Sunteți pe pagina 1din 16

Current Topics in Medicinal Chemistry 2005, 5, 15-30 15

Voltage Gated ion Channels: Targets for Anticonvulsant Drugs

Adam C. Errington1,3, Thomas Stöhr2 and George Lees1,3,*

1
Department of Pharmacology and Toxicology, Otago School of Medical Sciences, University of Otago, PO Box 913,
Dunedin, New Zealand, 2Schwarz Biosciences GmBH, Alfred Nobel Strasse 10, 40789 Monheim am Rhein, Germany,
3
Sunderland Pharmacy School, University of Sunderland, Chester Road Campus, Sunderland SR1 3SD, UK

Abstract: Epilepsy is one of the most prevalent neurological syndromes in the world today. Epilepsy describes a group of
brain disorders whose symptoms and causes are diverse and complicated, but all share a common behavioural
manifestation: the seizure. Seizures result from the abnormal discharge of groups of neurons within the brain, usually
within a focal point, that can result in the recruitment of large brain regions into epileptiform activity. Although the range
of explanations for the development of seizures can be as varied as genetic composition to acute head trauma, the net
result is often similar. The excitability of neurons is governed by the input they receive from their neighbours and the
intrinsic excitability of the neuron. In this review we focus on elements that are crucial to determining the intrinsic
excitability of neurons in the CNS, the voltage gated ion channels (VGICs).
VGICs as well as being important for physiological function are critical in producing hyperexcitability such as that
associated with seizure discharges. Many drugs routinely used in the clinical setting, as well as several novel experimental
drugs, have shown interactions with VGICs that underpin, at least in part, their anticonvulsant action. We review the
physiological roles of voltage gated ion channels that are selective for sodium, potassium and calcium conductance and
attempt to highlight their role in the pathology of epilepsy. This is supplemented by the mechanisms of drug action at
these important anticonvulsant targets for classical and clinically relevant compounds (e.g. phenytoin, ethosuximide) as
well as some important second generation drugs (e.g. gabapentin, levetiracetam) and novel experimental agents (e.g.
retigabine, losigamone, safinamide). We also briefly discuss the urgent need for new drugs in this arena and the potential
of combinatorial methods and recombinant screening to identify leads.
Key Words: Voltage gated ion channels; epilepsy; anticonvulsants

INTRODUCTION the electrogenic properties intrinsic to each neuron. These


are determined to a great extent by the complement and
Epilepsy is one of the most prevalent neurological localization of functional voltage gated ion channels
disorders with current estimates approximating between 0.5 expressed in the cell membrane [1]. It is intuitive, therefore,
– 2% of the global population being affected. The use of the
that these determinants of physiological excitability would
term epilepsy is to some extent misleading as it actually
be crucially involved in the pathological hyperexcitability
refers to a collection of neurophysiological disorders that are
occurring during the seizure discharges characteristic of the
diverse in terms of both their etiology and symptomology.
epilepsies. Indeed both excitatory and inhibitory neurotrans-
The typical behavioural manifestation common to all forms
mission as well as voltage-gated channels (that can be
of epilepsy is the aberrant synchronized discharge of
modulated selectively by natural or synthetic toxins), have
neuronal populations termed the ‘seizure’. Seizures result
been shown to be crucial for seizure like activity in
from phasic changes in the firing properties of groups of
acute chemoconvulsant models in rodent brain slices and in
neurons, usually within a discrete focal point, to an vivo [2].
intermittent high-frequency burst-firing mode. Induction of a
population of neurons into a pattern of burst-firing resulting It is not surprising, therefore, that many of the drugs used
in synchronized disruptive seizure discharges is determined in the treatment of seizures and epilepsy target the channels
by changes in excitability of neurons that are essentially and receptors that are vital for the initiation and spread of
governed by two major factors. The first determinant of seizure activity. In this review we focus specifically upon the
neuronal excitability in any given neuron, is the balance of voltage gated ion channels, discussing their function both
synaptic input from neighboring cells within the network in physiologically and pathologically and evaluate their
the form of chemical transmission (through ligand gated ion relevance as anticonvulsant drug targets by outlining how
channels and G-protein coupled metabotropic receptors) and they can be manipulated for therapeutic benefit.
aplastic but extremely rapid electrical transmission (through
gap juctions). Secondly, and perhaps most importantly are ELECTROPHYSIOLOGICAL PROPERTIES AND
MOLECULAR BIOLOGY OF VOLTAGE GATED
SODIUM CHANNELS (VGSCS)
*Address correspondence to this author at Chair & Head of Department of
Pharmacology & Toxicology, Otago School of Medical Sciences, University The action potential is the cellular mechanism by which
of Otago, PO Box 913, Dunedin, New Zealand; Tel: 64 3 4797265; Fax: 64 excitable cells within the nervous system communicate with
3 4799777; E-mail: george.lees@stonebow.otago.ac.uk one another. Action potentials propagating in large
1568-0266/05 $50.00+.00 © 2005 Bentham Science Publishers Ltd.
16 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

pyramidal cells of the cortical surface of the brain constitute 1.9 [9], have been characterised. cDNA clones have been
the unitary events in an EEG. At the cellular level, using the isolated from rat brain cDNA libraries and show that the
newly developed voltage clamp technique in the 1950s major central nervous system (CNS) channels are encoded
Hodgkin and Huxley showed that a transient voltage for by four genes entitled Scn1a, Scn2a, Scn3a and Scn8a [9-
activated sodium current (INa) was responsible for the rapid 11]. The corresponding protein products of these genes (or
rising phase of this crucial intracellular communication splice variants of them) form the major brain sodium channel
process. Although the techniques required to elucidate the α-subunits classified as NaV1.1-1.3 which are blocked by the
molecular correlate for this current were not available in the puffer fish ‘fugu’ poison (Tetrodotoxin, TTX). Several
1950s these seminal studies identified three essential features mutations of the Scn1a and the Scn2a genes have been
that have come to characterize the voltage gated sodium identified in individuals suffering from monogenetic epilepsy
channel (VGSC) family, namely: voltage dependent syndromes such as generalized epilepsy with febrile seizures
activation, rapid inactivation and selective ion conductance plus and severe myoclonic epilepsy of infancy (table 1). The
[3-5]. Following this series of elegant and pioneering gene Scn8a encodes the primarily CNS Nav 1.6 α-subunit
experiments a large number of detailed electrophysiological (formerly SkM2) and a further CNS channel resistant to
and molecular studies have elucidated much about the TTX, Scn5a (Nav1.5, formerly NaCh6), has also been
structure and function of the channel(s) responsible for INa. described. Crystal structures for these large homo-oligomeric
Molecular biology has determined the major structural pores remain elusive but the fine structure of the channel
component of the channel to be a protein of approximately complex has been determined by means of cryo-electron
260 kDa, called the α-subunit, which has four repeated microscopy and image reconstruction analysis. This reveals
(named I-IV) structural motifs consisting of six α-helical water filled pores (thought to contain the gating charges)
transmembrane spanning domains separated by intra and deep within the membrane plane and multiple intracellular
extracellular loops [6]. These four repeated domains fold vestibules which constitute a likely pathway for the permeant
together to form a central pore and it is their structural ions [12].
components that determine selectivity and conductance of
Functionally, VGSCs can be simply thought to exist in
the ion channel. Although α subunits alone can form
three conformational states and it is the transition between
functional channels co-expression of accessory β (33 kDa)
these states that allows selective and temporally regulated
subunits is most commonly required to confer the correct
ionic conductance. At the resting potential of most CNS
gating properties found in native channels. Channels formed
neurons, VGSCs can be found in a closed non-conducting
purely from α subunits exhibit slower inactivation and
positive shifts in the voltage dependence of activation state commonly referred to as the ‘resting’ state (Fig. 1C). In
response to change in membrane potential, such as that
compared to native neurons when expressed in Xenopus
provided by excitatory synaptic input [13], the channels
oocytes. To date three β (β1−3) accessory subunits
undergo a physical conformational change. Activation occurs
have been cloned from rat and human and their effects
within a fraction of a millisecond and as predicted by
upon channel gating have been extensively characterised
Hodgkin & Huxley [14] is due to movement of gating
[6-8].
charges within the membrane electric field. These so called
Based upon sequence homology, several different sodium ‘gating currents’ have been measured electrophysiologically
channel α subunits, recently named by IUPHAR as NaV1.1- [15,16] and attributed to sequences of positively charged

Table 1. Voltage Gated ion Channel Genes Bearing Mutations which have been Linked to Inherited Epilepsy.

Voltage-gated ion channel family Gene name Syndrome Reference

Potassium channels KCNQ2 BFNC [113,119]


KCNQ3 BFNC [119,120]
KCNQ3 JME [121]

Sodium channels SCN1A GEFS+ [122-124]


SCN1A SMEI [125,126]
SCN2A GEFS+ [127]
SCN2A BFNIC [128]
SCN1B GEFS+ [129]

Calcium channels CACNA1A IGE [130]

CACNA1H CAE [131]


BFNC: Benign familial neonatal convulsions
BFNIC: Benign familial neonatal/infantile convulsions
CAE: Childhood absence epilepsy
GEFS+: Generalized epilepsy with febrile seizures plus
IGE: Idiopathic generalised epilepsy
JME: Juvenile myoclonic epilepsy
SMEI: Severe myoclonic epilepsy of infancy
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 17

Fig. (1). (A) Structures of anticonvulsant drugs that act upon VGSCs. (B) Diagram of the subunit structure of VGSC α and β subunits
showing the pore forming region, the voltage sensor and inactivation gate. (C) Gating conformations of the voltage gated subunit.

