A summary of mechanistic hypotheses of gabapentin pharmacology

Page created by Carmen Juarez
 
CONTINUE READING
Epilepsy Research 29 (1998) 233 – 249

             A summary of mechanistic hypotheses of gabapentin
                             pharmacology

            Charles P. Taylor a,*, Nicolas S. Gee d, Ti-Zhi Su b, Jeffery D. Kocsis e,
            Devin F. Welty c, Jason P. Brown d, David J. Dooley a, Philip Boden d,
                                        Lakhbir Singh d
a
  Department of Neuroscience Therapeutics, Parke – Da!is Pharmaceutical Research, Di!ision of Warner– Lambert Co., Ann Arbor,
                                                        MI 48105, USA
     b
       Department of Molecular Biology, Parke–Da!is Pharmaceutical Research, Di!ision of Warner–Lambert Co., Ann Arbor,
                                                        MI 48105, USA
c
  Department of Pharmacokinetics and Drug Metabolism, Parke– Da!is Pharmaceutical Research, Di!ision of Warner– Lambert Co.,
                                                   Ann Arbor, MI 48105, USA
    d
      Parke – Da!is Neuroscience Research Centre, Cambridge Uni!ersity For!ie Site, Robinson Way, Cambridge, CB2 2QB, UK
        e
          Neuroscience and Regeneration Research Center A127A, Veterans Affairs Medical Center, Building 34, Room 123,
                                        950 Campbell A!e., West Ha!en, CT 06516, USA

                    Received 28 July 1997; received in revised form 1 October 1997; accepted 8 October 1997

Abstract

   Although the cellular mechanisms of pharmacological actions of gabapentin (Neurontin®) remain incompletely
described, several hypotheses have been proposed. It is possible that different mechanisms account for anticonvulsant,
antinociceptive, anxiolytic and neuroprotective activity in animal models. Gabapentin is an amino acid, with a
mechanism that differs from those of other anticonvulsant drugs such as phenytoin, carbamazepine or valproate.
Radiotracer studies with [14C]gabapentin suggest that gabapentin is rapidly accessible to brain cell cytosol. Several
hypotheses of cellular mechanisms have been proposed to explain the pharmacology of gabapentin: 1. Gabapentin
crosses several membrane barriers in the body via a specific amino acid transporter (system L) and competes with
leucine, isoleucine, valine and phenylalanine for transport. 2. Gabapentin increases the concentration and probably
the rate of synthesis of GABA in brain, which may enhance non-vesicular GABA release during seizures. 3.
Gabapentin binds with high affinity to a novel binding site in brain tissues that is associated with an auxiliary subunit
of voltage-sensitive Ca2 + channels. Recent electrophysiology results suggest that gabapentin may modulate certain
types of Ca2 + current. 4. Gabapentin reduces the release of several monoamine neurotransmitters. 5. Electrophysiol-
ogy suggests that gabapentin inhibits voltage-activated Na + channels, but other results contradict these findings. 6.
Gabapentin increases serotonin concentrations in human whole blood, which may be relevant to neurobehavioral

    * Corresponding author.

0920-1211/98/$19.00 © 1998 Elsevier Science B.V. All rights reserved.
PII S 0 9 2 0 - 1 2 1 1 ( 9 7 ) 0 0 0 8 4 - 3
234                             C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

actions. 7. Gabapentin prevents neuronal death in several models including those designed to mimic amyotrophic
lateral sclerosis (ALS). This may occur by inhibition of glutamate synthesis by branched-chain amino acid
aminotransferase (BCAA-t). © 1998 Elsevier Science B.V. All rights reserved.

1. Introduction                                                gabapentin was originally modeled after the struc-
                                                               ture of GABA, it does not modulate GABA
   Gabapentin, 1-(aminomethyl)cyclohexaneacetic                receptor function like conventional GABAergic
acid (Neurontin®) is a novel anticonvulsant drug               drugs, and it is inactive at GABA receptors. This
that is active in a variety of animal seizure models           review outlines several potential mechanisms of
(Taylor, 1995) and prevents partial seizures and               pharmacological action of gabapentin. The vari-
generalized tonic-clonic seizures in several                   ous hypotheses are considered in approximate
placebo-controlled clinical studies, both in add-on            order of their discovery, and each is considered
and monotherapy (Andrews et al., 1990; McLean                  critically, with references to the published litera-
et al., 1993; Marson et al., 1996; Beydoun et al.,             ture. Although a consensus has not yet been
1997; Burgey et al., 1997). Clinical use of                    reached, it seems likely that several different sites
gabapentin has been associated with several side               of action may be necessary to account for all of
effects, but it is generally well tolerated (Ramsey,           the pharmacological actions of gabapentin.
1995). Its pharmacokinetic profile, and use in
combination with other medications has been de-
scribed (McLean, 1995). Gabapentin was origi-                  2. Gabapentin and system L amino acid
nally synthesized to treat spasticity and to reduce            transporters
polysynaptic spinal reflexes. It is active in several
animal models of spasticity.                                      Gabapentin is an amino acid that exists at
   Recently, it has been shown in animal models                physiological pH as a zwitterion, and since it is
that gabapentin prevents nociceptive responses                 doubly-charged, its native permeability to mem-
from hyperalgesia in animal models (Everhart et                brane barriers within the body is low. However,
al., 1997; Field et al., 1997a,b; Gillin and Sorkin,           like several other amino acids, gabapentin is a
1997; Hunter et al., 1997; Hwang and Yaksh,                    substrate of the so-called system L transporter of
1997; Jun and Yaksh, 1997; Singh et al., 1996;                 gut (Stewart et al., 1993a) and of neurons and
Xiao and Bennett, 1997) and also has analgesic                 astrocytes (Su et al., 1995). This property allows
actions in clinical reports (Rosner et al., 1996;              gabapentin molecules to cross membrane barriers
Backonja et al., 1997; Mellick and Mellick, 1997;              more easily. In addition to the facilitated trans-
McGraw and Kosek, 1997). Other studies suggest                 port across cell membranes, there is a smaller
that gabapentin has anxiolytic-like effects in ani-            non-saturable component of transport (Stewart et
mal models (Singh et al., 1996). Furthermore,                  al., 1993a; Su et al., 1995) that is likely due to
gabapentin treatment prevents motoneuron de-                   passive diffusion. Due to differences in the influx
generation in an in vitro model of amyotrophic                 and efflux rate of gabapentin via system L in
lateral sclerosis (ALS) (Rothstein and Kuncl,                  cultured cells, it accumulates in cytosol to greater
1995) and delays death in a transgenic animal                  concentrations than in the bathing medium (Su et
model of ALS (Gurney et al., 1996). Results in a               al., 1995). These transport properties of
preliminary placebo-controlled clinical trial of               gabapentin probably account for the access of
gabapentin for treatment of ALS were not statisti-             gabapentin to brain cytosol (Vollmer et al., 1986),
cally significant (Miller et al., 1996), but suggested         where it is present at about ten-fold higher con-
that additional clinical studies are warranted.                centrations than in the brain extracellular space
   Gabapentin was designed as a structural analog              (Welty et al., 1993). The delayed anticonvulsant
of the inhibitory neurotransmitter !-aminobutyric              action of gabapentin in rats after a bolus intra-
acid (GABA) (Satzinger, 1994). Although                        venous dose (Welty et al., 1993) may be explained
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                                  235

Fig. 1. Schematic diagram of GABAergic inhibitory synapse in brain. In presynaptic ending, glutamate is synthesized primarily from
glutamine (by glutaminase) and free GABA is synthesized from glutamate by the enzyme glutamic acid decarboxylase (GAD).
GABA is degraded by the enzyme GABA-transaminase. The irreversible GABA-transaminase inhibitor, vigabatrin, is an anticonvul-
sant. GABA is packaged into synaptic vesicles, where it is released in response to presynaptic calcium influx. GABA also can be
released from the cytosol by reversal of the GABA transporter (GABA uptake). Post-synaptically, GABA activates GABAA
receptors, which are modulated by anticonvulsant benzodiazepines and barbiturates. GABAB receptors are also activated by
synaptic release of GABA or by the GABAB agonist, baclofen. GABA is taken up from the extracellular space by a specific
transporter that is blocked by the anticonvulsant tiagabine. Nipecotic acid is both a blocker and substrate for the GABA
transporter.

