract:

Background: The aim of this study was to determine the effects of N-(morpholinomethyl)-p-isopropoxy-phenylsuccinimide (MMIPPS) on the protective action of four classical antiepileptic drugs (AEDs: carbamazepine [CBZ], phenobarbital [PB], pheny- toin [PHT] and valproate [VPA]) against maximal electroshock (MES)-induced seizures in mice.

Methods: Tonic hind limb extension (seizure activity) was evoked in adult male albino Swiss mice by a current (sine-wave, 25 mA, 500 V, 50 Hz, 0.2 s stimulus duration) delivered via auricular electrodes. Total brain concentrations of AEDs were measured to deter- mine the characteristics of interaction between MMIPPS and classical AEDs in the mouse MES model.

Results: MMIPPS administered intraperitoneally (ip) at 100 mg/kg significantly elevated the threshold for electroconvulsions in mice (p < 0.01). MMIPPS at doses of 25 and 50 mg/kg had no impact on the threshold for electroconvulsions in mice. Moreover, MMIPPS (50 mg/kg) significantly enhanced the anticonvulsant activity of PB and VPA (p < 0.05), but not that of CBZ or PHT, in the MES test in mice. Pharmacokinetic studies revealed that MMIPPS (50 mg/kg) did not alter total brain concentrations of PB, but sig- nificantly elevated total brain concentrations of VPA in mice (p < 0.05).

Conclusions: The enhanced anticonvulsant action of PB by MMIPPS in the mouse MES model and lack of any pharmacokinetic inter- action between drugs make the combination of MMIPPS with PB of pivotal importance for further experimental and clinical studies.

Pharmacokinetic increase in total brain VPA concentration seems to be responsible for the enhanced anticonvulsant action of VPA by MMIPPS in the mouse MES model. The combinations of MMIPPS with CBZ and PHT are neutral from a preclinical viewpoint.

Key words:

antiepileptic drugs, maximal electroshock-induced seizures, pharmacokinetic/pharmacodynamic interaction, p-isopropoxyphenylsuccinimide derivative

Pharmacological Reports 2013, 65, 389–398 ISSN 1734-1140

Copyright © 2013 by Institute of Pharmacology Polish Academy of Sciences

(2)

Epilepsy affects more than 50 million people world- wide [22]. According to WHO reports, approximately 30% of patients suffering from epilepsy do not re- spond to currently available antiepileptic treatment.

Even though the cause of drug resistance is still un- known, numerous neurological disorders such as cor- tical dysplasia, hippocampal sclerosis, mutations in ion channels and receptors, neuroinflammation and some autoimmune processes are linked to the devel- opment of pharmacoresistance in epilepsy [22].

Therefore, there is a constant demand for novel thera- peutics and more efficacious treatment options in the field of epilepsy. At present, the search for novel an- tiepileptic drugs (AEDs) more efficacious and less toxic than classical AEDs is continuing [23, 25].

Nowadays, researchers and clinicians are trying to find some compounds that in the future could become the efficacious AEDs [16, 17, 20].

Growing experimental evidence indicates that some succinimide derivatives possess anticonvulsant properties in in vivo screening tests in rodents [5, 7, 14, 26]. For instance, 3-cyclohexylsuccinimides [5], N-morpholinomethyl derivative of m-bromophenyl- succinimide [7], N-pyridyl-substituted succinimides [26], N-(anilinomethyl)-p-isopropoxyphenylsuccini- mide (AMIPPS) [14], p-isopropoxyphenylsuccini- mide monohydrate (IPPS) [13], N-(o-carboxyanilino- methyl)-p-isopropoxyphenylsuccinimide (o-CAMIPPS), N-(m-carboxyanilinomethyl)-p-isopropoxyphenylsuci- nimide (m-CAMIPPS), and N-(p-carboxyanilinome- thyl)-p-isopropoxyphenylsuccinimide (p-CAMIPPS) [12], and N-hydroxymethyl-p-isopropoxyphenylsuc- cinimide (HMIPPS) [15], exhibited potent anticonvul- sant effects in the maximal electroshock-induced sei- zure (MES) test, recognized as the most widely em- ployed animal seizure model for early identification of candidate AEDs.

In our pilot study, we found that N-(morpholi- nomethyl)-p-isopropoxyphenylsuccinimide (MMIPPS) exerts the anticonvulsant properties by suppressing tonic-clonic seizures in the mouse MES test (Fig. 1).

Therefore, the aim of the presented study was to evaluate the effect of MMIPPS on the threshold for electroconvulsions, and to assess its influence on the protective activity of four various classical AEDs (carbamazepine [CBZ], phenobarbital [PB], pheny- toin [PHT], and valproate [VPA]) in the mouse MES-

secondary generalization in humans [10]. Both these tests are used as standard screening procedures for po- tentially anticonvulsant agents, and additionally help to evaluate their effects on classical AEDs which are fully effective in the suppression of tonic-clonic sei- zures in humans [10]. Therefore, these animal seizure models were selected for evaluation of the effects of MMIPPS alone, as well as in combination with four different classical AEDs.