arginine and lysine residues in the S4 regions of the α membrane potential and time and is a mechanism that
subunit [6] (Fig. 1B). The movement of the S4 region ensures adequate time for recovery before reopening of the
perpendicular to the plane of the membrane results in channel [22].
opening of the activation gate and allows sodium flux. For
The properties of rapid activation and inactivation of
the VGSC it was estimated that the movement of
VGSCs are essential determinants in neuronal firing
approximately six charges across the entire membrane would
frequency. As such they are also crucial for signal
be required for gating [7]. Neutralization of the charged
transmission in neuronal networks including recruitment of
residues in the S4 region markedly alters the voltage
oscillatory activity of putative importance for both cognition
dependence of activation [17]. Upon opening of the
and consciousness (i.e. γ frequency) [23]. However the
activation gate, the channel pore (formed by a re-entrant loop
ability of sodium channels to rapidly cycle through conform-
between transmembrane segments V and VI) selectively
ational states and facilitate high frequency neuronal firing is
conducts sodium ions along their electrochemical gradient
also indispensible for the generation of synchronous bursting
from the extracellular space to the cell interior thus
activity that is characteristic of epilepsy. Indeed computer
depolarizing further the neuronal transmembrane potential.
models of normal and disruptive (epileptic) neuronal
However, within a few milliseconds of opening of the network oscillations require the inclusion of the fast transient
activation gate, sustained depolarization results in sodium current (INa) carried by neuronal VGSCs [1,23].
termination of the sodium conductance by a process known
as inactivation. Inactivation usually (but not always, as As previously stated, sodium channel inactivation is
discussed below) occurs rapidly, attenuating the majority of often thought of as a single process but it is increasingly
the sodium current and for this reason is often referred to as clear that there are at least two distinct kinetic classes of
‘fast’ inactivation. The process of fast inactivation has been inactivation (ultra-slow activation is discussed later). The
strongly linked to the short intracellular loop between second type of sodium channel inactivation, termed slow, is
domains III and IV (the so-called III-IV linker) [18,19] (Fig. postulated as a cellular back-up mechanism that prevents INa
1B) which is also known as the inactivation gate. from becoming fully available rapidly. This type of
Enzymatically cutting this loop [17], deleting amino acids in inactivation appears to be independent of the III-IV linker
the linker [18], or directing antibodies against it [19], all (crucial for fast inactivation) and has recovery time constants
produce a marked slowing or disruption of the inactivation in the order of several hundreds of milliseconds to seconds
process. It is hypothesised that the intracellular portion of the [22]. Recruitment of VGSCs into the slowly inactivated
protein may bind to a so called ‘inactivation gate receptor’ conformation appears to proceed only under conditions of
on the cytoplasmic side of the IVS6 domain using a structural prolonged or high frequency depolarization [24-26].
motif comprising a hydrophobic group of isoleucine, Furthermore, there is an apparent proportional relationship
phenylalanine and methionine (IFM) [20,21] (Fig. 1C). between the time taken to recover from the slowly
Entry of sodium channels into the fast inactivated-state is inactivated state and the duration of depolarizing challenge
highly voltage dependent and becomes significantly more [27]. Type II and IIA sodium channels expressed in the
pronounced upon depolarization. The ability of channels to Xenopus oocyte expression system showed the time constant
recover from the fast inactivated state is dependent upon of recovery from slow inactivation was intrinsically related
18 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

to the duration of previous depolarizations by a power law: subunits. In functional studies activation of PKA resulted in
τ(t) = p•tD (where τ is the recovery constant for the channel a 25-40% reduction in peak Na+ current mediated by NaV1.2
from slow inactivation, t is the duration of previous channels expressed in Xenopus oocytes [41] or CNaIIA-1
depolarization, p is a constant kinetic setpoint and D a [42] (CHO cell derived stable line expressing NaV1.2) cells
positive scaling power). This type of inactivation process is without shifting voltage dependence of activation or
especially pertinent to epileptiform activity as the neuronal inactivation.
networks involved are often exposed to periods of prolonged
To complement this evidence it has also been shown that
high frequency neuronal activity [28-31] that may be
protein kinase C (PKC) is able to phosphorylate sodium
‘damped down’ by slow inactivation. It appears therefore
channel α subunits. The amplitude of sodium current is
that pharmacological enhancement or stabilization of the
slowly inactivated conformation of VGSCs may be useful in reduced and the rate of inactivation slowed in neuroblastoma
disrupting prolonged neuronal seizure discharges. treated with fatty acids such as OAG (1-oleoyl-2-acetyl-sn-
glycerol) or DOG (1,2-dioctanoyl-rac-glycerol) that activate
Although I Na is the principal sodium current responsible PKC by mimicking the endogenous second messenger
for neuronal activity the existence of a second type of molecule DAG. Slowed inactivation of sodium channels
sodium current, termed the persistent current (INaP), has been produced by activation of PKC results from phosphorylation
confirmed in a number of neuronal cell types [32]. This of the site situated on the inactivation gate that does not
current, although only representing 1-3% of the total peak appear to be targeted by PKA. In Xenopus oocytes
current is thought to be crucial for determination of spike transiently expressing rat brain Na+ channels the same
threshold in neurons [32]. The mechanism behind these effects as described above can be achieved using phorbol
persistent currents is not clearly defined but has been esters. In our laboratory we have shown that OAG can
variously attributed to the failure of a fraction of channels reduce sodium currents in voltage clamped N1E-115
(responsible for conducting INa) to inactivate or the ability to neuroblastoma and also inhibits sustained repetitive firing in
switch to an ‘ultra-slow’ inactivating gating conformation rat cultured cortical neurons (Fig. 2AB).
[32-35]. The voltage dependence of activation of INaP differs
from that of INa. Persistent Na+ currents are activated at Furthermore, activation of G-protein coupled receptors,
potentials below spike threshold (-60mV) and reach peak at including the muscarinic acetylcholine (mAChR) receptor
and the dopamine (D1) receptor that are coupled to the PKC
between –40 and –35mV with V50 for activation at around
and PKA pathways, respectively, also results in reduction of
–50mV [33,36-38]. INaP has been implicated in the fine
Na+ currents. The effects of the reduced Na+ current and
control of neuronal excitability based upon the sub-threshold
slowing of inactivation produced by activation of PKC are
activation kinetics and lack of rapid inactivation. As
evident in pyramidal neurons of the hippocampus. Activation
previously highlighted, synchronous neuronal network
of mAChRs using carbachol converts intrinsically burst
activity (like that associated with seizures) is strongly
firing CA1 neurons into a pattern of regular spiking [43,44].
dependent upon the ability of neurons to fire bursts of action
potentials. INaP has been shown to be crucial in both This is achieved by a reduction in not only the transient Na+
current but also the persistent current crucial for determining
hippocampal CA1 neurons and layer V neocortical neurons
spike threshold. Similar effects can be seen in hippocampal,
in the induction of burst firing modes. Evidence exists that
striatal and cortical neurons where activation of the D1
INaP is also thought to, at least in part, contribute to rhythmic
receptor pathway reduces the number of spikes in an evoked
sub-threshold oscillations that can determine behaviour of
burst.
entire neuronal networks in a number of structures including
the neocortex and entorhinal cortex [23]. It appears that INaP Neuromodulation of sodium channel function, particularly
is acting as a mechanism for ‘fine-tuning’ neuronal somatic channels responsible for spike generation, may have
excitability. Selective modulation of this current may an important influence upon the firing patterns of many
represent an attractive target for future anticonvulsant drug neurons. The ability to activate PKC or PKA directly by
development. It is possible that a ligand selective for INaP mimicking endogenous second messengers or indirectly via
may be able reduce hyperexcitability without the drawbacks GPCRs to produce reduction in both persistent and transient
associated with attenuating INa. Na+ currents may prove an exciting target for future AED
A new and exciting putative target for future design. Many large drug companies now use high-
throughput combinatorial screens to highlight new leads and
anticonvulsant drug intervention has come to light relatively
recently and is based upon a somewhat unexpected form of kinase effectors are the focus of much attention in a variety
of therapeutic areas. To date, however, there are no known
plasticity displayed by VGSCs (see refs [7,39] for detailed
review). Biochemical experiments including two dimensional clinical or experimental agents that exploit such pathways to
produce anticonvulsant effects.
phosphopeptide mapping have shown purified sodium
channel α subunits have five distinct phosphorylation sites
throughout the glycopeptide. Four of these sites are found on DRUGS ACTING UPON VGSCs
serine residues (573, 610, 623 and 687) in the large A number of anticonvulsant molecules in the pharma-
intracellular loop connecting domains I and II (LI-II) and a copoeia today have primary modes of action that involve
further one is located of the domain III-IV linker which modulation of neuronal VGSCs. The classical compound in
constitutes the inactivation gate [40]. In intact synaptosomes this class was discovered by Merritt and Putnam [45] for its
the Na + flux induced by veratridine is reduced by circa 26% ability to inhibit maximal electroshock-induced seizures
when activators of cAMP dependent protein kinase (PKA) (MES) in rodents. Since its discovery in 1938, phenytoin
are concurrently applied due to rapid phosphorylation of α [5,5-diphenylhydantoin, DPH] (Fig. 1A) and the pro-drug
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 19

Fig. (2). (A) Sustained repetitive firing in current clamped cultured rat cortical neurons evoked by somatic current injection. The PKC
activator OAG (20µM) markedly reduces the frequency of action potentials within the burst. (B) Sodium currents from voltage clamped
mouse N1E-115 neuroblastoma cells are attenuated by application of 10µM OAG for five minutes. Cells were voltage clamped at –100mV
and test pulses were delivered to 0mV.

Primidone have become first line treatments for partial and given potential [49,50,52]. The binding of DPH to the fast
generalized tonic clonic seizures due to their ability to reduce inactivated conformation of the sodium channels is relatively
seizures without producing marked sedation [46]. Much is slow and this, along with the strong use and voltage
now known about the mechanism(s) of action of DPH and it dependence, explains how the molecule is able to selectively
has become clear that the biophysical characteristics of its inhibit high frequency epileptiform discharges whilst leaving
interaction with sodium channels are central to its efficacy as normal cellular activity unaffected.
an anticonvulsant. In addition it has been reported that DPH is able to
Early evidence upon the mechanism of action of DPH reduce I NaP in dissociated cortical neurons [53], and that this
was obtained in experiments using supramaximal stimulation translates to a reduction in cellular excitability in layer V
of frog sciatic nerve. It was discovered that DPH produced neurons maintained in an intact slice (it is noteworthy that
no effect upon an initial action potential triggered by DPH concentrations were lower than those required to
stimulation but blocked a second rebound spike [47]. This inhibit INa [54]). Single channel experiments revealed that
appeared to suggest DPH would selectively inhibit high DPH preferentially impairs late channel openings rather than
frequency neuronal firing, a hypothesis that has been those occurring immediately in response to a voltage step
confirmed in several studies since. Notably high frequency which may explain the reduction of persistent currents. A
evoked action potentials in mouse spinal cord neurons were more recent study however disputes this claim showing that
inhibited by DPH without impairment of spontaneous low low micromolar concentrations of DPH produced little or no
frequency action potentials [48]. effect upon INaP [55].
Further and more extensive voltage clamp studies have Carbamazepine [5H-Dibenz[b,f]azepine-5-caboxamide,
delineated much about the biophysical interaction of DPH CBZ] (Fig. 1A) is an iminostilbene derivative of tricyclic
with sodium channels. Several studies using mouse antidepressants (structurally it is completely different to
neuroblastoma have shown that DPH produces tonic block of DPH in that it does not contain the ureide moeity
the transient sodium current INa and that this block is highly traditionally thought to be important to anticonvulsant
voltage dependent [49,50]. For example DPH has been molecules) [56]. However CBZ demonstrated shared
reported to have IC50 = 120µM at –85mV, but IC50 = 10µM anticonvulsant properties in vivo with DPH in potently
at –60mV [51]. Indeed the block of sodium currents by DPH inhibiting hindlimb extension in the MES seizure test whilst
in N18 mouse neuroblastoma could be reversed by up to proving relatively ineffective against pentylenetetrazol (PTZ)
95% if cells were hyperpolarized to –120mV [49]. It was induced epileptiform activity [57]. To complement this pre-
predicted that because the block of sodium channels clinical data, since its therapeutic launch CBZ has been
followed the voltage dependence of inactivation of INa that successfully used in the control of generalised (tonic-clonic)
DPH inhibition was related to the inactivated state. Indeed convulsive seizures and partial (focal motor or complex
DPH does shift the voltage dependence of steady state partial) seizures, a niche common with DPH [58].
inactivation in the hyperpolarizing direction and thus reduces Like DPH early clues towards the mechanism of
the population of channels available for activation at any action of CBZ were derived from its ability (or 10,11-
20 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

epoxycarbamazepine) to inhibit, at clinically relevant Differences in the binding kinetics of DPH and CBZ to
concentrations, repetitive firing in cultured mammalian CNS the sodium channel have been described with the latter
neurons [59]. Further and more detailed voltage clamp binding 5 times faster than the former but with three fold
studies in mouse neuroblastoma cells [49,52] showed CBZ lower affinity [60]. This may give CBZ an advantage in the
produced tonic block of INa that was highly voltage treatment of seizures characterized by comparatively brief
dependent; with approximately 40% of INa blocked by CBZ depolarizing discharges and may explain differential
(20µM) at a holding potential of –75mV, but virtually zero responses within certain patient sub groups to these agents.
block when held at –120mV [49]. These studies also showed Although it is still commonly used in the clinic, toxicological
that, like DPH, the effects of CBZ were use dependent and drawbacks related to CBZ (including hepatic toxicity) have
block was accumulated with repetitive stimulation (Fig. 3). led to the development of structural congeners that maintain
anticonvulsant efficacy whilst eliminating, to some extent,
CBZ produces shifts in the voltage dependence of steady the toxicity. Oxcarbazepine [OXC] (Fig. 1A) shares with
state fast inactivation (in the hyperpolarzing direction) by CBZ the dibenzazpine nucleus bearing the 5-carboxamide
stabilizing the inactivated conformation of the sodium substituent but is structurally different at the 10,11- position.
channel and thus reduces the ability of the channel to revert It appears that this substitution results in altered
the resting state (Fig. 3). The time constant for recovery from biotransformation when compared to CBZ bypassing the
fast inactivation is also prolonged by CBZ. 10,11-epoxide metabolite thought to be responsible for the

