Pharmacological Reports Copyright © 2013 2013, 65, 787–801 by Institute of Pharmacology ISSN 1734-1140 Polish Academy of Sciences

Review

Researchadvancesinbasicmechanisms ofandantiepilepticdrugaction

W³adys³awLasoñ1,Ma³gorzataChlebicka2,KonradRejdak2

1 DepartmentofExperimentalNeuroendocrinology,InstituteofPharmacology,PolishAcademyofSciences, Smêtna12,PL31-343Kraków,Poland

2 DepartmentofNeurology,MedicalUniversityofLublin,Jaczewskiego8,PL20-090Lublin,Poland

Correspondence: W³adys³awLasoñ,e-mail:[email protected]

Abstract: is a common neurological disease but the mechanism of generation has been only partially unraveled. Furthermore, almost 30% of epileptic patients are resistant to pharmacological treatment. Therefore, elucidation of the basic mechanism of sei- zures and search for new antiepileptics in order to treat the drug-resistant form of epilepsy and to improve the efficacy of current therapies seem justified. The aim of this overview is a brief presentation of some new concepts and research directions in pathogene- sis and pharmacotherapy of epilepsy. Development of ideas on the mechanisms of seizures and antiepileptic drugs reflects the prog- ress in our understanding of the central nervous system physiology, particularly of neurotransmission. Hyperactivity of excitatory amino acid systems, insufficient GABAA receptor-mediated neurotransmission, and disturbances in intrinsic properties of neuronal membranes are still regarded as the most important mechanisms of seizures. New data add to the complexity of GABA-glutamate in- teraction showing both excitatory and inhibitory role of GABA and glutamatergic in the central nervous system. Moreover, besides synaptic NMDA and GABAA receptors, also extrasynaptic receptors for the amino acid transmitters have been recently im- plicated in the pathomechanism of epilepsy. Changes in expression, polymorphisms, lost- or gain-function mutations as well as cel- lular energetic imbalance can contribute to the disturbed function of the - and voltage-dependent sodium, potassium, chloride and calcium channels, resulting in epileptiform activity. Voltage-dependent sodium and calcium channel blockers, and GABA mi- metics are the most clinically useful groups of antiepileptic drugs and the newest research in this field is focused on more selective and subtle regulations of their molecular targets. Of interest is an emerging role of extrasynaptic GABAA receptors, various kinds of potassium ion channels, hyperpolarization-activated cyclic nucleotide gated (HCN) channels, acid-sensing ion channels, and gap junctions in the regulation of neuronal excitability and seizures. Iono- and metabotropic glutamate receptors used to be viewed as an attractive target for new , however, opinions are now less enthusiastic, since their competitive and non-competitive antagonists possess undesired side effects. Positive or negative allosteric modulators of glutamate receptors with fewer side-effects can be more promising. The introduction of new compounds acting through novel pharmacological mechanisms gives hope that the proportion of patients with uncontrolled epilepsy will substantially decrease. However, this may be possible if molecular back- groundofthepharmacoresistanceinepilepsyisdeciphered.

Keywords: seizures,epilepsy,antiepilepticdrugs,molecularmechanisms

Pharmacological Reports, 2013, 65, 787–801 787 Introduction ligand- or voltage-gated sodium and calcium chan- nels. Deficiency in the inhibitory processes is linked mainly with insufficient GABAA receptor-mediated Epilepsy is regarded as a multifactor and sympto- neurotransmission and extracellular potassium cur- matologically highly diversified neurological disorder rents. Changes in expression, polymorphisms, lost- or characterized by recurrent unprovoked seizures. gain-function mutation of the ligand- and voltage- Physiological studies show that seizures reflect tran- dependent sodium, potassium, chloride and calcium sient, abnormal and synchronous hyperactivity of channels as well as cellular energetic imbalance and a neuronal population in the . Such brain dys- inflammatory processes can contribute to neuronal function can be accompanied by motor, sensory and hiperexcitabilityandseizures[3,37,77,116,125]. autonomic disturbances depending on brain region in- volved in the origin and/or spread of seizures. The re- lationships between cortical EEG, and extracellular and intracellular recordings from the focus of seizures TheGABAergicsystem evoked by local administration of a convulsant to the mammalian cortex was described by Ayala et al. [6]. The simplest form of epileptic activity which can be The g-aminobutyric acid (GABA) since long has been recorded in cortical or hippocampal neurons is the in- considered to be the principal inhibitory neurotrans- terictal spike, a synchronized burst of action poten- mitter in the mammalian brain. Enhancers of GABA- tials evoked by recurrent excitation. The depolariza- ergic transmission comprise a large group of classical tion underlying the interictal spike named a paroxys- and new generation antiepileptic drugs [30]. It has mal depolarization shift is followed by a prolonged been firmly established that GABA is produced by in- potassium current-dependent after-hyperpolarization. traneuronal decarboxylation of glutamate, and after The cellular mechanisms of seizure generation in- being released, its intrasynaptic action is quickly ter- volve rhythmic or tonic “runaway” excitation or the minated by and presynaptic neuronal uptake in- synchronized and rhythmic interplay between excita- volving the high-affinity GABA transporters. There tory and inhibitory neurons and membrane conduc- are four GABA transporters expressed in the human tances [76]. Seizures are classified into partial (simple brain that are named GAT-1, GAT-2, GAT-3 and or complex) and generalized categories including ab- betaine-GABA transporter BGT1. The glial and neu- sence, tonic, clonic, tonic-clonic, myoclonic, and feb- ronal GAT-1 received particular attention, as besides rile seizures. It has been firmly accepted that seizures the GAT-3, Gat-1 predominantly participates in can be generated in response to a loss of balance be- GABA uptake and is the target for the efficient anti- tween excitatory and inhibitory influences which re- epileptic drug [110]. The GABA uptake by sults in tonic depolarizations or repetitive, rhythmic GAT-1 was also shown to be compromised in the ge- burst discharges [76]. Experimentally, epileptiform netic model of absence epilepsy [27], however, in the burst discharges can be evoked in neuronal cultures light of differential involvement of GABA in the by removing extracellular magnesium [80], increase regulation of convulsive and nonconvulsive seizures, in extracellular potassium concentration [118], inhibi- significance of this phenomenon remains uncertain. tion of the sodium pump [8] or antagonizing GABAA There is a growing interest in amino acid transporters receptors [45]. The most common animal models of located extrasynaptically such as BGT1, as the inhibi- seizures comprise chemically induced seizures (pen- tor of both GAT-1 and BGT1 showed tylenetetrazole, kainic acid, pilocarpine), maximal effects in animal models of epilepsy [69, 110]. GABA electroshock and electrical or chemical kindling. Neu- is intracellularly catabolized by GABA transaminase rochemical approach strongly supports the electro- and the inhibition of this markedly increases physiological findings that seizures can be generated the GABA level in the brain tissue, which correlates from excessively enhanced excitatory processes in with an elevated seizure threshold. The antiepileptic a given population of neuronal cells in the brain or drug is an irreversible GABA transaminase from hypoactivity of neuronal inhibition. The former inhibitor, but the alternative mechanism of action of mechanism comprises hyperactivity of glutamatergic this drug was proposed to involve reverse transport of transmission and functional disturbances of the GABA by GAT-1 and GAT-3 transporters [83]. Be-

788 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

sides the impressive progress in knowledge on GABA brain, GABA depolarizes cells, which is likely to be production and in the central nervous sys- due to a high intracellular chloride level as compared tem, large body of evidence has been recently accu- with adult tissue [10]. The ability of GABA to hyper- mulated on the role of GABA receptors in the mecha- polarize neurons probably depends on higher expres- nism of seizures and in the action of antiepileptic sion of the chloride-extruding transporter KCC2 in drugs. GABA exerts its action via ionotropic GABAA late stages of brain development and decrease in ex- and the metabotropic GABAB receptors. The pen- pression of NKCC1 which transports chloride into tameric GABAA receptor complex is composed of cells [34, 102]. Interestingly, NKCC1 expression is two a and two b subunits and one g or d subunit elevated in patients with hippocampal sclerosis and which form ligand-gated chloride channel and de- cortical dysplasia suggesting its role in the pathogene- pending on the subunit composition of the receptor, it sis of refractory human epilepsy [113]. The widely ac- mediates tonic or phasic inhibition [83]. The GABAA cepted view on the role of GABA in mediating pre- receptor configuration also determines the affinity of dominantly inhibitory processes in the central nervous its agonists, antagonists and allosteric modulators. system has been challenged by some data from ex- Activation of GABAA receptor in the brain generates perimental studies. In particular, it has been reported fast inhibitory postsynaptic potentials (IPSP) and the that initiation and maintenance of epileptiform syn- key role of this receptor complex in prevention and chronization in the hippocampal-entorhinal cortex inhibition of seizures (except for the absence noncon- slices perfused by artificial cerebrospinal fluid devoid vulsive seizures) is commonly accepted [32]. The an- of magnesium ions depends on GABAA receptor ac- tagonists of this receptor (e.g., bicuculline, picrotoxin, tivity [4]. The ability of GABAA receptor-mediated or pentetrazole) belong to the most potent convul- mechanism to synchronize neuronal networks con- sants, whereas GABAA receptor agonists or positive necting the hippocampus, the entorhinal and perirhi- modulators (benzodiazepines, barbiturates, some neu- nal cortices, or the amygdala, may play a role in gen- rosteroids) inhibit seizures in both experimental ani- erating the recurrent limbic seizures which are charac- mals and in humans. Like deficit in GABA synthesis teristic of [5]. Taking as an or loss of GABA interneurons, also some changes in example the status epilepticus-induced reorganization expression or mutations of the GABAA receptor of GABAergic/glutamatergic network in the hippo- subunits can lead to pathological neuronal discharges. campus, Ben-Ari has postulated that the increase in Indeed, impairment of GABAergic transmission is the glutamatergic drive and the loss of the GABA- likely to contribute to seizure susceptibility observed mediated inhibitory drive in the dendrites of the prin- in several genetic and acquired animal models of epi- cipal cells in the hippocampus accompanied by a per- lepsy [91, 120]. The maintaining of the proper brain sistent increase in intracellular chloride concentration excitability level appears to depend on the balance be- contribute to seizure generation. The author stressed tween synaptic excitation and inhibition. Although in that seizure propagation to the other hippocampus de- the mammalian cortex probably only 10–20% of neu- pended on high-frequency oscillation which required rons synthesize GABA and ca. 17% of re- active NMDA and GABAA receptors [10]. The switch leases this transmitter, the GABA interneurons via GABAA receptor mode of action from inhibitory to GABAA receptors efficiently control excitatory amino excitatory during seizure activity, that might account acid transmission in the cortex. The functional bal- for intractability of some , was also reported ance between inhibitory and excitatory neurotrans- by other investigators [48]. Interestingly, experiments mission can be attributed to the ability of a single on brain tissue from patients with mesial temporal GABA interneuron to inhibit many glutamatergic lobe epilepsy demonstrated that only the subicular cells (divergence) and to converge a number of pyramidal cells which lacked expression of the potas- GABA interneurons on the same glutamatergic neu- sium-chloride KCC2, revealed GABAA ron in local cortical circuits. However, the relation- receptor-mediated depolarizing postsynaptic events, ship between GABAergic and glutamatergic neuro- further confirming the significance of disturbance in transmission can be more complex because the releas- chloride ion homeostasis in the pathomechanism of able pool of both GABA and glutamate can be epilepsy [47]. Disequilibrium of the chloride distribu- co-localized in the same neurons. Moreover, during tion was observed in some patients with intractable early stages of CNS development in contrast to adult epilepsy [44] and disturbance in chloride ion homeo-