in part by delayed distribution of drug to the                      3. Gabapentin and function of GABA systems in
brain compartment, with maximal concentrations                      brain
reached approximately 60 min after administra-
tion. Radiotracer studies confirm this idea (Welty                     Numerous reports in the literature (Meldrum,
et al., 1997), but anticonvulsant results suggest                   1985; Tunnicliff and Raess, 1991; Bowery, 1993;
that there may be an additional delay caused by                     Macdonald and Olsen, 1994) indicate that !-
biochemical events within brain tissue prior to                     aminobutyric acid (GABA) is a major inhibitory
anticonvulsant action (Welty et al., 1993, 1997).                   neurotransmitter in mammalian brain and that
However, recent data from models of hyperalgesia                    seizures occur if GABA synapses are impaired
in rats (Everhart et al., 1997; Field et al., 1997b)                (Tunnicliff and Raess, 1991). A variety of GABA-
indicate that the analgesic action of gabapentin is                 enhancing drugs such as GABAA agonists,
attained rapidly after an intrathecal injection of                  GABAA modulators (e.g. benzodiazepines), drugs
drug, suggesting that either analgesia and anticon-                 converted metabolically to GABA, GABA uptake
vulsant actions have different mechanisms, or that                  inhibitors (e.g. tiagabine (Gram et al., 1989)), and
the delay observed in anticonvulsant experiments                    inhibitors of GABA degradation (e.g. vigabatrin
with animals may be an over-estimate.                               (Meldrum, 1985)) prevent seizures in animal mod-
   The competition of gabapentin with transport                     els or in clinical use (Suzdak and Jansen, 1995;
of L-leucine, L-valine and L-phenylalanine in vitro                 Tunnicliff and Raess, 1991) (see Fig. 1). The
(Su et al., 1995) raises the possibility that some of               similarity of chemical structures between GABA
the pharmacological properties of gabapentin                        and gabapentin also suggests a functional rela-
arise from changes in cytosolic concentrations of                   tionship.
endogenous branched-chain amino acids. How-                            Gabapentin does not alter radioligand binding
ever, no studies to date support this hypothesis.                   at GABAA or GABAB receptors at concentrations
236                           C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

up to 100 "M (Taylor, 1995) nor does it alter                this action is probably not relevant for anticon-
[3H]GABA uptake into neuronal or glial cultures              vulsant activity (Goldlust et al., 1995).
(Su et al., 1995). In addition, gabapentin does not          Gabapentin in vitro increases the activity of par-
alter neuronal responses to the application of               tially-purified glutamic acid decarboxylase
GABA in electrophysiological experiments (Rock               (GAD), suggesting that gabapentin treatment
et al., 1993). These studies indicate that                   might increase the synthesis of GABA from gluta-
gabapentin does not interact directly with either            mate in brain tissues (Taylor et al., 1992). GAD
GABAA or GABAB receptors, nor with high-                     exists in two isoforms with different molecular
affinity Na + -dependent GABA transporters.                  and functional properties. GAD67 has a larger
Therefore, gabapentin’s actions are distinct from            molecular weight, and exists mostly in neuronal
those of several drugs that directly modulate                cell bodies; GAD65 is localized more in nerve
GABAA receptor function (Macdonald and                       endings and synaptosomes (Erlander and Tobin,
Olsen, 1994) such as benzodiazepines or barbitu-             1991; Erlander et al., 1991; Martin et al., 1991).
rates. Furthermore, the actions of gabapentin are            The in vitro activity of GAD65 is increased seven-
distinct from those of GABA uptake inhibitors                fold in saturating concentrations of pyridoxal 5"-
(Suzdak and Jansen, 1995) and from drugs that                phosphate, while GAD67 is only modulated 2-fold
alter GABAB receptor function (Bowery, 1993).                (Erlander and Tobin, 1991). Gabapentin may
   In vivo results suggest an interaction of                 modulate GAD activity in vivo. Gabapentin treat-
gabapentin with GABA systems. Gabapentin                     ment (25 mg/kg IP) increases brain concentrations
treatment causes decreased firing rates in rat sub-          of GABA in rats pretreated with aminooxyacetic
stantia nigra pars reticulata GABA neurons                   acid (AOAA) (Löscher et al., 1991). AOAA in-
(Bloms-Funke and Löscher, 1996). Spontaneous                hibits GABA transaminase and prevents GABA
firing rates are reduced substantially by other              degradation. Therefore, changes in brain GABA
GABAergic drugs such as benzodiazepines and                  concentrations after AOAA treatment suggest
muscimol. In animals, tonic extensor convulsions             that gabapentin enhances the GABA synthesis
caused by the GABA synthesis inhibitor thiosemi-             rate by 50–100% in several brain regions in vivo.
carbazide are blocked more potently than seizures            A recent study (Leach et al., 1997) with mice
from several other convulsant agents (Bartoszyk              given single or twice-daily doses of gabapentin
et al., 1986; Taylor, 1995). Thiosemicarbazide is            showed some significant changes in brain gluta-
an inhibitor of pyridoxyl 5"-phosphate (Tapia and            mate and glutamine concentrations and also
Salazar, 1991), an enzymatic cofactor needed for             changes in GABA-T activity, suggesting alter-
GABA synthesis by the cellular enzyme glutamic               ations in brain amino acid metabolism. In vivo
acid decarboxylase (GAD).                                    NMR spectroscopy indicates that brain GABA
   Thiosemicarbazide acts to trap pyridoxyl 5"-              concentrations are elevated in human patients
phosphate by combining with the free carbonyl                taking gabapentin, and that elevation of GABA is
group to form an inert complex. Clonic convul-               related to seizure control (Petroff et al., 1996;
sions caused by antagonism of GABA receptors                 Mattson et al., 1997). Recent results with NMR
are prevented by gabapentin less potently than               spectroscopy of rat brain tissues indicates that
convulsions from inhibition of GABA synthesis.               gabapentin treatment elevates brain GABA con-
Gabapentin does not prevent clonic convulsions               centration and also decreases brain glutamate
from antagonism of spinal glycine receptors.                 concentration (Petroff et al., 1997).
These results suggest that gabapentin may selec-                Gabapentin increases GABA release from rat
tively counteract GAD inhibitors. The results also           striatal brain slices in vitro (Götz et al., 1993), and
suggest that gabapentin does not interact directly           this action is prevented by the GABAA antago-
with GABA receptors or with glycinergic in-                  nist, bicuculline. Although the mechanism of the
hibitory synapses of spinal cord.                            bicuculline-sensitive effect is not clear, GABA can
   Gabapentin is a mixed-type inhibitor of                   be released from neuronal tissues by either Ca2 + -
GABA-transaminase at high concentrations, but                dependent or Ca2 + -independent mechanisms, the
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                        237

