1-s2.0-S1059131113001143-main

12
Review Molecular mechanisms of antiseizure drug activity at GABA A receptors L. John Greenfield Jr.* Dept. of Neurology, University of Arkansas for Medical Sciences, 4301W. Markham St., Slot 500, Little Rock, AR 72205, United States Seizures frequently result from an imbalance of excitation and inhibition due to a failure of inhibitory neurotransmission. Most agents that enhance GABA A receptor (GABA A R) function have antiseizure properties due to their ability to increase inhibitory neurotransmitter tone. The evidence linking epilepsy with dysfunction of GABAergic inhibition is substantial, and has been extensively reviewed 1–4 . 1. GABA A Rs and epilepsy GABA A Rs are pharmacologically complex, with binding sites for benzodiazepines (BZs), barbiturates, neurosteroids, general anes- thetics, loreclezole, and the convulsant toxins, picrotoxin and bicuculline. Protein subunits from seven different subunit fami- lies 5 assemble to form pentameric 6 transmembrane chloride channels (Fig. 1). In mammals, 16 subunit subtypes have been cloned, including 6 a, 3 b and 3 g subtypes, as well as d, p 7 , e 8 and u 9 and alternatively spliced variants of the b2 and g2 subtypes. Subunit expression is regulated by region, cell type 10 and developmental stage 11 , reducing the number of isoforms expressed in specific brain regions and individual neurons. The most common composition includes two a1, two b2 and a single g2 subunit; the d subunit is found instead of g in receptors expressed extrasynaptically. The subunits are arranged around a central water-filled pore, which gates to conduct Cl ions when GABA is bound (Fig. 1). Individual subunit subtypes confer different sensitivities to GABA A R modulators including BZs 12 , loreclezole 13 and zinc ions 14 (Table 1). GABA A Rs are the target not only of the BZs, but other ASDs including barbiturates and agents like tiagabine and vigabatrin 1 that increase GABA concentration at the synapse. Bicuculline and picrotoxin, which are GABA A R antagonists, induce seizures in animals and epileptiform activity in brain slice preparations. Several animal models of epilepsy have altered GABA A R number or function 1,2,15 . GABA A R subunit expression is altered in the hippocampi of experimental animals with recurrent seizures 16 and in patients with temporal lobe epilepsy 17,18 . Angelman’s syndrome is a human neurodevelopmental disorder associated with severe mental retardation and epilepsy, which is linked to a deletion mutation on chromosome 15q11-13 20 in a region encoding the GABA A R b3 subunit 21 . Two mutations in the g2 subunit that impair GABA A R function 22 , K289 M 23 and R43Q 24 , have been linked to a human syndrome of childhood absence epilepsy and febrile seizures, and a loss-of-function mutation in the a1 subunit causes autosomal dominant Juvenile Myoclonic Epilepsy 25 . The R43Q mutation in the g2 subunit reduces BZ sensitivity 26 by altering GABA A R assembly 27–29 and trapping the receptor in the endoplasmic reticulum 30 . Other point mutations in GABA A R subunits have also been associated with generalized epilepsies (see Fig. 2). Hence, GABA A R modulation by GABAergic ASDs is likely critical to their antiseizure activity. Seizure 22 (2013) 589–600 A R T I C L E I N F O Article history: Received 6 February 2013 Received in revised form 16 April 2013 Accepted 17 April 2013 Keywords: Inhibition Epilepsy Antiepileptic drugs GABA receptor Seizures Chloride channel A B S T R A C T The GABA A receptor (GABA A R) is a major target of antiseizure drugs (ASDs). A variety of agents that act at GABA A Rs s are used to terminate or prevent seizures. Many act at distinct receptor sites determined by the subunit composition of the holoreceptor. For the benzodiazepines, barbiturates, and loreclezole, actions at the GABA A R are the primary or only known mechanism of antiseizure action. For topiramate, felbamate, retigabine, losigamone and stiripentol, GABA A R modulation is one of several possible antiseizure mechanisms. Allopregnanolone, a progesterone metabolite that enhances GABA A R function, led to the development of ganaxolone. Other agents modulate GABAergic ‘‘tone’’ by regulating the synthesis, transport or breakdown of GABA. GABA A R efficacy is also affected by the transmembrane chloride gradient, which changes during development and in chronic epilepsy. This may provide an additional target for ‘‘GABAergic’’ ASDs. GABA A R subunit changes occur both acutely during status epilepticus and in chronic epilepsy, which alter both intrinsic GABA A R function and the response to GABA A R-acting ASDs. Manipulation of subunit expression patterns or novel ASDs targeting the altered receptors may provide a novel approach for seizure prevention. ß 2013 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved. * Tel.: +1 501 686 7236; fax: +1 501 686 7850. E-mail address: ljgreenfi[email protected] Contents lists available at SciVerse ScienceDirect Seizure jou r nal h o mep age: w ww.els evier .co m/lo c ate/ys eiz 1059-1311/$ see front matter ß 2013 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.seizure.2013.04.015

description

tugas

Transcript of 1-s2.0-S1059131113001143-main

  • ru

    Slot

    Seizure 22 (2013) 589600

    AR)

    min

    of t

    the

    siga

    llo

    f g

    eak

    cha

    BAe

    c ep

    Contents lists available at SciVerse ScienceDirect

    Seizu

    .eSeizures frequently result from an imbalance of excitation andinhibition due to a failure of inhibitory neurotransmission. Mostagents that enhance GABAA receptor (GABAAR) function haveantiseizure properties due to their ability to increase inhibitoryneurotransmitter tone. The evidence linking epilepsy withdysfunction of GABAergic inhibition is substantial, and has beenextensively reviewed14.

    1. GABAARs and epilepsy

    GABAARs are pharmacologically complex, with binding sites forbenzodiazepines (BZs), barbiturates, neurosteroids, general anes-thetics, loreclezole, and the convulsant toxins, picrotoxin andbicuculline. Protein subunits from seven different subunit fami-lies5 assemble to form pentameric6 transmembrane chloridechannels (Fig. 1). In mammals, 16 subunit subtypes have beencloned, including 6 a, 3 b and 3 g subtypes, as well as d, p7, e8 andu9 and alternatively spliced variants of the b2 and g2 subtypes.Subunit expression is regulated by region, cell type10 anddevelopmental stage11, reducing the number of isoformsexpressed in specic brain regions and individual neurons. Themost common composition includes two a1, two b2 and a singleg2 subunit; the d subunit is found instead of g in receptorsexpressed extrasynaptically. The subunits are arranged around a

    central water-lled pore, which gates to conduct Cl ions whenGABA is bound (Fig. 1). Individual subunit subtypes conferdifferent sensitivities to GABAAR modulators including BZs

    12,loreclezole13 and zinc ions14 (Table 1).

    GABAARs are the target not only of the BZs, but other ASDsincluding barbiturates and agents like tiagabine and vigabatrin1

    that increase GABA concentration at the synapse. Bicuculline andpicrotoxin, which are GABAAR antagonists, induce seizures inanimals and epileptiform activity in brain slice preparations.Several animal models of epilepsy have altered GABAAR number orfunction1,2,15. GABAAR subunit expression is altered in thehippocampi of experimental animals with recurrent seizures16

    and in patients with temporal lobe epilepsy17,18. Angelmanssyndrome is a human neurodevelopmental disorder associatedwith severe mental retardation and epilepsy, which is linked to adeletion mutation on chromosome 15q11-1320 in a regionencoding the GABAAR b3 subunit

    21. Two mutations in the g2subunit that impair GABAAR function

    22, K289 M23 and R43Q24,have been linked to a human syndrome of childhood absenceepilepsy and febrile seizures, and a loss-of-function mutation inthe a1 subunit causes autosomal dominant Juvenile MyoclonicEpilepsy25. The R43Q mutation in the g2 subunit reduces BZsensitivity26 by altering GABAAR assembly

    2729 and trapping thereceptor in the endoplasmic reticulum30. Other point mutations inGABAAR subunits have also been associated with generalizedepilepsies (see Fig. 2). Hence, GABAAR modulation by GABAergicASDs is likely critical to their antiseizure activity.

    GABAAR-acting ASDs. Manipulation of subunit expression patterns or novel ASDs targeting the altered

    receptors may provide a novel approach for seizure prevention.

    2013 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.

    * Tel.: +1 501 686 7236; fax: +1 501 686 7850.

    E-mail address: [email protected]

    1059-1311/$ see front matter 2013 British Epilepsy Association. Published by Elsevier Ltd. All rights reserved.http://dx.doi.org/10.1016/j.seizure.2013.04.015Review

    Molecular mechanisms of antiseizure d

    L. John Greeneld Jr.*

    Dept. of Neurology, University of Arkansas for Medical Sciences, 4301W. Markham St.,

    A R T I C L E I N F O

    Article history:

    Received 6 February 2013

    Received in revised form 16 April 2013

    Accepted 17 April 2013

    Keywords:

    Inhibition

    Epilepsy

    Antiepileptic drugs

    GABA receptor

    Seizures

    Chloride channel

    A B S T R A C T

    The GABAA receptor (GABA

    GABAARs s are used to ter

    the subunit composition

    actions at the GABAAR are

    felbamate, retigabine, lo

    antiseizure mechanisms. A

    led to the development o

    synthesis, transport or br

    chloride gradient, which

    additional target for GA

    epilepticus and in chroni

    jou r nal h o mep age: w wwg activity at GABAA receptors

    500, Little Rock, AR 72205, United States

    is a major target of antiseizure drugs (ASDs). A variety of agents that act at

    ate or prevent seizures. Many act at distinct receptor sites determined by

    he holoreceptor. For the benzodiazepines, barbiturates, and loreclezole,

    primary or only known mechanism of antiseizure action. For topiramate,

    mone and stiripentol, GABAAR modulation is one of several possible

    pregnanolone, a progesterone metabolite that enhances GABAAR function,

    anaxolone. Other agents modulate GABAergic tone by regulating the

    down of GABA. GABAAR efcacy is also affected by the transmembrane

    nges during development and in chronic epilepsy. This may provide an

    rgic ASDs. GABAAR subunit changes occur both acutely during status

    ilepsy, which alter both intrinsic GABAAR function and the response to

    re

    l s evier . co m/lo c ate /ys eiz

  • L.J. Greeneld Jr. / Seizure 22 (2013) 5896005902. Benzodiazepines

    BZs were initially developed as anxiolytic agents in the 1950s.Chlordiazepoxide was introduced in 1960, followed by diazepam31

    Fig. 1. Model of a GABAA receptor in the plasma membrane. A. A space-lling model of thwith the nicotinic acetylcholine receptor. There are two binding sites for GABA, betwee

    subunits (arrow). C. A schematic view shows the topology of each subunit with a large ex

    the second of which forms the chloride ion channel. Binding of GABA allows the channel

    (IPSP). D. Putative arrangement of 5 subunits to form a pentamer with central chloride

    Database PDB ID: 2BG9 from Unwin, N. (2005)198. Images modied from en.wikipedia

    Table 1Antiseizure Drugs and their GABAAR Effects.