Fig. (3). Voltage-gated Na+ channels are important targets for a number of clinically important anticonvulsant drugs including
carbamazepine (100 µM throughout).
A. Voltage clamped currents showing tonic block exerted by CBZ at two different holding potentials (test pulse applied to elicit the peak
inward current at 0.5 Hz). As shown in the graph on the right, the proportion of the current blocked is increased with depolarization (note
that, over the time course of these experiments, the currents had a slight tendency to increase or “run-up” due to dephosphorylation of the
Na+ channel complex). B. The tonic block can be enhanced by activating the channels at higher frequencies across the physiological range
(up to 200Hz). The marked facilitation of block is clearly shown by progressively diminishing currents in the train (overlays are shown in
inset) evoked in this experiment at 10 Hz for 3s. C. Paired pulse protocols (top right) can be used to show that CBZ generates a large
hyperpolarizing shift in the steady-state inactivation profile for the Na+ current and (not shown) it retards recovery from the fast-inactivated
state.
CBZ, LTG and DPH exert a broadly similar effect (exhibiting subtle differences in kinetics, voltage-dependence or frequency dependence).
The concensus interpretation of these data are that the anticonvulsant drugs bind preferentially to (and stabilize) inactivated channel states.
This preferential interaction with deploarised, ectopic and rapidly firing channels (in diseased tissue) underpins the therapeutic ratio of these
clinically effective anticonvulsants.
All experiments illustrated above (Errington and Lees Unpublished) were conducted on NIE115 murine neuroblastoma (in which native
evoked Na+ currents are tetrodotoxin sensitive) . Data are presented as Mean with SEM represented by vertical error bars (.n=3-4).
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 21

toxicity associated with the parent molecule [61]. Further resultant sodium channels are far less susceptible to voltage
modification has led to (S)-(-)-(10)-10-acetoxy-10,11- and frequency dependent block by LAs [69,70]. The
dihydro-5H-dibenz/b,f/azepine-5-carboxamide (BIA 2-093) similarity between the blocking action of the LAs and the
(Fig. 1A) which was found to be particularly effective in anti-epileptic drugs (AEDs) suggested that this binding site
preventing MES induced seizures with similar potency to may be shared across both classes of molecules and indeed
CBZ but with fewer cognitive and motor deficits. Like CBZ the F1764A and Y1771A mutants showed greatly attenuated
and OXC, BIA 2-093 has been shown to inhibit VGSCs in a block by DPH and LTG [11]. Furthermore Kuo [56] has
frequency and voltage dependent manner in N1E-115 cells shown that applications of mixtures of LTG, DPH and CBZ
confirming a shared mechanism of action with the former cannot produce double occupancy of the channel. He
[62]. Secondary to the state dependent block of VGSCs concludes that despite dissimilarity in structure, these drugs
CBZ, OXC and BIA 2-093 have all been shown to inhibit (and probably CBZ derivatives OXC and BIA 2-093) likely
veratrine induced neurotransmitter (glutamate, aspartate, share the same or very similar receptor site on the sodium
GABA, dopamine) release in rat striatal slices. channel α subunit.
Lamotrigine [3,5-diamino-6-(2,3-dichloro-phenyl)-1,2,4- Despite the effectiveness of the aforementioned AEDs in
triazine , LTG] (Fig. 1A) is the first of a new generation of controlling seizures, in patients with generalized convulsive
drugs developed in the 1980’s and 90’s to have sodium seizures, some patient groups are still found to be refractory
channel inhibition as its primary mechanism of action. to existing therapies. This has led to further development of
Although between 60-80% of patients responded well to anticonvulsant compounds despite negative market forces
existing agents some patients did not have seizures including long lead times and brief patent life of new
effectively controlled. The observation that the folates molecules. Some of these newer drugs, discovered by
possessed convulsant activity in animals led to the screening screening large libraries of compounds in various animal
of putative antifolates as potential anticonvulsant candidates. models, have been shown to act (to some degree) on VGSCs.
From these screens LTG emerged as the lead compound Topiramate [2,3:4,5-bis-O-(1-methyl-ethylidene)-36-D-
showing similar pharmacological properties as CBZ and fructopyranose sulfamate, TPM] (Fig. 1A) is an
DPH [63-65]. Early evidence implicating the VGSC as anticonvulsant drug which was licensed for clinical use in
the primary target for LTG was derived from neurochemical many European countries in the late 1990’s. In the traditional
studies in which glutamate and GABA release evoked by animal models (MES, PTZ) TPM showed a profile similar to
veratridine (but not potassium) were suppressed in slices of that of DPH [71] and it has therefore become clinically
rat cortex [64]. This was later confirmed by whole cell patch established as an add-on therapy in simple or complex partial
clamp [66] studies on cultures of rat cortical neurons in seizures with or without secondary generalization. The
which LTG reduced the incidence of synaptic events mechanism of action of TPM, although still unclear appears
significantly without affecting evoked postsynaptic to exhibit three facets, namely: blockade of kainate evoked
responses. It was also demonstrated that while LTG did not excitatory currents [72], enhancement of inhibition through
attenuate single action potentials it did markedly reduce GABAergic chloride channels [73] and depression of sodium
burst firing [67-68]. Voltage clamp studies in N4TG1 mouse currents [74]. TPM significantly reduced sustained repetitive
neuroblastoma confirm that LTG exhibits similar biophysical firing in cultured hippocampal pyramidal cells at
interactions with VGSCs compared to CBZ and DPH. LTG therapeutically relevant (10-30µM) concentrations and this
tonically inhibits INa in a concentration dependent manner was later confirmed to be due to attenuation of INa. In
from a holding potential of –80mV but like CBZ and DPH cerebellar granule cells [75] and dissociated neorcortical
the block is highly voltage dependent and can be reversed by neurons [76], TPM produced voltage dependent tonic block
hyperpolarization. The voltage dependence of steady state of I Na and shifted the steady state inactivation curve to more
inactivation is also shifted in the hyperpolarizing direction to negative values in a fashion previously described for LTG,
a similar extent to CBZ and DPH [52]. It has been proposed CBZ, DPH, OXC and BIA 2-093. TPM has also been
that LTG may be unique in acting on the slow inactivated reported to inhibit sustained sodium conductances (INaP) in
state of the channel, rather than classical fast inactivation evoked in neocortical neurons in response to slowly
gating, but whether this truly represents the physiological depolarizing membrane potential ramps at concentrations
slow inactivated state or slow binding kinetics with the fast lower than those required to inhibit INa. In this way TPM
inactivated state is unclear. Like DPH, LTG has been shown mimics DPH and, furthermore, it is conceivable that the
to block INaP in rat neocortical neurons, but the degree of inhibition of the persistent current may represent a major
block is less than 10% at a concentration of 10µM. contribution to the reduction in cellular excitability produced
Interestingly two derivatives of LTG (sipatrigine, 202W92) by all these drugs.
that are putative neuroprotective agents, are able to inhibit
INaP far more profoundly with 70 and 90% inhibition The tetronic acid derivative Losigamone [(±)-5(R,S)-5-
respectively at 10µM [55]. (2- chlorophenyl)hydroxymethyl) - 4 – methoxy (5H0 -furanone,
LSG] (Fig. 1A) is an experimental anticonvulsant currently
There is striking and widely accepted evidence that all undergoing phase II/III clinical trials in patients with partial
(PHT, CBZ & LTG) these anticonvulsants and another group and secondary generalised seizures. LSG has been shown to
of drugs that modulate the VGSCs, the local anesthetics produce a reduction in spontaneous synaptic activity in
(LAs), do so by binding to a receptor site inside the cultured hippocampal neurons and a marked suppression of
cytoplasmic vestibule of the ion conducting pore of the sustained repetitive firing evoked by somatic current
channel. More specifically if either F1764 or Y1771 are injection [77]. However the effects of LSG are apparently
mutated to alanine in transmembrane segment IVS6 the different from many classical sodium channel-affecting
22 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