latter depending upon reversal of the GABA up-                      gyrus (Xiong and Stringer, 1997). At first glance,
take carriers (Szerb, 1982; Pin and Bockaert,                       decreased paired-pulse inhibition suggests a de-
1989). Gabapentin increases electrophysiological                    crease in GABA function, but similar results were
responses caused by the non-vesicular release of                    obtained in another study with vigabatrin (Sayin
GABA in rat optic nerves in a sucrose-gap ap-                       et al., 1997) (a known inhibitor of GABA degra-
paratus (Kocsis and Honmou, 1994). Very similar                     dation). The vigabatrin results were partially re-
increases in GABA responses were seen upon                          versed by co-application of a GABAB blocker,
inward current in voltage-clamped pyramidal neu-                    suggesting that higher extracellular GABA con-
rons in rat hippocampal slices in vitro (Honmou                     centrations activate presynaptic GABAB receptors
et al., 1995a,b, see Fig. 2. In both experiments,                   (Sayin et al., 1997).
GABA release was triggered by nipecotic acid, a                        Measurements of GABA release have been
substrate and competitive inhibitor of GABA                         confirmed in studies of [3H]GABA release from
transport that is not active at GABA receptors.                     brain slices (Fichter et al., 1996). Rat striatal
Application of nipecotic acid disrupts the normal                   brain slices were preincubated with either
equilibrium of the GABA transporter, and causes                     [3H]GABA or [3H]glutamine to differentially label
the release of cytosolic GABA by hetero-exchange                    two GABA pools. GABA release was measured
(Szerb, 1982; Kocsis and Honmou, 1994). The                         by scintillation counting (for pre-loaded
responses are due to activation of GABAA recep-                     [3H]GABA) or by cation-exchange chromatogra-
tors, and are blocked by bicuculline.                               phy followed by scintillation counting (for
   In addition, in vivo, gabapentin treatment de-                   [3H]GABA synthesized from [3H]glutamine).
creases paired-pulse inhibition in the rat dentate                  [3H]GABA release after preloading with
                                                                    [3H]glutamine was caused by application of
                                                                    nipecotic acid (1 mM) and was enhanced approxi-
                                                                    mately 50% by gabapentin pretreatment. In con-
                                                                    trast, [3H]GABA release after preloading with
                                                                    [3H]GABA was not altered by gabapentin. These
                                                                    results are consistent with the idea that
                                                                    gabapentin increases synthesis of GABA via
                                                                    GAD. Furthermore, since results were obtained in
                                                                    the presence of the GABA-transaminase inhibitor
                                                                    AOAA, the action of gabapentin is not likely to
                                                                    be caused by GABA-T inhibition. The electro-
                                                                    physiological and biochemical results with
                                                                    gabapentin on GABA systems (Kocsis and Hon-
                                                                    mou, 1994; Honmou et al., 1995a,b; Fichter et al.,
                                                                    1996) are consistent with increased GAD activity,
                                                                    since hippocampal tissues (Tapia and Salazar,
Fig. 2. Gabapentin enhances the nipecotic acid-induced release      1991), optic nerves (Ochi et al., 1993) and striatal
of GABA in rat hippocampal slices in vitro. A whole-cell            tissues each contain significant amounts of GAD.
voltage clamp recording was made from a single CA1 pyrami-
                                                                       Although nipecotic acid normally is not present
dal neuron, and nipecotic acid (10 mM) was applied for 2.0 s
by a pressure pulse to a nearby micropipette. The nipecotate        in brain, it is a convenient probe for the GABA
caused non-vesicular GABA to be released from neighboring           uptake transporter. [3H]Nipecotic acid (During et
cellular elements, resulting in a bicuculline-sensitive inward      al., 1995) or [3H] derivatives of other selective
current that was measured at intervals of 5 min. The applica-       GABA uptake inhibitors (Suzdak et al., 1994)
tion of gabapentin (100 "M, triangles) at time zero caused a
                                                                    have been used to study the localization and
significant increase in inward current in comparison to control
experiments with no drug present (circles); N = 5 for control       function of GABA transporters. The stoichiome-
experiments, N =7 for gabapentin experiments. Reproduced            try of GABA transporters has been described
from Honmou et al. (1995a) with permission.                         (Keynan and Kanner, 1988; Liron et al., 1988)
238                                  C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

Fig. 3. Gabapentin labels a high-affinity binding site in brain tissues. This autoradiograph was made by incubating a frozen section
of rat brain tissue with [3H]gabapentin and a film emulsion. After incubation, the film was developed, revealing areas of dense
[3H]gabapentin binding sites (shown in white). Abbreviations show brain regions: 1,2 are superficial layers of neocortex; CA3, CA2,
CA1 and dg (dentate gyrus) are areas of hippocampus; cg is central gray; ls is lateral septum; gr, mol and w are granule cell layer,
molecular layer and white matter of the cerebellum.

and changes in the cellular microenvironment                         4. Receptor binding studies with [3H]gabapentin
would cause reversal of the normal inward flux of
GABA via transporters. Either cytosolic Na +                           Gabapentin does not affect ligand binding at a
loads, cellular depolarization, or particularly acti-                wide variety of commonly-studied drug and neu-
vation of glutamate receptors (which causes both                     rotransmitter binding sites and voltage-activated
depolarization and Na + loading) cause net efflux                    ion channels including GABA, glutamate, and
of GABA by reversed transport (Pin and Bock-                         glycine receptors of several types (Taylor, 1995).
aert, 1989). Glutamate release and cellular depo-                    However, gabapentin itself has been used to
larization occur during seizures, and a recent                       define a novel binding site in brain tissues.
report (During et al., 1995) suggests that in hu-                      Tritiated gabapentin binds with high affinity to
man epileptic brain tissues, non-vesicular GABA                      a single population of sites in rat (KD 38 nM),
release is reduced while calcium-dependent GABA                      mouse (KD 14 nM) and pig (KD 17 nM) synaptic
release occurs normally. Therefore, gabapentin                       plasma membranes prepared from cerebral cortex
may compensate for a pathological reduction in                       (Suman et al., 1993; Thurlow et al., 1993). The
transport-mediated GABA release in epileptic                         maximum binding capacity of [3H]gabapentin re-
brain Kocsis and Mattson, 1996). This hypothesis                     ported for rat brain membranes is 4.6 pmol/mg
remains to be tested critically, but experiments                     (Suman et al., 1993), and similar values have been
with gabapentin using microdialysis to measure                       obtained for mouse and pig tissue (Thurlow et al.,
synaptic and non-vesicular GABA release might                        1993). Mapping of [3H]gabapentin binding sites in
help address this idea.                                              rat brain has been achieved by autoradiographic
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                        239

studies (Hill et al., 1993) (Fig. 3). The highest             [3H]gabapentin labels a novel pharmacological
levels of specific binding were detected in the               site.
outer layers of the cerebral cortex. Binding sites in            Compounds from two chemical series have
the hippocampus were highest in the dendritic                 been identified as potent inhibitors of
region of the CA1 pyramidal cell layer and molec-             [3H]gabapentin binding. Several 3-substituted ana-
ular layer of the dentate gyrus. Binding appeared             logues of GABA, most notably (SR)-3-isobutyl-
to be localized to areas of excitatory input com-             GABA (IC50 = 80 nM), were found to be active
monly associated with seizure activity. In the cere-          (Suman et al., 1993). The S( +)-enantiomer of
bellum, sites were concentrated in the molecular              3-isobutyl-GABA was 10-fold more potent than
layer, around regions of excitatory input. While              the R(−)-enantiomer (Taylor et al., 1993). The
the distribution of [3H]gabapentin binding sites              rank order of potency of gabapentin and the two
was qualitatively similar to that of the NMDA                 enantiomers of 3-isobutyl GABA were the same
receptor, as defined by strychnine-insensitive                both in the [3H]gabapentin binding assay and in
[3H]glycine binding, regression analysis yielded a            animal seizure models (Taylor et al., 1993). This
correlation coefficient of only 0.48. Lesion studies          observation suggests that the binding of
showed that [3H]gabapentin binding sites were                 gabapentin to the [3H]gabapentin binding site
most probably localized on neurons rather than                may be important to the antiepileptic activity of
on glia.                                                      the drug. Potent and stereospecific displacement
   A variety of compounds have been tested for                of [3H]gabapentin binding was also apparent with
inhibition of specific [3H]gabapentin binding to              several large neutral amino acids (Thurlow et al.,
brain membranes. Inactive compounds (IC50 ! 1                 1993). For each amino acid tested, the L-enan-
mM) included those that interact with GABA                    tiomer was 100–1000-fold more potent than the
receptors (kojic amine, muscimol, bicuculline,                corresponding D-enantiomer. These findings to-
isonipecotic acid), NMDA receptors (glutamate,                gether with the observation that [3H]gabapentin is
glycine, D-serine, N-methyl-D-aspartic acid, 7-               transported across membranes by a system L
chlorokynurenic acid and trans-crotonic acid) and             transporter (Stewart et al., 1993a,b; Su et al.,
the GABA transporter (nipecotic acid and THPO                 1995) led to the hypothesis that [3H]gabapentin
(4,5,6,7-tetrahydroisoxazolo[4,5-c]pyridin-3-ol))             may label the recognition site of a neuronal trans-
(Suman et al., 1993). The low affinities of GABA,             porter similar to system L (Thurlow et al., 1993).
baclofen and (+ )-MK-801 for the [3H]gabapentin
binding site suggest that [3H]gabapentin does not             4.1. Purification and identification of the
interact with either GABA or NMDA receptors                   [ 3H]Gabapentin binding protein
(Suman et al., 1993). Mg2 + and the polyamines,
spermine and spermidine, which are known al-                     To clarify the molecular nature of the
losteric modulators of NMDA receptors (Foster                 [3H]gabapentin binding protein (GBP), it was
and Fagg, 1987), inhibited [3H]gabapentin binding             purified from pig brain. Preliminary biochemical
in a non-competitive manner with IC50 values of               studies suggested that the GBP was a membrane-
27, 12 and 15 "M, respectively. Maximal inhibi-               associated protein. Brain membranes were solubi-
tion with Mg2 + or polyamines was only about                  lized with a non-ionic detergent, Tween 20, and
60%. A combination of 3 mM MgCl2 and 3 mM                     the GBP was purified to near-homogeneity by
spermine did not inhibit [3H]gabapentin binding               sequential chromatography over six matrices (Gee
to a greater extent than either compound alone,               et al., 1996). Electrophoretic analysis revealed the
suggesting the possibility of a common site of                purified GBP to have a subunit molecular weight
action. A range of anticonvulsant drugs including             of approximately 130 000. The partial N-terminal
diazepam, carbamazepine, phenobarbital, pheny-                sequence of the purified protein was identical to
toin, pentobarbital, sodium valproate, ethosux-               that reported for the $2% subunit of the L-type
imide and #-hydroxy-GABA were all inactive                    voltage-dependent Ca2 + channel from skeletal
(IC50 ! 1 mM) supporting the idea that                        muscle (Hamilton et al., 1989). Binding of
240                           C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