Additionally, combinations of MMIPPS with four classical AEDs were investigated in relation to their possible effects on motor coordination, long-term memory and muscular strength by use of the chimney test, step-through passive avoidance task and grip- strength test, respectively. Finally, total brain AED concentrations were measured with fluorescence po- larization immunoassay to ascertain whether any ob- served effects were consequent to a pharmacody- namic and/or a pharmacokinetic interaction.

Fig. 1. Dose-response relationship and time course of the anticonvul- sant action of N-(morpholinomethyl)-p-isopropoxyphenylsuccin- imide (MMIPPS) against MES-induced seizures in mice. Columns are presented as median effective doses (ED50in mg/kg ± SE as error bars) of MMIPPS, protecting 50% of animals tested against MES- induced hind limb extension. MMIPPS was administered ip at various pretreatment times (15, 30, 60 and 120 min) in the MES-induced sei- zure test

(3)

Materials and Methods

Animals and experimental conditions

Adult male Swiss mice (weighing 22–26 g) that were kept in colony cages with free access to food and tap water, housed under standardized housing conditions (natural light-dark cycle, temperature of 23 ± 1°C, and relative humidity of 55 ± 5%), were used. After 7 days of adaptation to laboratory conditions, the ani- mals were randomly assigned to experimental groups, each comprised of 8 mice. Each mouse was used only once and all tests were performed between 08:00–

15:00 h. Procedures involving animals and their care were conducted in accordance with current European Community and Polish legislation on animal experi- mentation. Additionally, all efforts were made to minimize animal suffering and to use only the number of animals necessary to produce reliable scientific data. The experimental protocols and procedures de- scribed in this manuscript were approved by the First Local Ethics Committee at the Medical University of Lublin (License No.: 18/2006) and the Second Local Ethics Committee at the University of Life Sciences in Lublin (License Nos.: 79/2009 and 15/2012), and complied with the European Communities Council Directive of 24 November 1986 (86/609/EEC).

Drugs

The following drugs were used: N-(morpholi- nomethyl)-p-isopropoxyphenylsuccinimide (MMIPPS – C18H24N2O4– molecular weight = 332.390; synthe- sized by Dr. S.L. Kocharov, Mndjoyan’s Institute of Fine Organic Chemistry of the National Academy of Sciences of the Republic of Armenia, Yerevan, Arme- nia), carbamazepine (CBZ – a gift from Polpharma, Starogard Gdañski, Poland), phenobarbital (PB – Polfa, Kraków, Poland), phenytoin (PHT – Polfa, Warszawa, Poland), and valproate (VPA – magnesium salt – kindly donated by ICN-Polfa S.A., Rzeszów, Poland). All drugs, except for VPA, were suspended in a 1% solution of Tween 80 (Sigma, St. Louis, MO, USA) in distilled water, while VPA was directly dis- solved in distilled water. All drugs were administered intraperitoneally (ip), in a volume of 5 ml/kg body weight, as follows: PHT – 120 min, PB – 60 min, CBZ, VPA and MMIPPS – 30 min before electrocon- vulsions and brain sampling for the measurement of

AED concentrations. The pretreatment times before testing of the AEDs were based on information about their biological activity from the literature [10] and our previous experiments [11–18]. The times to the peak of maximum anticonvulsant effects for all AEDs were used as the reference times in all behavioral tests and pharmacokinetic estimation of total brain AED concentrations. The pretreatment time (30 min) before testing MMIPPS was established in our pilot study as the time to peak of maximum anticonvulsant activity of MMIPPS (Fig. 1).

Electroconvulsions

Electroconvulsions were induced by applying an alter- nating current (50 Hz; 500 V) via ear-clip electrodes from a rodent shocker generator (type 221; Hugo Sachs Elektronik, Freiburg, Germany). The stimulus duration was 0.2 s. Tonic hind limb extension was used as the endpoint. This apparatus was used to induce seizures in two methodologically different experimental ap- proaches: maximal electroshock seizure threshold (MEST) test and MES test [10].

Maximal electroshock seizure threshold (MEST) test

The MEST test was first used to assess the anticon- vulsant effects of MMIPPS administered alone. In this test, at least 4 groups of control mice, each consisting of 8 animals, were challenged with currents of vary- ing intensities ranging between 5–8 mA so that 10–30, 30–50, 50–70 and 70–90% of animals exhibited the endpoint. After establishing the current intensity- effect curve (i.e., current intensity in mA vs. percent- age of mice convulsing) for each dose of MMIPPS tested, the electroconvulsive threshold was calculated according to the log-probit method of Litchfield and Wilcoxon [8]. The electroconvulsive threshold was expressed as the median current strength value (CS50

in mA) predicted to produce tonic hind limb extension in 50% of the animals tested. This experimental pro- cedure was performed for various increasing doses of MMIPPS (25, 50 and 100 mg/kg), until the CS50value of MMIPPS-injected animals was statistically differ- ent from that of the control animals. The doses of MMIPPS were empirically selected when testing the threshold for electroconvulsions in mice. Only doses of MMIPPS that did not significantly affect the sei- zure threshold in the MEST test were selected for test-

Interaction of MMIPPS with antiepileptic drugs

Dorota ¯ó³kowska et al.