    Anti-seizure drug Subunit specicity Site of action

    Benzodiazepines a13, a5; g a/g interface,a1(H101)

    Zolpidem a1 > a2,3; g a/g interface Barbiturates b subunits? M3 residues?

    Loreclezole b2, b3 B2(N289) Ganaxolone a, b, d, # with e a1(Q241)

    Topiramate a6 > a4 > a1-a2? unknown

    Felbamate unknown unknown

    Retigabine unknown unknown

    Losigamone unknown unknown

    Stiripentol a3, # with b1, e unknown Vigabatrin GABA transaminase

    Gabapentin,

    pregabalin

    Glutamic acid decarboxylase

    Valproic Acid Glutamic acid decarboxylase

    Tiagabine GAT-1 (GABA transporter) and nitrazepam.32 In 1965, diazepam was rst used to treat statusepilepticus in humans.33 Clonazepam was introduced in the 1970sprimarily as an ASD,34 and clobazam, a 1,5 benzodiazepine, waslater developed as an ASD with less sedative effect.35 However, the

    e pentomer in side view (A) and top view (B) based on the high sequence homology

    n a and b subunits (bent arrows), and one for BZs between the alpha and gammatracellular loop containing a cysteine loop and 4 transmembrane domains (M1-M4),

    to open and conduct Cl ions, resulting in the fast inhibitory post-synaptic potentialchannel lined by the M2 subunit. [Derived from the published structure: RCSB PDB

    .org/wiki/GABA_A_receptor, used with permission (public domain)].

    GABAAR action Other mechanisms

    Left shift of GABA

    C/R curve, "open frequency

    Possible increased

    GABAAR channel conductance

    " channel open time &burst duration

    Use-dependent Na+ channel block

    Barbiturate-like? inhibits GABAAR at " concentrationBarbiturate-like, also "open frequency

    Open channel block at high conc.

    Enhanced GABA currents Na+/Ca2+ channel block, AMPA/kainate

    receptor block

    Barbiturate-like Blocks NMDA receptor currents

    Increased GABA IPSCs Opens KCNQ2/3 potassium channels

    Enhanced GABA

    receptor current

    Use-dependent Na+ channel block

    Barbiturate-like CYP450 inhibition

    increases [GABA]

    " GABA synthesis? a2-d subunit of voltage-gated Ca2+ channel

    " GABA synthesis? block of Na+, T-type Ca2+ channelsBlocks GABA reuptake

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600 591induction of tolerance limits their use as ASDs. The 1,5 BZs mayproduce less tolerance than the 1,4 BZs, but tolerance to 1,5 BZsdoes occur.36

    BZ activity at GABAARs is a function of the drugs afnity for theBZ binding site and its intrinsic allosteric effect on the GABAAR. Theefcacy of individual compounds varies widely. Most BZs inclinical use are full agonists that maximally enhance GABAARactivity. Flumazenil, a competitive antagonist used to reverse BZ-induced sedation,37 binds to the BZ site without affecting GABAARfunction. Abecarnil,39 imidazenil,40 and bretazenil41 are partialagonists at the BZ site which have antiseizure efcacy in animalmodels and appear less prone to tolerance.42 Several b-carbolinesare inverse BZ agonists that inhibit GABA binding43 and caninduce seizures or anxiety.44

    Antiseizure Activity. BZs are effective against most experimentalseizure types, but individual drugs vary in their potency/efcacy inspecic seizure models and their other clinical effects.45 BZs areparticularly effective against convulsive seizures induced bypentylenetetrazol46 and less effective against tonic seizuresinduced by maximal electroshock.47 BZs also slow the develop-ment of kindling.48

    GABAAR Subunits and BZ Pharmacology. BZ augmentation ofGABAAR currents requires a g subunit, and the selectivity of BZresponsiveness is determined by which a subunits are present.5,49

    The BZ binding site is located in a cleft between the extracellularamino termini of the a and g subunits.51 The a1 subunit results in a

    Fig. 2. Model of a prototype GABAAR subunit (based on a1 subunit diagram from Olsen &generalized epilepsies (see also Macdonald et al., 2012200) in black, and locations of poreceptor with high afnity for the hypnotic, zolpidem, dening theBZ-1 (or V-1) receptor type.50 The a2 and a3 subunits result inreceptors with moderate zolpidem afnity, termed BZ-2 receptors.GABAARs containing the a5 subunit and/or the g3 subunit aresensitive to diazepam but not to zolpidem and are termed BZ-3receptors. GABAARs with the a4 or a6 subunits are insensitive tomost BZs.

    The subunit composition not only determines the afnity forparticular BZs, but also the clinical/behavioral effect of the BZ atthat receptor. The role of the a subunits in BZ pharmacology wasrevealed by the discovery of a single histidine (H) residue found inall BZ-sensitive a subunits (H101 in the rat a1 subunit), but not inthe BZ-insensitive a4 or a6 subunits, which instead have acharged arginine (R) residue. This H residue was discovered in astrain of alcohol-non-tolerant rats, which were found to have aspontaneous point mutation in the a6 subunit (R100Q) that madetheir a6-containing GABAARs (found mostly in the cerebellum)diazepam-sensitive, accounting for their ethanol and BZ intoler-ance.53 Mutation of R100 to H in a6 dramatically increased BZbinding in this normally insensitive subunit, while mutation ofH101 to R in a1 reduced BZ sensitivity.52 BZ-insensitive a subunitmutations were subsequently knocked-in to identify BZactions at receptors containing that subunit. In homozygousa1(H101R) knock-in mice, the anxiolytic effect was intact, butBZs were not protective against pentylenetetrazol-inducedconvulsions and did not produce sedation or amnesia, suggesting

    Tobin, 1990)199 showing approximate locations of point mutations associated with

    int mutations associated with ASD sites of action. See text for details.

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600592that binding to the (wild type) a1 subunit is responsible forsedative, amnestic and antiseizure actions.54 Moreover, thesedative-hypnotic, zolpidem, showed no sedative effect ina1(H101R) mice.55 Unfortunately, these ndings underscorethe association between sedative and antiseizure efcacy at a1-containing GABAARs. Similarly, the anxiolytic

    56 and myorelax-ant57 properties of BZs appear to derive from a2- and a3-containing GABAARs, while the a5 subunit was critical foramnestic effects.58 BZs may also have a true analgesic effectindependent of their sedative and anxiolytic actions, associatedwith the a2 and a3 more than a5 subunits.59 Since there is noevidence of biophysically distinct effects of BZs on receptorscomposed of different a subunits, the different behavioral effectsare likely due to the brain regions and neuronal populationsexpressing these specic GABAAR isoforms. New a2/a3 subunit-selective BZs appear to be anxiolytic but not sedating,60 and non-sedating antiseizure BZs that do not induce tolerance61 may alsobe possible.

    BZ Actions at GABAARs. BZs increase the amplitude62 or decay

    time63 of GABA-mediated inhibitory post-synaptic potentials(IPSPs). Current noise uctuation analysis64 and single channelstudies65 demonstrated that the BZs increased the openingfrequency of the GABAAR chloride channel. In patch-clamprecordings of CNS neurons, BZs produce a leftward shift of theconcentration-response curve for GABA,66 due to an increase in theafnity for GABA at its binding site, with no change in the kineticsof channel gating65 or single channel conductance. In contrast, aninverse agonist at the BZ site reduced the channel openingfrequency for a given GABA concentration. These ndings areconsistent with binding studies showing the allosteric interactionbetween the BZ and GABA binding sites. By increasing the afnityof the receptor for GABA through slowing the unbinding rate, theBZs increase the current produced by low GABA concentrations,but not by high GABA concentrations at which receptor binding issaturated, as observed in the synaptic cleft67. Thus, BZs generallydo not increase the amplitude of miniature inhibitory postsynapticcurrents (mIPSCs) from individual synapses, but instead prolongthe mIPSC decay phase68 by slowing the dissociation of GABA fromthe receptor.69 Prolongation of the mIPSC increases temporal andspatial summation of multiple synaptic inputs, which in turnincreases the amplitude of stimulus-evoked polysynaptic IPSCs.The BZs thus increase the inhibitory tone of GABAergic synapses,which reduces the hypersynchronous ring of neuron populationsthat underlies seizures.1

    An alternative mechanism for BZ enhancement of GABAARcurrents has been proposed based on the controversial ndingthat BZs progressively increase GABAAR single channel conduc-tance.70 Unlike prior reports of a 27 pS main conductance leveland a 19 pS subconductance level,65 Eghbali et al.70 foundconductance levels ranging from 8 to 53 pS in response to GABAalone, and 7080 pS in the presence of diazepam, with up to 7-foldincrease in conductance observed when diazepam was added. Theincreases in conductance were seen predominantly in cell-attached patches onto neurons expressing native receptors, butwere also observed in outside-out patches. They found similarincreases in channel conductance induced by pentobarbital,71

    neuroactive steroids,72 propofol,73 and GABA itself.74 The samegroup75 subsequently found that high conductance (>40 pS)GABAAR channels were not observed in recombinant a1-b1-g2GABAARs expressed in L929 cells, nor was conductance increasedby diazepam unless the GABAAR-associated protein (GABARAP)was also co-expressed, apparently facilitating clustering ofGABAAR proteins through its interaction with the cytoplasmicloop of the g2 subunit as occurs at synapses with native receptors.Since peptides mimicking the intracellular g2 loop (g2 381403)self-associate, and application of this peptide to the cytoplasmicsurface of inside-out patches prevented the diazepam-inducedincrease in conductance, they hypothesized that the apparentsingle channel conductance changes induced by BZs are due tosynchronized openings of multichannel clustered GABAARs viaconcerted action through the interacting cytoplasmic loops ofconjoined receptors. Such interactions might occur throughshared transmembrane domains between interacting receptors,as observed in G-protein-coupled receptors.76 This hypothesiscould be addressed using concatenated subunits, chimeras andother strategies. However, synchronization of multiple identicalchannels by such a mechanism should still require a discrete 9 pSunitary conductance of which the larger conductance states areinteger multiples, which has not been reported.

    3. Barbiturates

    Phenobarbital was synthesized by Emil Fischer at Bayer in 1911and introduced as an ASD by Alfred Hauptmann in 1912. Itstendency to produce sedation and cognitive slowing or confusion,as well as paradoxical hyperactivity in children, has curtailed itsuse in favor of more modern alternatives.77 In addition to theirenhancement of GABAAR currents, barbiturates also inhibitrepetitive action potential ring at neuronal sodium channels78

    by reducing fractional open time and shifting the potential of half-maximal opening towards hyperpolarized potentials.79 Theseactions contribute to both their antiseizure action and theiradverse effect prole.