[3H]gabapentin to COS-7 cells transiently express-           larly or extracellularly disposed (see Fig. 4). Hy-
ing rabbit skeletal muscle $2% cDNA and to par-              drophilicity plots suggest that the $2 polypeptide
tially purified muscle Ca2 + channel subunits                contains two transmembrane domains (Ellis et al.,
confirmed that the GBP and the $2% Ca2 + chan-               1988) but biochemical data support a model
nel subunit are the same protein (Gee et al., 1996).         where the $2 polypeptide is wholly extracellular
Gabapentin is the first ligand described that inter-         (Jay et al., 1991; Brickley et al., 1995). The pres-
acts with the $2% subunit of a Ca2 + channel.                ence of a single transmembrane anchor in the %
                                                             polypeptide is widely accepted.
4.2. Structure of !oltage-dependent calcium
channels                                                     4.3. Roles of the subunits in calcium channel
                                                             function
   Ca2 + channels are multi-subunit complexes
found not only in the brain but also in peripheral              Only the N-type channel from rabbit brain
tissues such as skeletal muscle, heart and lung              (Witcher et al., 1993) and the L-type Ca2 + chan-
(Catterall,    1995).     The     distribution   of          nel from skeletal muscle (Leung et al., 1988; Jay et
[3H]gabapentin binding sites in rat peripheral tis-          al., 1991) have been purified along with their
sues is consistent with an interaction of                    associated auxiliary subunits. Thus, the particular
gabapentin at Ca2 + channels (Gee et al., 1996).             subunit combinations that occur in vivo for other
Ca2 + channels consist of at least three subunits            channels are unclear. This should be borne in
(for review, see Dolphin (1995)): $1 (the pore-              mind when considering the results of ‘mix and
forming subunit), $2% and #. Skeletal muscle                 match’ heterologous expression studies with
Ca2 + channels also have a ! subunit. High-                  cloned Ca2 + channel subunits. Most of these
threshold Ca2 + channels are classified as L-, N-,           studies have utilized the Xenopus lae!is oocyte
P-, or R-types on the basis of their sensitivity to          expression system. Functional channels require
various organic ligands and several neurotoxins.             only the $1 subunit (Mori et al., 1991). However,
Apart from gabapentin, all Ca2 + channel ligands             significant changes to the electrophysiology of the
appear to bind to $1 subunits. Molecular cloning             channel are effected by co-expression with the
studies have revealed at least six genes encoding            auxiliary subunits. A peak barium current (IBa) of
$1 subunits, and a number of splice variants have            31 nA was recorded for the N-type $1B subunit
also been identified (for review, see Hofmann et             alone, whereas expression with the skeletal muscle
al. (1994)). The # subunit is a hydrophilic protein          $2%A or skeletal muscle #1 yielded a peak IBa of 89
that interacts with the $1 subunit (Pragnall et al.,         and 664 nA, respectively. Significant co-operativ-
1994). Four genes encode # subunits and splice               ity of the auxiliary subunits was demonstrated by
variants for three of them have been described               co-expression of all three subunits ($1B$2%A#1)
(Perez-Reyes and Schneider, 1994). A single gene             which yielded an IBa of ! 6 "A (Mori et al.,
encodes the $2% subunit (DeJongh et al., 1990)               1991). Several other groups also report a co-oper-
and a number of splice variants have been found              ative effect of $2% and # on $1 (Singer et al., 1991;
(Perez-Reyes and Schneider, 1994). The $2% sub-              Williams et al., 1992). Shistik et al. (1995) have
unit is synthesized as a pre-protein that undergoes          dissected the electrophysiological and biochemical
extensive post-translational modifications. The              mechanisms of modulating Ca2 + channel current
membrane targeting signal sequence is removed                characteristics by auxiliary subunits. Whole-cell
and a second proteolytic cleavage event generates            calcium channel currents of $1C (cardiac) were
a small C-terminal fragment (%) that remains                 increased 8–10 fold by either $2% or #2 alone but
attached to the larger fragment ($2) by a disulfide          100-fold with both subunits. They found that the
bridge. The protein is also heavily glycosylated             increase in the current by $2% resulted both from
(Sharp and Campbell, 1989; Jay et al., 1991). The            a three-fold increase in the amount of $1C present
membrane topology of the $2% subunit is unclear,             in the membrane and from modulation of the
thus the [3H]gabapentin site could be intracellu-            gating characteristics of $1C. On the other hand,
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                                       241

Fig. 4. Schematic diagram of the structure of the Ca2 + channel $1, $2 and # subunits. Taken with permission from Gurnett et al.
(1996). The cell plasma membrane is shown as a lipid bilayer with intracellular and extracellular media labeled. Small branched
structures indicate extracellular sites of glycosylation (sugar polymers attached to protein). ‘S – S’ denotes sites of putative cysteine
linkages. The main voltage-sensing and ion-conducting subunit (central part of $1) is shown in its postulated conformation spanning
the lipid bilayer. The gabapentin binding protein ($2%) is shown in its presumed conformation anchored in the lipid bilayer and
associating with the $1 subunit, but with most amino acids exposed to the extracellular fluid. Recent experiments with site-directed
deletions of $2% Gurnett et al. (1996) indicate that the extracellular region of this protein enhances Ca2 + current amplitude and the
intramembrane portion interacts directly with the $1 subunit (arrows). It is not yet known what part of the $2% protein gabapentin
binds to.

#2 alone affected only the gating characteristics.                      in a subtle manner. It is possible that inhibition of
Few electrophysiological studies on the effects of                      monoamine neurotransmitter release (Reimann,
gabapentin on Ca2 + channels have been reported                         1983; Schlicker et al., 1985; Dooley et al., 1996) is
and further work in this area is required.                              caused by an interaction of gabapentin with Ca2 +
  However, several observations are consistent                          channels. Monoamine release is never inhibited by
with the idea that gabapentin modulates Ca2 +                           more than 50% by gabapentin (Reimann, 1983;
channels, particularly if channels are modulated                        Schlicker et al., 1985; Dooley et al., 1996). Re-
242                           C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

cently, voltage-clamp records of high-threshold              it alter sustained firing of action potentials in
Ca2 + currents from rat neocortical pyramidal                cultured neurons (Rock et al., 1993). However,
neurons show that gabapentin (10 – 100 "M) re-               with longer incubation periods in vitro, it de-
versibly reduces the nitrendipine-sensitive compo-           creases sustained firing of Na + -dependent action
nent of current by about 35% (Stefani et al.,                potentials (Wamill and McLean, 1994). In addi-
1997). However, another study with acutely-iso-              tion, a recent report suggests that gabapentin has
lated human dentate granule neurons showed no                other electrophysiological actions that may ac-
effect of gabapentin on total cellular Ca2 + cur-            count for reduced excitability (Kawasaki et al.,
rents (Schumacher et al., 1997). These findings              1995). It is not yet clear whether these in vitro
suggest that gabapentin may have subtle actions              findings are relevant for anticonvulsant or other
on Ca2 + channels that are only apparent in cer-             pharmacological actions of gabapentin in vivo.
tain sub-populations or experimental protocols.