Pharmacological Reports, 2013, 65, 787–801 789 stasis and GABAergic transmission facilitate epileptic In contrast to the well-recognized role of ionotropic discharges [46]. Results from animal models and hu- GABAA receptor in the pathomechanism of seizures, man studies indicate that changed expression of less consistent data have been obtained on engage- GABAA receptor subunits may contribute to the ment of metabotropic GABAB receptor in the regula- pathomechanism of intractable epilepsy [19, 63]. tion of brain excitability. GABAB receptors mediate Laschet et al. [56] found that maintaining the GABAA slow inhibition and are localized presynaptically, thus, receptor in a phosphorylated state was necessary to they can inhibit release of both excitatory and inhibi- sustain efficient GABAergic inhibition in the epilep- tory [53]. Agonists of GABAB re- togenic cortex. These authors suggested the existence ceptors show pro- or anticonvulsant effect in pentetra- of a functional link between the regional cerebral glu- zole-induced seizures in immature animals [71] and cose hypometabolism in patients with partial epilepsy inhibit cocaine-induced seizures [41]. Agonists and and dysfunction of GABAergic mechanism due to de- antagonists of GABAB promote and suppress general- ficiency in GABAA receptor phosphorylation. This is ized absence seizures in genetic animal models of an interesting hypothesis, especially if one takes into this disorder, respectively. Peripherally administered account that the majority of antiepileptic drugs de- GABA enhancers in a majority of cases suppress con- crease metabolic processes in the brain tissue. Of vulsive seizures in both animals and humans, but pro- great interest is an emerging role of extrasynaptic mote the occurrence of spike and wave discharges in GABAA receptors in the regulation of neuronal excit- thalamo-cortical circuits that are typical of general- ability and seizures. Using the pilocarpine model of ized absence epilepsy [22, 32]. The GABA receptor- temporal lobe epilepsy in mice, Zhang et al. showed mediated hyperpolarization of relay nuclei of the that surprisingly, tonic inhibition in dentate granule thalamus de-inactivates low-threshold calcium cur- cells was observed in epileptic mice and that these rents and facilitates rhythmic activity in thalamo- animals had altered expression of GABAA receptor corticalcircuit[32]. subunits on dentate granule cell dendrites and the g2 subunit was shifted from synaptic to perisynaptic lo- cation [140]. It is now well established that there are not only synaptic, but also extrasynaptic high affinity Theglutamatergicsystem GABAA receptors differentially distributed in the brain. The latter receptors which are activated by ex- trasynaptic GABA and some endogenous modulators Glutamic acid is the main excitatory amino acid neu- are responsible for persistent tonic inhibition and are rotransmitter in the central nervous system. It acti- likely to control susceptibility to seizures [127]. In- vates both ionotropic receptors named by their ago- deed, it has been demonstrated in some models of sei- nists NMDA (N-methyl-D-aspartic acid), AMPA zures that the tonic inhibition is maintained in epilep- (a-amino-3-hydroxy-5-methyl-isoxazolopropionic acid), tic tissue, in contrast to defective function of synaptic and kainate receptors, as well as metabotropic gluta- GABAA receptors and diminished synaptic inhibition mate receptors, which via G-proteins affect intracellu- [92]. It has been also suggested that the decrease in lar processes and ion channel activities [123]. Iono- neocortical phasic GABA inhibition and enhancement tropic glutamate receptor agonists show potent procon- of tonic inhibition in the thalamo-cortical circuits are vulsant properties and are often used in experimental likely to be involved in the pathomechanism of ab- modelsofseizures. sence epilepsy as demonstrated in both pharmacologi- The NMDA receptors are tetramers, composed of cal and genetic models of this disorder [29]. In addi- NR1 subunit in various alternative splicing forms and tion, selective activation of extrasynaptic GABAA re- NR2A – NR2D subunits. The subunit composition de- ceptors in the thalamus was sufficient to induce termines ligand affinity, conductance for Ca2+ ions absence seizures in non-epileptic animals [27]. It is and kinetics of NMDA-gated ion channel [89]. The believed that the extrasynaptic GABAA receptors, NMDA receptor activation requires simultaneous which are sensitive to ambient concentration of extra- binding of glutamate and /D-serine and AMPA cellular GABA can be promising targets for antiepi- receptor-mediated initial depolarization for removing leptic, antiparkinsonian, antipsychotic and procogni- magnesium ions which block NMDA receptors at tivedrugs[18]. resting membrane potentials. Glutamate binds with

790 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

a higher affinity to NMDA receptors than to AMPA stration of glycine and a polyamine site antagonists receptors and the NMDA-gated channel remains open had beneficial effects in experimental seizure models. even for a couple of seconds after agonist binding It should also be mentioned here that through the acti- have taken place. Also the conductance of NMDA re- vation of GABAergic interneurons, NMDA receptors ceptors for calcium ions is higher than that of other can suppress circuit level neural activity, especially the cation-conducting ionotropic receptors. Depending on limbic circuits [38]. Epileptic seizures are associated the concentration of calcium ions entering the cells with a drop of brain tissue pH, which should lead to determining frequency of synaptic stimulation, one proton inhibition of NMDA receptor activity. On the can observe activation of protein kinases (when the other hand, inhibitors of acid-sensing ion channels de- intracellular calcium level is low) or phosphatases crease seizure-like bursting in hippocampal slices sug- (when calcium level is high). These phenomena may gesting a new target for antiepileptic therapy [137]. be responsible for processes of synaptic potentiation In contrast to the well-recognized involvement of or depression via the regulation of AMPA receptor synaptic NMDA receptors in the regulation of brain ex- phosphorylation. The NMDA receptor activity is citability, less is known about functional role of extra- regulated by a number of agents, such as polyamines, synaptic NMDA receptors in pathomechanism of sei- zinc ions, nitric oxide or steroid hormones. Experi- zures. These extrasynaptic receptors, which can be acti- mental data provided a strong evidence for the key vated by excitatory amino acid synaptic spillover, have role of NMDA receptors in neuronal plasticity, but different subunit composition and couple with distinct also in seizures and neuronal damage. In experimental signaling pathways than the synaptic sites [59]. It has animals NMDA receptor antagonists, e.g., dizocilpine been postulated that extrasynaptic NMDA receptor acti- or ketamine, inhibit seizures evoked by pentetrazole, vation may lead to neuronal damage [59, 95], and in this pilocarpine, maximal electroshock or sensory stimula- way may contribute to development of seizures. tion. These NMDA antagonists appear to delay devel- The AMPA receptor which is responsible for gen- opment of kindling but have a weaker effect on the eration of fast excitatory postsynaptic potentials fully developed kindling [61]. Unfortunately, both (EPSP) in the central nervous system forms channels competitive and non-competitive NMDA receptor an- permeable to sodium, potassium and, with some limi- tagonists showed in experimental and clinical studies tation, calcium ions. In the presence of an agonist, the serious undesired effects, such as motor coordination AMPA receptor desensitizes and the desensitization is and memory impairment and disorientation. More- inhibited by positive allosteric modulators of this re- over, these unwanted symptoms are aggravated in ani- ceptor. Negative modulators of AMPA receptors, e.g., mal models of epilepsy. Therefore, some allosteric 2,3-benzodiazepines were also synthesized. The AMPA modulators of NMDA receptors, especially those receptor is composed of four homologous proteins which interact with glycine and polyamine binding GluR1–4 (or GluRA–D) in various configurations. sites seemed more promising as potential antiepileptic Each of these proteins can exist in two alternative- drugs [86]. Antagonists of glycine binding sites show splice variants, named “flip” and “flop”. The presence affinity for NR1/NR2A subunits, whereas ligands of of GluR2 subunit in AMPA receptor complex deter- polyamine site bind to NR1A/NR2B subunit com- mines low permeability to calcium ions. Those AMPA plexes [42]. The anticonvulsant effect of the partial receptors that do not possess GluR2 show a two-fold agonist of the glycine-binding site, D-, higher permeability to calcium ions than to sodium probably involves desensitization of the NMDA re- ones. The AMPA receptor is phosphorylated by sev- ceptor. Moreover, D-cycloserine potentiates anticon- eral protein kinases, and this process is often initiated vulsant effects of some antiepileptic drugs and in low by NMDA receptor-dependent intracellular flow of doses has a positive influence on memory processes calcium ions. Phosphorylation of AMPA receptor fur- [134, 135]. The new generation antiepileptic drug, ther enhances ion flow and can lead to pathological harcoseride is an antagonist of the glycine binding site hyperactivity of glutamatergic transmission and epi- on NMDA receptor complex. Also felbamate, besides leptic discharges. Contrariwise, calcineurin-induced blocking voltage-dependent sodium channels and en- dephosphorylation of AMPA receptor decreases its hancing GABAergic transmission, showed antagonis- activity. tic activity at glycine binding site in the NMDA re- AMPA and kainate receptor antagonists showed ceptor. It is noteworthy that also concomitant admini- anticonvulsant effects in genetic and chemical models