    When co-applied with low concentrations of GABA, barbitu-rates increased the mean open time, but had no effect on thelengths of 3 distinct open state durations.80 The increase in meanopen time resulted from fewer openings in the shorter twodurations (O1 and O2) and more long duration (O3) openings.There was also an increase in long burst durations, but no changein the single channel opening frequency or in the closedfrequency duration histogram. The increase in channel opentime results in greater chloride current ux and increasedlikelihood that channel openings will summate, producing largerinhibitory currents. In the absence of GABA, high concentrationsof pentobarbital (EC50 0.33 mM) and phenobarbital (EC50 3 mM)directly activated GABAAR chloride currents with lower efcacy(smaller maximal currents) than GABA82. Barbiturate-activatedGABAAR currents were blocked by bicuculline and picrotoxin,and at high concentrations both phenobarbital and pentobarbi-tal produced open channel block that rapidly terminated theinduced currents. The concentrations involved in both directactivation and open channel block are far higher than usualtherapeutic levels, thus the main antiseizure mechanism is likelythe GABAAR allosteric effect in concert with synaptic andextrasynaptic GABA.

    Unlike the BZs, barbiturate sensitivity does not require aspecic subunit composition. Homomeric b1 receptors expressedin Xenopus oocytes were directly activated by pentobarbital eventhough they were not responsive to GABA; these currents wereblocked by picrotoxin or penicillin but not bicuculline.83

    Pentobarbital also induced current in both a1-b3 and b3homomeric GABAARs.

    84 At saturating GABA concentrations,pentobarbital markedly potentiated a1-b3-d currents, increasingdesensitization and single channel open duration,85 suggestingthat barbiturates may be particularly effective at enhancing tonicGABAAR currents at extrasynaptic sites activated by low GABAconcentrations. The e subunit, which increases spontaneousGABAAR channel openings, signicantly reduced

    86 but did notcompletely eliminate87 barbiturate responsiveness when co-expressed with a and b subunits. These ndings suggest thatbarbiturate sensitivity does not require either an a or a g subunitas it is present in b homomeric receptors; however, presence of

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600 593the d or e subunits that substitute for g can dramatically modifybarbiturate responsiveness by altering the efcacy of GABA foractivating the channel.

    Chimera and mutagenesis studies have identied domains andresidues that contribute to barbiturate action, though in less detailthan for the BZs. Mutation of the proline (P228A) in the rsttransmembrane domain of the b1 subunit reduced barbiturateenhancement of GABA-evoked currents in a1-b1-g2L GABAARswhile increasing apparent GABA afnity, without otherwisealtering single channel kinetics.88 A chimera study usingconstructs created from the amino terminal end of the b3 subunitand the carboxyl terminal end of the r1 subunit, which formshomomeric GABAC receptors that are insensitive to barbiturates,found that residues of the b3 subunit involved in pentobarbitalbinding to GABAARs are located downstream from the middle ofthe M2 region.89 Similarly, mutation of a r1 tryptophan (W328) inthe third transmembrane domain to a hydrophobic residueproduced both allosteric and direct channel activation bypentobarbital.90

    4. Loreclezole

    Loreclezole has antiseizure activity in a variety of seizuremodels, acting more like a barbiturate than a BZ in that theincrease in seizure threshold produced by loreclezole waspotentiated rather than blocked by the BZ antagonist, umaze-nil.91 In the hippocampal slice, loreclezole, potentiated pairedpulse inhibition92 and inhibited epileptiform discharges inducedby low Ca2+ or low Mg2+. Loreclezole strongly potentiatedrecombinant GABAARs containing a b2 or b3 subunit but didnot enhance currents from b1-containing receptors.93 A singleasparagine residue (b2(N289) or b3(N290)) at the cytoplasmicend of the 2nd transmembrane domain confers sensitivity toloreclezole; this amino acid is a serine in the b1 subunit.94

    Mutation of b1S290 to N conferred loreclezole sensitivity to b1-containing GABAARs, while mutation of b2N289 or b3N290 to Seliminated loreclezole enhancement. When both b1 and b3 wereco-expressed in the same receptor, loreclezole sensitivity wasabolished, suggesting a dominant effect of the b1 subunit.95

    Coexpression with different alpha subunits altered the degree ofloreclezole potentiation (a1 = a2 = a3 > a5 > a4);96 expressionof a5 and b3 with the p subunit reduced loreclezole potentia-tion,97 while a1-b3-e receptors showed normal loreclezoleenhancement.87

    At higher concentrations (> 6 mM) loreclezole causedconcentration-dependent inhibition of GABAAR currents byenhancing the rate of apparent desensitization.98 The effectwas inconsistent with open channel block as it was voltageindependent, non-competitive, and increased with increasingGABA concentration. The BZ site was not involved as theinhibition was not antagonized by umazenil and did not requirea g subunit. This nding has important clinical implications, sinceinadvertent high levels of drug could inhibit rather than enhanceGABAAR function and potentially trigger seizure activity.Loreclezole inhibited a1-b1-g2L GABAAR currents that werenot potentiated by low concentrations of loreclezole, suggestingseparate sites for enhancement and inhibition. At the singlechannel level, high loreclezole concentrations decreased a1-b1-g2L mean open time by decreasing the average durations of theopen states, and also increased the occurrence of a 20 ms closedstate. Loreclezole inhibition was equally effective when appliedto the intracellular side of the receptor, suggesting that itsinhibitory binding site was accessible from both sides of themembrane, and pre-application of loreclezole prior to GABAinhibited the subsequent GABAAR current, indicating thatbinding did not require an open channel. Loreclezole was alsofound to inhibit r1 homomeric GABAC receptors with an IC50 of0.5 mM, which was proposed as a rapid means of pharmacologicalidentication of these receptors.99

    5. Ganaxolone

    Ganaxolone (3a-OH-3b-methyl-5a-pregnan-20-one) is the3b-methylated synthetic analog of the neurosteroid, allopregna-nolone, a natural metabolite of progesterone that allostericallyenhances GABAAR current. It has a broad range of antiseizureactivity in animal epilepsy models100 including seizures inducedby bicuculline, t-butylbicyclophosphorothionate (TBPS, a highafnity ligand for the GABAAR picrotoxin site), aminophylline andcorneal kindling, and is well tolerated and effective againstseizures in humans. Ganaxolone is currently under evaluation forpartial onset seizures. It may have special utility in women withcatamenial epilepsy who have increased seizures during periods oflow progesterone in the menstrual cycle, as well as in children withinfantile spasms.101

    The antiseizure effect of neurosteroids is not due to action at theprogesterone receptor (PR), as exogenous allopregnanolone pre-vented seizures in PR knockout mice.102 Moreover, progesteronesantiseizure effect requires metabolism to allopregnanolone, as itwas blocked by nasteride, a 5a-reductase inhibitor that blocksallopregnanolone synthesis from progesterone. Hence, the antisei-zure effect of allopregnanolone (and ganaxolone) is mediated byGABAARs.

    Specic neurosteroids can allosterically modulate GABAARactivity, either positively or negatively, via distinct binding siteson GABAARs.

    104 Since a variety of GABAAR-interacting steroidhormones are synthesized in the brain,105 these agents representendogenous modulators of GABAAR function. Allopregnanolone issynthesized in two steps from progesterone: 5a-reductaseconverts progesterone to 5a-dihydroprogesterone, then 3-ahydroxysteroid oxidoreductase reversibly converts 5a-dihydro-progesterone to 3a-OH-5a-pregnan-20-one (allopregnanolone).Addition of the 3b-methyl group on ganaxolone does not alterbinding to GABAARs, but prevents further metabolism of the 3a-OH group and thus prolongs its GABAAR-modulating actions.

    100

    Ganaxolone enhanced GABA and BZ binding via positive allostericmodulation of GABAAR activity, and at nanomolar concentrationspotentiated GABA-evoked currents at a1-b1-g2L, a2-b1-g2L ora3-b1-g2L GABAARs expressed in Xenopus oocytes, while directactivation of chloride ux occurred to a limited extent only atmicromolar concentrations.100

    The effects of GABAAR-enhancing neurosteroids on singlechannel GABAAR currents were studied using androsterone(5a-androstan-3a-ol-17-one) and pregnanolone (5b-pregnan-3a-ol-20-one) but likely apply to other positive steroidGABAAR modulators including ganaxolone. Like the barbitu-rates and loreclezole, these agents increased the proportion ofopenings to the two longer open states (O2 and O3) withoutaltering the intrinsic durations of those states, and producedlonger burst durations.106 There was no change in singlechannel conductance. Unlike the barbiturates, however, theneurosteroids also increased single channel opening frequencyand reduced closed time durations in all but the shortest closedtime distributions (thought to represent intraburst closures).At high concentrations (10 mM), both agents reduced openchannel durations consistent with open channel (ickering)block.

    The subunit selectivity of GABAAR-enhancing neurosteroidswas initially controversial. An early study107 of recombinantreceptors expressed from combinations of a1, a6, b3, g2 and dshowed loss of neurosteroid responsiveness with combinationscontaining the delta subunit. There was also reduced sensitivity of

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600594cerebellar granule neurons to neurosteroids later in in vitrodevelopment, when d subunit expression increases. These ndingswere interpreted as showing inhibition of neurosteroid respon-siveness by inclusion of the d subunit. However, mice in which thed subunit was knocked out showed decreased behavioralsensitivity to neurosteroids.108 Moreover, in receptors composedof a1 or a6, b3 and either g2L or d, the greatest potentiation bytetrahydrodeoxycorticosterone (THDOC) was seen in a1-b3-dGABAARs.