anticonvulsants previously described. In the majority of central nervous system. Calcium influx into the cellular
cellular studies inhibition of SRF can be closely correlated to interior occurring in response to channel gating is essential
impairment of INa. In whole cell recordings made from for the translation of transient electrical signals into
hippocampal neurons in brain slice preparations LSG did not biochemical events, both acutely, as in neurotransmitter
inhibit INa at relevant concentrations. LSG did however release, and more chronically, as in regulating gene
produce inhibition of INaP and it has been suggested that this expression and even cell-death. VGCCs were initially
may prove to be crucial to its ability to decrease neuronal described by Paul Fatt and Bernard Katz in crustacean
hyperexcitability [78]. In this respect LSG may represent the muscle but have subsequently been shown to exist in an
first compound to be used in epilepsy that inhibits INaP array of mammalian cells types including, crucially, CNS
without significant effect on transient sodium currents. neurons. VGCCs were first classified according to their
Another new anticonvulsant ligand which acts upon pharmacological and biophysical characteristic into P/Q, N,
voltage gated sodium channels is currently in phase III trials. L, R (high voltage activated, HVA) and T (low voltage
Safinamide [Propanamide, 2-[[[4-[(3-fluorophenyl)methoxy] activated, LVA) types [80]. HVA channels have been
phenyl]methyl]amino]-, (2S)-monomethanesulfonate, PNU- extensively characterised biochemically revealing that
151774E, NW-1015, SAF] (Fig. 1A) produced inhibition of VGCCs are complex heteromeric structures. VGCCs have
sustained repetitive firing, inhibition of TTX sensitive been demonstrated to be formed by at least three main
sodium currents and displayed higher affinity for the Na+ subunits namely α1, α2δ and β, as well as, in some cases a γ
channel [3H] batrachotoxin receptor site (site 2) than subunit. The main component of the VGCC is the
phenyoin or lamotrigine (EC50: 8.2µM) [79]. Although the approximately 2000 amino acid 190-250kDa α subunit. The
actions of SAF appear to be complex and multifaceted, VGCC α subunit is structurally similar to VGSC α subunits,
including inhibition of L and N-type calcium channels, the with four repeated domains each being produced by six
compound nonetheless represents another molecule whose transmembrane spanning segments. As with its sodium and
anticonvulsant actions are conferred by interaction with potassium conducting counterparts it is predicted that the
VGSCs. calcium channel voltage sensor is the S4 membrane spanning
segment and the pore is formed by a re-entrant loop between
Clearly the VGSC represents a major ion channel target the S5 and S6 segments (Fig. 4B). Ten VGCC α subunits
for both existing clinical and experimental anticonvulsant have thus far been identified through homology screening
molecules. It is apparent that despite varied chemical and this has led to reclassification of the different Ca2+
structures many compounds can interact to produce state channel types. The cloned subunits are know divided into
dependent block of INa and that in some cases there is three functionally related families based upon their
overlapping reduction of INaP providing a dual mechanism biophysical and pharmacological characteristics [81].
for reducing hyperexcitability. Cav1.1-1.4 subunits form the L-type channels whilst Cav2.1,
Cav2.2 and Ca v2.3 form the P/Q, N and R types respectively.
STRUCTURE, FUNCTION AND ANTICONVULSANT
The LVA T-type channels include an α subunit from the
PHARMACOLOGY OF VOLTAGE GATED
Cav3.1-3.3 types [80].
CALCIUM CHANNELS
The voltage gated calcium channels (VGCCs) are critical The first of the calcium currents to be described was the
for the proper functioning of excitable cells within the HVA L-type (because the current is long lasting, particularly

Fig. (4). (A) Structures of anticonvulsant drugs acting at VGCCs. (B) Diagram of the subunit structure of the VGCC α subunit and the
accessory β, α2 and δ subunits.
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 23

when Ba2+ is used as the permeant ion [82]) current in associated with epileptic phenotypes. For example the
smooth and skeletal muscle. The L-type channels are stargazer mouse has shown that mutations of γ subunits
characterized by slow voltage dependent inactivation, high result in loss of γ2 subunits without compensatory up
voltage of activation, modulation by cAMP dependent regulation of other γ subunit types. It has been suggested by
phosphorylation pathways and inhibition by dihydro- some authors that this may contribute to the pathogenesis in
pyridines [83]. The T-type LVA calcium channels that were this mutant mouse strain that results in spike wave seizures
discovered in cerebellar purkinje neurons [84] but characteristic of absence epilepsy [87]. This is still
characterised in detail in dorsal root ganglion neurons [82]. somewhat controversial however as some groups implicate
The T-type designation is based upon the rapid inactivation the mutant γ subunit analogue (stargazin protein) an
of this type of channel resulting in transient currents. important role in molecular trafficking and cell surface
Furthermore the T-type channels are activated at much more expression of AMPA receptors [88].
negative membrane potentials, have small single channel
conductances and are insensitive to block by The LVA T-type VGCC has been implicated in playing a
dihydropyridines. significant role in controlling rhythmic discharges in
thalamocortical neurons that display epileptiform spike wave
A further type of Ca2+ current was revealed based upon discharges associated with absence seizures. 2-Ethyl-2-
kinetic and pharmacological divergence from the existing L methylsuccinimide [Ethosuximide, ESM] (Fig. 4A) is an
and T-type currents. Nowycky et al. showed using whole AED that has an unusual clinical profile. ESM shows
cell and single-channel recording from dorsal root ganglion efficacy against absence seizures but not against partial or
neurons a calcium current that activated at more negative generalized tonic-clonic seizures. This has been shown to be
potentials than did the L-type but more positive than the T- due to the ability of ESM to partially inhibit T-type calcium
type [82]. The new current, which became known as the N- channels at therapeutically relevant concentrations and hence
type, also showed more rapid inactivation than L but slower reduce the hyperexcitability of thalamocortical neurons that
inactivation than the transient type. However it is the is associated specifically with absence seizures [89].
sensitivity to block by the snail peptide ω-conotoxin GVIA Furthermore it has also been demonstrated that methyl-
that is the crucial distinguishing factor for identification of phenyl suximide (the active metabolite of the related
N-type channels [85]. The remaining P, Q and R-type compound methsuximide) but not its inactive analog also
calcium channels were identified in a range of neuronal types produces inhibition of T-type currents in thalamic neurons
based upon sensitivity to other peptide toxins. P-type [90]. To complement these findings a recent study has shown
currents are highly sensitive to block by the spider toxin ω- that both of these agents are able to inhibit cloned human T-
agatoxin IVA and were first recorded in purkinje neurons type VGCCs expressed in HEK293 cells whereas non-
whilst the Q-type were first found in cerebellar granule anticonvulsant congeners were ineffective [91].
neurons [86] and display much lower affinity for the toxin.
Finally the R-type were found also in cerebellar granule A recent and interesting development in the field of
VGCC anticonvulsant pharmacology are the novel drugs 1-
neurons and take their name from the fact they are resistant
(aminomethyl) cyclohexanacetic acid [Gabapentin, GBP]
to the subunit specific organic and peptide Ca2+ channel
and S(+)-3-isobutyl-gamma-amniobutyric acid (Pregabalin,
blockers [86].
PGL) (Fig. 4A). GBP has proven to be effective against a
As described above, VGCCs are formed by a pore wide range of seizure models in animals and clinically useful
forming α1 subunit linked with several accessory subunits. for the treatment of partial seizures. It has been approved as
As with the VGSC these accessory subunits are not required an add-on therapy in the U.K. since 1992 and the U.S. since
for the formation of functional ion conducting pores but are 1993. GBP was designed to be a CNS penetrating GABA
essential for conferring correct gating kinetics and cell analog but was later found to have no effect at the GABA
surface expression of the ion channel. The VGCC β subunits receptors, enzymes or transporters.
(β1-4) are 53-65kDa proteins found within the intracellular
compartment which induce large changes to the voltage A putative binding site for GBP was first identified on
dependence of activation and the rate of inactivation. the α2δ subunit of HVA VGCCs using [3H]-gabapentin on
purified porcine brain [92]. The α2 and δ components were
VGCCs also include an α2δ (α2δ 1-4) subunit which are 123-
found to both be required for binding of GBP although the
129kDa dimers that are linked to the α1 subunit by a
dispulphide link. The α2 component is a membrane spanning disulphide link between the two did not necessarily need to
protein whilst the δ component is found extracellularly (Fig. be intact. Binding of GBP to these subunits also appears to
be subunit specific with higher affinity binding to α2δ-1 than
4B). The α2δ subunit is also thought to modulate channel
α2δ-2 and no binding at α2δ-3 and α2δ-4. This binding
gating kinetics and regulate levels of expression but to a
lesser degree than the β subunit [80]. Furthermore several evidence is supported by electrophysiological data in a
25-36kDa γ (γ1-5) subunits have been identified but the number of different preparations that show inhibition of
calcium currents. Several studies report inhibition of HVA
expression and functional status of this subunit is still
calcium channels in dissociated rat brain neurons and dorsal
relatively unknown.
root ganglion cells [93,94], reduction of Ca2+ influx into
There is considerable evidence available (reviewed synaptosomes through P/Q-type channels and decreased
recently by Jones [87]) concerning the role of VGCC in glutamate release [95]. Recent experiments by Bayer and
epileptogenesis although there are few available AEDs that colleagues have shown that GBT selectively blocks P/Q-type
target the VGCC to exert their anticonvulsant effects. In Ca2+ channels in dorsal horn neurons (Fig. 5). The authors
particular genetic mutations in human calcium channel genes suggest that this may result from interaction of the GBP
(table 1) or those of several mouse species have been binding α2δ-1 or α2δ-2 subunits with the α1A pore forming
24 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

Fig. (5). (A) Postsynaptic responses to exogenously applied glutamate (1mM) and glycine (1mM) to whole cell patch clamped dorsal root
neurons. Gabapentin (1µM) does not significantly effect the amplitude or duration of either the evoked excitatory or inhibitory currents. (B)
AMPA-EPSCs and GLY-IPSCs evoked by extracellular stimulation are significantly attenuated by gabapentin (1µM) when N-type channels
are blocked but not when P/Q-type channels are blocked. This demonstrates gabapentin is able to preferentially block P/Q-type calcium
channels in sensory neurons (from ref. [95]).

subunit of the P/Q-type channels [96]. Although other of K+ ions into the cell rather than outward (Kir); and the
potential mechanisms have been proposed for GBP including recently discovered twin pore K channels (KCNK, TWIK,
enhancement of the GABA synthesizing enzyme, glutamic TRAAK and related channels) that appear to generate
acid decarboxylase (GAD), it is now generally accepted that neuronal resting membrane potential. The largest of these
actions of GBP and that of its follow-up compound PGL are “superfamilies”, and also the first to be discovered, are the
at least in part mediated by interaction with α2δ−1/2 subunits channels that give rise to IKv (and IKA) currents. The Kv
of VGCCs. The clinically important AEDs LTG and TPM channel sub-units are the best characterised of the K+
have also shown some activity against VGCCs although channels in terms of both structure and function. The first of
modulation of VGSC is widely accepted as their primary the K v channels to be cloned were those from the Drosophila
mode of action. Patch clamp experiments have shown that fruit fly voltage gated K+ channel genes Shaker, Shal, Shab
TPM is able to produce inhibition of L-type VGCCs in and Shaw but since, a further 29 related members of the Kv
dentate granule cells [97] whilst in rat cortical neurons LTG family have been discovered. These have been divided into
produces concentration dependent reduction in N and P but eight gene families each having several members {KCNA
not L-type currents [98]. (Kv1.1-1.8); KCNB (Kv2.1-2.3); KCNC (Kv3.1-3.4); KCND
(Kv4.1-4.3); KCNF (Kv5.1); KCNG (Kv6.1-6.4); KCNS
STRUCTURE, FUNCTION AND ANTICONVULSANT (Kv9.1-9.3); KCNV (Kv8.1-8.3)} and can form homo or
PHARMACOLOGY OF VOLTAGE GATED heteromeric channels (Kv1, Kv2, Kv3 and Kv4) with other
POTASSIUM CHANNELS subunits within their own family.
Potassium channels that are gated by changes in As previously discussed, Kv channels, are typically
membrane potential represent a functionally and structurally formed by tetrameric assemblies of Kv α−subunits [99] each
diverse family of ion channels that are crucial for many key of which having six transmembrane spanning domains (S1-
cellular processes including setting resting membrane S6) much like a single sodium channel repeated motif (Fig.
potential, determining firing threshold, action potential 6C). The S4 transmembrane spanning segment provides the
repolarization [14] and determination of postsynaptic voltage sensing capacity [100] whilst the pore of the
excitability. They represent one of the greatest unexploited complete channel is formed by the re-entrant loop between
targets for future AED development: only a few S5-S6 (P-loop). The P-loop contains the structural motif
experimental compounds evidently target these channels as GYG which forms the basis of the selectivity filter [101] and
their primary mode of action. Several families of mammalian makes the channel selective for K+ ion conductance.
K+ channels exist, including those gated by a change in MacKinnon’s group has made a huge contribution to our
membrane potential (Kv); those which favor the movement knowledge of the fine molecular structure of voltage gated
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 25

Fig. (6). (A) Structures of anticonvulsant drugs acting at VGKCs. (B) Diagram showing the structure of potassium channel α subunits
including the voltage gated (Kv), KCNQ and the twin pore KCNK type channels. (C) Differences in subunit composition of complete ion
channels. Na+ and Ca2+ channels are homomers comprising a single α subunit whilst potassium channels are formed by heteromeric
assembly of four smaller α subunits.