Pharmacological Reports, 2013, 65, 787–801 791 of seizures. Their adverse effects on memory and mo- are localized postsynaptically on both glutamatergic tor coordination in experimental animals are less pro- hippocampal and cortical cells and GABAergic inter- found than those evoked by the NMDA receptor an- neurons. Their activation leads to phosphorylation tagonists [105, 107]. Moreover, the AMPA and kai- and inactivation of several types of potassium chan- nate receptor antagonists enhance efficacy of classical nels, which leads to depolarization of neuronal cells. antiepileptic drugs and their interactions with pheno- The group III mGluRs are presynaptic inhibitory barbital and appear to be especially posi- autoreceptors localized on glutamatergic cell endings tive. Among antiepileptic drugs, barbiturates and topi- and presynaptic inhibitory heteroreceptors on some ramate showed ability to block the AMPA receptors GABAergicneuronalcells. [67, 108]. Also another potential antiepileptic drug, Pharmacological studies show that agonists of talampanel, is a noncompetitive AMPA receptor an- group I mGluR induce seizures in experimental ani- tagonist with broad spectrum of anticonvulsant activ- mals and prolong duration of interictal and ictal dis- ity. Kainate receptors are several times less abundant charges in hippocampoal slices, whereas antagonists in the brain tissue than AMPA receptors, but their of these receptors suppress seizures. In contrast, the widespread distribution suggests that they are present stimulation of group II and III mGluRs prevents sei- in the majority of neuronal cells. Cloning studies zures. Furthermore, the group II mGluR agonist showed, that kainate receptors could be composed of LY354740 significantly enhanced anticonvulsant ef- GluR5, GluR6, GluR7 and KA1 and KA2 subunits. fect of diazepam while the mGluR5 antagonist MPEP The GluR5 and GluR6 can form homomeric channels, showed a positive interaction with some conventional whereas other subunits are a part of heteromeric com- antiepileptic drugs in the amygdala-kindling model of binations with the GluR5 and GluR6 proteins. Long- complex partial seizures [16, 52]. Metabotropic gluta- lasting depolarization of neurons by postsynaptically mate receptors can be also involved in the regulation localized kainate receptors leads to intracellular flow of non-convulsive seizures because ligands of these of calcium ions via the voltage-dependent calcium receptors suppress seizure activity by the selective re- channels, and this effect may be of significance in duction of hyperactive glutamatergic synaptic com- synaptic plasticity, but also in seizurogenic and neuro- munication within the cortex and thalamus [2, 85]. toxic effects of kainate [14, 126]. The well-established Therefore, group I mGluR antagonists and group II convulsant and neurotoxic effects of kainate can be and III mGluR agonists can be regarded as potential also associated with presynaptic inhibition of GABA antiepileptic and neuroprotective drugs, provided that release. This assumption is supported by observation theylackseriousundesiredeffects[72,81]. that kainate diminishes GABA-dependent inhibitory The process of glutamate release and uptake in synaptic potentials in hippocampal CA1 region. An- brain tissue engages several membrane transporters. other interesting hypothesis is that presynaptic kainate There are 5 glutamic acid transporters (GLAST, receptors are involved in enhancement of glutamate re- GLT-1, EAAC1, EA4 and EAAT5) which are local- lease, as the activation of these receptors increases the ized in membranes of and neurons. The concentration of calcium ions in hippocampal synapto- synthesis and activity of glutamate transporters is somes. The above-mentioned facts suggest that some regulated by the concentration of glutamic acid, as the selective modulators of kainate receptors can be con- main signal, whereas inactivation of these proteins re- sidered as potential antiepileptic drugs. sultsinseizuresandneurotoxicity[122,123,130]. The family of metabotropic glutamate receptors A decrease in hyperactivity of glutamatergic trans- comprises 8 subtypes of receptors, mGluR1 – mission in order to inhibit seizures can be achieved via: mGluR8, which were divided into 3 groups on the ba- 1) inhibition of glutamic acid synthesis using azaser- sis of differences in amino acid sequence, pharmacol- ine or glutaminase inhibitors, e.g., 6-diazo-5-oxy-1- ogical characteristic and intracellular signaling path- norleucine. Among antiepileptic drugs, gabapentin at- ways. The activation of mGluR belonging to Group I tenuates glutamic acid synthesis through the inhibi- (mGluR1 and mGluR5) stimulates phosphatidylinosi- tion of BCAA aminotransferase, whereas vigabatrin tol (IP3) synthesis and release of intracellular cal- decreases both glutamic acid and D-aspartic acid lev- cium, whereas the stimulation Group II (mGluR2 and els in brain tissue. Also valproate reduces the concen- mGluR3) and group III (mGluR4, mGlu6-8) mGluRs tration of the excitatory amino acid, aspartate in nerve inhibits adenylate cyclase [25]. The group I mGluRs terminals[82];

792 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

2) decrease in synaptic release of glutamic acid tween segments 5 and 6 of each domain forms the ion through modulation of its presynaptic receptors or selectivity filter [24, 139]. The a-subunits NaV1.1, calcium-dependent neurotransmiter release. Lamo- NaV1.2, NaV1.3, and NaV1.6 are predominantly ex- trigine is an antiepileptic drug which, besides blocking pressed in the central nervous system. The b-subunits voltage-dependent sodium channels, inhibits in mi- consist of one N-terminal domain, one transmembra- cromolar concentrations glutamic acid and D-aspartic ne domain, and one C-terminal domain. The b-sub- acidrelease; units can regulate the expression of VGSC, increasing 3)enhancementofglutamatere-uptake; sodium channel density and neuronal excitability 4) attenuation of postsynaptic glutamate effects using an- [90]. The voltage-dependent sodium channels play the tagonists of ionotropic and group I metabotropic gluta- key role in generating both normal action potentials mate receptors, which induce desensitization or de- and seizures [54]. Epileptic seizures are associated crease density of glutamate receptors. Among antiepi- with modest sustained depolarization below the inac- leptic drugs which in their mechanism of action show tivation threshold for action potential generating so- antagonistic component at glutamate receptors, one can dium channels. The depolarization enables synchro- mention felbamate, topiramate and phenobarbital. nous, high-frequency neuronal firing, the electrocorti- Some data suggest that when analyzing glutamate cographic correlate of which comprises ictal epileptic role in seizure phenomena, one should take into ac- field potentials. Antagonists of voltage-dependent so- count the complex and only partly recognized interac- dium channels decrease the maximal amplitude of so- tions between glutamatergic neurons, glia and extra- dium current and prolong the time of the channel in- cellular matrix [60, 62]. In fact, an interesting hy- activation. In this manner they decrease availability of pothesis was formulated suggesting that epileptic sodium channels during high-frequency epileptic dis- discharges resulted from pathological high-frequency charges [108]. Inactivation refers to depolarization- calcium waves produced by syncytia, which induced conformational changes from an open- led to synchronization of neuronal discharges. These channel to its non-conducting state. Usually, the iso- processes are accompanied by an augmented release forms Nav1.1–Nav1.4, Nav1.6, and Nav1.7 have fast of excitatory neurotransmitters which further facili- inactivation kinetics, whereas Nav1.5, Nav1.8, and tate generation of calcium waves in astrocytes [23, Nav1.9 have slower inactivation [109]. Most of the 58]. Adenosine is an endogenous anticonvulsive and traditional antiepileptic drugs which are sodium chan- neuroprotective agent of glial origin which is re- nel antagonists stabilize mainly the fast inactivation garded as a promising target for antiepileptic therapy of the voltage-gated sodium channels, whereas laco- [15, 33, 60]. Gap junctions responsible for electro- samide acts on the process of slow inactivation. Thus, tonic coupling between neurons and astrocytes and phenytoin carbamazepine, lamotrigine, felbamate, synchronization of epileptoidal discharges are also topiramate, oxcarbazepine, zonisamide, rufinamide, considered as a potential target for future antiepileptic eslicarbazepine acetate, and probably gabapentin, and drugs. The role of the extracellular matrix in the sodium valproate bind to the fast-inactivated state of mechanism of seizures has been only recently taken the voltage-gated sodium channel and produce a tran- into account [66, 138]. Correspondingly, metallo- sient, voltage- and frequency-dependent reduction in proteinase-9 inhibition was shown to attenuate experi- channel conductance [28, 77]. A new generation an- mental epileptogenesis [133], but these results have to tiepileptic drug, lacosamide binds to the slow- besupportedbyfurtherstudies. inactivated state of the voltage-gated sodium channel and evokes a long-lasting, voltage- and frequency- dependentdecreaseinchannelconductance[9,119].