    109 At high concentrations (1 mM), THDOC inhibited thisisoform. There is currently consensus that d subunit-containingreceptors are more sensitive to neurosteroid enhancement, thoughother subunits are also involved in mediating neurosteroid actions.Presence of the a6 subunit reduced neurosteroid sensitivity.110

    THDOC (1 mM) enhanced a1-b3-d more than a6-b3-d currents,but increased the extent of desensitization and prolongeddeactivation for both receptor isoforms; a1-a6 and a6-a1chimeras (spliced in transmembrane domain M1) suggested thatdifferences in deactivation rate and its voltage-dependencecorrelated with N-terminal domains, while the extent of desensi-tization and its voltage-dependence correlated with C-terminaldomains.111 In dentate granule cells from epileptic animals, anincrease in a4 subunit expression was associated with decreasedneurosteroid enhancement.112 In receptors composed of one asubunit (from a2 through a5), b2 and g2S, neurosteroidpotentiation was dependent on the conserved glutamine residue(a1(Q241)) in the rst transmembrane domain of the respective asubunit.113 The d subunit did not affect neurosteroid binding butlikely inuenced the efcacy of neurosteroid potentiation. Muta-tion of a1Q241 to L abolished neurosteroid potentiation, whilemutation to W mimicked the effect of steroids and preventedfurther augmentation; the neighboring S240 residue also partici-pated in steroid binding.114 A subsequent study using concatenat-ed subunits demonstrated that a single functional binding site (notdisrupted by the Q241 mutation) was sufcient for neurosteroidmodulation.115

    6. GABAAR effects of other ASDs

    6.1. Topiramate

    Topiramate is a heterotricyclic sulfamate with several mecha-nisms of action including inhibition of sodium and calciumchannels, inhibition of carbonic anhydrase, and augmentation ofGABA-evoked currents. Topiramates effects on GABAARs maycontribute both to its antiseizure efcacy and its side effect proleincluding memory problems, fatigue and psychomotor slowing. Itis approved for partial and generalized seizures, the LennoxGastaut syndrome, and migraine.

    Topiramate inhibited voltage-gated sodium channels, with aleft shift of the steady state inactivation curve116 resulting inintermittent blockade of sustained repetitive action potentialring during prolonged depolarization.117 It also blocked repetitivering in hippocampal CA3 neurons from spontaneously epilepticrats, and reduced excitatory post-synaptic potentials andresponses to bath-applied glutamate, suggesting blockade ofpost-synaptic glutamate receptors,118 affecting kainate-evokedbut not NMDA-evoked currents.119 Topiramate also inhibited bothL-type and non L-type high voltage activated calcium currents120

    and reduced bicarbonate production via inhibition of carbonicanhydrase,121 which may contribute to its antiseizure effect.

    Topiramate (10 mM) enhanced chloride ux into cerebellargranule neurons stimulated by 10 mM GABA, but did notsignicantly increase chloride inux alone.122 It also enhancedGABA-evoked currents in cultured cortical neurons that wereinsensitive to the BZ, clonazepam, and clonazepam-potentiatedGABA currents in topiramate-insensitive neurons, conrming thattopiramates site of action on GABAARs is independent of the BZsite.123 Subunit selectivity studies have been inconsistent.Topiramate (1100 mM) reversibly inhibited Cl currents evokedby 110 mM GABA in Xenopus oocytes expressing a1-b2-g2S anda2-b2-g2S GABAARs, and reduced the apparent desensitization(current-fading rate) in a1-b2-g2S-expressing oocytes, butpotentiated GABA-evoked Cl currents and increased the desensi-tization rate in a6-b2-g2S GABAARs, with no effect on a4-b2-g2Sreceptors or mixed population GABAARs expressed from rat brainmRNA.124 In contrast, another study found that topiramate couldboth potentiate and directly activate b2 or b3-containingheteromeric GABAARs, with greatest effect on a4-b3-g2S;

    125

    positive or negative effects on b1-containing GABAARs dependedon the co-expressed alpha subunit. Additional studies are neededto clarify the site and mechanism of action.

    6.2. Felbamate

    Felbamate was launched as a promising novel ASD in 1993 withan uncertain mechanism of action, but its use was curtailed afterearly reports of aplastic anemia126 and hepatic failure.127

    Felbamate inhibited binding of the GABA antagonist [3H]T-BOBwith a regional pattern different from that produced by GABAagonists, bicuculline, zinc or neurosteroids,128 and enhancedGABA-elicited Cl currents in cultured cortical neurons. Felbamateenhancement was not blocked by umazenil, and felbamate didnot affect pentobarbital potentiation or PTX inhibition of GABA-evoked currents, suggesting an independent site of action. Itprolonged the mean burst duration of GABA-activated singlechannel currents, suggesting a barbiturate-like effect.129 Felba-mate also blocked N-methyl-D-aspartate (NMDA) receptor cur-rents, an alternative possible antiseizure mechanism.130 Derivativecompounds including uorofelbamate and carisbamate that arenot associated with hematopoietic or hepatic toxicity may reviveinterest in felbamatelike agents.131

    6.3. Ezogabine

    Ezogabine (also known as retigabine) is a novel ASD effective ina variety of animal models. Its primary effect is enhancedactivation of heteromeric potassium channels composed of theKCNQ2 and KCNQ3 subunits132 which underlie the M currentthat is a major determinant of resting membrane potential andneuronal excitability. However, ezogabine also dose-dependentlyand reversibly potentiated GABAAR-dependent IPSC peak ampli-tude, decay times and total charge transfer.133 EPSCs wereunaffected, and paired pulse depression was unchanged, suggest-ing a post-synaptic effect at GABAARs. Ezogabine potentiatedGABA-induced currents in rat cortical neurons in a concentration-dependent fashion at 10 mM and above.132 This action was notantagonized by umazenil, indicating a non-BZ site of action.Subunit dependence and binding site are not known.

    6.4. Losigamone

    Losigamone is a novel ASD which inhibited the persistentcomponent of sodium currents in hippocampal neurons atdepolarized potentials, suppressed sustained repetitive ring134

    and decreased the frequency of spontaneous action potentialswithout affecting miniature post-synaptic current amplitudes.135

    Losigamone stimulated 36Cl inux into spinal cord neurons in theabsence of GABA, and potentiated 36Cl inux stimulated bysubmaximal GABA concentrations; both effects were blocked bybicuculline or picrotoxin.136 Losigamone did not affect the specicbinding of [3H]GABA, [3H]unitrazepam, or [35S]t-butyl-bicyclo-phosphorothionate (TBPS) to their receptors, and there was

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600 595no difference in the effect of the + or stereoisomers or the racemicmixture. The site of action at GABAARs is unknown.

    6.5. Stiripentol

    Stiripentol is as an adjunct ASD which was thought to act byinhibiting cytochrome P450 enzymes involved in metabolism ofconventional ASDs. Recently, stiripentol was found to enhancerecombinant GABAAR currents, with greater potentiation of a3-containing receptors and reduced potentiation with the b1 or esubunits.137 It caused a leftward shift in the GABA concentration-response relationship without increasing maximal GABA-evokedcurrents, and did not involve sites associated with neurosteroid orloreclezole potentiation.137 Saturating barbiturate sites withpentobarbital occluded stiripentol enhancement, and stiripentolincreased the duration but not the frequency of opening of GABAARchannels, suggesting a barbiturate-like mechanism.138

    6.6. GABA-enhancing agents

    An additional mechanism for enhancing GABAAR-mediatedinhibition involves increasing the concentration or duration ofGABA in the synaptic cleft and perisynaptic/extrasynaptic sites.Enhancing GABA synthesis or blocking its reuptake or catabolismcould prolong GABA IPSPs and increase perisynaptic spill-over,increasing tonic/extrasynaptic GABA currents which likely play amajor role in seizure prevention.

    6.7. Gabapentin and pregabalin

    Gabapentin was designed as a GABA analog, and some studieshave suggested that it modulates the action of the GABA syntheticenzyme, glutamic acid decarboxylase (GAD) and the glutamatesynthesizing enzyme, branched-chain amino acid transaminase,resulting in increased GABA synthesis.139 Gabapentin increasesnon-synaptic GABA responses from neuronal tissues in vitro andincreases GABA levels in brain.140 Its other (likely primary) mode ofaction involves binding to the a2-d binding site on L- or P/Q-typepresynaptic voltage-gated calcium channels, presumably inhibit-ing excessive neurotransmitter release141 by interfering withcalcium channel functional expression or trafcking142. Pregaba-lin, which binds to the a2-d calcium channel subunit with higherpotency, may have similar mechanisms of action. Most studiesemphasize the calcium channel as the primary site,143 though acontributory effect on GABA metabolism is possible.144

    6.8. Valproic acid

    Valproic acid may affect GABA production, among other ASDmechanisms. Valproate caused non-signicant increases in cere-bral GABA levels but elevated brain GAD activity signicantly.145

    Valproic acid increased GABA synthesis and release in brainregions including substantia nigra,146 which is thought to beinvolved in the control of seizure generation and propagation.147 Italso reduced the release of gamma-hydroxybutyric acid andattenuated neuronal excitation induced by NMDA-type glutamatereceptors.147 Additional ASD mechanisms include enhancingsodium channel inactivation (like phenytoin) and reducing T-typeCa2+ channel currents (like ethosuximide),148 perhaps explainingits utility against both partial onset and absence seizures.

    6.9. Tiagabine

    Tiagabine is a derivative of nipecotic acid that binds to thepresynaptic GAT-1 GABA transporter and blocks GABA reuptake,without acting as a false neurotransmitter. In controlhippocampal slices, tiagabine alone induced a signicantchloride conductance, suggesting that GAT-1 activity controlsthe basal level of extracellular GABA.149 Tiagabine prolongedhippocampal IPSC duration in a lamina-specic fashion, withgreater effect in stratum radiatum (167%) than stratum oriens(115%).150 Tiagabine reduced the frequency of epileptiformdischarges in hippocampal slices exposed to low Mg2+ or 4-aminopyridine.151 The ability of tiagabine to prolong GABAcurrents was preserved in hippocampal slices from pilocarpine-treated epileptic rats, hence the GAT-1 transporter remains afunctional target for regulating GABA levels in this model oftemporal lobe epilepsy.152 Tiagabine was effective as an adjunctagent against partial onset seizures, though its prolongedtitration period, requirement for multiple daily doses and CNSside effect prole (dizziness, tremor, somnolence, mood dis-turbances and rare psychotic symptoms) have limited its use.153

    6.10. Vigabatrin

    Vigabatrin (g-vinyl-GABA) has a unique mechanism of action,functioning as a suicide inhibitor of GABA transaminase, theprimary enzyme for GABA catalysis. This leads to permanentinhibition of the affected enzyme, requiring synthesis of newGABA transaminase and hence prolonging the biological half-lifebeyond its pharmacokinetic half-life of 8 hours.154 It is effectiveagainst complex partial seizures,155 LennoxGastaut syndromeand West Syndrome, for which it has become an alternative toACTH156 particularly in children with tuberous sclerosis.157

    Vigabatrin increases the extracellular concentrations of GABAboth in vitro158 and in vivo.159 The anti-seizure effects ofincreasing GABA concentration may be complex. Biphasicresponses to vigabatrin administration have been observed, withan early proconvulsant effect.160 A biphasic effect was also seen onafter-discharge duration with an early facilitation of the limbicpattern of epileptiform discharge and later suppression at higherdoses.161 Elevated GABA concentrations may increase neuronalsynchronization, particularly of thalamic neurons in absenceepilepsy, and there have been several reports of vigabatrinworsening absence seizures or precipitating absence statusepilepticus.162