                                                             7. Gabapentin and serotonin concentration in
5. Gabapentin and other neurotransmitters                    whole blood

   Gabapentin has actions on monoamine neuro-                   Gabapentin given to healthy human volunteers
transmitter release in vitro. Gabapentin causes              increases the concentration of serotonin in whole
significant decreases (10 – 15% blockade) in the             blood (Rao et al., 1988). These authors speculate
electrically- or 20– 50 mM K + -evoked release of            that increased serotonin might be due to changes
noradrenaline, dopamine and serotonin from                   in serotonin metabolism or uptake in platelets.
brain slices (Reimann, 1983; Schlicker et al., 1985;         However, this has not been studied in vitro. They
Dooley et al., 1996). The inhibitory action of               also speculate that changes in serotonin could
gabapentin on striatal dopamine release is clearly           explain changes that were observed in sleep pat-
different from that of the GABAB agonist ba-                 terns (increased duration of stages 3 and 4 with-
clofen (Reimann, 1983). Reduced monoamine re-                out changes in total sleep time, REM sleep time
lease may relate either to an action on Ca2 +                or REM latency).
channels (see section above) or to changes in
monoamine metabolism. Recent results indicate
that pretreatment of rats with gabapentin signifi-           8. Gabapentin and neuroprotection (glutamate
cantly reduces the augmented noradrenaline and               metabolism)
dopamine turnover caused by systemic adminis-
tration of 3,4-diaminopyridine (Pugsley et al.,                 Although seizures induced by glutamate ago-
1997). These results indirectly suggest that                 nists are not prevented by gabapentin, high doses
gabapentin may alter the function of Ca2 + chan-             significantly delay such seizures, suggesting that
nels involved with monoamine release (see section            gabapentin might interact with glutamatergic
above). Although changes in monoamine function               synapses (Bartoszyk et al., 1986; Taylor, 1995).
induced by gabapentin might not relate directly to           Studies with gabapentin in an in vitro model
anticonvulsant effects, alterations in monoamine             meant to mimic some aspects of motor neuron
neurotransmission might underlie behavioral ef-              disease (amyotrophic lateral sclerosis or ALS) in-
fects (anxiolytic-like action or analgesia).                 dicate that neuronal cell death is prevented by
                                                             gabapentin treatment (Rothstein and Kuncl,
                                                             1995). The model utilizes the application of a
6. Gabapentin and voltage-sensitive Na + channels            selective inhibitor of glutamate uptake, and neu-
                                                             ronal death is reduced by selective antagonists of
  Gabapentin does not alter voltage-clamped                  the AMPA type of glutamate receptor. Therefore,
sodium currents in the manner of phenytoin, car-             it is reasonable to postulate that protective effects
bamazepine or lamotrigine (Taylor, 1993), nor did            of gabapentin might arise from changes in gluta-
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                       243

mate metabolism or release. Additionally, studies            1990). This hypothesis for the action of
with a transgenic mouse model of ALS (Gurney et              gabapentin by enhanced degradation of glutamate
al., 1994) indicate that chronic treatment with              also remains to be tested in vivo.
high dosages of gabapentin significantly delays                 Gabapentin in vitro decreases the tissue content
paralysis and death in transgenic mice (Gurney et            of glutamine (a metabolic precursor of glutamate
al., 1996).                                                  and GABA) in isolated hippocampal tissue
   A recent paper (Welty et al., 1995) details two           (Kapetanovic et al., 1995). Although the
hypotheses for the neuroprotective action of                 metabolic pathway involved with this effect has
gabapentin. One pathway for glutamate synthesis              not been described, changes in glutamine
in brain tissues arises from transamination of               metabolism or transport might be relevant for
$-ketoglutarate by branched-chain amino acids                decreased glutamate synthesis.
(leucine, isoleucine, valine) by the enzyme
branched-chain amino acid aminotransferase
(BCAA-T). Gabapentin is a competitive inhibitor              9. Effects in animal models of anxiety
of BCAA-T with inhibition constants (Ki =0.6–
1.2 mM) equivalent to the substrate affinity for                Gabapentin was investigated in several animal
endogenous branched-chain amino acids (Km =                  models that predict utility for the treatment of
0.4–1.2 mM) (Goldlust et al., 1995). Recent in               anxiety. These include the marmoset human
vitro studies show that this action of gabapentin            threat test, the rat elevated X-maze and the rat
is selective for the isoform of BCAA-T found in              conflict test (Singh et al., 1996). In all of these
brain cytosol (neuronal form), with little effect on         models, gabapentin produced anxiolytic-like ef-
the mitochondrial form of BCAA-T (astroglial                 fects with minimum effective doses ranging from 3
form) (Hutson et al., 1995). Since the therapeutic           to 30 mg/kg (Singh et al., 1996). In all three
range of concentrations for gabapentin (10 – 100             models, gabapentin produced activity with similar
"M) is similar to the endogenous concentrations              efficacy to that of benzodiazepines. Most non-
of branched-chain amino acids, gabapentin may                benzodiazepine drugs produce much weaker activ-
significantly inhibit the action of BCAA-T in vivo,          ity than gabapentin in the rat elevated X-maze
and this may result in decreased cytosolic concen-           and the marmoset human threat test. For exam-
trations of glutamate that would reduce gluta-               ple, gabapentin’s activity is more pronounced
mate-dependent cell death. Decreased synthesis of            than that of buspirone or other experimental com-
glutamate from labeled L-leucine in the presence             pounds such as cholecystokinin CCKB antago-
of gabapentin has been demonstrated in vitro,                nists (Singh et al., 1991) or serotonin 5-HT3
although total tissue content of glutamate was not           receptor antagonists (Broekkamp et al., 1989). A
altered (Kapetanovic et al., 1995). Decreases in             recent study of the antidepressant, phenylzine
whole-brain glutamate of 20% after gabapentin                (Paslawski et al., 1996) suggests that elevation of
treatment have been demonstrated by NMR spec-                whole brain GABA concentration (see above)
troscopy of rat brain extracts (Petroff et al.,              could be related to antianxiety effects.
1997). A decrease in the synthesis of glutamate
from L-leucine after administration of gabapentin
in vivo remains to be demonstrated.                          10. Effects in animal models of pain
   In addition, gabapentin in vitro stimulates the
activity of the catabolic enzyme glutamate dehy-                Gabapentin administration to rats or mice does
drogenase (GDH) at high concentrations (Gold-                not alter acute responses to thermal or chemical
lust et al., 1995). It is possible that gabapentin           stimulation. However, delayed pain responses in
administration would enhance GDH activity in                 several animal models are reduced. Formalin and
vivo, as has been proposed for the treatment of              carrageenan are two chemical irritants that are
ALS with high-dose administration of endoge-                 widely used in studies of tonic pain and hyperal-
nous branched-chain amino acids (Plaitakis,                  gesia from peripheral inflammation. The subcuta-
244                            C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