Thevoltage-gatedsodiumchannels

Voltage-gated sodium channels are integral mem- Thevoltage-gatedcalciumchannels brane glycoproteins composed of an a-subunit that is associated with b-subunits [24, 109]. The a-subunit contains four multispanning homologous domains Voltage gated calcium channels are heteromeric com- that form the central pore of the channel. The loop be- plexes with the pore-forming a1 subunit and acces-

Pharmacological Reports, 2013, 65, 787–801 793 sory subunits. Their molecular structure and mecha- can be classified in four big subfamilies, i.e., inwardly + + nism of inactivation are similar to the voltage-gated rectifying K channels (Kir), two-pore K channels + sodium channels. The neuronal voltage-gated calcium (K2P), calcium-activated K channels (KCa) and volt- channels are divided into L-, P/Q-, N- and T-type age-gated channels (Kv) [131, 136]. Presently, 12 channels. The L-type calcium channels are predomi- subfamilies of voltage-gated potassium channels nantly localized postsynaptically and belong to the Kv1–Kv12 with various homology and ability to form slowly inactivating category. They mediate sustained tetramers and encoded by 40 genes in humans have calcium ion influx in postsynaptic neurons, which been identified [50]. The subfamily of voltage- further augments neuronal depolarization. L-type cal- dependent potassium channels KCNQ (Kv7) com- cium channel antagonists show antiepileptogenic prises: KCNQ1/KCNE1 responsible for action poten- properties but they can aggravate absence epilepsy tial repolarization in the cardiac muscle, KCNQ2/3 [55, 124]. N-type and P/Q-type channels are ex- underlying M-current, which controls resting mem- pressed presynaptically and are involved in neuro- brane potentials and frequency of action potentials in transmitter exocytosis. The T-type channels open with neurons, KCNQ4 expressed in the inner ear and the slight depolarization, and are quickly inactivated. The KCNQ5 which forms complexes with the KCNQ3 2+ low-threshold Ca current regulates oscillatory activ- [111]. Functional significance of the various voltage- ity of thalamic neurons (pacemaker) and probably dependent potassium channels is supported by genetic participates in generating generalized absence sei- studies. The mutation of KCNQ1/KCNE1 genes re- zures characterized by 3 Hz spike-wave discharges in sults in the long QT syndrome (LQTS), mutation of the EEG recordings. Indeed, polymorphisms or muta- KCNQ2/3 is associated with the benign familial neo- tions in the CACNA1H gene coding for the voltage natal convulsions (BFNC), whereas mutation of 2+ gated T-type Ca channel can be associated with KCNQ4 is linked with the congenital deafness [70]. pathogenesis of some generalized epilepsies, e.g., the The KCNQ2 and KCNQ3 genes encode proteins childhood absence epilepsy [1]. Voltage-gated cal- which are important regulators of neuronal membrane cium channels can be divided into low-voltage- excitability and, generally, the activation of Kv7 activated channels and high-voltage-activated chan- channels inhibits depolarization of neuronal cells [21, nels. The low-voltage-activated (T-type) calcium 78, 79]. The KCNQ (Kv7) channels regulate the re- channel blockers, such as ethosuximide, zonisamide, sponse of neurons to excitation and form a potent in- and probably sodium valproate prevent synchronized hibitory mechanism in the brain. In 2000, it was dis- depolarization of neurons, particularly in the thalamo- covered that KCNQ potassium channels were a mo- cortical circuits. The high-voltage-activated calcium lecular target for retigabine [78]. Thus, retigabine was channel blockers, such as lamotrigine (N-type, P/Q- the first drug that was recognized as a selective acti- type) felbamate, gabapentin (a2b-1 subunit), topira- vator of neuronal KCNQ2-5 channels, but devoid of mate, levetiracetam (N-type), pregabalin (a2b-1 affinity for KCNQ1 channels in the cardiac muscle. subunit), like phenobarbital, prevent The KCNQ2/3 potassium channels are key players in release (N- and P/Q-type) and post-synaptic depolari- the mechanism of inhibition of neuronal excitability zation(L-type)[74,77]. so, one can postulate that retigabine influences one of the most fundamental mechanisms of epilepsy [106]. It has been pointed out that even a slight reduction in KCNQ2/KCNQ3 function can be epileptogenic and The voltage-dependent potassium channel mutations in either KCNQ2 or KCNQ3 cause BFNC epilepsy [12]. The defective functioning of the volt- Potassium channels form the largest and the most di- age dependent potassium channel leads to the loss of verse family of ion channels involved in the regula- control over excitability. In contrast, heteromerization tion of a variety of physiological functions. The of KCNQ2 and KCNQ3 strongly potentiates the M- subunits of potassium channels are encoded in hu- current resulting in anti-epileptogenic effects [36]. mans by 78 genes, however, their alternative splicing Modulation of M-currents has a great impact on neu- and heteromerization add to the complexity of the ronal excitability because these currents are the only functional protein complexes. Taking into account the ones which are maintained during initiation of action structure and mode of activation, potassium channels potential and have ability to inhibit epileptic dis-

794 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

charges. M channels are not involved in repolarization milial neonatal convulsions shows spontaneous gen- of individual action potentials but exert a dampening eralized tonic-clonic seizures neuronal plasticity, but, effect on repetitive discharges and excitability of neu- like in human BFNC, without neuronal loss and hip- rons [21]. Retigabine binds to activation gate region pocampal mossy fiber sprouting [115]. The Szt-1 interacting with a specific amino acid fragment and mice with a spontaneous mutation involving KCNQ2 stabilizing the neuronal KCNQ (Kv7) channels in an (Kv7.2) have a reduced baseline seizure threshold and open conformation. Specifically, it has been proposed lower sensitivity to an M-current enhancer [88]. On that retigabine binds to a hydrophobic pocket formed the other hand, in two animal models of temporal lobe upon channel opening between the cytoplasmic parts epilepsy (amygdalar kindling and electrical stimula- of S5 and S6 segments of Kv7.2 involving Trp236 tion of the hippocampus) the KCNQ2 immunoreactiv- and the channel’s gate, which explains the large hy- ity was increased in the basolateral amygdala. It was perpolarizing shift in voltage-dependent activation concluded that the KCNQ2 subunit upregulation [136]. Such characteristic amino acid pocket to which could be an adaptive response to counteract the limbic retigabine binds in neuronal cells is not present in the seizures [94]. The Kv7.2/Kv7.3 channels are targets cardiac muscle cells. Thus, retigabine activates for pharmacotherapy of epilepsy, stroke, migraine, de- KCNQ2/KCNQ3 potassium channel shifting their mentia, bipolar depression, anxiety and neuropathic voltage-dependence to more negative values [132]. pain. The non-inactivating K+ M-current is known to By opening the Kv7 channels, retigabine prevents so- be slowly activated during depolarization to- dium depolarizing currents and occurrence of epilep- wards the threshold for action potential hyperpolariz- tic discharges. Retigabine action seems to be not re- ing the cell and decreasing neuronal excitability. Nu- stricted to hyperpolarization of the resting potential of merous second messengers converging pathways and neuronal membranes and enhancement of neuronal intracellular calcium control the M-current, and the resistance against generation of epileptiform activity. activation of various Gq/G11-coupled receptors pre- It has been reported that through the activation of pre- dominantly inhibits this current [21]. The M-current synaptic M-currents, retigabine can also inhibit the appears to play a key role in controlling neuronal ex- depolarization-evoked excitatory amino acid release citability because it is the only sustained current from cerebrocortical synaptosomes [65, 132]. The which counteracts depolarization near the threshold of broad antiepileptic activity of retigabine was proved action potential [20, 73]. Therefore, the M-current is in several experimental models of chemical, electrical thought to act as an efficient brake for repetitive ac- and genetic models of seizures and was confirmed in tion potential firing. M-current inhibition by muscar- clinical trials [13]. One can expect that retigabine can inic receptor agonists enhances neuronal excitability, be a prototype drug for a new generation of antiepi- as shown in the cells derived from both peripheral and leptic drugs. Indeed, some benzanilide- and adamant- central nervous system. This is in line with procon- ane-derived compounds with agonistic activity at vulsant effects of cholinergic receptor agonists, e.g., KCNQ2/KCNQ3 channels and possessing antiepilep- systemic administration of pilocarpine in high doses tic properties have recently been presented [39, 104, produces recurrent limbic seizures in rodents widely 132]. Several other newly identified compounds dis- accepted as a model of temporal lobe epilepsy [121]. played isoform-specific prolongation of Kv7 potas- The blockade of M-current in the hippocampus- sium channel opening and induced significant reduc- entorhinal cortex slices of neonatal and adult rats pro- tion in activation that differed from that of retigabine duced epileptiform activity with a developmental pat- [40]. KCNQ2 and KCNQ3 genes encode Kv7.2 and tern resembling BNFC [93]. This is in agreement with Kv7.3 potassium channel subunits, respectively. the study that showed that M-current inhibition in the Kv7.2 and Kv7.3 form heteromeric voltage-gated po- Mg2+-free seizure model in hippocampal slices of im- tassium channels, which are responsible for the gen- mature rats readily transited the interictal bursting to eration of M-current which regulates repeated neu- sustained depolarization [98]. The significance of the ronal discharges [128]. Mutations of KCNQ2 and M-current in seizure prevention was supported by in- KCNQ3 genes that reduce the current-M amplitude troduction of a new antiepileptic drug to the clinic, cause the rare hereditary benign familial neonatal sei- that is retigabine which acts mainly, if not exclusively, zures [26]. It has been found that the mouse model of through a positive modulation of Kv7.2–7.5 channels. human KCNQ2 and KCNQ3 mutations for benign fa- Muscarinic receptor stimulation with linopiridine in-