(4)

vulsant efficacy of MMIPPS in the effects observed in combination with the AEDs in the MES test.

Maximal electroshock seizure (MES) test

In the MES test, mice were challenged with a current of a fixed intensity (25 mA) that was 4–5-fold higher than the CS50 value in vehicle-treated control mice [10]. These parameters of stimulation (maximal elec- troshock) typically result in all mice responding with tonic hind limb extension immediately after stimula- tion. The AEDs administered alone and their combi- nation with MMIPPS were tested for their ability to increase the number of animals not responding with tonus (i.e., protected from tonic hind limb extension) after stimulation. Again, at least 4 groups of mice, each consisting of 8 animals and treated with a differ- ent dose of the AEDs alone or in combination with MMIPPS, were challenged with a current of 25 mA to yield 10–30, 30–50, 50–70 and 70–90% of animals protected from tonic seizures. After constructing a dose-effect curve (i.e., dose in mg/kg vs. percentage of mice protected), the protective median effective dose (ED50) value of the AED tested was calculated according to a log-probit method according to Litchfield and Wilcoxon [8]. Each ED50 value repre- sented a dose of the AED (in mg/kg) predicted to pro- tect 50% of mice tested against MES-induced exten- sion of the hind limbs. MMIPPS was tested for its ability to affect the anticonvulsive potency of AEDs.

As mentioned earlier, MMIPPS was administered in doses that per se had no effect on the seizure thresh- old in the MEST test. In this experimental protocol, an increase in the anticonvulsant potency of the AED tested in combination with MMIPPS would be re- flected by a lower ED50 value of the test AED (i.e., lower dose of test drug was necessary to protect 50%

of the mice challenged). In the presented study, CBZ and PHT were administered at doses ranging between 6–14 mg/kg, PB at doses ranging between 15–30 mg/

kg and VPA at doses ranging between 150–300 mg/

kg. These AED doses suppressed tonic seizures in 10–90% of mice subjected to the MES test.

centrations was performed only for those combina- tions of MMIPPS with AEDs for which the anticon- vulsant effect in the MES test was significantly greater than that for control (an AED + vehicle- treated) animals. Thus, the measurements of total brain concentrations of PB and VPA were undertaken at the doses that corresponded to their ED50 values from the MES test. Specifically, mice pretreated with a given AED alone or in combination with MMIPPS were decapitated at times reflecting the peak of maxi- mum anticonvulsant effects for the drugs in the MES test. The whole brains of mice were removed from skulls, weighed, harvested and homogenized using Abbott buffer (1 : 2 w/v; Abbott Laboratories, North Chicago, IL, USA) in an Ultra-Turrax T8 homo- genizer. The homogenates were then centrifuged at 10,000 × g for 10 min and the supernatant samples of 100 µl were collected and analyzed for AED content.

Total brain concentrations of PB and VPA were meas- ured by a fluorescence polarization immunoassay us- ing an analyzer (Abbott TDx) and manufacturer- supplied reagent kits (Abbott Laboratories, North Chicago, IL, USA). Total brain AED concentrations were expressed in µg/ml of brain supernatants as the means ± standard error (SE) of at least 8 separate brain preparations.

Step-through passive avoidance task

Each animal was administered an AED either singly or in combination with MMIPPS (50 mg/kg), at doses corresponding to their ED50values from the MES test, on the first day before training. The time before com- mencement of the training session (after drug admini- stration) was identical to that for the MES test. Subse- quently, the animals were placed in an illuminated box (10 × 13 × 15 cm) connected to a larger dark box (25 × 20 × 15 cm) equipped with an electric grid floor.

Entrance of the animals to the dark box was punished by an adequate electric footshock (0.6 mA for 2 s).

The animals that did not enter the dark compartment were excluded from subsequent experimentation. On the following day (24 h later), the pre-trained animals were placed again into the illuminated box and ob- served for up to 180 s. Mice that avoided the dark compartment for 180 s were considered as having

(5)

remembered the task. The time the mice took to enter the dark box was noted and the median latencies (retention times) with 25th and 75th percentiles were calculated. The step-through passive avoidance task gives information about ability to acquire a task (learning) and to recall a task (retrieval). Therefore, it may be regarded as a measure of long-term memory [24].

Grip-strength test

The effects of combinations of MMIPPS (50 mg/kg) with different classical AEDs at doses corresponding to their ED50 values from the MES test on skeletal muscular strength in mice were quantified by the grip-strength test of Meyer et al. [19]. The grip- strength apparatus (BioSeb, Chaville, France) com- prised of a wire grid (8 × 8 cm) connected to an iso- metric force transducer (dynamometer). The mice were lifted by the tails so that their forepaws could grasp the grid. The mice were then gently pulled backward by the tail until the grid was released. The maximal force exerted by the mouse before losing grip was recorded. The mean of 3 measurements for each animal was calculated, and subsequently the mean maximal force of 8 animals per group was de- termined. The muscular strength in mice is expressed in N (newtons) as the means ± SE of at least 8 deter- minations.