neous administration of formalin into the plantar             glutamate receptors on dorsal horn neurons of the
surface of the rodent paw produces a biphasic                 spinal cord. It remains to be seen whether
nocifensive behavioral response. The early phase              gabapentin alters NMDA responses in the spinal
consists of intense licking and biting of the in-             cord, but NMDA antagonists have similar actions
jected paw and lasts up to 10 min but a second                to gabapentin in the formalin test.
late phase of licking and biting occurs from 10 to               The antihyperalgesic action of gabapentin does
60 min after injection (Dubuisson and Dennis,                 not depend on activation of opiate receptors, and
1977). The late phase is a state of facilitated pain          is not altered by the opiate antagonist, naloxone
response (hyperalgesia) associated with inflamma-             (Field et al., 1997b). Unlike morphine, gabapentin
tion. This behavioral response has been shown to              does not reduce gut motility in rats. Sedation and
correlate with a biphasic increase in the activity of         ataxia are caused by gabapentin only at doses ten
C-fiber primary afferent neurons after formalin               times higher than those preventing pain responses.
injection (McCall et al., 1996). Carrageenan elicits          Repeated administration of gabapentin does not
little or no immediate pain response (Wheeler-                result in tolerance to antihyperalgesia. Morphine
Aceto et al., 1990), but causes hyperalgesic re-              tolerance does not cross-generalize to gabapentin
sponses to thermal or mechanical stimuli with a               (Field et al., 1997b). Therefore, gabapentin’s anal-
maximum 3–4 h after injection into the footpad                gesic actions are distinct from those of opiate
(Hargreaves et al., 1988). The formalin and car-              analgesics.
rageenan behavioral tests involve sensitization of               Results of an open-label clinical study in pa-
sensory neurons of the spinal dorsal horn in re-              tients with reflex sympathetic dystrophy (RSD)
sponse to injury or intense artificial activation of          suggest that gabapentin may reduce neuropathic
C-fiber afferents (Woolf and Wall, 1986).                     pain (Mellick et al., 1995). RSD is characterized
Gabapentin (30–300 mg/kg) selectively blocks the              by burning pain, allodynia, hyperpathia, vasomo-
tonic phase of formalin nociception without                   tor and sudomotor disturbances, edema, and
changing the peripheral swelling caused by car-               trophic changes to bone, skin and soft tissues.
rageenan, but gabapentin reduces the mechanical               Satisfactory (scored good to excellent) pain relief
and thermal hyperalgesia from carrageenan                     was obtained in all eight patients given 300 or 600
(Singh et al., 1996). Therefore, gabapentin may               mg gabapentin daily. Gabapentin treatment cor-
act within the spinal cord or brain to reduce                 rected skin temperature and color, and reduced
sensitization of dorsal horn sensory neurons.                 allodynia, hyperalgesia and hyperpathia in most
   Gabapentin reduces pain responses from neu-                patients. These results suggest that a placebo-con-
ropathy produced by chronic constriction of the               trolled clinical study of gabapentin for RSD is
sciatic nerve (Hunter et al., 1997; Xiao and Ben-             warranted. In addition, results of a double-
nett, 1997) or by ligation of spinal nerves at the            blinded clinical study of gabapentin for pain from
L5 and L6 levels (Hunter et al., 1997; Hwang and              diabetic neuropathy showed a significant reduc-
Yaksh, 1997). These results in various animal                 tion of pain scores in comparison to placebo
models show that gabapentin reduces mechanical                (Backonja et al., 1997).
hyperalgesia (pin prick response), mechanical al-                In summary, gabapentin is active in animal
lodynia (Von Frey nylon monofilament), thermal                models that require sensitization of pain responses
hyperalgesia (from radiant heat) and thermal allo-            but is not active in transient models of pain.
dynia (from cold water). The intrathecal adminis-             Therefore, it may not reduce immediate pain from
tration of gabapentin blocked thermal and                     injury, but it appears to reduce abnormal hyper-
mechanical hyperalgesia (Hwang and Yaksh,                     sensitivity (allodynia and hyperalgesia) induced by
1997; Xiao and Bennett, 1997), suggesting that                inflammatory responses or nerve injury. When
gabapentin may work from a spinal site of action.             considered with gabapentin’s relative lack of un-
Thermal hyperalgesia is mediated primarily by                 desirable side effects, gabapentin may eventually
C-fiber sensory afferents, which produce their ac-            be shown to improve treatment for several
tions in the spinal cord mainly via NMDA-type                 chronic pain syndromes; three additional placebo-
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                                    245

controlled clinical studies for various pain syn-           dogenous amino acids) interact with an auxiliary
dromes are presently underway.                              subunit of voltage-gated Ca2 + channels. Addi-
                                                            tional studies are needed to establish which of the
                                                            various potential mechanisms account for activity
11. Electrophysiology studies of glutamate                  of gabapentin in anticonvulsant, antinociceptive,
responses                                                   anxiolytic and neuroprotective models.

  Rock et al. (1993) used electrophysiological
recordings from single cultured neurons from rat
                                                            References
cortex, mouse spinal cord and rat sensory neurons
to evaluate potential actions of gabapentin on              Andrews, J., Chadwick, D., Bates, D., 1990. Gabapentin in
neurotransmitter responses and voltage-dependent                partial epilepsy. Lancet 335, 1114 – 1117.
ion channels. Gabapentin (concentrations up to              Backonja, M., Hes, M.S., LaMoreaux, L.K., Garofalo, E.A.,
500 "M) had no effects on repetitive sodium                     Koto, E.M. and the US Gabapentin Study Group 210,
                                                                1997. Gabapentin reduces pain in diabetics with painful
action potentials nor on isolated calcium currents
                                                                peripheral neuropathy: results of a double-blind, placebo-
(whole-cell currents included components of both                controlled clinical trial (945-210) [Abstract], American Pain
high N/L- and low threshold T-type currents).                   Society, 16th Annual Meeting, Abstracts, p. 108.
Inhibitory responses produced by the ion-                   Bartoszyk, G.D., Meyerson, N., Reimann, W., Satzinger, G.,
tophoretic application of GABA or glycine were                  von Hodenberg, A., 1986. Gabapentin. In: Meldrum, B.S.,
                                                                Porter, R.J. (Eds.). New Anticonvulsant Drugs. John
not altered by gabapentin. The only effects of
                                                                Libbey, London, pp. 147 – 163.
gabapentin observed were on NMDA responses                  Beydoun, A., Fischer, J., Labar, D.R., Harden, C., Cantrell,
in cortical neurons. Gabapentin (100 "M) en-                    D., Uthman, B.M., Sackellares, J.C., Abou-Khalil, B.,
hanced the sustained portion of NMDA responses                  Ramsay, R.E., Hayes, A., Greiner, M., Garofalo, E.,
in seven out of 18 neurons in the absence of                    Pierce, M., 1997. Gabapentin monotherapy: II. A 26-week
                                                                double-blind, dose-controlled, multicenter study of conver-
exogenous glycine. No effects of gabapentin were
                                                                sion from polytherapy in outpatients with refractory com-
seen when experiments were repeated in the pres-                plex-partial or secondarily generalized seizures. Neurology
ence of excess glycine, nor were any effects of                 49, 746 – 752.
gabapentin reported on single NMDA channels                 Bloms-Funke, P., Löscher, W., 1996. The anticonvulsant
recorded from outside-out membrane patches                      gabapentin decreases firing rates of substantia nigra pars
                                                                reticulata neurons. Eur. J. Pharmacol. 316, 211 – 218.
taken from cortical neurons, either in the absence
                                                            Bowery, N.G., 1993. GABA-B receptor pharmacology. Annu.
or presence of glycine. No effects of gabapentin                Rev. Pharmacol. Toxicol. 33, 109 – 147.
were seen with currents induced by the excitatory           Brickley, K., Campbell, V., Berrow, N., Leach, R., Norman,
amino acids, kainate and quisqualate.                           R.I., Wray, D., Dolphin, A.C., Baldwin, S.A., 1995. Use of
                                                                site-directed antibodies to probe the topography of the $2
                                                               subunit of voltage-gated Ca2 + channels. FEBS Lett. 364,
                                                                129 – 133.
12. Conclusions                                             Broekkamp, C.L., Berendsen, H.H., Jenck, F., VanDelft,
                                                                A.M., 1989. Animal models for anxiety and response to
   Although many studies have attempted to es-                  serotonergic drugs. Psychopathology 22, 2 – 21.
tablish the cellular and molecular targets of the           Burgey, G.K., Morris, H.H., Rosenfeld, W., Blume, W.T.,
                                                                Penovich, P.E., Morrell, M.J., Liederman, D.B., Crockatt,
actions of gabapentin, a clear consensus still has
                                                                J.G., Lamoreaux, L., Garofalo, E., Pierce, M., 1997.
not been obtained. It is quite likely that several              Gabapentin monotherapy: I. An 8-day double-blind, dose-
different cellular actions account for various as-              controlled, multicenter study in hospitalized patients with
pects of gabapentin pharmacology. At present                    refractory complex-partial or secondarily generalized
several laboratories have demonstrated that                     seizures. Neurology 49, 739 – 745.
                                                            Catterall, W.A., 1995. Structure and function of voltage-gated
gabapentin treatment alters the metabolism or
                                                                ion channels. Ann. Rev. Biochem. 64, 493 – 531.
concentrations of glutamate, glutamine or GABA              DeJongh, K.S., Warner, C., Catterall, W.A., 1990. Subunits of
in brain tissues. Several laboratories also have                purified calcium channels: $2 and % are encoded by the
demonstrated that gabapentin (and several en-                   same gene. J. Biol. Chem. 265, 14738 – 14741.
246                                   C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249