Pharmacological Reports, 2013, 65, 787–801 795 creased neuronal firing in slices from human epilepto- LQTS type 2 and for many years this has been a major genic cortex, whereas retigabine had opposite effects focus of clinical and research interest. However, only [43]. It is of note that the activation of K+ M current recently a clear link was identified between KCNH2 can play also an important role in the mechanism of mutations and epilepsy in humans [51, 87]. It was ob- action of neuropeptides with anticonvulsant activity, served that LQT2-causing perturbations in the such as dynorphin and somatostatin [68, 75, 112]. In KCNH2-encoded potassium channel might result in low concentrations, the endogenous k opioid receptor susceptibility to recurrent seizure activity. Addition- agonists dynorphin A and dynorphin B augmented ally, ERG potassium channels blockade in astrocytes M-current in CA3 neurons of the rat hippocampus, changed potassium concentrations extraneuronally, but in high concentration, dynorphin A suppressed the and such changes were shown to be epileptogenic M-current. Dynorphin was found to selectively aug- [49]. This has clinical implications as post-traumatic ment the M-current in the hippocampal CA1 neurons changes in hippocampal glia mimic the effects of by an opiate receptor mechanism [68]. Qiu et al. [99] acute blockade by external antagonists, including loss reported that somatostatin receptor subtype 4 cou- of K conductance with subsequent failure of glial po- pling to M-channels was critical to the inhibition of tassium homeostasis, which in turn promotes abnor- epileptiform activity. They found that the somatosta- mal neuronal excitation [31]. Further research should tin receptor 4 knockout mice showed shorter latencies focus on this interesting mechanism as it might prove and increased seizure severity when compared with to be an important component of the complex patho- wild-type mice in both kainate and pentetrazole sei- physiology of drug refractoriness in epilepsy. In addi- zuremodels[9]. tion, ion channels co-expressed in the brain and the Pharmacological and molecular data indicate that, heart, such as potassium channels are logical candi- besides KCNQ2/KCNQ3, also other potassium chan- dates as risk factors for sudden unexplained death in nels, e.g., KATP, Kv1.1 and GIRK2 regulate neuronal epilepsy (SUDEP) because defects in intrinsic mem- excitability and can be involved in pathomechanism brane excitability could underlie both epilepsy and of seizures [132]. Specifically, the Kv1channels are cardiacarrhythmiasthatcausedeath[84]. involved in regulating the action potential firing pat- Hyperpolarization-activated cyclic nucleotide gated terns and neuronal excitability [103]. Mutations (HCN) channels structurally resemble voltage-gated Kcna1 and Kcna2 are associated with epilepsy and potassium channels. Altered expression levels of the ataxia in humans, whereas mice devoid of Kv1.1 or HCN1 subunit have been shown in the hippocampus Kv1.2 genes showed high susceptibility to seizures of patients with temporal lobe epilepsy as well as in [17, 117]. Developmental seizure susceptibility of animal models of this disorder [101]. On the other Kv1.1 potassium channel knockout mice has been de- hand, the HCN2 subunit is abundant in the thalamus, scribed by Rho et al. [100] as a clinically relevant and silencing of the gene coding for this protein leads model of early-onset epilepsy. The a-dendrotoxin- to non-convulsive seizures in mice. These observa- -induced inhibition of these channels results in sei- tions suggest that HCN could play a role in both lim- zures, whereas compounds that prevent Kv1.1 inacti- bic and absence epilepsy and can be a potential target vation decreased pentetrazole- and maximal electric for antiepileptic therapy [96, 114]. Lamotrigine and shock-evokedseizuresinmice[7,64]. gabapentin were shown to activate the hippocampal Another subtype of the potassium channel has an HCN channels, but it is premature to draw any con- additional link to CNS diseases. This is KCNH2 clusion, to what extent this effect might contribute to (HERG1; human ether-à-go-go-related gene) which antiepilepticpropertiesofthesedrugs[97]. was initially discovered in the hippocampus [129], and it was later demonstrated that this gene was ex- pressed in many regions of the central nervous sys- tem. Further studies reported that hippocampal ex- Concludingremarks pression of the ERG family of potassium channels was distributed preferentially in hippocampal astro- cytes that may regulate neuronal excitability [35]. There is general consensus that seizures result from Later, the mutations in that gene, causing the loss of excessively enhanced excitatory processes in a given its function, were found to be responsible for the population of neuronal cells in the brain or from defi-

796 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

cient neuronal inhibition. Traditionally, the above 2. AlexanderGM,GodwinDW:Metabotropicglutamate mechanism was thought to comprise hyperactivity of receptorsasastrategictargetforthetreatmentofepi- lepsy.EpilepsyRes,2006,71,1–22. glutamatergic transmission and insufficient GABAA 3. ArmijoJA,ValdizánEM,DeLasCuevasI,CuadradoA: receptor-mediated neurotransmission. New data add Advancesinthephysiopathologyofepileptogenesis: to the complexity of GABA-glutamate interaction molecularaspects.RevNeurol,2002,34,409–429. 4. showing both excitatory and inhibitory role of GABA AvoliM,D’AntuonoM,LouvelJ,KöhlingR,BiaginiG, PumainR,D’ArcangeloG,TancrediV:Networkand and glutamatergic neurons in the central nervous sys- pharmacologicalmechanismsleadingtoepileptiform tem. Moreover, besides synaptic NMDA and GABAA synchronizationinthelimbicsysteminvitro.ProgNeu- receptors, also extrasynaptic receptors for the amino robiol,2002,68,167–207. 5. acid transmitters have been recently implicated in the AvoliM,deCurtisM:GABAergicsynchronizationin thelimbicsystemanditsroleinthegenerationofepilep- pathomechanism of seizures. Changes in gene expres- tiformactivity.ProgNeurobiol,2011,95,104–132. sion, polymorphisms, lost- or gain-function mutation 6. AyalaGF,DichterM,GumnitRJ,MatsumotoH,Spencer as well as cellular energetic imbalance can contribute WA:Genesisofepilepticinterictalspikes.Newknowl- to disturbed function in the ligand- and voltage- edgeofcorticalfeedbacksystemssuggestsaneuro- physiologicalexplanationofbriefparoxysms.BrainRes, dependent sodium, potassium, chloride and calcium 1973,52,1–17. channels,resultinginseizureactivity. 7. BagettaG,NisticóG,DollyJO:Productionofseizures The principal targets for antiepileptic drugs com- andbraindamageinratsby a-dendrotoxin,aselective + prise voltage-dependent sodium, potassium and cal- K channelblocker.NeurosciLett,1992,139,34–40. 8. BalestrinoM,YoungJ,AitkenP:Blockof(Na+,K+)- cium channels, GABAA receptors, GABA-meta- ATPasewithouabaininducesspreadingdepression-like bolizing and GABA transporters. Excitatory depolarizationinhippocampalslices.BrainRes,1999, amino acid receptors and some synaptic proteins can 838,37–44. also be involved in the mechanism of antiepileptic 9. BecerraJL,OjedaJ,CorrederaE,RuizGiménezJ: drug action [11, 57, 77]. Looking for future directions Reviewoftherapeuticoptionsforadjuvanttreatmentof of therapy of epilepsy, one may stress significance of focalseizuresinepilepsy:focusonlacosamide.CNS Drugs,2011,25,Suppl1,3–16. molecularly characterized new drug targets, drugs act- 10. Ben-AriY:Seizuresbegetseizures:thequestforGABA ing in direct vicinity of seizure focus (focal method of asakeyplayer.CritRevNeurobiol,2006,18,135–144. drug delivery), and gene and cell therapy. A signifi- 11. BialerM:Chemicalpropertiesofantiepilepticdrugs cant progress has been made in elucidating the role of (AEDs).AdvDrugDelivRev,2012,64,887–895. 12. amino acid receptor subunits, various kinds of potas- BiervertC,SchroederBC,KubischC,BerkovicSF, ProppingP,JentschTJ,SteinleinOK:A potassiumchan- sium ion channels, HCN channels, acid-sensing ion nelmutationinneonatalhumanepilepsy.Science,1998, channels, and gap junctions in the regulation of neu- 279,403–406. ronal excitability. There is a great hope that molecular 13. Blackburn-MunroG,Dalby-BrownW,MirzaNR,Mik- genetics and proteomics strategy will help in creating kelsenJD,Blackburn-MunroRE:Retigabine:chemical new, effective anticonvulsants. However, it seems that synthesistoclinicalapplication.CNSDrugRev,2005, 11,1–20. the real breakthrough in our understanding of the 14. BlossEB,HunterRG:Hippocampalkainatereceptors. mechanism of epilepsy and antiepileptic drug action VitamHorm,2010,82,167–184. will depend on integration of molecular and func- 15. BoisonD:Adenosinedysfunctioninepilepsy.Glia, tional studies and on proper transformation and pres- 2012,60,1234–1243. 16. entation of data, which should facilitate the substan- BorowiczKK,£uszczkiJJ,CzuczwarSJ:2-Methyl-6- phenylethynyl-pyridine(MPEP),anon-competitive tive discussion between specialists of various fields of mGluR5antagonist,differentiallyaffectstheanticonvul- experimentalandclinicalepileptology. santactivityoffourconventionalantiepilepticdrugs againstamygdala-kindledseizuresinrats.Pharmacol Rep,2009,61,621–630. 17. BrewHM,GittelmanJX,SilversteinRS,HanksTD, DemasVP,RobinsonLC,RobbinsCA etal.:Seizures andreducedlifespaninmicelackingthepotassium References: channelsubunitKv1.2,buthypoexcitabilityandenlarged Kv1currentsinauditoryneurons.JNeurophysiol,2007, 98,1501–1525. 1. AdamsPJ,SnutchTP:Calciumchannelopathies: 18. BrickleySG,ModyI:ExtrasynapticGABAA receptors: voltage-gatedcalciumchannels.SubcellBiochem,2007, theirfunctionintheCNSandimplicationsfordisease. 45,215–251. Neuron,2012,73,23–34.