Chimney test

The chimney test of Boissier et al. [1] was used to quantify the adverse effect potential of classical AEDs administered in combination with MMIPPS. In this test, the animals had to climb backwards up a plastic tube (3 cm inner diameter, 30 cm long) and impairment of motor performance was indicated by the inability of the mice to climb backward up the transparent tube within 60 s. The acute adverse-effect potentials for the combinations of classical AEDs with MMIPPS were determined for the AEDs admin- istered at doses corresponding to their ED50 values from the MES test when combined with MMIPPS (50 mg/kg).

Statistics

Both CS50and ED50values with their 95% confidence limits were calculated by computer-assisted log-

probit analysis according to Litchfield and Wilcoxon [8]. Subsequently, the respective 95% confidence lim- its were transformed to SE as described previously [11]. Statistical analysis of data from the MEST test was performed with one-way analysis of variance (ANOVA) followed by the post-hoc Tukey-Kramer test for multiple comparisons among four CS50values.

Statistical analysis of data from the MES test was per- formed with one-way ANOVA followed by the post- hoc Tukey-Kramer test for multiple comparisons among 3 ED50values. Total brain AED concentrations were statistically compared using the unpaired Stu- dent’s t-test. The results obtained in the step-through passive avoidance task were statistically evaluated us- ing Kruskal-Wallis nonparametric ANOVA. The re- sults from the grip-strength test were verified with one-way ANOVA. The data from the chimney test were statistically analyzed with the Fisher’s exact probability test. Differences among values were con- sidered statistically significant if p < 0.05. All statisti- cal tests were performed using commercially avail- able GraphPad Prism version 4.0 for Windows (GraphPad Software, San Diego, CA, USA).

Results

Influence of MMIPPS on the threshold for electroconvulsions

MMIPPS administered systemically (ip, 30 min prior to the MEST test) at a dose of 100 mg/kg significantly elevated the threshold for electroconvulsions in mice from 6.11 ± 0.45 mA to 8.23 ± 0.40 mA (p < 0.01;

Tab. 1). The experimentally-derived CS50 values for animals receiving MMIPPS at doses of 25 and 50 mg/kg did not significantly differ from that for control animals subjected to the MEST test (Tab. 1).

Effects of MMIPPS on the protective action of CBZ, PB, PHT and VPA in the mouse MES model

All investigated classical AEDs (CBZ, PB, PHT and VPA) administered alone exhibited a clear anticonvul- sant activity in the MES test in mice (Tab. 2). When MMIPPS (50 mg/kg) was co-administered with PB, it significantly enhanced the anticonvulsant action of the latter drug in the MES test by reducing the ED50value

Interaction of MMIPPS with antiepileptic drugs

Dorota ¯ó³kowska et al.

(6)

of PB from 23.25 ± 1.83 mg/kg to 18.01 ± 1.12 mg/kg (p < 0.05; Tab. 2). In contrast, MMIPPS (25 mg/kg) had no significant impact on the anticonvulsant action of PB against MES-induced seizures (Tab. 2). Moreo- ver, when MMIPPS (50 mg/kg) was co-administered with VPA, it significantly enhanced the anticonvul- sant action of the latter drug by reducing the ED50 value of VPA from 259.3 ± 10.87 mg/kg to 215.3 ± 14.80 mg/ kg (p < 0.05; Tab. 2). MMIPPS at the dose of 25 mg/kg had no impact on the anticonvulsant ac- tion of VPA against MES-induced seizures in mice (Tab. 2). Similarly, MMIPPS at both doses of 25 and 50 mg/kg did not significantly alter the anticonvulsant action of CBZ or PHT in the MES test in mice (Tab. 2).

Effect of MMIPPS on total brain antiepileptic drug concentrations

As determined by the fluorescence polarization im- munoassay method, MMIPPS (50 mg/kg) did not sig- nificantly affect the total brain concentration of PB co-administered at a dose of 18.1 mg/kg (Tab. 3). In contrast, MMIPPS (50 mg/kg) significantly elevated (by 29%) the total brain concentration of VPA

Treatment (mg/kg) ED50(mg/kg) n

CBZ + vehicle 10.77 ± 0.87 16

CBZ + MMIPPS (25) 9.13 ± 1.22 8

CBZ + MMIPPS (50) 8.61 ± 0.83 16

F (2, 37) = 1.691; p = 0.1983

PB + vehicle 23.25 ± 1.83 16

PB + MMIPPS (25) 19.42 ± 1.20 24

PB + MMIPPS (50) 18.01 ± 1.12* 16

F (2, 53) = 3.344; p = 0.0429

PHT + vehicle 9.87 ± 0.86 24

PHT + MMIPPS (25) 9.58 ± 0.97 32

PHT + MMIPPS (50) 8.53 ± 0.96 16

F (2, 69) = 0.4043; p = 0.6690

VPA + vehicle 259.3 ±10.87 32

VPA + MMIPPS (25) 230.4 ± 11.31 16

VPA + MMIPPS (50) 215.3 ± 14.80* 32

F (2, 77) = 3.330; p = 0.0410

Results are presented as median effective doses (ED50in mg/kg

± SE) of AEDs, protecting 50% of animals tested against MES- induced hind limb extension. Each experimental group consisted of 8 animals. To determine each ED50value, 4–5 groups of animals (8 mice per group) were used. All AEDs were administered ip: PHT – 120 min, PB – 60 min, CBZ and VPA – 30 min prior to the MES test.