K+ channels through X-ray crystallographic studies of Kv3 channels in cortical interneurons where it has been
related bacterial channels [102] correlated to the short duration action potentials that allow
very high firing frequencies. The significant role the delayed
Due to the diversity of K+ channels brought about by
rectifier channels play in controlling cellular excitability and
many gene families, splice variation and heteromeric
burst firing frequency makes them an ideal therapeutic target
assembly (see subunit structures in Fig 6C) the range of
that may be exploited for anticonvulsant benefit.
physiological properties and roles of these channels are
extremely diverse. The Kv channels can however be Levetiracetam [(S)-α-ethyl-2-oxo-pyrrolidine acetamide,
classified into three broad groups based upon their voltage LEV] (Fig 6A) is a pyrrolidine derivative that is a well-
dependence of activation and the extent to which they tolerated adjunctive therapy for refractory partial seizures in
inactivate. At resting membrane potential Kv channels reside adults. Despite its clinical success the mechanism of action
in a closed state and are activated in response to of LEV has remained elusive with GABAergic facilitation,
depolarization whereupon they allow K+ ions to flow from inhibition of Na+ /low voltage activated Ca2+ current and
the cellular interior resulting in hyperpolarization of inhibition of excitatory ionotropic conductances being ruled
membrane potential. The first functional class of K v channels out as potential candidates [103,104]. Complementing this
are the so-called low voltage activated channels (LVA, e.g. lack of activity at established molecular target sites for
Kv1.1, KCNA). These are non-inactivating channels that anticonvulsants in vitro, is the notable observation that LEV
produce sustained outward potassium currents in response to differs from existing anticonvulsants in showing lack of
relatively low changes in membrane potential away from rest activity against MES and PTZ induced seizures in vivo.
(~ -60mV). LVA channels activate and deactivate slowly and Recently however Madeja and colleagues [105] have
as such are often known as delayed rectifier channels. The postulated that the anticonvulsant properties of LEV may be
slow kinetic features associated with LVA channels means substantially contributed to by a reduction in the
they are important in determining spike frequency adaptation conductance of delayed rectifier K+ channels. The authors of
but have little effect upon single spikes or the duration or this study demonstrated using acutely isolated rat and guinea
rise time of the first spike within a burst. The second major pig CA1 neurones, and the whole cell patch clamp
subtype of the delayed rectifier family, are the high voltage technique, that a 26% reduction in the delayed rectifier K+
activated channels (HVA, e.g. Kv3, KCNC). HVA activate current is produced by 100µM LEV. This inhibition was
and deactivate more rapidly than LVA channels but demonstrated to be sufficient to produce marked impairment
activation only occurs in response to large depolarization, of repetitive action potential firing. This study implicates
such as that brought about by an action potential. HVA LEV as the first compound to have its primary mode of
channels are therefore mostly responsible for the repolariz- action as a blocker of delayed rectifier channels although no
ation phase of the action potential and allowing high distinction is made between the LVA and HVA types.
frequency neuronal firing. This is typified by expression of
26 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

The final major functional type of Kv channels are those Retigabine [(D-23129, N-(2-amino-4-(4-fluorobenzylamino)-
channels which are capable of inactivation and give rise to phenyl) carbamic acid ethyl ester, RTG] (Fig 6A) is
the so called transient outward K+ current or A currents. structurally unrelated to any currently marketed anticon-
These channels activate more rapidly than the delayed vulsant drug and has been shown to exert anticonvulsant
rectifier type but also rapidly and spontaneously deactivate effects in broad range of seizure models. It was first found
owing to a ‘ball and chain’ or slower C-type inactivation that RTG activated a K+ conductance in marginally
mechanism depending upon the subunit composition of the depolarized NG108-15 neuronal cells at concentrations
channel. There is no evidence to date that any existing approximating to therapeutic plasma levels [115]. Although
clinical or experimental drugs exploit modulation of A- it was unknown at the time of this study it was later observed
currents as their primary mode of action. However it has that the pharmacological profile of the RTG induced current
been reported that LTG and CBZ are able to potentiate was consistent with that of the M-current. It has since been
outward KA conductance at concentrations similar to those at confirmed in Chinese hamster ovary (CHO) cells transfected
which they exert sodium channel inhibition [106,107]. 4-AP with either KCNQ2 (homomer) or KCNQ2/KCNQ3
models owing to the occlusion of a primary target by the (heteromer) that RTG is indeed that first molecule in a new
convulsant toxin. class of KCNQ K+ channel opening drugs [116] (Fig. 7). The
direct interaction of Retigabine with the KCNQ channels that
The M-current (IM) is a slowly activating, non-
are responsible for BFNC may be able to overcome the small
inactivating and slowly deactivating current that was first
loss of function underlying this rare epileptic condition and
described in frog sympathetic ganglia by Brown and Adams
illustrates the importance of molecular biology and genetics
in 1980 [108]. The name is derived from the ability of to the development of therapeutic agents for the treatment of
muscarinic receptor stimulation to inhibit the K+ current and idiopathic syndromes.
this has been shown to be present in a number of CNS
neurons [109]. IM has an important role in determining A further important family of potassium channels has
neuronal excitability as it represents the only sustained been recognized only within the last eight to ten years. Leak
inhibitory current in the range of potentials around spike currents (resting or background conductances) have been
threshold. The slow activation and deactivation kinetics of I M described in physiology for many years but only in 1996 was
mean that it plays a significant part in controlling resting it discovered that these currents were carried by discrete
membrane potential, responsiveness to synaptic input and as molecular entities within the cell membrane. The first of
a braking mechanism on repetitive firing. The molecular these ion channel structures was cloned from the
identity of the M-current was unknown until it was neuromuscular junction of Drosophila and became known as
discovered that currents induced by heteromers of KCNK0. Later in the same year the first mammalian gene for
KCNQ2/KCNQ3 potassium channel subunits shared similar this type of channel subunit, KCNK1, was also discovered
pharmacological and biophysical properties to native IM although this was found to be physiologically non-
[110]. The KCNQ genes encode a growing family of K+ functional. Although the differences in structural and
channel products that are structurally related to the Kv functional properties associated with KCNK channels are too
channel α subunits and like the classical Kv channels are extensive to cover fully in this review (reviewed in detail by
gated by changes in membrane potential. They have six Goldstein et al. [117]) there is some interesting pharma-
transmembrane spanning domains, a single pore forming cology related to these channels that deserves attention.
loop that forms the selectivity filter and sequences of Briefly the KCNK family of K+ channels subunits differ in
charged amino acids in the S4 domain that probably that they have two pore forming or P domains and four
represent the voltage sensor (Fig. 6BC). Although it has not (rather than six) transmembrane spanning segments (Fig.
been shown directly, KCNQ subunits, of which there are five 6B). They are open at rest and are subject to modulation by a
known types (KCNQ1, KCNQ2, KCNQ3, KCNQ4 and number of physiologically pertinent agents including cyclic
KCNQ5), likely form homo or heterotetramers to produce nucleotides, noradrenaline, serotonin and fatty acids. This
functional channels [111]. The significance of these channels type of ligand modulation at KCNK channels is capable of
in the control of cellular excitability is demonstrated by the influencing channel gating and conductance properties and
observation that mutations of the KCNQ 2/3 channels result as such is predicted to contribute to control of membrane
in the rare condition of benign familial neonatal convulsions potential and cellular excitability.
(BFNC, table 1) [112,113]. BNFC mutations in either
KNCQ2 or KNCQ3 may only produce a reduction of The neuroprotective agent sipatrigine (STG), which is
chemically related to LTG, has been shown to potently
between 20-30% [114] in the current amplitude of the
inhibit at least two of the two pore domain channels (that are
expressed heteromeric channels but this is often sufficient to
known to be sensitive to arachidonic acid), namely KCNK2
induce tonic-clonic seizures in the neonate.
(also known as TREK-1) and KCNK4 (also known as
Owing to the clear link between, an albeit rare idiopathic TRAAK) when they are expressed in HEK293 cells [118]. If
form of epilepsy, KCNQ2/3 mutations and the correlation one assumes that the KCNK channels are the main targets
between these channel subunits and the M-current (that is so for the neuroprotective actions of STG it may be expected
crucial for determining cellular excitability), it would appear that inhibition of the leak conductance would result in
that K+ channel opening represents an ideal therapeutic depolarization and increased excitability/excitotoxicity.
target for novel anticonvulsant ligands. Indeed an However, if KCNK2/4 were preferentially expressed on
experimental anticonvulsant ligand already undergoing GABAergic interneurones, reduced net excitability may be
Phase II clinical trials has been shown to act, at least in part, produced by an increase in polysynaptic inhibitory tone.
through opening KCNQ2/KCNQ3 potassium channels. Alternatively chronic depolarization resulting from the
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 27

Fig. (7). The anticonvulsant agent retigabine is mechanistically novel. It produces (A) a concentration dependent increase in potassium
conductance through KNCQ2/KNCQ3 heteromers expressed in CHO cells by directly opening the channel. (B) Retigabine also shifts the
voltage dependence of activation to more negative potentials in a reversible fashion. This results in the cellular potential being taken further
from the threshold potential for spike firing (from ref. [115]).