    A major limitation to the use of vigabatrin is the relatively highincidence of vision disturbance associated with chronic use. In 21of 30 children treated with vigabatrin who were screened for visiondisturbances despite no report of visual symptoms, 4 had visualeld constrictions, which did not improve after discontinuation ofthe drug.163 One study associated visual disturbances withinfantile spasms rather than the drug.164 The prevalence of visionproblems ranged from 10 to 40% of pediatric and adult patientsexposed to vigabatrin.165 This adverse effect may be directlyrelated to elevated GABA concentrations, as hippocampal neuronsgrown in depolarizing conditions (25 mM KCl) showed 20% loss ofMAP-2-positive neurons in the presence of vigabatrin, which wasmimicked by GABA (100 mM) and blocked by bicuculline orpicrotoxin.166 The cellular mechanism of this toxicity is notentirely clear, but in situations in which GABA is depolarizing,excessive GABA could promote calcium entry through L-type(slowly desensitizing) voltage-gated calcium channels,167 which inturn could result in activation of caspases and cell death.168

    7. Excitatory GABAA currents

    Early in CNS development, neurons express the Na+/K+/Cl

    cotransporter, NKCC1, rather than the K+/Cl cotransporter, KCC2,which is expressed in adult neurons. NKCC1 increases intracellularCl resulting in a depolarizing Cl reversal potential, while KCC2exports Cl yielding the hyperpolarizing Cl reversal potential

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600596found in adult neurons.169 As a result, activation of GABAARs maybe excitatory during early development.170 Excitatory GABAARcurrents may play a trophic role in neuronal migration andconnectivity,171 but may also contribute to epileptogenesis.172

    Endogenous GABA appears to be proconvulsant in early postnatalrat hippocampal slices, as GABAA antagonists blocked epileptiformactivity induced by depolarization with high external [K+]173.However, BZ and barbiturate antiseizure efcacy appear to beintact, likely because persistent opening of GABAAR channels (inthe presence of allosteric agents) may reduce the depolarizingchloride reversal potential, resulting in shunt inhibition.Alternatively, subthreshhold GABA-evoked depolarization mayinactivate sodium channels and prevent action potential ring.174

    The current through GABAAR channels can also be altered bychanges in intracellular bicarbonate, [HCO3

    ],169 which can owthrough the channel.175 Changes in [HCO3

    ] may underlie reducedsynaptic GABA currents during development of BZ tolerance.176

    Depolarizing GABAAR currents may also be a source of interictalspike activity, as observed in epileptic subiculum neurons inhippocampal brain slices from patients with temporal lobeepilepsy.177 Changes in the GABA current reversal potential mightalso explain why diazepam can be less effective in children withepileptic encephalopathies,178 and rarely can cause status epilep-ticus in patients with the Lennox Gastaut syndrome.179

    Bumetanide, an inhibitor of NKCC-1 used clinically as a potentloop diuretic, has been proposed as an adjunctive agent to reduceintraneuronal chloride accumulation and reverse the depolarizingchloride gradient that makes GABAARs excitatory, particularly inthe neonatal setting.170 Bumetanide improved the responsivenessof hippocampal slices to phenobarbital in reducing epileptiformdischarges induced by low Mg2+,180 and also suppressed electro-graphic seizures in neonatal rats.172 Depolarizing GABA currentsmay occur in cortical more than subcortical brain regions, possiblyexplaining the electroclinical uncoupling that occurs with pheno-barbital in neonatal seizures.181

    Bumetanide has been used successfully in a single reportedhuman neonate with seizures,182 and a clinical trial(NCT00830531) is in progress. However, elimination of thedepolarizing Cl gradient during development may have adverseconsequences; a permanent reduction in AMPAR-mediatedexcitatory neurotransmission and sensorimotor gating decitswere observed after bumetanide exposure in neonatal rats.183 Suchpotential adverse effects will have to be weighed against thebenets associated with seizure termination or prevention.

    8. GABAAR subunit changes and ASD efcacy in epilepsy andstatus epilepticus

    There is considerable evidence that GABAAR composition andfunction change with epilepsy, both acutely in the setting of statusepilepticus and in chronic epilepsy. During status epilepticus, thereis a rapid change in GABAAR function that reduces BZ sensitivity,

    184

    apparently due to activity-dependent internalization of GABAARsand replacement with BZ-insensitive receptors.185 Kainic acid-induced status epilepticus in young (postnatal day 9) rats alteredthe normal developmental pattern of GABAAR subunit matura-tion,186 preventing the normal switch from a2 to a1-containingGABAARs. These changes correlate with the nding in animalmodels15,184,187 and humans188,189 that BZs are effective early inthe course of status epilepticus but lose their potency and efcacyfor termination of status with longer seizure duration. Thesendings increase the urgency for treatment of status epilepticusearly in its course, and also suggest the need to nd alternativetreatments that retain efcacy in refractory status epilepticus, ormethods to prevent or reverse the GABAAR changes that occur withprolonged seizures.In the pilocarpine model of temporal lobe epilepsy, the a1subunit in dentate granule neurons was downregulated and a4subunit was upregulated,190 while in younger (postnatal day 20)animals, the a1 subunit was increased after status epilepticus.191

    GABAAR subunit changes after pilocarpine-induced status areassociated with altered physiological properties including reducedneurosteroid sensitivity and an increase in a4-containing recep-tors colocalized with synapses rather than extracellular sites.112

    Such changes might contribute to the propensity for spontaneousseizures or alternatively represent a functional adaptation toreduce seizure expression. Substitution of a1 with a4 mightreduce synaptic (phasic) but increase extrasynaptic (tonic)inhibition, and BZ sensitivity would be lost in a4-containingreceptors. Using an adenovirus vector to insert the a1 subunit genedriven by the a4 promoter resulted in a decrease in seizurefrequency, suggesting that the GABAAR subunit changes werecontributory toward epileptogenesis.192 Hence, altering GABAARsubunit expression in brain regions critical to seizure developmentmay provide a new therapeutic strategy for seizure prevention.Alternatively, novel ASDs targeting the specic epilepsy-associatedsubunit composition might provide a less invasive approachtoward seizure control.

    Long term exposure to the GABAAR allosteric agentsthemselves can alter GABAAR composition and function. In ratpups exposed to therapeutic concentrations of diazepam orphenobarbital from postnatal day 10 through 40, then taperedfor 2 weeks and euthanized on postnatal day 90 showedincreased mRNA expression in dentate granule neurons forGAT-1, 3 and 4, GABAAR subunits a4, a6, b3, d and u and GABABreceptor subunit R1, and decreased mRNA expression for GAD65,GAD67 and GABAAR subunits a1 and a3.

    193 Tolerance- anddependence-associated changes in GABAAR subunit compositionwith chronic use of BZs194,195 and barbiturates196,197 have beenreported, and may reect tolerance to either the antiseizure oradverse CNS effects. However, these ndings suggest that notonly seizures, but their prolonged treatment with GABAergicagents, can alter inhibitory neuronal function for extendedperiods or even permanently, with possible cognitive andbehavioral consequences that must be considered whencontemplating chronic GABAergic ASD use.

    References

    1. Treiman DM. GABAergic mechanisms in epilepsy. Epilepsia 2001;42(Suppl3):812.

    2. Olsen RW, DeLorey TM, Gordey M, Kang MH. GABA receptor function andepilepsy. Advances in Neurology 1999;79:499510.

    3. Ben-Ari Y. Seizures beget seizures: the quest for GABA as a key player. CriticalReviews in Neurobiology 2006;18:13544.

    4. Sperk G, Furtinger S, Schwarzer C, Pirker S. GABA and its receptors in epilepsy.Advances in Experimental Medicine and Biology 2004;548:92103.

    5. Macdonald RL, Olsen RW. GABAA receptor channels. Annual Reviews of Neuro-science 1994;17:569602.

    6. Nayeem N, Green TP, Martin IL, Barnard EA. Quaternary structure of the nativeGABAA receptor determined by electron microscopic image analysis. Journal ofNeurochemistry 1994;62:8158.

    7. Hedblom E, Kirkness EF. A novel class of GABAA receptor subunit in tissues ofthe reproductive system. Journal of Biological Chemistry 1997;272:1534650.

    8. Davies PA, Hanna MC, Hales TG, Kirkness EF. Insensitivity to anesthetic agentsconferred by a class of GABAA receptor subunit. Nature 1997;385:8203.

    9. Bonnert TP, McKernan RM, Le Bourdelles B, Smith DW, Hewson L, Rigby MR,Sirinathsinghji DJ, Brown N, Wafford K, Whiting PJ. Q, a novel g-aminobutyricacid type A subunit. Proceedings of the National Academy of Sciences of the UnitedStates of America 1999;96:98916.

    10. Wisden W, Laurie DJ, Monyer H, Seeburg PH. The distribution of 13 GABAAreceptor subunit mRNAs in the rat brain. I. Telencephalon, diencephalon,mesencephalon. Journal of Neuroscience 1992;12:104062.

    11. Brooks-Kayal AR, Jin H, Price M, Dichter MA. Developmental expression ofGABAA receptor subunit mRNAs in individual hippocampal neurons in vitroand in vivo. Journal of Neurochemistry 1998;70:101728.

    12. Pritchett DB, Sontheimer H, Shivers BD, Ymer S, Kettenmann H, Schoeld PR,Seeburg PH. Importance of a novel GABAA subunit for benzodiazepine phar-macology. Nature 1989;338:5825.

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600 59713. Wingrove PB, Wafford KA, Bain C, Whiting PJ. The modulatory action ofloreclezole at the g-aminobutyric acid type A receptor is determined by asingle amino acid in the b2 and b3 subunit. Proceedings of the National Academyof Sciences of the United States of America 1994;91:456973.

    14. Draguhn A, Verdorn TA, Ewert M, Seeburg PH, Sakmann B. Functional andmolecular distinction between recombinant rat GABAA receptor subtypes byZn2+. Neuron 1990;5:7818.

    15. Jones-Davis DM, Macdonald RL. GABA(A) receptor function and pharmacologyin epilepsy and status epilepticus. Current Opinion in Pharmacology 2003;3:128.

    16. Kokaia M, Pratt GD, Elmer E, Bengzon J, Fritschy JM, Kokaia Z, Lindvall O,Mohler H. Biphasic differential changes of GABAA receptor subunit mRNAlevels in denate gyrus granule cells following recurrent kindling-inducedseizures. Molecular Brain Research 1994;23:32332.

    17. Brooks-Kayal AR, Shumate MD, Jin H, Rikhter TY, Coulter DA. Selective changesin single cell GABAA receptor subunit expression and function in temporal lobeepilepsy. Nature Medicine 1998;4:116672.