MMIPPS was administered systemically (ip) at 30 min before the MES test. Statistical analysis of data was performed with log-probit method and one-way ANOVA followed by the post-hoc Tukey- Kramer test for multiple comparisons. n – total number of animals used at doses where anticonvulsant effects ranged between 4–6 pro- bits. However, log-probit method considered only those groups of animals in which anticonvulsant effects ranged between 16–84%.

This is the reason for the total number of animals (n) being different for various ED50 values. F – value of F-statistics from one-way ANOVA; p – value of probability from one-way ANOVA; CBZ – carba- mazepine, PB – phenobarbital, PHT – phenytoin, and VPA – val- proate. * p < 0.05 vs. control (AED + vehicle-treated) animals

Vehicle 6.11 ± 0.45 24

MMIPPS (25) 6.75 ± 0.53 24

MMIPPS (50) 7.30 ± 0.44 16

MMIPPS (100) 8.23 ± 0.40** 32

F (3, 92) = 4.380; p = 0.0063

Data are presented as median current strengths (CS50values ± SE) required to produce tonic hind limb extension in 50% of animals tested in the maximal electroshock-induced seizure threshold (MEST) test. Each experimental group consisted of 8 animals. To de- termine each CS50value, 4 groups of animals (8 mice per group) were used. MMIPPS was administered ip 30 min before the test. Sta- tistical evaluation of the data was performed with log-probit method [8] and one-way ANOVA, followed by post-hoc Tukey-Kramer test for multiple comparisons. n – Number of animals tested at current strength intensities, in which seizure effects ranged between 16–84%.

However, log-probit method considered only those groups of ani- mals whose seizure effects ranged between 16–84%. This is the rea- son for the total number of animals (n) being different for various CS50 values. F – value of F-statistics from one-way ANOVA; p – probability from one-way ANOVA. ** p < 0.01 vs. the control (vehicle-treated) animals

(7)

coadministered at a dose of 215.3 mg/kg (p < 0.05;

Tab. 3).

Effects of MMIPPS and its combination with various antiepileptic drugs on motor performance, long-term memory and skeletal muscular strength of animals in the chimney, step-through passive avoidance and grip-strength tests MMIPPS administered alone at a dose of 50 mg/kg did not impair long-term memory in animals chal- lenged with the step-through passive avoidance task (Tab. 4). Similarly, MMIPPS at 50 mg/kg neither af- fected muscular strength in mice in the grip-strength

test, nor impaired motor coordination in mice sub- jected to the chimney test (Tab. 4). When MMIPPS (50 mg/kg) was administered in combination with CBZ, PB, PHT and VPA at doses corresponding to their ED50values from the MES test, motor perform- ance as assessed by the chimney test was unaffected (Tab. 4). Similarly, MMIPPS (50 mg/kg) concomi- tantly administered with the classical AEDs had no significant impact on skeletal muscular strength of the animals, as assessed by the grip-strength test (Tab. 4).

Furthermore, none of the combinations studied im- paired long-term memory, as determined in the pas- sive avoidance test (Tab. 4). As regards the AEDs ad- ministered alone at doses corresponding to their ED50

values from the MES-induced seizure test, the AEDs had no significant impact on long-term memory, mus- cular strength and motor performance in mice (results not shown).

Discussion

The results of the presented study indicate that MMIPPS, in a dose-dependent manner, elevated the threshold for electroconvulsions in mice. Moreover, the agent at the sub-protective dose of 50 mg/kg (i.e., the dose that by itself did not significantly affect the threshold for electroconvulsions) potentiated the anti- convulsant activity of PB and VPA against MES- induced seizures in mice. In contrast, MMIPPS at the sub-protective dose of 50 mg/kg had no significant im-

Interaction of MMIPPS with antiepileptic drugs

Dorota ¯ó³kowska et al.

Tab. 4. Effects of N-(morpholinomethyl)-p-isopropoxyphenylsuccinimide (MMIPPS) and its combinations with four classical antiepileptic drugs on long-term memory, muscular strength and motor performance in mice

Treatment (mg/kg) Retention time (s) Grip-strength (N) Motor coordination impairment (%)

Vehicle 180 (180; 180) 0.927 ± 0.054 0

MMIPPS (50) + vehicle 180 (150.8; 180) 0.888 ± 0.056 0

CBZ (8.6) + MMIPPS (50) 180 (180; 180) 0.909 ± 0.057 0

PB (18.0) + MMIPPS (50) 180 (165.8; 180) 0.896 ± 0.057 12.5

PHT (8.5) + MMIPPS (50) 180 (180; 180) 0.915 ± 0.056 0

VPA (215.3) + MMIPPS (50) 175.5 (145; 180) 0.890 ± 0.056 12.5

Results are presented as: 1) median retention times (in seconds; with 25thand 75thpercentiles in parentheses) from the passive avoidance task, assessing long-term memory in mice; 2) mean grip-strengths (in newtons ± SE) from the grip-strength test, assessing muscular strength in mice; and 3) percentage of animals showing motor coordination impairment in the chimney test in mice. Each experimental group consisted of 8 animals, and 3 different groups of animals were used to study behavioral effects of each combination in the grip-strength, chimney and step-through passive avoidance tests in mice. Statistical analysis of data from the passive avoidance task was performed with non-parametric Kruskal-Wallis ANOVA test, whereas those from the grip-strength test were analyzed with one-way ANOVA. The Fisher’s exact probability test was used to analyze the results from the chimney test. All drugs were administered ip at times scheduled from the MES test, and at doses corre- sponding to their ED50values against MES-induced seizures in mice (for more details see legend to Tab. 2)