decreased leak conductance may cause reduction in excitatory refractory patients: there is a surprisingly large incidence of
neurotransmitter release by depolarization mediated block of sudden death in epilepsy patients (SUDEP) and a clear
glutamatergic neurons. Although the anticonvulsant clinical need for new drugs. The market analysts appear to be
compound LTG produces only <10% block of this current at ignoring completely the obvious promise that such drugs
10µM [118] it is important to consider the other mechanisms hold in the massive pharmaceutical market of neuropathic
by which the anticonvulsant is known to act. A small pain. These chronic pain syndromes are notoriously difficult
reduction in the KCNK2/4 current produced by LTG to treat but licensed anticonvulsants (even those with overt
application may, on its own, produce only insignificant side effects in the form of sedation, impairment of hepatic
changes in excitability but the resulting depolarization would function and serious blood dyscrasias are often useful in the
undoubtedly recruit more inactivation of both sodium treatment of chronic pain). Gabapentin has become the gold-
channel and calcium channels. It is known that LTG binds standard in the treatment of conditions like post-herpetic and
potently to the inactivated conformation of the sodium trigeminal neuralgia. The NIH in the USA continue to fund
channel (as above) to reduce electrogenesis and also reduces drug discovery research in epilepsy and it seems likely that
excitatory neurotransmitter release that likely results from novel drugs in the future will exploit the superfamilies of
reduction in availability of calcium channels in the ion-selective pores reviewed here.
presynaptic terminal. In these terms, what looks like a
paradoxical excitant or pro-convulsant surrogate effect of the REFERENCES
drug may represent a synergistic pathway to promote its [1] Traub, R. D.; Borck, C.; Colling, S. B.; Jefferys, J. G. On the
potency in blocking epileptiform activity. This type of structure of ictal events in vitro. Epilepsia 1996, 37, 879-891.
structure-dependent, non-selective cross talk with other [2] Jefferys, J. G. R. Models and Mechanisms of Experimental
channels may explain the diverse pharmacological profiles of Epilepsies. Epilepsia 2003, 44, 44-50.
[3] Hodgkin, A. L.; Huxley, A. F. The dual effect of membrane
DPH, CBZ and LTG in the living brain (despite their potential on sodium conductance in the giant axon of Loligo. J.
commonality at the VGSCs). Physiol. (London) 1952, 116, 497-506.
[4] Hodgkin, A. L.; Huxley, A. F. The components of membrane
Clearly the functional diversity amongst the K+ channels conductance in the giant axon of Loligo. J. Physiol (London) 1952,
make them prospective targets for future AED development. 116, 473-496.
The production of selective modulators of the various K+ [5] Hodgkin, A. L.; Huxley, A. F. Currents carried by sodium and
channel types may serve to further relieve the symptoms of potassium ions through the membrane of the giant axon of Loligo.
J. Phsiol. (London) 1952, 116, 473-506.
those patients with refractory or idiopathic epilepsies whose [6] Catterall, W. A. Cellular and molecular biology of voltage-gated
conditions are poorly controlled by the existing compounds. Na+ channels. Physiol. Rev. 1992, 72, S15-S48.
[7] Catterall, W. A. Molecular properties of brain sodium channels: an
CONCLUSION important target for anticonvulsant drugs. Adv. Neurol. 1999, 79,
441-456.
To sum up we wish to stress that voltage-gated ion [8] Qu, Y.; Curtis, R.; Lawson, D.; Gilbride, K.; Ge, P.; DiStefano, P.
S.; Silos-Santiago, I.; Catterall, W. A.; Scheuer, T. Differential
channels have been crucial in the treatment of epilepsy (often modulation of sodium channel gating and persistant sodium
their mode of action emerged after serendipitous discovery currents by the β1, β2 and β3 subunits. Mol. Cell. Neurosci. 2001,
of their clinical promise). One by one the large multi- 18, 570-580.
national pharmaceutical companies are withdrawing from [9] Goldin, A. L.; Barchi, R. L.; Caldwell, J. H.; Hofmann, F.; Howe,
biorational drug discovery in the epilepsy field (because of J. R.; Hunter, J. C.; Kallen, R. G.; Mandel, G.; Meisler, M. H.;
Netter, Y. B.; Noda, M.; Tamkun, M. M.; Waxman, S. G.; Wood, J.
the prohibitive development costs and the long lead times). N.; Catterall, W. A. Nomenclature of voltage-gated sodium
This is clearly detrimental to the significant number of channels. Neuron 2000, 28, 365-368.
28 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

[10] Noda, M.; Ikeda, T.; Kayano, T.; Suzuki, H.; Takeshima, M.; [34] Brown, A. M.; Schwindt, P. C.; Crill, W. E. Different voltage
Kurasaki, H.; Takahashi, S.; Numa, S. Existence of distinct sodium dependence of transient and persistant Na+ currents is compatible
channel messenger RNAs in rat brain. Nature 1986, 320, 121-127. with modal gating hypothesis for sodium channels. J.
[11] Ragsdale, D. S.; Avoli, M. Sodium channels as molecular targets Neurophysiol. 1994, 71, 2562-2565.
for antiepileptic drugs. Brain Res. Rev. 1998, 26, 16-28. [35] Huguenard, J. R.; Hamill, O. P.; Prince, A. Developmental changes
[12] Catterall, W. A. A 3D view of sodium channels. Nature 2001, 409, in Na + conductance in rat cortical neurons: appearance of a slowly
988-991. inactivating component. J. Neurophysiol. 1988, 59, 778-795.
[13] Hille, B. Ion channels of excitable membranes; Sinauer Assoc.: [36] Crill, W. E. Persistant sodium current in mammalian central
Sunderland, MA., 1992. nervous system. Annu. Rev. Physiol. 1996, 58, 349-362.
[14] Hodgkin, A. L.; Huxley, A. F. A quantative description of [37] Stafstrom, C. E.; Schwindt, P. C.; Chubb, M. C.; Crill, W. E.
membrane current and its application to conduction and excitation Properties of persistant sodium conductance and calcium
in nerve. J Physiol (Lond) 1952, 117, 500-544. conductance of layer V neurons from cat sensorimotor coretx in
[15] Armstrong, C. M.; Bezanilla, F. Charge movement associated with vitro. J. Neurophysiol. 1985, 53, 152-170.
the opening and closing of the activation gates of Na channels. J. [38] Stafstrom, C. E.; Schwindt, P. C.; Crill, W. E. Negative slope
Gen. Physiol. 1974, 63, 533-552. conductance due to a persistant subthreshold sodium current in cat
[16] Armstrong, C. M.; Bezanilla, F. Currents related to the movement neocortical neurons in vitro. Brain Res. 1982, 236, 221-226.
of the gating particles of the sodium channels. Nature 1973, 242, [39] Cantrell, A. R.; Catterall, W. A. Neuromodulation of Na+ channels:
459-461. an unexpected form of cellular plasticity. Nat. Rev. Neurosci. 2001,
[17] Stümer, W.; Conti, F.; Suzuki, X.; Wang, M.; Noda, M.; Yahadi, 2, 397-407.
N.; Kubo, H.; Numa, S. Structural parts involved in activation and [40] Murphy, B. J.; Rossie, S.; DeJongh, K. S.; Catterall, W. A.
inactivation of the voltage gated sodium channel. Nature 1989, Identification of sites of selective phosphorylation and
339, 597-603. dephosphorylation of the rat brain sodium channel α subunit by
[18] Patton, D. E.; West, J. W.; Catterall, W. A.; Goldin, A. L. Amino cAMP dependent protein kinase and phosphoprotein phosphatases.
acid residues required for fast Na+ channel inactivation: charge J. Biol. Chem. 1993, 268, 27355-27362.
neutralizations and deletions in the III-IV linker. Proc. Nat. Acad. [41] Smith, R. D. Goldin, A. L. Phosphorylation of brain sodium
Sci. U. S. A. 1992, 89, 10905-10909. channels in the I-II linker modulates function in Xenopus oocytes.
[19] Vassilev, P. M.; Scheuer, T.; Catterall, W. A. Identification of an J. Neurosci. 1996, 16, 1965-1974.
intracellular peptide segment involved in sodium channel [42] Li, M.; West, J. W.; Lai, Y.; Scheuer, T.; Catterall, W. A.
inactivation. Science 1988, 241, 1658-1661. Functional modulation of brain sodium channels by cAMP-
[20] West, J. W.; Patton, D. E.; Scheuer, T.; Wang, Y.; Goldin, A. L.; dependent phosphorylation. Neuron 1992, 8, 1151-1159.
Catterall, W. A. A cluster of hydrophobic amino acid residues [43] Alroy, G.; Su, H.; Yaari, Y. Protein kinase C mediates muscarinic
required for fast Na+ channel inactivation. Proc. Nat. Acad. Sci. block of intrinsic bursting in rat hippocampal neurons. J. Physiol.
U. S. A. 1992, 89, 10910-10914. 1999, 518, 71-79.
[21] McPhee, J. C.; Ragsdale, D. S.; Scheuer, T.; Catterall, W. A. A [44] Azouz, R.; Jensen, M. S.; Yaari, Y. Muscarinic modulation of of
critical role for transmembrane segment IVS6 of the sodium intrinsic burst firingi in rat hippocampal neurons. Eur. J. Neurosci.
channel α subunit in fast inactivation. J. Biol. Chem. 1995, 270, 1994, 6, 961-966.
12025-12034. [45] Merritt, H. H.; Putnam, T. J. Sodium diphenylhydantoinate in the
[22] Goldin, A. L. Mechanisms of sodium channel inactivation. Curr. treatment of convulsive disorders. J. Am. Med. Assoc. 1938, 111,
Opin. Neurobiol. 2003, 13, 284-290. 1068-1073.
[23] Traub, R. D.; Jefferys, G. R.; Whittington, M. A. Fast Oscillations [46] Brodie, M. J.; Dichter, M. A. Antiepileptic drugs. N. Engl. J. Med.
in Cortical Circuits.; The MIT Press: Cambridge, MA, 1999. 1996, 334, 168-175.
[24] Featherstone, I. A.; Richmond, J. E.; Ruben, P. C. Interaction [47] Toman, J. E. P. The neuropharmacology of antiepileptics.
between fast and slow inactivation in Skm1 sodium channels. Electroencephalogr. Clin. Neurophysiol. 1949, 1, 33-44.
Biophys. J. 1996, 71, 3098-3109. [48] McLean, M. J.; McDonald, R. L. Multiple actions of phenytoin on
[25] Richmond, J. E.; Featherstone, I. A.; Hartmann, H. A.; Ruben, P. mouse spinal cord neurons in cell culture. J. Pharmacol. Exp. Ther.
C. Slow inactivation in human cardiac sodium channels. Biophys. 1986, 237, 779-789.
J. 1998, 74, 2945-2952. [49] Willow, M.; Gonoi, T.; Catterall, W. A. Voltage clamp analysis of
[26] Fleidervish, I. A.; Friedman, A.; Gutnick, M. J. Slow inactivation the inhibitory actions of diphenylhydantoin and carbamazepine on
of Na+ current and slow cumulative spike adaptation in mouse and voltage-sensitive sodium channels in neuroblastoma cells. Mol.
guinea pig neocortical neurones in slices. J. Physiol. 1996, 493, 83- Pharmacol. 1985, 27, 549-558.
97. [50] Matsuki, N.; Quandt, F. N.; Ten Eick, R. E.; Yeh, J. Z.
[27] Toib, A.; Lyakhov, V.; Marom, S. Interaction between duration of Characterization of the block of sodium channels by phenytoin in
activity and time course of recovery from slow inactivation in mouse neuroblastoma cells. J. Pharmacol. Exp. Ther. 1984, 228,
mammalian brain Na+ channels. J. Neurosci. 1998, 18, 1893-1903. 523-530.
[28] Avoli, M.; Barbarosie, M.; Lucke, A.; Nagao, T.; Lopantsev, V.; [51] Taylor, C. P.; Meldrum, B. S. Na+ channels as targets for
Kohling, R. Synchronous GABA-mediated potentials and neuroprotective drugs. Trends Pharmacol. Sci. 1995, 16, 309-315.
epileptiform discharges in the rat limbic system in vitro. J. [52] Lang, D. G.; Wang, C. M.; Cooper, B. R. Lamotrigine, Phenytoin
Neurosci. 1996, 16, 3912-3924. and Carbamazepine interactions on the sodium current present in
[29] Lopantsev, V.; Avoli, M. Participation of GABAA-mediated N4TG1 mouse neuroblastoma cells. J. Pharmacol. Exp. Ther.
inhibition in ictal-like discharges in the rat entorhinal cortex. J. 1993, 266, 829-834.
Neurophysiol. 1998, 79, 352-360. [53] Chao, T. I.; Alzheimer, C. Effects of phenytoin on the persistent
[30] Avoli, M.; D'Antuono, M.; Louvel, J.; Kohling, R.; Biagini, G.; Na+ current of mammalian CNS neurones. Neuroreport 1995, 6,
Pumain, R.; D'Arcangelo, G.; Tancredi, V. Network and 1778-1780.
pharmacological mechanisms leading to epileptiform [54] Lampl, I.; Schwindt, P.; Crill, W. Reduction of Cortical Pyramidal
synchronization in the limbic system in vitro. Prog. Neurobiol. Neuron Excitability by the Action of Phenytoin on Persistent Na+
2002, 68, 167-207. Current. J. Pharmacol. Exp. Ther. 1998, 284, 228-237.
[31] Dougalis, A.; Lees, G.; Ganellin, C. R. The sleep lipid oleamide [55] Spadoni, F.; Hainsworth, A. H.; Mercuri, N. B.; Caputi, L.;
may represent and endogenous anticonvulsant: an in vitro Martella, G.; Lavaroni, F.; Bernardi, G.; Stefani, A. Lamotrigine
comparative study in the 4-aminopyridine rat brain slice model. derivatives and riluzole inhibit I NaP in cortical neurons. Neuroreport
Neuropharmacology 2003. 2002, 13, 1167-1170.
[32] Taylor, C. P. Na+ currents that fail to inactivate. Trends Neurosci. [56] Kuo, C. C. A Common Anticonvulsant Binding Site for Phenytoin,
1993, 16, 455-460. Carbamazepine, and Lamotrigine in Neuronal Na+ Channels. Mol.
[33] Alzheimer, C.; Schwindt, P. C.; Crill, W. E. Modal gating of Na+ Pharmacol. 1998, 54, 712-721.
channels as a mechanism of peristant Na+ current in pyramidal [57] Krall, R. L.; Penry, J. K.; White, B. G.; Kupferberg, H. J.;
neurons from rat and cat sensorimotor coretx. J. Neurosci. 1993, Swinyard, E. A. Anti-epileptic drug development. II.
13, 660-673. Anticonvulsant drug screening. Epilepsia 1978, 19, 404-428.
Voltage Gated Ion Channels: Targets for Anticonvulsant Drugs Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 29