Dolphin, A.C., 1995. Voltage-dependent calcium channels and          Gram, L., Larsson, O.M., Johnsen, A., Schousboe, A., 1989.
    their modulation by neurotransmitters and G-proteins.                Experimental studies of the influence of vigabatrin on the
    Exp. Physiol. 80, 1–36.                                              GABA system. Br. J. Clin. Pharmacol. 27, 13S – 18S.
Dooley, D.J., Suman-Chauhan, N., Madden, Z., 1996. Inhibi-           Gurnett, C.A., DeWaard, M., Campbell, K.P., 1996. Dual
    tion of K + -evoked [3H]noradrenaline release from rat               function of the voltage-dependent Ca2 + channel $2% sub-
    neocortical slices by the anticonvulsant gabapentin [ab-             unit in current stimulation and subunit interaction. Neuron
    stract]. Soc. Neurosci. Abstr. 22, 1992.                             16, 431 – 440.
Dubuisson, D., Dennis, S.G., 1977. The formalin test: a quan-        Gurney, M.E., Pu, H., Chiu, A.Y., Dal Canto, M.C., Pol-
    titative study of analgesic effects of morphine, meperidine,         chow, C.Y., Alexander, D.D., Caliendo, J., Hentati, A.,
    and brain stem stimulation on rats and cats. Pain 4,                 Kwon, Y.W., Deng, H.-X., Chen, W., Zhai, P., Sufit, R.L.,
    161 – 174.                                                           Siddique, T., 1994. Motor neuron degeneration in mice
During, M., Ryder, K.M., Spencer, D.D., 1995. Hippocampal                expressing a human Cu, Zn superoxide dismutase muta-
    GABA transporter function in temporal-lobe epilepsy. Na-             tion. Science 264, 1772 – 1775.
    ture 376, 174 – 177.                                             Gurney, M.E., Cutting, F.B., Zhai, P., Doble, A., Taylor,
Ellis, S.B., Williams, M.E., Ways, N.R., Brenner, R., Sharp,             C.P., Andrus, P.K., Hall, E.D., 1996. Benefit of vitamin E,
    A.H., Leung, A.T., Campbell, K.P., McKenna, E., Koch,                riluzole and gabapentin in a transgenic model of familial
    W.J., Hui, A., 1988. Sequence and expression of mRNAs                amyotrophic lateral sclerosis. Ann. Neurol. 39, 147 – 157.
    encoding the $1 and $2 subunits of a DHP-sensitive cal-          Hamilton, S.L., Hawkes, M.J., Brush, K., Cook, R., Chang,
    cium channel. Science 241, 1661–1664.                                R.J., Smilowitz, H.M., 1989. Subunit composition of the
Erlander, M.G., Tobin, A.J., 1991. The structural and func-              purified dihydropyridine binding protein from skeletal
    tional heterogeneity of glutamic acid decarboxylase: A               muscle. Biochemistry 28, 7820 – 7828.
    review. Neurochem. Res. 16, 215–226.                             Hargreaves, K.R., Dubner, F., Brown, C., Flores, A.S., Joris,
Erlander, M.G., Tillakaratne, N.J., Feldblum, S., Patel, N.,             J., 1988. A new and sensitive method for measuring ther-
    Tobin, A.J., 1991. Two genes encode distinct glutamate
                                                                         mal nociception in cutaneous hyperalgesia. Pain 32, 77 – 84.
    decarboxylases. Neuron 7, 91–100.
                                                                     Hill, D.R., Suman Chauhan, N., Woodruff, G.N., 1993. Lo-
Everhart, A.W., Willis, W.D., Hulsebosch, C.E., 1997.
                                                                         calization of [3H]-gabapentin to a novel site in rat brain:
    Gabapentin inhibits mechanical and thermal allodynia in a
                                                                         autoradiographic studies. Eur. J. Pharmacol. 244, 303 –
    rodent model of chronic central pain following spinal
                                                                         309.
    hemisection [abstract]. Soc. Neurosci. Abstr. 23, 1812.
                                                                     Hofmann, F., Biel, M., Flockerzi, V., 1994. Molecular basis
Fichter, N., Taylor, C.P., Feuerstein, T.J., 1996. Nipecotate-
                                                                         for Ca2 + channel diversity. Annu. Rev. Neurosci. 17,
    induced GABA release from slices of the rat caudato-puta-
                                                                         399 – 418.
    men: effects of gabapentin N–S [abstract]. Arch.
                                                                     Honmou, O., Kocsis, J.D., Richerson, G.B., 1995a.
    Pharmacol. 354, R35.
                                                                         Gabapentin potentiates the conductance increase induced
Field, M.J., Holloman, E.F., McCleary, S., Hughes, J., Singh,
                                                                         by nipecotic acid in CA1 pyramidal neurons in vitro.
    L., 1997a. Evaluation of gabapentin and S-( +)-3-isobutyl-
    GABA in a rat model of postoperative pain. J. Pharmacol.             Epilepsy Res. 20, 193 – 202.
    Exp. Ther. 282, 1242–1246.                                       Honmou, O., Oyelese, A.A., Kocsis, J.D., 1995b. The anticon-
Field, M.J., Oles, R.J., Lewis, A.S., McCleary, S., Hughes, J.,          vulsant gabapentin enhances promoted release of GABA in
    Singh, L., 1997b. Gabapentin (Neurontin) and S-( +)-3-               hippocampus: a field potential analysis. Brain Res. 692,
    isobutyl GABA represent a novel class of selective antihy-           273 – 277.
    peralgesic agents. Br. J. Pharmacol. 121, 1513–1522.             Hunter, J.C., Gogas, K.R., Hedley, L.R., Jacobson, L.O.,
Foster, A.C., Fagg, G.E., 1987. Taking apart NMDA recep-                 Kassotakis, L., Thompson, J., Fontana, D.J., 1997. The
    tors. Nature 329, 395–396.                                           effect of novel anti-epileptic drugs in rat experimental
Gee, N.S., Brown, J.P., Dissanayake, V.U.K., Offord, J.,                 models of acute and chronic pain. Eur. J. Pharmacol. 324,
    Thurlow, R., Woodruff, G.N., 1996. The novel anticonvul-             153 – 160.
    sant drug, gabapentin (Neurontin), binds to the $2% sub-         Hutson, S.M., Drown, P., I’lyosova, D., Reinhart, G.D., 1995.
    unit of a calcium channel. J. Biol. Chem. 271, 5768–5776.            Role of gabapentin and branched chain aminotransferase
Gillin, S., Sorkin, L.S., 1997. Gabapentin reverses the allody-          isoenzymes in astrocyte neurotransmitter metabolism [ab-
    nia produced by the administration of anti-GD2 gan-                  stract]. J. Neurochem. 66, S76.
    glioside, an immunotherapeutic agent. Anesth. Analg. (in         Hwang, J.H., Yaksh, T.L., 1997. Effect of subarachnoid
    press).                                                              gabapentin on tactile-evoked allodynia in a surgically-in-
Goldlust, A., Su, T., Welty, D.F., Taylor, C.P., Oxender,                duced neuropathic pain model in the rat. Reg. Anesthesia.
    D.L., 1995. Effects of the anticonvulsant drug gabapentin            22, 249 – 256.
    on enzymes in the metabolic pathways of glutamate and            Jay, S.D., Sharp, A.H., Kahl, S.D., Vedvick, T.S., Harpold,
    GABA. Epilepsy Res. 22, 1–11.                                        M.M., Campbell, K.P., 1991. Structural characterization
Götz, E., Feuerstein, T.J., Meyer, D.K., 1993. Effects of               of the dihydropyridine-sensitive calcium channel $2-subunit
    gabapentin on release of !-aminobutyric acid from slices of          and the associated % peptides. J. Biol. Chem. 266, 3287 –
    rat neostriatum. Drug Res. 43, 636–638.                              3293.
C.P. Taylor et al. / Epilepsy Research 29 (1998) 233–249                                   247