Tab. 3. Brain concentrations of antiepileptic drugs administered sin- gly or in combination with N-(morpholinomethyl)-p-isopropoxy- phenylsuccinimide (MMIPPS)

Treatment (mg/kg) Brain concentration (µg/ml) PB (18.1) + vehicle 11.53 ± 0.91 PB (18.1) + MMIPPS (50) 12.38 ± 0.85 VPA (215.3) + vehicle 147.51 ± 13.98 VPA (215.3) + MMIPPS (50) 190.64 ± 14.27*

Data are presented as the mean concentrations (in µg/ml ± SE of 8 determinations) of AEDs in brain tissue. Statistical evaluation of data was performed with unpaired Student’s t-test. Brain tissue samples were taken at times scheduled for the MES test, and total brain AED concentrations were quantified using fluorescence polarization im- munoassay. For more details see legend to Table 2. * p < 0.05 vs.

control group (AED + vehicle-treated animals)

(8)

nature because MMIPPS did not significantly alter to- tal brain PB concentrations in the experimental ani- mals. In contrast, MMIPPS pharmacokinetically ele- vated the total brain VPA concentrations in the experi- mental animals. It is noteworthy that in this study, total brain AED concentrations were verified with fluorescence polarization immunoassay technique because, as reported earlier, only total brain AED con- centrations provide the exact classification and char- acterization of interactions between AEDs in preclini- cal studies [2, 18]. Moreover, in the presented study, total brain concentrations of CBZ or PHT were not measured because MMIPPS did not potentiate the anticonvulsant action of these AEDs in the mouse MES model.

The selective pharmacodynamic enhancement of the anticonvulsant action of PB in the mouse MES model by MMIPPS, one can try to explain in part by consider- ing the molecular mechanisms of action of the exam- ined drugs. As for PB, the drug binds to an allosteric regulatory site on the GABAA-benzodiazepine recep- tor-chloride ionophore complex, and enhances the GABAAreceptor-mediated currents that hyperpolarize postsynaptic neuronal cell membranes, thus impeding the epileptic activity transmission. PB blocks excita- tory responses induced by glutamate thereby the AMPA receptors. At high concentrations, PB blocks L- and N-type calcium currents [for review see 3, 6, 21]. In the case of MMIPPS, its molecular mecha- nism(s) of action is/are unknown. However, it is highly likely that this p-isopropoxyphenylsuccinimide derivative exerts its anticonvulsant action through the blockade of voltage-gated sodium channels [for more details see 15].

Taking into consideration the fact that MMIPPS could be a voltage-gated sodium channel blocker (like PHT and CBZ), it is highly likely that MMIPPS would potentiate the anticonvulsant action of PB, whose molecular mechanisms of action are different than the blockade of sodium channel conductance in neurons. Generally, it is accepted that AEDs with dif- ferent mechanisms of action can mutually enhance the protective action of these drugs in experimental stud- ies [4]. Of note, MMIPPS did not potentiate the anti- convulsant action of CBZ and PHT whose molecular mechanisms of action are mainly related to the block-

readily explain the observed interaction between MMIPPS and classical AEDs in the mouse MES- induced seizure model. In the case of the interaction between MMIPPS and VPA, it should be highlighted that MMIPPS elevated total brain concentration of VPA (by 29%), which resulted in a 17% reduction of ED50value for VPA in the mouse MES model. Thus, it can be suggested that the pharmacokinetic increase in total brain VPA concentrations was entirely respon- sible for the observed enhancement of the anticonvul- sant action of VPA in the mouse MES model. As re- gards molecular mechanisms of action of VPA, the AED also blocks sodium channels [for review see 9], and it seems that MMIPPS competes with VPA.

Considering the anticonvulsant profile of p-iso- propoxyphenylsuccinimide derivatives and their ef- fects on classical AEDs in the mouse MES model, it should be highlighted that p-isopropoxyphenylsucci- nimide monohydrate (IPPS) potentiated the anticon- vulsant action of PHT and VPA, but not that of CBZ and PB [13]. On the other hand, o-CAMIPPS attenu- ated the anticonvulsant action of CBZ and, simultane- ously, had no significant impact on the protective ac- tion of PHT, PB and VPA against MES-induced sei- zures in mice [12]. In the case of HMIPPS, it potentiated the anticonvulsant action of PB and VPA, but not that of CBZ and PHT in the mouse MES model [15]. On the other hand, a comparison of effects exerted by MMIPPS with those reported earlier for N-(anilinomethyl)-p-isopropoxyphenylsuccinimide (AMIPPS), confirms that both, N-(anilinomethyl)- and N-(morpholinomethyl)-derivatives of p-isoprop- oxyphenylsuccinimide significantly enhanced the an- ticonvulsant action of PB and VPA in the mouse MES model. Unfortunately, in both cases, the observed in- teractions of AMIPPS and MMIPPS with VPA were complicated by significant pharmacokinetic increases in total brain VPA concentrations [14]. The results of the presented study on MMIPPS, and reported earlier for AMIPPS indicate that both N-anilinomethyl and N-morpholinomethyl derivatives of p-isopropoxy- phenylsuccinimide possess the same pattern profile on the action of classical AEDs in the mouse MES model.