    18. Loup F, Weiser HG, Yonekawa Y, Aguzzi A, Fritschy JM. Selective alterations inGABAA receptor subtypes in human temporal lobe epilepsy. Journal of Neuro-science 2000;20(14):540119.

    20. Matsumoto A, Kumagai T, Miura K, Miyazaki S, Hayakawa C, Yamanaka T.Epilepsy in Angelman syndrome associated with chromosome 15q deletion.Epilepsia 1992;33:108390.

    21. DeLorey TM, Handforth A, Anagnostaras SG, Homanics GE, Minassian BA,Asatourian A, Fanselow MS, Delgado-Escueta A, Ellison GD, Olsen RW. Micelacking the beta3 subunit of the GABAA receptor have the epilepsy phenotypeand many of the behavioral characteristics of Angelman syndrome. Journal ofNeuroscience 1998;18:850514.

    22. Bianchi MT, Song L, Zhang H, Macdonald RL. Two different mechanisms ofdisinhibition produced by GABAA receptor mutations linked to epilepsy inhumans. Journal of Neuroscience 2002;22(13):53217.

    23. Baulac S, Huberfeld G, Gournkel-An I, Mitropoulou G, Beranger A, Prud-homme J-F, Baulac M, Brice A, Bruzzone R, LeGuern E. First genetic evidence ofGABAA receptor dysfunction in epilepsy: a mutation in the g2-subunit gene.Nature Genetics 2001;28:468.

    24. Wallace RH, Marini C, Petrou S, Harkin LA, Bowser DN, Panchal RG, WilliamsDA, Sutherland GR, Mulley JC, Scheffer IE, Berkovic SF. Mutant GABA(A)receptor gamma2-subunit in childhood absence epilepsy and febrile seizures.Nature Genetics 2001;28:4952.

    25. Cossette P, Liu L, Brisebois K, Dong H, Lortie A, Vanasse Ml, Saint-Hilaire JM,Carmant L, Verner A, Lu WY, Wang YT, Rouleau GA. Mutation of GABRA1 in anautosomal dominant form of juvenile myoclonic epilepsy. Nature Genetics2002;31:1849.

    26. Bowser DN, Wagner DA, Czajkowski C, Cromer BA, Parker MW, Wallace RH,Harkin LA, Mulley JC, Marini C, Berkovic SF, Williams DA, Jones MV, Petrou S.Altered kinetics and benzodiazepine sensitivity of a GABAA receptor subunitmutation [gamma 2(R43Q)] found in human epilepsy. Proceedings of theNational Academy of Sciences of the United States of America 2002;99:151705.

    27. Frugier G, Coussen F, Giraud MF, Odessa MF, Emerit MB, Boue-Grabot E, GarretM. A gamma 2(R43Q) mutation, linked to epilepsy in humans, alters GABAAreceptor assembly and modies subunit composition on the cell surface.Journal of Biological Chemistry 2007;282:381928.

    28. Hales TG, Tang H, Bollan KA, Johnson SJ, King DP, McDonald NA, Cheng A,Connolly CN. The epilepsy mutation, gamma2(R43Q) disrupts a highly con-served inter-subunit contact site, perturbing the biogenesis of GABAA recep-tors. Molecular and Cellular Neuroscience 2005;29:1207.

    29. Sancar F, Czajkowski C. A GABAA receptor mutation linked to human epilepsy(gamma2R43Q) impairs cell surface expression of alphabetagamma receptors.Journal of Biological Chemistry 2004;279:470349.

    30. Kang JQ, Macdonald RL. The GABAA receptor gamma2 subunit R43Q mutationlinked to childhood absence epilepsy and febrile seizures causes retention ofalpha1beta2gamma2S receptors in the endoplasmic reticulum. Journal ofNeuroscience 2004;24:86727.

    31. Sternbach LH, Reeder E. Quinazolines and 1, 4-benzodiazepines, IV: transfor-mations of 7-chloro-2-methylamino-5-phenyl-3H-1,4-benzodiazepine-4-ox-ide. Journal of Organic Chemistry 1961;26:493641.

    32. Sternbach LH, Fryer RI, Keller O, et al. Quinazolines and 1, 4-benzodiazepines,X: nitro-substituted 5-phenyl-1,4-benzodiazepine derivatives. Journal of Me-dicinal Chemistry 1963;6:2615.

    33. Gastaut H, Naquet R, Poire R, Tassarini CH. Treatment of status epilepticuswith diazepam (valium). Epilepsia 1965;6:16782.

    34. Sato S. Benzodiazepines, clonazepam. In: Levy RH, Mattson RH, Meldrum BS,editors. Antiepileptic drugs. New York: Raven Press; 1995 . p. 72534.

    35. Chapman AG, Horton RW, Meldrum BS. Anticonvulsant action of a1,5-benzodiazepine, clobazam, in reex epilepsy. Epilepsia 1978;19:2939.

    36. Munn R, Farrell K. Open study of clobazam in refractory epilepsy. PediatricNeurology 1993;9:4659.

    37. Shannon M, Albers G, Burkhardt K. Safety and efcacy of umazenil in thereversal of benzodiazepine-induced conscious sedation. Journal of Pediatrics1997;131:5826.

    39. Turski L, Stephens DN, Jensen LH, Petersen EN, Meldrum BS, Patel S,Hansen JB, Loscher W, Schneider HH, Schmiechen R. Anticonvulsantaction of the beta-carboline abecarnil: studies in rodents and baboon,Papio papio. Journal of Pharmacology and Experimental Therapeutics1990;253:34452.40. Zanotti A, Mariot R, Contarino A, Lipartiti M, Giusti P. Lack of anticonvulsanttolerance and benzodiazepine receptor downregulation with imidazenil inrats. British Journal of Pharmacology 1996;117:64752.

    41. Rundfeldt C, Wlaz P, Honack D, Loscher W. Anticonvulsant tolerance andwithdrawal characteristics of benzodiazepine receptor ligands in differ-ent seizure models in mice. Comparison of diazepam, bretazenil andabecarnil. Journal of Pharmacology and Experimental Therapy 1995;275:693702.

    42. Hernandez TD, Heninger C, Wilson MA, Gallager DW. Relationship of agonistefcacy to changes in GABA sensitivity and anticonvulsant tolerance followingchronic benzodiazepine ligand exposure. European Journal of Pharmacology1989;170(3):14555.

    43. Haefely W, Kyburz E, Gerecke M, Mohler H. Recent advances in the molecularpharmacology of benzodiazepine receptors and in the structure-activity rela-tionships of their agonists and antagonists. Advances in Drug Research1985;14:165322.

    44. Polc P. Electrophysiology of benzodiazepine receptor ligands: multiple mech-anisms and sites of action. Progress in Neurobiology 1988;31:349423.

    45. Randall LO, Kappell B. Pharmacological activity of some benzodiazepines andtheir metabolites. In: Garattini S, Mussini E, Randall LO, editors. The benzo-diazepines. New York: Raven Press; 1973. p. 2751.

    46. Rogawski MA, Porter RJ. Antiepileptic drugs: pharmacological mechanismsand clinical efcacy with consideration of promising developmental stagecompounds. Pharmacological reviews 1990;42(3):22386.

    47. Swinyard EA, Castellion AW. Anticonvulsant properties of some benzodiaze-pines. Journal of Pharmacology and Experimental Therapeutics 1966;151:36975.

    48. Albertson TE, Stark LG, Derlet RW. Modication of amygdaloid kindling bydiazepam in juvenile rats. Brain Research Developmental Brain Research1990;51(2):24952.

    49. Verdoorn TA, Draguhn A, Ymer S, Seeburg PH, Sakmann B. Functional proper-ties of recombinant rat GABAA receptors depend upon subunit composition.Neuron 1990;4:91928.

    50. Luddens H, Korpi ER, Seeburg P. GABAA/benzodizaepine receptor heterogene-ity: neurophysiological implications. Neuropharmacology 1995;34(3):24554.

    51. Smith GB, Olsen RW. Functional domains of GABAA receptors. Trends inPharmacological Sciences 1995;16(5):1628.

    52. Dunn SMJ, Davies M, Muntoni AL, Lambert JJ. Mutagenesis of the rat a1subunit of the g-aminobutyric acidA receptor reveals the importance ofresidue 101 in determining the allosteric effects of benzodiazepine siteligands. Molecular Pharmacology 1999;56:76874.

    53. Korpi ER, Kleingoor C, Kettenmann H, Seeburg PH. Benzodiazepine-inducedmotor impairment linked to point mutation in cerebellar GABAA receptor.Nature 1993;361:3569.

    54. McKernan RM, Rosahl TW, Reynolds DS, Sur C, Wafford KA, Atack JR, Farrar S,Myers J, Cook G, Ferris P, Garrett L, Bristow L, Marshall G, Macaulay A, Brown N,Howell O, Moore KW, Carling RW, Street LJ, Castro JL, Ragan CI, Dawson GR,Whiting PJ. Sedative but not anxiolytic properties of benzodiazepines aremediated by the GABAA receptor a1 subunit. Nature Neuroscience2000;3(6):52930.

    55. Crestani F, Martin JR, Mohler H, Rudolph U. Mechanism of action of thehypnotic zolpidem in vivo. British Journal of Pharmacology2000;131(7):12514.

    56. Low K, Crestani F, Keist R, Benke D, Brunig I, Benson JA, Fritschy JM, Rulicke T,Bluethmann H, Mohler H, Rudolph U. Molecular and neuronal substrate for theselective attenuation of anxiety. Science 2000;290:1314.

    57. Crestani F, Low K, Keist R, Mandelli M, Mohler H, Rudolph U. Molecular targetsfor the myorelaxant action of diazepam. Molecular Pharmacology 2001;59:4425.

    58. Crestani F, Keist R, Fritschy JM, Benke D, Vogt K, Prut L, Bluthmann H, Mohler H,Rudolph U. Trace fear conditioning involves hippocampal alpha5 GABA(A)receptors. Proceedings of the National Academy of Sciences of the United States ofAmerica 2002;99:89805.

    59. Knabl J, Zeilhofer UB, Crestani F, Rudolph U, Zeilhofer HU. Genuine antihyper-algesia by systemic diazepam revealed by experiments in GABAA receptorpoint-mutated mice. Pain 2009;141:2338.

    60. Dias R, Sheppard WF, Fradley RL, Garrett EM, Stanley JL, Tye SJ, Goodacre S,Lincoln RJ, Cook SM, Conley R, Hallett D, Humphries AC, Thompson SA, WaffordKA, Street LJ, Castro JL, Whiting PJ, Rosahl TW, Atack JR, McKernan RM, DawsonGR, Reynolds DS. Evidence for a signicant role of alpha 3-containing GABAAreceptors in mediating the anxiolytic effects of benzodiazepines. Journal ofNeuroscience 2005;25:106828.