Pharmacological Reports, 2013, 65, 787–801 797 19. Brooks-KayalAR,ShumateMD,JinH,RikhterTY, 36. EtxeberriaA,Santana-CastroI,RegaladoMP,AivarP, CoulterDA:SelectivechangesinsinglecellGABAA re- VillarroelA:ThreemechanismsunderlieKCNQ2/3het- ceptorsubunitexpressionandfunctionintemporallobe eromericpotassiumM-channelpotentiation.JNeurosci, epilepsy.NatMed,1998,4,1166–1172. 2004,24,9146–9152. 20. BrownDA,AdamsPR:Muscarinicsuppressionof 37. FattoreC,PeruccaE:Novelmedicationsforepilepsy. anovelvoltage-sensitiveK+ currentinavertebrateneu- Drugs,2011,71,2151–2178. ron.Nature,1980,283,673–676. 38. FitzgeraldPJ:TheNMDA receptormayparticipatein 21. BrownDA,PassmoreGM:NeuralKCNQ(Kv7)chan- widespreadsuppressionofcircuitlevelneuralactivity,in nels.BrJPharmacol,2009,156,1185–1195. additiontoasimilarlyprominentroleincircuitlevelac- 22. BudziszewskaB,VanLuijtelaarG,CoenenAM, tivation.BehavBrainRes,2012,230,291–298. LeœkiewiczM,LasoñW:Effectsofneurosteroidson 39. Fritch PC, McNaughton-Smith G, Amato GS, Burns JF, spike-wavedischargesinthegeneticepilepticWAG/Rij Eargle CW, Roeloffs R, Harrison W et al.: Novel rat.EpilepsyRes,1999,33,23–29. KCNQ2/Q3 agonists as potential therapeutics for epilepsy 23. CarmignotoG,HaydonPG:Astrocytecalciumsignaling and neuropathic pain. J Med Chem, 2010, 53, 887–896. andepilepsy.Glia,2012,60,1227–1233. 40. GaoZ,ZhangT,WuM,XiongQ,SunH,ZhangY,ZuL 24. CatterallWA:Fromioniccurrentstomolecularmecha- etal.:Isoform-specificprolongationofKv7(KCNQ)po- nisms:thestructureandfunctionofvoltage-gatedso- tassiumchannelopeningmediatedbynewmolecularde- diumchannels.Neuron,2000,26,13–25. terminantsfordrug-channelinteractions.JBiolChem, 25. ConnPJ,PinJP:Pharmacologyandfunctionsofmeta- 2010,285,28322–2832. botropicglutamatereceptors.AnnuRevPharmacolToxi- 41. GasiorM,KaminskiR,WitkinJM:Pharmacological col,1997,37,205–237. modulationofGABAB receptorsaffectscocaine-induced 26. CooperEC:Potassiumchannels:howgeneticstudiesof seizuresinmice.Psychopharmacology(Berl),2004,174, epilepticsyndromesopenpathstonewtherapeutictar- 211–219. getsanddrugs.Epilepsia,2001,42,Suppl5,49–54. 42. GhasemiM,SchachterSC:TheNMDA receptorcom- 27. CopeDW,DiGiovanniG,FysonSJ,OrbánG,Errington plexasatherapeutictargetinepilepsy:areview.Epi- AC,LorinczML,GouldTMetal.:Enhancedtonic lepsyBehav,2011,22,617–640. GABAA inhibitionintypicalabsenceepilepsy.NatMed, 43. GigoutS,WierschkeS,LehmannTN,HornP,Dehnicke 2009,15,1392–1398. C,DeiszRA:Muscarinicacetylcholinereceptor- 28. CosfordND,MeinkePT,StaudermanKA,HessSD:Re- mediatedeffectsinslicesfromhumanepileptogeniccor- centadvancesinthemodulationofvoltage-gatedion tex.Neuroscience,2012,223,399–411. channelsforthetreatmentofepilepsy.CurrDrugTargets 44. GorjiA,StemmerN,RambeckB,JürgensU,MayT, CNSNeurolDisord,2002,1,81–104. PannekHW,BehneFetal.:Neocorticalmicroenviron- 29. CrunelliV,LerescheN,CopeDW:GABA-A Receptor mentinpatientswithintractableepilepsy:potassiumand FunctioninTypicalAbsenceSeizures.In:Jasper’sBasic chlorideconcentrations.Epilepsia,2006,47,297–310. MechanismsoftheEpilepsies[Internet].4th edn.Eds. 45. HablitzJJ,HeinemannU:Alterationsinthemicroenvi- NoebelsJL,AvoliM,RogawskiMA,OlsenRW, ronmentduringspreadingdepressionassociatedwith Delgado-EscuetaAV,NationalCenterforBiotechnology epileptiformactivityintheimmatureneocortex.Brain Information,Bethesda,MD,2012. ResDevBrainRes,1989,46,243–252. 30. CzuczwarSJ,PatsalosPN:Thenewgenerationof 46. HochmanDW,D’AmbrosioR,JanigroD,Schwartzkroin GABA enhancers.Potentialinthetreatmentofepilepsy. PA:Extracellularchlorideandthemaintenanceofspon- CNSDrugs,2001,15,339–350. taneousepileptiformactivityinrathippocampalslices. 31. D’AmbrosioR,MarisDO,GradyMS,WinnHR,Janigro JNeurophysiol,1999,81,49–59. D.ImpairedK+ homeostasisandalteredelectrophysio- 47. HuberfeldG,WittnerL,ClemenceauS,BaulacM,Kaila logicalpropertiesofpost-traumatichippocampalglia. K,MilesR,RiveraC:Perturbedchloridehomeostasis JNeurosci,1999,19,8152–8162. andGABAergicsignalinginhumantemporallobeepi- 32. DepaulisA,DeransartC,VergnesM,MarescauxC:GA- lepsy.JNeurosci,2007,27,9866–9873. BAergicmechanismsingeneralizedepilepsies:theneu- 48. IlieA,RaimondoJV,AkermanCJ:Adenosinerelease roanatomicaldimension.RevNeurol(Paris),1997,153, duringseizuresattenuatesGABAA receptor-mediatedde- Suppl1,S8–13. polarization.JNeurosci,2012,32,5321–5332. 33. DunwiddieTV:Adenosineandsuppressionofseizures. 49. JanigroD,GaspariniS,D’AmbrosioR,McKhannG, AdvNeurol,1999,79,1001–1010. 2nd,DiFrancescoD:ReductionofK+ uptakeingliapre- 34. DzhalaVI,TalosDM,SdrullaDA,BrumbackAC, ventslong-termdepressionmaintenanceandcausesepi- MathewsGC,BenkeTA,DelpireEetal.:NKCC1trans- leptiformactivity.JNeurosci,1997,17,2813–2824. porterfacilitatesseizuresinthedevelopingbrain.Nat 50. JentschTJ:NeuronalKCNQpotassiumchannels:physi- Med,2005,11,1205–1213. ologyandroleindisease.NatRevNeurosci,2000,1, 35. EmmiA,WenzelHJ,SchwartzkroinPA,TaglialatelaM, 21–30. CastaldoP,BianchiL,NerbonneJetal.:Dogliahave 51. JohnsonJN,HofmanN,HaglundCM,CascinoGD, heart?Expressionandfunctionalroleforether-a-go-go WildeAA,AckermanMJ:Identificationofapossible currentsinhippocampalastrocytes.JNeurosci,2000,20, pathogeniclinkbetweencongenitallongQT syndrome 3915–3925. andepilepsy.Neurology,2009,72,224–231.