While explaining the results obtained in this study, another fact is worth mentioning. It was found that

(9)

MMIPPS administered either alone or in combination with classical AEDs, at doses corresponding to their ED50values from the MES test, did not affect the mo- tor coordination of animals in the chimney test. Addi- tionally, MMIPPS had no impact on long-term mem- ory and muscular strength in mice subjected to the step-through passive avoidance task and grip-strength test, respectively. Consequently, it can be concluded that the lack of adverse effects when MMIPPS was combined with classical AEDs testifies to the low toxic potential of these drug combinations.

In conclusion, this study suggests that the co- administration of MMIPPS with classical AEDs, espe- cially with PB, might be a promising treatment when applied in clinical settings in patients with tonic-clonic seizure or partial convulsions, with or without secon- darily generalization. Further neurochemical and elec- trophysiological studies are needed to confirm that MMIPPS might be considered as a supplementary therapeutic agent in the treatment of epilepsy.

Acknowledgments:

This study was supported by grants from the Medical University and Institute of Rural Health in Lublin, Poland. Professor J.J.

£uszczki is a Member of the Academy of Young Scholars of the Polish Academy of Sciences (Warszawa, Poland). The authors are grateful for the generous gifts of carbamazepine from Polpharma SA in Starogard Gdañski, Poland, and valproate from ICN-Polfa SA, Rzeszów, Poland.

Disclosure of conflicts of interest:

The authors have no disclosures to declare.

References:

1.Boissier JR, Tardy J, Diverres JC: A simple novel method to explore tranquilizer activity: the chimney test (in French). Med Exp (Basel), 1960, 3, 81–84.

2.Cadart M, Marchand S, Pariat C, Bouquet S, Couet W:

Ignoring pharmacokinetics may lead to isoboles misin- terpretation: illustration with the norfloxacin-

theophylline convulsant interaction in rats. Pharm Res, 2002, 19, 209–214.

3.Czapiñski P, B³aszczyk B, Czuczwar SJ: Mechanisms of action of antiepileptic drugs. Curr Top Med Chem, 2005, 5, 3–14.

4.Deckers CL, Czuczwar SJ, Hekster YA, Keyser A, Kubova H, Meinardi H, Patsalos PN, et al.: Selection of antiepileptic drug polytherapy based on mechanisms of action: the evidence reviewed. Epilepsia, 2000, 41, 1364–1374.

5.Kamiñski K, Obniska J: Synthesis and anticonvulsant properties of new 1-(2-pyridinyl)- 3-substituted

pyrrolidine-2,5-dione derivatives. Acta Pol Pharm Drug Res, 2008, 65, 457–465.

6.Kwan P, Brodie MJ: Phenobarbital for the treatment of epilepsy in the 21st century: a critical review. Epilepsia, 2004, 45, 1141–1149.

7.Lange J, Rump S, Ga³ecka E, Ilczuk I, Lechowska- Postek M, Rabsztyn T: Synthesis and properties of new cyclic derivatives of succinic acid with anticonvulsant activity. Pharmazie, 1977, 32, 82–84.

8.Litchfield JT, Wilcoxon F: A simplified method of evalu- ating dose-effect experiments: J Pharmacol Exp Ther, 1949, 96, 99–113.

9.Löscher W: Basic pharmacology of valproate: a review after 35 years of clinical use for the treatment of epi- lepsy. CNS Drugs, 2002, 16, 669–694.

10.Löscher W, Fassbender CP, Nolting B: The role of techni- cal, biological and pharmacological factors in the labora- tory evaluation of anticonvulsant drugs. II. Maximal elec- troshock seizure models. Epilepsy Res, 1991, 8, 79–94.

11.£uszczki JJ, Antkiewicz-Michaluk L, Czuczwar SJ: Iso- bolographic analysis of interactions between 1-methyl- 1,2,3,4-tetrahydroisoquinoline and four conventional an- tiepileptic drugs in the mouse maximal electroshock- induced seizure model. Eur J Pharmacol, 2009, 602, 298–305.

12.£uszczki JJ, Cioczek JD, Kocharov SL, Andres-Mach M, Kominek M, ¯ó³kowska D: Effects of three N-(car- boxyanilinomethyl) derivatives of p-isopropoxyphenyl- succinimide on the anticonvulsant action of carba- mazepine, phenobarbital, phenytoin and valproate in the mouse maximal electroshock-induced seizure model.

Eur J Pharmacol, 2010, 648, 74–79.