Jun, J.H., Yaksh, T.L., 1997. Effect of intrathecal gabapentin        McLean, M.J., Ramsey, R.E., Leppik, I., Rowan, A.J., Shel-
    and 3-isobutyl GABA on the hyperalgesia observed after                lenberger, M.K., Wallace, J., US Gabapentin Study
    thermal injury in the rat. Anesth. Analg. (in press).                 Group, 1993. Gabapentin as add-on therapy in refractory
Kapetanovic, I.M., Taylor, C.P., Yonekawa, W.D., Kupfer-                  partial epilepsy: a double-blind, placebo-controlled, paral-
    berg, H.J., 1995. Effect of gabapentin (GBP) on amino                 lel-group study. Neurology 43, 2292 – 2298.
    acids in rat hippocampal slices in vitro [abstract]. Soc.         Meldrum, B.S., 1985. GABA and other amino acids. In: Frey,
    Neurosci. Abstr. 21, 2117.                                            M.M., Janz, D. (Eds.), Antiepileptic Drugs. Springer,
Kawasaki, H., Mattia, D., Zona, C., Avoli, M., 1995. Effects              Berlin, pp. 153 – 188.
    induced by gabapentin on the electrophysiological proper-         Mellick, G.A., Mellick, L.B., 1997. Reflex sympathetic dystro-
    ties of bursting neurons in the rat subiculum in vitro                phy treated with gabapentin. Arch. Phys. Med. Rehabil.
    [abstract]. Soc. Neurosci. Abstr. 21, 2117.                           78, 98 – 105.
Keynan, S., Kanner, B.I., 1988. !-aminobutyric acid transport         Mellick, G.A., Mellicy, L.B., Mellick, L.B., 1995. Gabapentin
    in reconstituted preparations from rat brain: coupled                 in the management of reflex sympathetic dystrophy. J. Pain
    sodium and chloride fluxes. Biochemistry 27, 12–17.                   Symptom Manage. 10, 265 – 266.
Kocsis, J.D., Honmou, O., 1994. Gabapentin increases                  Miller, R.G., Moore, D., Young, L.A., Armon, C., Barohn,
    GABA-induced depolarization in rat neonatal optic nerve.              R.J., Bromberg, M.B., Bryan, W.W., Gelinas, D.F., Men-
    Neurosci. Lett. 169, 181–184.                                         doza, M.C., Neville, H.E., Parry, G.J., Petajan, J.H., Rav-
Kocsis, J.D., Mattson, R.H., 1996. GABA levels in the brain:              its, J.M., Ringel, S.P., Ross, M.A., WALS Study Group,
    a target for new antiepileptic drugs. Neuroscientist 2,               Western Amyotrophic Lateral Sclerosis Study Group,
    326 – 334.                                                            1996. Placebo-controlled trial of gabapentin in patients
Leach, J.P., Sills, G.J., Butler, E., Forrest, G., Thompson,              with amyotrophic lateral sclerosis. Neurology 47, 1383 –
    G.G., Brodie, M.J., 1997. Neurochemical actions of                    1388.
    gabapentin in mouse brain. Epilepsy Res. 27, 175–180.             Mori, Y., Freidrich, T., Kim, M.S., Mikami, A., Nakai, J.,
Leung, A.T., Imagawa, T., Block, B., Franzini-Armstrong, C.,              Ruth, P., Bosse, E., Hofmann, F., Flockerzi, V., Furuichi,
    Campbell, K.P., 1988. Biochemical and ultrastructural                 T., 1991. Primary structure and functional expression from
    characterization of the 1,4-dihydropyridine receptor from             complimentary DNA of a brain calcium channel. Nature
    rabbit skeletal muscle. Evidence for a 52 000 Da subunit. J.          350, 398 – 402.
    Biol. Chem. 263, 994–1001.                                        Ochi, S., Lim, J.Y., Rand, M.N., During, M.J., Sakatani, K.,
Liron, Z., Wong, E., Roberts, E., 1988. Studies on uptake of              Kocsis, J.D., 1993. Transient presence of GABA in astro-
    !-aminobutyric acid by mouse brain particles; toward the              cytes of the developing optic nerve. Glia 9, 188 – 198.
    development of a model. Brain Res. 444, 119–132.                  Paslawski, T., Treit, D., Baker, G.B., George, M., Coutts,
Löscher, W., Honack, D., Taylor, C.P., 1991. Gabapentin                  R.T., 1996. The antidepressant drug phenelzine produces
    increases aminooxyacetic acid-induced GABA accumula-                  antianxiety effects in the plus-maze and increases in rat
    tion in several regions of rat brain. Neurosci Lett. 128,             brain GABA. Psychopharmacology 127, 19 – 24.
    150 – 154.                                                        Perez-Reyes, E., Schneider, T., 1994. Calcium channels: struc-
Macdonald, R.L., Olsen, R.W., 1994. GABAA receptor chan-                  ture, function and classification. Drug Devl. Res. 33, 295 –
    nels. Annu. Rev. Neurosci. 17, 569–602.                               318.
Marson, A.G., Kadir, Z.A., Chadwick, D.W., 1996. New                  Petroff, O.A., Rothman, D.L., Behar, K.L., Lamoureux, D.,
    antiepileptic drugs: a systematic review of their efficacy and        Mattson, R.H., 1996. The effect of gabapentin on brain
    tolerability [see comments]. Br. Med. J. 313, 1169–1174.              gamma-aminobutyric acid in patients with epilepsy. Ann.
Martin, D., Martin, S.B., Wu, S.J., Espina, N., 1991. Regula-             Neurol. 39, 95 – 99.
    tory properties of brain glutamate decarboxylase (GAD):           Petroff, O.A., Manor, D., Behar, K.L., 1997. Gabapentin
    The apoenzyme of GAD is present principally as the                    decreases cortical glutamate rapidly in a rat model.
    smaller of two molecular forms of GAD in brain. J.                    Epilepsy Res. (submitted).
    Neurochem. 11, 2725–2731.                                         Pin, J.P., Bockaert, J., 1989. Two distinct mechanisms, differ-
Mattson, R.H., Rothman, D.L., Behar, K.L., Petroff, O.A.C.,               entially affected by excitatory amino acids, trigger GABA
    1997. Gabapentin: a GABA active drug [abstract]. Epilep-              release from fetal mouse striatal neurons in primary cul-
    sia 38, 65 – 66.                                                      ture. J. Neurosci. 9, 648 – 656.
McCall, W.D., Tanner, K.D., Levine, J.D., 1996. Formalin              Plaitakis, A., 1990. Glutamate dysfunction and selective motor
    induces biphasic activity in C-fibers in the rat. Neurosci.           neuron degeneration in amyotrophic lateral sclerosis: A
    Lett. 208, 45 – 48.                                                   hypothesis. Ann. Neurol. 28, 3 – 8.
McGraw, T., Kosek, P., 1997. Erythromelalgia pain managed             Pragnall, M., De Waard, M., Mori, Y., Tanabe, T., Snutch,
    with gabapentin. Anesthesiol 86, 988–990.                             T.P., Campbell, K.P., 1994. Calcium channel # subunit
McLean, M.J., 1995. Gabapentin: chemistry, absorption, dis-               binds to a conserved motif in the I – II cytoplasmic linker
    tribution and excretion. In: Levy, R.H., Mattson, R.H.,               of the $1-subunit. Nature 368, 67 – 70.
    Meldrum, B.S. (Eds.), Antiepileptic Drugs. Raven, New             Pugsley, T.A., Whetzel, S.Z., Dooley, D.J., 1997. Reduction of
    York, pp. 843 – 849.                                                  3,4-diaminopyrdine-induced biogenic amine synthesis and
You can also read