798 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

52. K³odziñskaA,BijakM,Chojnacka-WójcikE,Kroczka 69. MadsenKK,WhiteHS,SchousboeA:Neuronaland B,SwiaderM,CzuczwarSJ,PilcA:RolesofgroupII non-neuronalGABA transportersastargetsforantiepi- metabotropicglutamatereceptorsinmodulationofsei- lepticdrugs.PharmacolTher,2010,125,394–401. zureactivity.NaunynSchmiedebergsArchPharmacol, 70. MaljevicS,WuttkeTV,SeebohmG,LercheH:Kv7 2000,361,283–288. channelopathies.PflugersArch,2010,460,277–288. 53. KohlMM,PaulsenO:TherolesofGABA Breceptorsin 71. MaresP,KubováH:Whatistheroleofneurotransmitter corticalnetworkactivity.AdvPharmacol,2010,58, systemsincorticalseizures?PhysiolRes,2008,57, 205–229. Suppl3,S111–20. 54. KöhlingR:Voltage-gatedsodiumchannelsinepilepsy. 72. MaresP,MikuleckáA,TicháK,Lojková-JaneckováD, Epilepsia2002,43,1278–1295. KubováH:Metabotropicglutamatereceptorsasatarget 55. Ku³ak W, Sobaniec W, Wojtal K, Czuczwar SJ: Calcium foranticonvulsantandanxiolyticactioninimmaturerats. modulation in epilepsy. Pol J Pharmacol, 2004, 56, 29–41. Epilepsia,2010,51,Suppl3,24–26. 56. LaschetJJ,KurcewiczI,MinierF,TrottierS,Khallou- 73. MarrionNV:ControlofM-current.AnnuRevPhysiol, LaschetJ,LouvelJ,GigoutSetal.:Dysfunctionof 1997,59,483–504. GABAA receptorglycolysis-dependentmodulationin 74. MartinDJ,McClellandD,HerdMB,SuttonKG,Hall humanpartialepilepsy.ProcNatlAcadSciUSA,2007, MD,LeeK,PinnockRD,ScottRH:Gabapentin- 104,3472–3477. mediatedinhibitionofvoltage-activatedCa2+ channel 57. LasoñW,Dudra-JastrzêbskaM,RejdakK,CzuczwarSJ: currentsinculturedsensoryneuronesisdependenton Basicmechanismsofantiepilepticdrugsandtheirphar- cultureconditionsandchannelsubunitexpression.Neu- macokinetic/pharmacodynamicinteractions:anupdate. ropharmacology,2002,42,353–366. PharmacolRep,2011,63,271–292. 75. MazaratiA,WasterlainCG:Anticonvulsanteffectsof 58. LeeTS,ManeS,EidT,ZhaoH,LinA,GuanZ,KimJH fourneuropeptidesintherathippocampusduringself- etal.:Geneexpressionintemporallobeepilepsyiscon- sustainingstatusepilepticus.NeurosciLett,2002, sistentwithincreasedreleaseofglutamatebyastrocytes. 331,123–127. MolMed,2007,13,1–13. 76. McCormickDA,ContrerasD:Onthecellularandnet- 59. LiST,JuJG:Functionalrolesofsynapticandextra- workbasesofepilepticseizures.AnnuRevPhysiol, synapticNMDA receptorsinphysiologicalandpatho- 2001,63,815–846. logicalneuronalactivities.CurrDrugTargets,2012,13, 77. MeldrumBS,RogawskiMA:Moleculartargetsforan- 207–221. tiepilepticdrugdevelopment.Neurotherapeutics,2007, 60. LiT,QuanLanJ,FredholmBB,SimonRP,BoisonD: 4,18–61. Adenosinedysfunctioninastrogliosis:causeforseizure 78. MiceliF,SoldovieriMV,MartireM,TaglialatelaM: generation?NeuronGliaBiol,2007,3,353–366. Molecularpharmacologyandtherapeuticpotentialof 61. LöscherW:Pharmacologyofglutamatereceptorantago- neuronalKv7-modulatingdrugs.CurrOpinPharmacol, nistsinthekindlingmodelofepilepsy.ProgNeurobiol, 2008,8,65–74. 1998,54,721–741. 79. MiceliF,VargasE,BezanillaF,TaglialatelaM:Gating 62. LosiG,CammarotaM,CarmignotoG:Theroleofastro- currentsfromKv7channelscarryingneuronalhyperex- gliaintheepilepticbrain.FrontPharmacol,2012,3,132. citabilitymutationsinthevoltage-sensingdomain.Bio- 63. LoupF,WieserHG,YonekawaY,AguzziA,Fritschy physJ,2012,102,1372–1382. JM:SelectivealterationsinGABAA receptorsubtypesin 80. ModyI,LambertJD,HeinemannU:Lowextracellular humantemporallobeepilepsy.JNeurosci,2000,20, magnesiuminducesepileptiformactivityandspreading 5401–5419. depressioninrathippocampalslices.JNeurophysiol, 64. LuQ,PeeveyJ,JowF,MonaghanMM,MendozaG, 1987,57,869–888. ZhangH,WuJetal.:DisruptionofKv1.1N-typeinacti- 81. 81.MoldrichRX,ChapmanAG,DeSarroG,Meldrum vationbynovelsmallmoleculeinhibitors(disinactiva- BS:Glutamatemetabotropicreceptorsastargetsfordrug tors).BioorgMedChem,2008,16,3067–3075. therapyinepilepsy.EurJPharmacol,2003,476,3–16. 65. LuisiR,PanzaE,BarreseV,IannottiFA,ViggianoD, 82. MorlandC,NordengenK,GundersenV:Valproate SecondoA,CanzonieroLMetal.:Activationofpre- causesreductionoftheexcitatoryaminoacidaspartatein synapticM-typeK+ channelsinhibits[3H]D-aspartatere- nerveterminals.NeurosciLett,2012,527,100–104. leasebyreducingCa2+ entrythroughP/Q-typevoltage- 83. MulaM:Newantiepilepticdrugs:moleculartargets. gatedCa2+ channels.JNeurochem,2009,109,168–181. CentNervSystAgentsMedChem,2009,2,79–86. 66. LukasiukK,WilczynskiGM,KaczmarekL:Extracellu- 84. NashefL,HindochaN,MakoffA:Riskfactorsinsudden larproteasesinepilepsy.EpilepsyRes,2011,96, deathinepilepsy(SUDEP):thequestformechanisms. 191–206. Epilepsia,2007,48,859–871. 67. £uszczkiJJ:Third-generationantiepilepticdrugs: 85. NgombaRT,SantoliniI,SaltTE,FerragutiF,Battaglia mechanismsofaction,pharmacokineticsandinterac- G,NicolettiF,vanLuijtelaarG:Metabotropicglutamate tions.PharmacolRep,2009,61,197–216. receptorsinthethalamocorticalnetwork:strategictargets 68. MadambaSG,SchweitzerP,SigginsGR:Dynorphinse- forthetreatmentofabsenceepilepsy.Epilepsia,2011, lectivelyaugmentstheM-currentinhippocampalCA1 52,1211–1222. neuronsbyanopiatereceptormechanism.JNeuro- 86. NicolsonA,LeachJP:Futureprospectsforthedrug physiol,1999,82,1768–1775. treatmentofepilepsy.CNSDrugs,2001,15,955–968.