13.£uszczki JJ, Kocharov SL, Czuczwar SJ: Effect of p-isopropoxyphenylsuccinimide monohydrate on the an- ticonvulsant action of carbamazepine, phenobarbital, phenytoin and valproate in the mouse maximal electroshock-induced seizure model. Pharmacol Rep, 2010, 62, 194–202.

14.£uszczki JJ, Kocharov SL, Czuczwar SJ: N-(anilino- methyl)-p-isopropoxyphenyl-succinimide potentiates the anticonvulsant action of phenobarbital and valproate in the mouse maximal electroshock-induced seizure model. Neurosci Res, 2009, 64, 267–272.

15.£uszczki JJ, Kominek M, Florek-£uszczki M,

Tchaytchian DA, Kocharov SL, ¯ó³kowska D: Influence of N-hydroxymethyl-p-isopropoxyphenylsuccinimide on the anticonvulsant action of different classical antiepilep- tic drugs in the mouse maximal electroshock-induced seizure model. Epilepsy Res, 2012, 100, 27–36.

16.£uszczki JJ, Plech T, Wujec M: Effect of 4-(4- bromophenyl)-5-(3-chlorophenyl)-2,4-dihydro- 3H-1,2,4-triazole-3-thione on the anticonvulsant action of different classical antiepileptic drugs in the mouse maximal electroshock-induced seizure model. Eur J Pharmacol, 2012, 690, 99–106.

17.£uszczki JJ, Plech T, Wujec M: Influence of 5-(3- chlorophenyl)-4-(4-methylphenyl)-2,4-dihydro-3H- 1,2,4-triazole-3-thione on the anticonvulsant action of 4 classical antiepileptic drugs in the mouse maximal electroshock-induced seizure model. Pharmacol Rep, 2012, 64, 970–978.

Interaction of MMIPPS with antiepileptic drugs

Dorota ¯ó³kowska et al.

(10)

19.Meyer OA, Tilson HA, Byrd WC, Riley MT: A method for the routine assessment of fore- and hindlimb grip strength of rats and mice. Neurobehav Toxicol, 1979, 1, 233–236.

20.Obniska J, Chlebek I, Kamiñski K, Bojarski AJ, Sata³a G: Synthesis, anticonvulsant activity and 5-HT1A/5-HT7 receptors affinity of 1-[(4-arylpiperazin-1-yl)-propyl]- succinimides. Pharmacol Rep, 2012, 64, 326–335.

21.Rogawski MA, Porter RJ: Antiepileptic drugs: pharma- cological mechanisms and clinical efficacy with consid- eration of promising developmental stage compounds.

Pharmacol Rev, 1990, 42, 223–286.

22.Sander JW: The epidemiology of epilepsy revisited. Curr Opin Neurol, 2003, 16, 165–170.

23.Stables JP, Kupferberg HJ: Chapter 16 – The NIH Anti- convulsant Drug Development (ADD) Program: preclini- cal anticonvulsant screening project. In: Molecular and

Morre M, Dodd RH, Rossier J: Benzodiazepine impairs and beta-carboline enhances performance in learning and memory tasks. Nature, 1986, 321, 864–866.

25.White HS, Woodhead JH, Wilcox KS, Stables JP, Kupferberg HJ, Wolf HH: Discovery and preclinical de- velopment of antiepileptic drugs. In: Antiepileptic drugs, 5th edn. Eds. Levy RH, Mattson RH, Meldrum BS, Perucca E, Lippincott Williams & Wilkins, Philadelphia, 2002, 36–48.

26.Zejc A, Obniska J, Wilimowski M, Rutkowska M, Witkowska M, Barczyñska J, Kêdzierska-GoŸdzik L et al.: Synthesis and anticonvulsant properties of some aryl- succinate methylpyridylimides. Pol J Pharmacol Pharm, 1990, 42, 69–77.

Received: July 30, 2012; in the revised form: November 6, 2012;

accepted: November 16, 2012.

Cytaty

Powiązane dokumenty

To separate the relationship between the different effects of morphine and the gene expression profiles in the striatum, we compared responses to acute and chronic drug treatment

In our study, the combined treatment with CDP and L-701,324 (a glycine B site antagonist) or D -cycloserine (a glycine site partial agonist), all at low, ineffective doses,

Results: Repeated flunitrazepam administration (1 mg/kg, sc and 2 mg/kg, ip) for 8 consecutive days induced tolerance to the motor impairing effects of flunitrazepam in mice, both

Anticonvulsant activity of the antiepileptic drugs was determined in mice pretreated with these drugs before MES as ED 50 values (i.e., doses of the drugs in mg/kg, protecting 50%

Background: The aim of the study was to analyze the influence of acute and chronic treatment with tianeptine, an antidepressant se- lectively accelerating presynaptic

Administration of propranolol to sham-operated rats did not affect cancellous bone of the femur, but increased the width of trabeculae of the femoral meta- physis in ovariectomized

The present results also showed that risperidone at a higher dose (0.1 mg/kg) inhibited the decrease in memory retention of the object recognition evoked by MK-801 (0.2 mg/kg) (F(1,

Background: The purpose of this study was to evaluate the effect of hydrochlorothiazide (HCTZ), a thiazide-type diuretic and an antihypertensive drug, on the anticonvulsant activity