[58] Meldrum, B. Do preclinical seizure models preselect certain mechanism of action of PNU-151774E, a novel antiepileptic
adverse effects of antiepileptic drugs. Epilepsy Res. 2002, 50, 33- compound. J. Pharmacol. Exp. Ther. 1999, 288, 1151-1159.
40. [80] Catterall, W. A. Structure and regulation of voltage-gated Ca2+
[59] McLean, M. J.; McDonald, R. L. Carbamazepine and 10,11- channels. Annu. Rev. Cell. Dev. Biol. 2000, 16, 521-555.
epoxycarbamazepine produce use and voltage dependent limitation [81] Ertel, E.; Campbell, K. P.; Harpold, M. M.; Hofmann, F.; Mori, Y.
of rapidly firing action potentials in mouse central neurons in cell Nomenclature of voltage-gated calcium channels. Neuron 2000, 25,
culture. J. Pharmacol. Exp. Ther. 1986, 238, 727-738. 533-535.
[60] Kuo, C. C.; Bean, B. P.; Chen, L. L.; Chen, R. C. Carbamzepine [82] Nowycky, M. C.; Fox, A. P.; Tsien, R. W. Three types of neuronal
inhibition of neuronal Na+ currents: quantitative distinction from calcium channel with different calcium agonist sensitivity. Nature
phenytoin and possible clinical implications. Mol. Pharmacol. 1985, 316, 440-443.
1977, 51, 1077-1083. [83] Reuter, H. Calcium channel modulation by neurotransmitters,
[61] Tecoma, E. S. Oxcarbazepine. Epilepsia 1999, 40, S37-S46. enzymes and drugs. Nature 1983, 301, 569-574.
[62] Bonifácio, M. J.; Sheridan, R. D.; Parada, A.; Cunha, R. A.; [84] Llinás, R. Yarom, Y. Electrophysiology of mammalian inferior
Patmore, L.; Soares-da-Silva, P. Interaction of the novel olive neurones in vitro. Different types of voltage dependent ionic
anticonvulsant, BIA 2-093, with voltage gated sodium channels: conductances. J. Physiol. 1981, 315, 569-584.
comparison with carbamazepine. Epilepsia 2001, 42, 600-608. [85] McCleskey, E. W.; Fox, A. P.; Feldman, D. H.; Cruz, L. J.;
[63] Leach, M. J.; Baxter, M. G.; Critchley, M. A. E. Neurochemical Olivera, B. M.; Tsien, R. W.; Yoshikami, D. ω-Conotoxin: direct
and behavioural aspects of Lamotrigine. Epilepsia 1991, 32, S4-S8. and persistant blockade of specific calcium channels in nuerons but
[64] Leach, M. J.; Marden, C. M.; Miller, A. A. Pharmacological studies not muscle. Proc. Nat. Acad. Sci. U. S. A. 1987, 84, 4327-4331.
on Lamotrigine, a novel potential antiepileptic drug. II. [86] Randall, A.; Tsien, R. W. Pharmacological dissection of multiple
Neurochemical studies on the mechanism of action. Epilepsia types of Ca2+ channel currents in rat cerebellar granule neurons. J.
1986, 27, 490-497. Neurosci. 1995, 15, 2995-3012.
[65] Miller, A. A.; Wheatly, P.; Sawyer, D. A.; Baxter, M. G.; Roth, B. [87] Jones, O. T. Ca2+ channels and epilepsy. Eur. J.Pharmacol. 2002,
Pharmacological studies on lamotrigine, a novel potential 447, 211-225.
antiepiletic drug: I. Anticonvulsant profile in mice and rats. [88] Chen, L.; El Husseini, A.; Tomita, S.; Bredt, D. S.; Nicoll, R. A.
Epilepsia 1986, 27, 483-489. Stargazin Differentially Controls the Trafficking of {alpha}-
[66] Hamill, O. P.; Marty, A.; Neher, E.; Sakmann, B.; Sigworth, F. J. Amino-3-hydroxyl-5-methyl-4-isoxazolepropionate and Kainate
Improved patch clamp techniques for high-resolution current Receptors. Mol. Pharmacol. 2003, 64, 703-706.
recordings from cells and cell free memebrane patches. Pflug. [89] Coulter, D. A.; Huguenard, J. R.; Prince, D. A. Specific petit mal
Arch. 1981, 391, 85-100. anticonvulsants reduce calcium currents in thalamic neurons.
[67] Lees, G.; Leach, M. J. Studies on the mechanism of action of the Neurosci. Lett. 1989, 98, 74-78.
novel anticonvulsant lamotrigine (Lamictal) using primary [90] Coulter, D. A.; Huguenard, J. R.; Prince, D. A. Differential effects
neurological cultures from rat cortex. Brain Research 1993, 612, of petit mal anticonvulsants and convulsants on thalamic neurons:
190-199. calcium current reduction. Br. J. Pharmacol. 1990, 100, 800-806.
[68] Wang, C. M.; Lang, D. G.; Cooper, B. R. Lamotrigine effects on [91] Gomora, J. C.; Daud, A. N.; Weiergraber, M.; Perez-Reyes, E.
ion channels in neuronal cells. Epilepsia 1993, 34, 117-118. Block of cloned human T-type calcium channels by succinimide
[69] Ragsdale, D. S.; McPhee, J. C.; Scheuer, T.; Catterall, W. A. antiepileptic drugs. Mol. Pharmacol. 2001, 60, 1121-1132.
Molecular determinants of state-dependent block of Na channels by [92] Gee, N. S.; Brown, J. P.; Dissanayake, V. U.; Offord, J.; Thurlow,
local anesthetics. Science 1994, 265, 1724-1728. R.; Woodruff, G. N. The novel anticonvulsant drug, gabapentin
[70] Ragsdale, D. S.; McPhee, J. C.; Scheuer, T.; Catterall, W. A. (neurontin), binds to the alpha2delta subunit of a calcium channel.
Common molecular determinants of local anesthetic, J. Biol. Chem. 1996, 271, 768-776.
antiarrhythmic and anticonvulsant block of voltage gated Na [93] Sutton, K. G.; Martin, D. J.; Pinnock, R. D.; Lee, K.; Scott, R. H.
channels. Proc. Nat. Acad. Sci. U. S. A. 1996, 93, 9270-9275. Gabapentin inhibits high-threshold calcium channel currents in
[71] Shank, R. P.; Gardocki, J. F.; Vaught, J. L.; Davis, C. B.; cultured rat dorsal root ganglion neurones. Br. J. Pharmacol. 2002,
Schupsky, J. J.; Raffa, R. B.; Dodgson, S. J.; Nortey, S. O.; 135, 257-265.
Maryanoff, B. E. Topiramate: preclinical evaluation of a [94] Stefani, A.; Spadoni, F.; Bernardi, G. Gabapentin inhibits calcium
structurally novel anticonvulsant. Epilepsia 1994, 35, 450-460. currents in isolated rat brain neurons. Neuropharmacology 1998,
[72] Gibbs, J. W. III; Sombati, S.; DeLorenzo, R. J.; Coulter, D. A. 37, 83-91.
Cellular actions of topiramate: blockade of kainate-evoked inward [95] Fink, K.; Dooley, D. J.; Meder, W. P.; Suman-Chauhan, N.; Duffy,
currents in cultured hippocampal neurons. Epilepsia 2000, 41, S10- S.; Clussman, H.; Göthert, M. Inhibition of neuronal Ca2+ influx by
S16. gabapentin and pregabalin in the human neocortex.
[73] Brown, S. D.; Wolf, H. H.; Swinyard, E. A.; Twyman, R. E.; Neuropharmacology 2001, 42, 229-236.
White, H. S. The novel anticonvulsant topiramate enhances [96] Bayer, K.; Ahmadi, S.; Zeilhofer, H. U. Gabapentin may inhibit
GABA-mediated chloride flux. Epilepsia 1993, 34, 123-124. synaptic transmission in the mouse spinal cord dorsal horn through
[74] DeLorenzo, R. J.; Sombati, S.; Coulter, D. A. Effects of topiramate a preferential block of P/Q-type Ca2+ channels.
on sustained repetitive firing and spontaneous recurrent seizure Neuropharmacology 2004, 46, 743-749.
discharges in cultured hippocampal neurons. Epilepsia 2000, 41, [97] Zhang, X.; Velumian, A. A.; Jones, O. T.; Carlen, P. L. Modulation
S40-S44. of high-voltage activated calcium channels in dentate granule cells
[75] Zona, C.; Ciotti, M. T.; Avoli, M. Topiramate attenuates voltage- by topiramate. Epilepsia 2000, 41, S52-S60.
gated sodium currents in rat cerebellar granule cells. Neurosci. Lett. [98] Stefani, A.; Spadoni, F.; Siniscalchi, A.; Benardi, G. Lamotrigine
1997, 231, 123-126. inhibits Ca2+ currents in cortical neurons: functional implications.
[76] Taverna, S.; Sancini, G.; Mantegazza, M.; Franceschetti, S.; Eur. J. Pharmacol. 1996, 307, 113-116.
Avanzini, G. Inhibition of transient and persistent Na+ current [99] MacKinnon, R. Determination of the subunit stoichiometry of a
fractions by the new anticonvulsant topiramate. J. Pharmacol. Exp. voltage-activated potassium channel. Nature 1991, 350, 232-235.
Ther. 1999, 288, 960-968. [100] Bezanilla, F. The voltage sensor in voltage-dependent ion channels.
[77] Draguhn, A.; Jungclaus, M.; Sokolowa, S.; Heinemann, U. Physiol. Rev. 2000, 80, 555-592.
Losigamone decreases spontaneous synaptic activity in cultured [101] Yellen, G. The voltage gated potassium channels and their
hippocampal neurons. Eur. J. Pharmacol. 1997, 325, 245-251. relatives. Nature 2002, 419, 35-42.
[78] Gebhardt, C.; Breustedt, J. M.; Noldner, M.; Chatterjee, S. S.; [102] MacKinnon, R. Potassium channels. FEBS Lett. 2003, 555, 62-65.
Heinemann, U. The antiepileptic drug losigamone decreases the [103] Zona, C.; Marchetti, C.; Margineau, D. G. The novel antiepileptic
persistent Na+ current in rat hippocampal neurons. Brain Res. drug candidate levetiracetam does not modify the biophysical
2001, 920, 27-31. porperties of the Na+ channel in rat cortical neurons in culture.
[79] Salvati, P.; Maj, R.; Caccia, C.; Cervini, M. A.; Fornaretto, M. G.; Society for neuroscience abstracts 1999, 25, 1868.
Lamberti, E.; Pevarello, P.; Skeen, G. A.; White, H. S.; Wolf, H.H.; [104] Zona, C.; Niespodziany, I.; Marchetti, C.; Klitgaard, H.; Bernardi,
Faravelli, L. Biochemical and electrophysiological studies on the G.; Margineanu, D. G. Levetiracetam does not modulate neuronal
30 Current Topics in Medicinal Chemistry, 2005, Vol. 5, No. 1 Lees et al.