    61. Rosenberg HC, Tietz EI, Chiu TH. Tolerance to anticonvulsant effects of diaze-pam, clonazepam, and clobazam in amygdala-kindled rats. Epilepsia1989;30:27685.

    62. Macdonald R, Barker JL. Benzodiazepines specically modulate GABA-medi-ated postsynaptic inhibition in cultured mammalian neurones. Nature1978;271:5634.

    63. Tietz EI, Zeng XJ, Chen S, Lilly SM, Rosenberg HC, Kometiani P. Antagonist-induced reversal of functional and structural measures of hippocampal ben-zodiazepine tolerance. Journal of Pharmacology and Experimental Therapeutics1999;291:93242.

    64. Study RE, Barker JL. Diazepam and ()-pentobarbital: uctuation analysisreveals different mechanisms for potentiation of g-aminobutyric acidresponses in cultured central neurons. Proceedings of the National Academyof Sciences of the United States of America 1981;78:71804.

  • L.J. Greeneld Jr. / Seizure 22 (2013) 58960059865. Rogers CJ, Twyman RE, Macdonald RL. Benzodiazepine and beta-carbolineregulation of single GABAA receptor channels of mouse spinal neurones inculture. Journal of Physiology (London) 1994;475:6982.

    66. Twyman RE, Rogers CJ, Macdonald RL. Differential regulation of g-aminobu-tyric acid receptor channels by diazepam and phenobarbital. Annals of Neu-rology 1989;25:21320.

    67. Bianchi MT, Botzolakis EJ, Lagrange AH, Macdonald RL. Benzodiazepine mod-ulation of GABA(A) receptor opening frequency depends on activation con-text: a patch clamp and simulation study. Epilepsy Research 2009;85:21220.

    68. Edwards FA, Konnerth A, Sakmann B. Quantal analysis of inhibitory synaptictransmission in the dentate gyrus of rat hippocampal slices: a patch-clampstudy. Journal of Physiology 1990;430:21349.

    69. Otis TS, Mody I. Modulation of decay kinetics and frequency of GABAAreceptor-mediated spontaneous inhibitory post-synaptic currents in hippo-campal neurons. Proceedings of the National Academy of Sciences of the UnitedStates of America 1992;78:71804.

    70. Eghbali M, Curmi JP, Birnir B, Gage PW. Hippocampal GABA(A) channelconductance increased by diazepam. Nature 1997;388:715.

    71. Eghbali M, Birnir B, Gage PW. Conductance of GABAA channels activated bypentobarbitone in hippocampal neurons from newborn rats. Journal of Physi-ology 2003;552:1322.

    72. Gaul S, Ozsarac N, Liu L, Fink RH, Gage PW. The neuroactive steroids alphax-alone and pregnanolone increase the conductance of single GABAA channels innewborn rat hippocampal neurons. The Journal of Steroid Biochemistry andMolecular Biology 2007;104:3544.

    73. Eghbali M, Gage PW, Birnir B. Effects of propofol on GABAA channel conduc-tance in rat-cultured hippocampal neurons. European Journal of Pharmacology2003;468:7582.

    74. Birnir B, Eghbali M, Cox GB, Gage PW. GABA concentration sets the conduc-tance of delayed GABAA channels in outside-out patches from rat hippocam-pal neurons. Journal of Membrane Biology 2001;181:17183.

    75. Everitt AB, Luu T, Cromer B, Tierney ML, Birnir B, Olsen RW, Gage PW.Conductance of recombinant GABA (A) channels is increased in cells co-expressing GABA(A) receptor-associated protein. Journal of Biological Chemis-try 2004;279:217016.

    76. Breitwieser GE. G protein-coupled receptor oligomerization: implications forG protein activation and cell signaling. Circulation Research 2004;94:1727.

    77. Kwan P, Brodie MJ. Phenobarbital for the treatment of epilepsy in the 21stcentury: a critical review. Epilepsia 2004;45:11419.

    78. Macdonald RL, McLean MJ. Anticonvulsant drugs: mechanisms of action. In:Delgado-Escueta AV, Ward Jr AA, Woodbury DM, Porter RJ, editors. Advances inneurology. New York: Raven Press; 1986. p. 71336.

    79. Rehberg B, Duch DS, Urban BW. The voltage-dependent action of pentobarbitalon batrachotoxin-modied human brain sodium channels. Biochimica et Bio-physica Acta 1994;1194:21522.

    80. Macdonald RL, Rogers CJ, Twyman RE. Barbiturate regulation of kinetic prop-erties of the GABAA receptor channel of mouse spinal neurones in culture.Journal of Physiology 1989;417:483500.

    82. Rho JM, Donevan SD, Rogawski MA. Direct activation of GABAA receptors bybarbiturates in cultured rat hippocampal neurons. Journal of Physiology1996;497(Pt 2):50922.

    83. Krishek BJ, Moss SJ, Smart TG. Homomeric beta 1 gamma-aminobutyric acid Areceptor-ion channels: evaluation of pharmacological and physiological prop-erties. Molecular Pharmacology 1996;49:494504.

    84. Davies PA, Kirkness EF, Hales TG. Modulation by general anaesthetics of ratGABAA receptors comprised of alpha 1 beta 3 and beta 3 subunits expressed inhuman embryonic kidney 293 cells. British Journal of Pharmacology1997;120:899909.

    85. Feng HJ, Bianchi MT, Macdonald RL. Pentobarbital differentially modulatesalpha1beta3delta and alpha1beta3gamma2L GABAA receptor currents. Mo-lecular Pharmacology 2004;66:9881003.

    86. Davies PA, Hanna MC, Hales TG, Kirkness EF. Insensitivity to anaesthetic agentsconferred by a class of GABA(A) receptor subunit. Nature 1997;385:8203.

    87. Neelands TR, Fisher JL, Bianchi M, Macdonald RL. Spontaneous and gamma-aminobutyric acid (GABA)-activated GABA(A) receptor channels formed byepsilon subunit-containing isoforms. Molecular Pharmacology 1999;55:16878.

    88. Greeneld Jr LJ, Zaman SH, Sutherland ML, Lummis SC, Niemeyer MI, BarnardEA, Macdonald RL. Mutation of the GABAA receptor M1 transmembraneproline increases GABA afnity and reduces barbiturate enhancement. Neuro-pharmacology 2002;42:50221.

    89. Serani R, Bracamontes J, Steinbach JH. Structural domains of the humanGABAA receptor 3 subunit involved in the actions of pentobarbital. Journal ofPhysiology 2000;524(Pt 3):64976.

    90. Amin J. A single hydrophobic residue confers barbiturate sensitivity to gam-ma-aminobutyric acid type C receptor. Molecular Pharmacology 1999;55:41123.

    91. Ashton D, Fransen J, Heeres J, Clincke GH, Janssen PA. In vivo studies on themechanism of action of the broad spectrum anticonvulsant loreclezole. Epi-lepsy Research 1992;11:2736.

    92. Zhang CL, Heinemann U. Effects of the triazole derivative loreclezole (R72063)on stimulus induced ionic and eld potential responses and on differentpatterns of epileptiform activity induced by low magnesium in rat entorhinalcortex-hippocampal slices. Naunyn-Schmiedebergs Archives of Pharmacology1992;346:5817.93. Wafford KA, Bain CJ, Quirk K, McKernan RM, Wingrove PB, Whiting PJ, KempJA. A novel allosteric modulatory site on the GABAA receptor beta subunit.Neuron 1994;12:77582.

    94. Wingrove PB, Wafford KA, Bain C, Whiting PJ. The modulatory action ofloreclezole at the gamma-aminobutyric acid type A receptor is determinedby a single amino acid in the beta 2 and beta 3 subunit. Proceedings of theNational Academy of Sciences of the United States of America 1994;91:456973.

    95. Fisher JL, Macdonald RL. Functional properties of recombinant GABA(A)receptors composed of single or multiple beta subunit subtypes. Neurophar-macology 1997;36:160110.

    96. Smith AJ, Alder L, Silk J, Adkins C, Fletcher AE, Scales T, Kerby J, Marshall G,Wafford KA, McKernan RM, Atack JR. Effect of alpha subunit on allostericmodulation of ion channel function in stably expressed human recombinantgamma-aminobutyric acid(A) receptors determined using (36)Cl ion ux.Molecular Pharmacology 2001;59:110818.

    97. Neelands TR, Macdonald RL. Incorporation of the pi subunit into functionalgamma-aminobutyric Acid(A) receptors. Molecular Pharmacology1999;56:598610.

    98. Donnelly JL, Macdonald RL. Loreclezole enhances apparent desensitization ofrecombinant GABAA receptor currents. Neuropharmacology 1996;35:123341.

    99. Thomet U, Baur R, Dodd RH, Sigel E. Loreclezole as a simple functional markerfor homomeric rho type GABA(C) receptors. European Journal of Pharmacology2000;408:R12.

    100. Carter RB, Wood PL, Wieland S, Hawkinson JE, Belelli D, Lambert JJ, White HS,Wolf HH, Mirsadeghi S, Tahir SH, Bolger MB, Lan NC, Gee KW. Characteriza-tion of the anticonvulsant properties of ganaxolone (CCD 1042; 3alpha-hydroxy-3beta-methyl-5alpha-pregnan-20-one), a selective, high-afnity,steroid modulator of the gamma-aminobutyric acid(A) receptor. Journal ofPharmacology and Experimental Therapeutics 1997;280:128495.

    101. Monaghan EP, Mcauley JW, Data JL. Ganaxolone: a novel positive allostericmodulator of the GABA(A) receptor complex for the treatment of epilepsy.Expert Opinion on Investigational Drugs 1999;8:166371.

    102. Reddy DS, Castaneda DC, OMalley BW, Rogawski MA. Anticonvulsant activityof progesterone and neurosteroids in progesterone receptor knockout mice.Journal of Pharmacology and Experimental Therapeutics 2004;310:2309.

    104. Reddy DS. Pharmacology of endogenous neuroactive steroids. Critical Reviewsin Neurobiology 2003;15:197234.

    105. Kimoto T, Tsurugizawa T, Ohta Y, Makino J, Tamura H, Hojo Y, Takata N,Kawato S. Neurosteroid synthesis by cytochrome p450-containing systemslocalized in the rat brain hippocampal neurons: N-methyl-D-aspartate andcalcium-dependent synthesis. Endocrinology 2001;142:357889.

    106. Twyman RE, Macdonald RL. Neurosteroid regulation of GABAA receptorsingle-channel kinetic properties of mouse spinal cord neurons in culture.Journal of Physiology 1992;456:21545.

    107. Zhu WJ, Wang JF, Krueger KE, Vicini S. Delta subunit inhibits neurosteroidmodulation of GABAA receptors. Journal of Neuroscience 1996;16:664856.