Pharmacological Reports, 2013, 65, 787–801 799 87. OmichiC,MomoseY,KitaharaS.CongenitallongQT 104. RoeloffsR,WickendenAD,CreanC,WernessS, syndromepresentingwithahistoryofepilepsy:misdiag- McNaughton-SmithG,StablesJ,McNamaraJOetal.: nosisorrelationshipbetweenchannelopathiesofthe InvivoprofileofICA-27243[N-(6-chloro-pyridin-3- heartandbrain?Epilepsia,2010,51,289–292. yl)-3,4-difluoro-benzamide],apotentandselective 88. OttoJF,YangY,FrankelWN,WhiteHS,WilcoxKS: KCNQ2/Q3 (Kv7.2/Kv7.3) activator in rodent anticonvul- A spontaneous mutation involving Kcnq2 (Kv7.2) reduces sant models. J Pharmacol Exp Ther, 2008, 326, 818–828. M-current density and spike frequency adaptation in 105. RogawskiMA:RevisitingAMPA receptorsasanantiepi- mouse CA1 neurons. J Neurosci, 2006, 26, 2053–2059. lepticdrugtarget.EpilepsyCurr,2011,11,56–63. 89. PaolettiP:MolecularbasisofNMDA receptorfunctional 106. RogawskiMA,BazilCW:Newmoleculartargetsfor diversity.EurJNeurosci,2011,33,1351– 1365. antiepilepticdrugs: a2d,SV2A,andKv7/KCNQ/Mpo- 90. PatinoGA,IsomLL:Electrophysiologyandbeyond: tassiumchannels.CurrNeurolNeurosciRep,2008,8, multiplerolesofNa+ channel b subunitsindevelopment 345–352. anddisease.NeurosciLett,2010,486,53–59. 107. RogawskiMA,DonevanSD:AMPA receptorsinepi- 91. PavlovI,HuuskoN,DrexelM,KirchmairE,SperkG, lepsyandastargetsforantiepilepticdrugs.AdvNeurol, PitkänenA,WalkerMC:Progressivelossofphasic,but 1999,79,947–963. nottonic,GABAA receptor-mediatedinhibitioninden- 108. RogawskiMA,LöscherW:Theneurobiologyofantiepi- tategranulecellsinamodelofpost-traumaticepilepsyin lepticdrugs.NatRevNeurosci,2004,5,553–564. rats.Neuroscience,2011,194,208–219. 109. Savio-GalimbertiE,GollobMH,DarbarD:Voltage- 92. PavlovI,WalkerMC:TonicGABAA receptor-mediated gatedsodiumchannels:biophysics,pharmacology,and signallingintemporallobeepilepsy.Neuropharmacol- relatedchannelopathies.FrontPharmacol,2012,3,124. ogy,2013,69,55–61. 110. SchousboeA,MadsenKK,WhiteHS:GABA transport 93. PeñaF,Alavez-PérezN:Epileptiformactivityinduced inhibitorsandseizureprotection:thepastandfuture.Fu- bypharmacologicreductionofM-currentinthedevelop- tureMedChem,2011,3,183–187. inghippocampusinvitro.Epilepsia,2006,47,47–54. 111. SchroederBC,HechenbergerM,WeinreichF,Kubisch 94. PenschuckS,BastlundJF,JensenHS,StensbolTB, C,JentschTJ:KCNQ5,anovelpotassiumchannel EgebjergJ,WatsonWP:ChangesinKCNQ2immunore- broadlyexpressedinbrain,mediatesM-typecurrents. activityintheamygdalaintworatmodelsoftemporal JBiolChem,2000,275,24089–24095. lobeepilepsy.BrainResMolBrainRes,2005,141, 112. SchweitzerP,MadambaSG,SigginsGR.Somatostatin 66–73. increasesavoltage-insensitiveK+ conductanceinrat 95. PetraliaRS:DistributionofextrasynapticNMDA recep- CA1hippocampalneurons.JNeurophysiol,1998,79, torsonneurons.ScientificWorldJ,2012,267120. 1230–1238. 96. PoolosNP:Hyperpolarization-ActivatedCyclic 113. SenA,MartinianL,NikolicM,WalkerMC,ThomM, Nucleotide-Gated(HCN)IonChannelopathyinEpilepsy. SisodiyaSM:IncreasedNKCC1expressioninrefractory In:Jasper’sBasicMechanismsoftheEpilepsies[Inter- humanepilepsy.EpilepsyRes,2007,74,220–227. th net].4 edn.Eds.NoebelsJL,AvoliM,RogawskiMA, 114. ShahMM,HuangZ,MartinelloK:HCNandKV7(M-) OlsenRW,Delgado-EscuetaAV,NationalCenterfor channelsastargetsforepilepsytreatment.Neuropharma- BiotechnologyInformation(US),Bethesda,MD,2012. cology,2013,69,75-81. 97. PosteaO,BielM:ExploringHCNchannelsasnovel 115. SinghNA,OttoJF,DahleEJ,PappasC,LeslieJD, drugtargets.NatRevDrugDiscov,2011,10,903–914. VilaythongA,NoebelsJL etal.:Mousemodelsofhu- 98. QiuC,JohnsonBN,TallentMK:K+ M-currentregulates manKCNQ2andKCNQ3mutationsforbenignfamilial thetransitiontoseizuresinimmatureandadulthippo- neonatalconvulsionsshowseizuresandneuronalplastic- campus.Epilepsia,2007,48,2047–2058. itywithoutsynapticreorganization.JPhysiol,2008,586, 99. QiuC,ZeydaT,JohnsonB,HochgeschwenderU, 3405–3423. deLeceaL,TallentMK:Somatostatinreceptorsubtype4 116. SinghR,GardnerRJ,CrosslandKM,ShefferIE, couplestotheM-currenttoregulateseizures.JNeurosci, BerkovicSF:Chromosomalabnormalitiesandepilepsy: 2008,28,3567–3576. areviewforcliniciansandgenehunters.Epilepsia,2002, 100. RhoJM,SzotP,TempelBL,SchwartzkroinPA:Devel- 43,127–140. opmentalseizuresusceptibilityofkv1.1potassiumchan- 117. SmartSL,LopantsevV,ZhangCL,RobbinsCA, nelknockoutmice.DevNeurosci,1999,21,320–327. WangH,ChiuSY,SchwartzkroinPA etal.:Deletionof 101. RichichiC,BrewsterAL,BenderRA,SimeoneTA, theKV1.1potassiumchannelcausesepilepsyinmice. ZhaQ,YinHZ,WeissJH,BaramTZ:Mechanismsof Neuron,1998,20,809–819. seizure-induced‘transcriptionalchannelopathy’ of 118. Somjen GG, Müller M: Potassium-induced enhancement of hyperpolarization-activatedcyclicnucleotidegated persistent inward current in hippocampal neurons in isola- (HCN)channels.NeurobiolDis,2008,29,297–305. tion and in tissue slices. Brain Res, 2000, 885, 102–110. 102. RiveraC,VoipioJ,PayneJA,RuusuvuoriE,LahtinenH, 119. StephenLJ,BrodieMJ:Pharmacotherapyofepilepsy: LamsaK,PirvolaU,SaarmaM,KailaK:TheK+/Cl– newlyapprovedanddevelopmentalagents.CNSDrugs, co-transporterKCC2rendersGABA hyperpolarizing 2011,25,89–107. duringneuronalmaturation.Nature,1999,397,251–255. 120. TreimanDM:GABAergicmechanismsinepilepsy.Epi- 103. RobbinsCA,TempelBL:Kv1.1andKv1.2:similar lepsia,2001,42,Suppl3,8–12. channels,differentseizuremodels.Epilepsia,2012,53, 121. TurskiL,IkonomidouC,TurskiWA,BortolottoZA, Suppl1,134–141. CavalheiroEA:Review:cholinergicmechanismsand

800 Pharmacological Reports, 2013, 65, 787–801 Basicmechanismsofseizures W³adys³aw Lasoñ et al.

epileptogenesis.Theseizuresinducedbypilocarpine: ofKCNQ2/Q3potassiumchannels.MolPharmacol, anovelexperimentalmodelofintractableepilepsy. 2000,58,591–600. ,1989,3,154–171. 133. WilczynskiGM,KonopackiFA,WilczekE,LasieckaZ, 122. UedaY,DoiT,TokumaruJ,YokoyamaH,NakajimaA, GorlewiczA,MichalukP,WawrzyniakMetal.:Impor- MitsuyamaY,Ohya-NishiguchiHetal.:Collapseofex- tantroleofmatrixmetalloproteinase9inepileptogene- tracellularglutamateregulationduringepileptogenesis: sis.JCellBiol,2008,180,1021–1035. down-regulationandfunctionalfailureofglutamate 134. WlaŸP:Anti-convulsantandadverseeffectsoftheglyci- transporterfunctioninratswithchronicseizuresinduced neBreceptorligands,D-cycloserineandL-701,324: bykainicacid.JNeurochem,2001,76,892–900. comparisonwithcompetitiveandnon-competitive 123. UrbanskaEM,CzuczwarSJ,KleinrokZ,TurskiWA: N-methyl-D-aspartatereceptorantagonists.BrainRes Excitatoryaminoacidsinepilepsy.RestorNeurolNeuro- Bull,1998,46,535–540. sci,1998,13,25–39. 135. WlaŸP,PoleszakE:Differentialeffectsofglycineon 124. vanLuijtelaarG,WiadernaD,ElantsC,ScheenenW: theanticonvulsantactivityofD-cycloserineand OppositeeffectsofT-andL-typeCa2+ channelsblockers L-701,324inmice.PharmacolRep,2011,63, ingeneralizedabsenceepilepsy.EurJPharmacol,2000, 1231–1234. 406,381–389. 136. WuttkeTV,SeebohmG,BailS,MaljevicS,LercheH: 125. VezzaniA,BartfaiT,BianchiM,RossettiC,FrenchJ: Thenewanticonvulsantretigabinefavorsvoltage- Therapeuticpotentialofnewantiinflammatorydrugs. dependentopeningoftheKv7.2(KCNQ2)channelby Epilepsia,2011,52,67–69. bindingtoitsactivationgate.MolPharmacol,2005,67, 126. VincentP,MulleC:Kainatereceptorsinepilepsyandex- 1009–1017. citotoxicity.Neuroscience,2009,158,309–323. 137. XiongZ-G.,PignataroG.,LiM.,ChangSY.,SimonRP: 127. WalkerMC,SemyanovA:Regulationofexcitabilityby Acid-sensingionchannels(ASICs)aspharmacological extrasynapticGABAA receptors.ResultsProblCellDif- targetsforneurodegenerativediseases.CurrOpinPhar- fer,2008,44,29–48. macol,2008,8,25–32. 128. WangHS,PanZ,ShiW,BrownBS,WymoreRS, 138. YinP,YangL,ZhouHY,SunRP:Matrixmetalloprotei- CohenIS,DixonJE,McKinnonD:KCNQ2andKCNQ3 nase-9maybeapotentialtherapeutictargetinepilepsy. potassiumchannelsubunits:molecularcorrelatesofthe MedHypotheses,2011,76,184–186. M-channel.Science,1998,282,1890–1893. 139. YuFH,CatterallWA:Overviewofthevoltage-gatedso- 129. WarmkeJW,GanetzkyB:A familyofpotassiumchannel diumchannelfamily.GenomeBiol,2003,4,207. genesrelatedto eag in Drosophila andmammals.Proc 140. ZhangN,WeiW,ModyI,HouserCR:Alteredlocaliza- NatlAcadSciUSA,1994,91,3438–3442. tionofGABAA receptorsubunitsondentategranulecell 130. WernerFM,CoveñasR:Classicalneurotransmittersand dendritesinfluencestonicandphasicinhibitionina neuropeptidesinvolvedingeneralizedepilepsy:afocus mousemodelofepilepsy.JNeurosci,2007,27, onantiepilepticdrugs.CurrMedChem,2011,18, 7520–7531. 4933–4948. 131. WickendenAD:Potassiumchannelsasanti-epileptic drugtargets.Neuropharmacology,2002,43,1055–1060. 132. WickendenAD,YuW,ZouA,JeglaT,WagonerPK: Received: January29,2013; intherevisedform: March4,2013; Retigabine,anovelanti-convulsant,enhancesactivation accepted: March11,2013.

Pharmacological Reports, 2013, 65, 787–801 801