voltage-gated Na+and T-type Ca2+currents. Seizure 2001, 10, 279- of KCNQ3 (c.925T-->C) in a Japanese family with benign familial
286. neonatal convulsions. Ann. Neurol. 2000, 47, 822-826.
[105] Madeja, M.; Georg Margineanu, D.; Gorji, A.; Siep, E.; Boerrigter, [121] Vijai, J.; Kapoor, A.; Ravishankar, H. M.; Cherian, P. J.; Girija, A.
P.; Klitgaard, H.; Speckmann, E. J. Reduction of voltage-operated S.; Rajendran, B.; Rangan, G.; Jayalakshmi, S.; Mohandas, S.;
potassium currents by levetiracetam: a novel antiepileptic Radhakrishnan et, a. Genetic association analysis of KCNQ3 and
mechanism of action? Neuropharmacology 2003, 45, 661-671. juvenile myoclonic epilepsy in a South Indian population. Human
[106] Zona, C.; Tancredi, V.; Longone, P.; D'Arcangelo, G.; D'Antuono, Genetics 2003, 113, 461-463.
M.; Manfredi, M.; Avoli, M. Neocortical potassium currents are [122] Wallace, R. H.; Scheffer, I. E.; Barnett, S.; Richards, M.; Dibbens,
enhanced by the antiepileptic drug lamotrigine. Epilepsia 2002, 43, L.; Desai, R. R.; Lerman-Sagie, T.; Lev, D.; Mazarib, A.; Brand,
685-690. N.; Ben-Zeev, B.; Goikhman, I.; Singh, R.; Kremmidiotis, G.;
[107] Zona, C.; Tancredi, V.; Palma, E.; Pirrone, G. C.; Avoli, M. Gardner, A.; Sutherland, G. R.; George, A. L. J.; Mulley, J. C.;
Potassium currents in rat cortical neurons in culture are enhanced Berkovic, S. F. Neuonal sodium-channel alpha1-subunit mutations
by the antiepileptic drug carbamazepine. Can. J. Physiol. in generalized epilepsy with febrile seizures plus. Am. J. Hum.
Pharmacol. 1990, 68, 545-547. Genet. 2001, 68, 859-865.
[108] Brown, D. A.; Adams, P. R. Muscarinic supression of a novel [123] Spampanato, J.; Escayg, A.; Meisler, M. H.; Goldin, A. L.
voltage-sensitive K+ current in a vertebrate neuron. Nature 1980, Functional effects of two voltage gated sodium channel mutations
283, 673-676. that cause generalized epilepsy with febrile seizures plus type 2. J.
[109] Halliwell, J. V.; Adams, P. R. Voltage clamp analysis of Neurosci. 2001, 21, 7481-7490.
muscarinic excitation in hippocampal neurons. Brain Res. 1982, [124] Ito, M.; Nagafuji, H.; Okazawa, H.; Yamakawa, K.; Sugawara, T.;
250, 71-92. Mazaki-Miyazaki, E.; Hirose, S.; Fukama, G.; Mitsudome, A.;
[110] Wang, H. S.; Pan, Z.; Shi, W.; Brown, B. S.; Wymore, R. S.; Wada, K.; Kaneko, S. Autosomal dominant epilepsy with febrile
Cohen, I. S.; Dixon, J. E.; McKinnon, D. KNCQ2 and KNCQ3 seizures plus with missense mutations of the (Na+)-channel alpha 1
potassium channels subunits: molecular correlates of the M- subunit gene, SCN1A. Epilepsy Res. 2002, 48, 15-23.
channel. Science 1998, 282, 1890-1893. [125] Ceulemans, B. P.; Claes, L. R.; Lagae, L. G. Clinical correlations
[111] Robbins, J. KCNQ potassium channels: physiology, of mutations in the SCN1A gene: from febrile seizures to severe
pathophysiology, and pharmacology. Pharmacol. Ther. 2001, 90, myoclonic epilepsy in infancy. Pediatr. Neurol. 2004, 30, 236-243.
1-19. [126] Sugawara, T.; Tsurubuchi, Y.; Fujiwara, T.; Mazaki-Miyazaki, E.;
[112] Charlier, C.; Singh, N. A.; Ryan, S. G.; Lewis, T. B.; Reus, B. E.; Nagata, K.; Montal, M.; Inoue, T.; Yamakawa, K. Nav1.1 channels
Leach, R. J.; Leppert, M. A pore mutation in an idiopathic epilepsy with mutations of severe myoclonic epilepsy in infancy display
family. Nat. Genet. 1998, 18, 53-55. attenuated currents. Epilepsy Res. 2003, 54, 201-207.
[113] Biervert, C.; Schroeder, B. C.; Kubisch, C.; Berkovic, S. F.; [127] Sugawara, T.; Tsurubuchi, Y.; Agarwala, K. L.; Ito, M.; Fukuma,
Propping, P.; Jentsch, T. J.; Steinlein, O. K. A potassium channel G.; Mazaki-Miyazaki, E.; Nagafuji, H.; Noda, M.; Imoto, K.;
mutation in neonatal human epilepsy. Science 1998, 279, 403-406. Wada, K.; Mitsudome, A.; Kaneko, S.; Montal, M.; Nagata, K.;
[114] Schroeder, B. C.; Kubisch, C.; Stein, V.; Jensch, T. J. Moderate Hirose, S.; Yamakawa, K. A missense mutation of the Na+ channel
loss of function of cyclic-AMP-modulated KNCQ2/KNCQ3 K+ alpha II subunit gene Na(v)1.2 in a patient with febrile and afebrile
channels causes epilepsy. Nature 1998, 396, 687-690. seizures causes channel dysfunction. Proc. Nat. Acad. Sci. U. S. A.
[115] Rundfeldt, C. The new anticonvulsant retigabine (D-23129) acts as 2001, 98, 6384-6389.
an opener of K+ channels in neuronal cells. Eur. J. Pharmacol. [128] Berkovic, S. F.; Heron, S. E.; Giordano, L.; Marini, C.; Guerrini,
1997, 336, 243-249. R.; Kaplan, R. E.; Gambardella, A.; Steinlein, O. K.; Grinton, B.
[116] Rundfeldt, C.; Netzer, R. The novel anticonvulsant retigabine E.; Dean, J.T.; Bordo, L. Hodgson, B.L.; Yamamoto, T.; Mulley,
activates M-currents in Chinese hamster ovary-cells tranfected with J.C.; Zara, F.; Scheffer, I.E. Benign familial neonatal-infantile
human KCNQ2/3 subunits. Neurosci. Lett. 2000, 282, 73-76. seizures: characterization of a new sodium channelopathy. Ann.
[117] Goldstein, S. A. N.; Bockenhauer, D.; O'Kelly, I.; Zilberberg, N. Neurol. 2004, 55, 550-557.
Potassium leak channels and the KNCK family of two-P-domain [129] Wallace, R. H.; Scheffer, I. E.; Parasivam, G.; Barnett, S.; Wallace,
subunits. Nat. Rev. Neurosci. 2001, 2, 175-184. G. B.; Sutherland, G. R.; Berkovic, S. F.; Mulley, J. C. Generalized
[118] Meadows, H. J.; Chapman, C. G.; Duckworth, D. M.; Kelsell, R. epilepsy with febrile seizures plus: mutation of the sodium channel
E.; Murdock, P. R.; Nasir, S.; Rennie, G.; Randall, A. D. The subunit SCN1B. Neurology 2002, 58, 1426-1429.
neuroprotective agent sipatrigine (BW619C89) potently inhibits the [130] Chioza, B.; Osei-Lah, A.; Nashef, L.; Suarez-Merino, B.; Wilkie,
human tandem pore-domain K+ channels TREK-1 and TRAAK. H.; Sham, P.; Knight, J.; Asherson, P.; Makoff, A. J. Haplotype and
Brain Res. 2001, 892, 94-101. linkage disequilibrium analysis to characterise a region in the
[119] Singh, N. A.; Westenskow, P.; Charlier, C.; Pappas, C.; Leslie, J.; calcium channel gene CACNA1A associated with idiopathic
Dillon, J.; Anderson, V. E.; Sanguinetti, M. C.; Leppert, M. F. generalised epilepsy. Eur. J. Hum. Genet.: EJHG 2002, 10, 857-
KCNQ2 and KCNQ3 potassium channel genes in benign familial 864.
neonatal convulssions: expansion of the functional mutation [131] Khosravani, H.; Altier, C.; Simms, B.; Hamming, K. S.; Snutch, T.
spectrum. Brain 2003, 126, 2726-2737. P.; Mezeyova, J.; McRory, J. E.; Zamponi, G. W. Gating effects of
[120] Hirose, S.; Zenri, F.; Akiyoshi, H.; Fukama, G.; Iwata, H.; Inoue, mutations in the Cav3.2 T-type calcium channel associated with
T.; Yonetani, M.; Tsutsumi, M.; Muranaka, H.; Kurokawa, T.; childhood absence epilepsy. J. Biol. Chem. 2004, 279, 9681-9684.
Hanai, T.; Wada, K.; Kaneko, S.; Mitsudome, A. A novel mutation

S-ar putea să vă placă și