    108. Mihalek RM, Banerjee PK, Korpi ER, Quinlan JJ, Firestone LL, Mi ZP, Lagenaur C,Tretter V, Sieghart W, Anagnostaras SG, Sage JR, Fanselow MS, Guidotti A,Spigelman I, Li Z, DeLorey TM, Olsen RW, Homanics GE. Attenuated sensitivityto neuroactive steroids in gamma-aminobutyrate type A receptor delta sub-unit knockout mice. Proceedings of the National Academy of Sciences of theUnited States of America 1999;96:1290510.

    109. Wohlfarth KM, Bianchi MT, Macdonald RL. Enhanced neurosteroid potentia-tion of ternary GABA(A) receptors containing the delta subunit. Journal ofNeuroscience 2002;22:15419.

    110. Puia G, Ducic I, Vicini S, Costa E. Does neurosteroid modulatory efcacydepend on GABAA receptor subunit composition? Receptors Channels1993;1:13542.

    111. Bianchi MT, Haas KF, Macdonald RL. Alpha1 and alpha6 subunits specifydistinct desensitization, deactivation and neurosteroid modulation ofGABA(A) receptors containing the delta subunit. Neuropharmacology2002;43:492502.

    112. Sun C, Mtchedlishvili Z, Erisir A, Kapur J. Diminished neurosteroid sensitivityof synaptic inhibition and altered location of the alpha4 subunit of GABA(A)receptors in an animal model of epilepsy. Journal of Neuroscience2007;27:1264150.

    113. Hosie AM, Clarke L, da SH, Smart TG. Conserved site for neurosteroid modu-lation of GABA A receptors. Neuropharmacology 2009;56:14954.

    114. Akk G, Li P, Bracamontes J, Reichert DE, Covey DF, Steinbach JH. Mutations ofthe GABA-A receptor alpha1 subunit M1 domain reveal unexpected complex-ity for modulation by neuroactive steroids. Molecular Pharmacology2008;74:61427.

    115. Bracamontes JR, Steinbach JH. Steroid interaction with a single potentiatingsite is sufcient to modulate GABA-A receptor function. Molecular Pharmacol-ogy 2009;75:97381.

    116. Zona C, Ciotti MT, Avoli M. Topiramate attenuates voltage-gated sodiumcurrents in rat cerebellar granule cells. Neuroscience Letters 1997;231:1236.

    117. McLean MJ, Bukhari AA, Wamil AW. Effects of topiramate on sodium-depen-dent action-potential ring by mouse spinal cord neurons in cell culture.Epilepsia 2000;41(Suppl 1):S214.

    118. Hanaya R, Sasa M, Ujihara H, Ishihara K, Serikawa T, Iida K, Akimitsu T, Arita K,Kurisu K. Suppression by topiramate of epileptiform burst discharges inhippocampal CA3 neurons of spontaneously epileptic rat in vitro. BrainResearch 1998;789:27482.

  • L.J. Greeneld Jr. / Seizure 22 (2013) 589600 599119. Gibbs III JW, Sombati S, DeLorenzo RJ, Coulter DA. Cellular actions of topir-amate: blockade of kainate-evoked inward currents in cultured hippocampalneurons. Epilepsia 2000;41(Suppl 1):S106.

    120. Zhang X, Velumian AA, Jones OT, Carlen PL. Modulation of high-voltage-activated calcium channels in dentate granule cells by topiramate. Epilepsia2000;41(Suppl 1):S5260.

    121. Mirza N, Marson AG, Pirmohamed M. Effect of topiramate on acid-basebalance: extent, mechanism and effects. British Journal of Clinical Pharmacology2009;68:65561.

    122. White HS, Brown SD, Woodhead JH, Skeen GA, Wolf HH. Topiramate enhancesGABA-mediated chloride ux and GABA-evoked chloride currents in murinebrain neurons and increases seizure threshold. Epilepsy Research1997;28:16779.

    123. White HS, Brown SD, Woodhead JH, Skeen GA, Wolf HH. Topiramate mod-ulates GABA-evoked currents in murine cortical neurons by a nonbenzodia-zepine mechanism. Epilepsia 2000;41(Suppl 1):S1720.

    124. Gordey M, DeLorey TM, Olsen RW. Differential sensitivity of recombinantGABA(A) receptors expressed in Xenopus oocytes to modulation by topira-mate. Epilepsia 2000;41(Suppl 1):S259.

    125. Simeone TA, Wilcox KS, White HS. Subunit selectivity of topiramate modula-tion of heteromeric GABA(A) receptors. Neuropharmacology 2006;50:84557.

    126. Pennell PB, Ogaily MS, Macdonald RL. Aplastic anemia in a patient receivingfelbamate for complex partial seizures. Neurology 1995;45:45660.

    127. ONeil MG, Perdun CS, Wilson MB, McGown ST, Patel S. Felbamate-associatedfatal acute hepatic necrosis. Neurology 1996;46:14579.

    128. Kume A, Greeneld Jr LJ, Macdonald RL, Albin RL. Felbamate inhibits [3H]t-butylbicycloorthobenzoate (TBOB) binding and enhances Cl- current at thegamma-aminobutyric AcidA (GABAA) receptor. Journal of Pharmacology andExperimental Therapeutics 1996;277:178492.

    129. Rho JM, Donevan SD, Rogawski MA. Barbiturate-like actions of the propane-diol dicarbamates felbamate and meprobamate. Journal of Pharmacology andExperimental Therapeutics 1997;280:138391.

    130. Subramaniam S, Rho JM, Penix L, Donevan SD, Fielding RP, Rogawski MA.Felbamate block of the N-methyl-D-aspartate receptor. Journal of Pharmacol-ogy and Experimental Therapeutics 1995;273:87886.

    131. Landmark CJ, Johannessen SI. Modications of antiepileptic drugs for im-proved tolerability and efcacy. Perspectives in Medicinal Chemistry2008;2:2139.

    132. Rundfeldt C, Netzer R. Investigations into the mechanism of action of the newanticonvulsant retigabine. Interaction with GABAergic and glutamatergicneurotransmission and with voltage gated ion channels. Arzneimittel-For-schung 2000;50:106370.

    133. Otto JF, Kimball MM, Wilcox KS. Effects of the anticonvulsant retigabine oncultured cortical neurons: changes in electroresponsive properties and syn-aptic transmission. Molecular Pharmacology 2002;61:9217.

    134. Gebhardt C, Breustedt JM, Noldner M, Chatterjee SS, Heinemann U. Theantiepileptic drug losigamone decreases the persistent Na+ current in rathippocampal neurons. Brain Research 2001;920:2731.

    135. Draguhn A, Jungclaus M, Sokolowa S, Heinemann U. Losigamone decreasesspontaneous synaptic activity in cultured hippocampal neurons. EuropeanJournal of Pharmacology 1997;325:24551.

    136. Dimpfel W, Chatterjee SS, Noldner M, Ticku MK. Effects of the anticonvulsantlosigamone and its isomers on the GABAA receptor system. Epilepsia1995;36:9839.

    137. Fisher JL. The anti-convulsant stiripentol acts directly on the GABA(A) receptoras a positive allosteric modulator. Neuropharmacology 2009;56:1907.

    138. Quilichini PP, Chiron C, Ben Ari Y, Gozlan H. Stiripentol, a putative antiepilepticdrug, enhances the duration of opening of GABA-A receptor channels. Epilepsia2006;47:70416.

    139. Taylor CP. Mechanisms of action of gabapentin. Revue Neurologique (Paris)1997;153(Suppl 1):S3945.

    140. Loscher W, Honack D, Taylor CP. Gabapentin increases aminooxyacetic acid-induced GABA accumulation in several regions of rat brain. NeuroscienceLetters 1991;128:1504.

    141. Dooley DJ, Taylor CP, Donevan S, Feltner D. Ca2+ channel alpha2delta ligands:novel modulators of neurotransmission. Trends in Pharmacological Sciences2007;28:7582.

    142. Hendrich J, Van Minh AT, Heblich F, Nieto-Rostro M, Watschinger K, StriessnigJ, Wratten J, Davies A, Dolphin AC. Pharmacological disruption of calciumchannel trafcking by the alpha2delta ligand gabapentin. Proceedings of theNational Academy of Sciences of the United States of America 2008;105:362833.

    143. Sills GJ. The mechanisms of action of gabapentin and pregabalin. CurrentOpinion in Pharmacology 2006;6:10813.

    144. Nemeroff CB. The role of GABA in the pathophysiology and treatment ofanxiety disorders. Psychopharmacology Bulletin 2003;37:13346.

    145. Loscher W. Anticonvulsant and biochemical effects of inhibitors of GABAaminotransferase and valproic acid during subchronic treatment in mice.Biochemical Pharmacology 1982;31:83742.

    146. Loscher W. Valproate enhances GABA turnover in the substantia nigra. BrainResearch 1989;501:198203.

    147. Loscher W. Valproate: a reappraisal of its pharmacodynamic properties andmechanisms of action. Progress in Neurobiology 1999;58:3159.

    148. Macdonald RL, Kelly KM. Antiepileptic drug mechanisms of action. Epilepsia1995;36(Suppl 2):S212.

    149. Frahm C, Engel D, Draguhn A. Efcacy of background GABA uptake in rathippocampal slices. Neuroreport 2001;12:15936.150. Engel D, Schmitz D, Gloveli T, Frahm C, Heinemann U, Draguhn A. Laminardifference in GABA uptake and GAT-1 expression in rat CA1. Journal ofPhysiology 1998;512(Pt 3):6439.

    151. Richter D, Luhmann HJ, Kilb W. Intrinsic activation of GABA receptors sup-presses epileptiform activity in the cerebral cortex of immature mice. Epilepsia2010.

    152. Frahm C, Stief F, Zuschratter W, Draguhn A. Unaltered control of extracellularGABA-concentration through GAT-1 in the hippocampus of rats after pilocar-pine-induced status epilepticus. Epilepsy Research 2003;52:24352.

    153. Adkins JC, Noble S. Tiagabine. A review of its pharmacodynamic and phar-macokinetic properties and therapeutic potential in the management ofepilepsy. Drugs 1998;55:43760.

    154. Richens A. Pharmacology and clinical pharmacology of vigabatrin. Journal ofChild Neurology 1991;Suppl 2:S710.

    155. Gram L, Klosterskov P. Dam M. gamma-vinyl GABA: a double-blind placebo-controlled trial in partial epilepsy. Annals of Neurology 1985;17:2626.

    156. Riikonen R. ACTH therapy of West syndrome: Finnish views. Brain andDevelopment 2001;23:6426.

    157. Parisi P, Bombardieri R, Curatolo P. Current role of vigabatrin in infantilespasms. The European Journal of Paediatric Neurology 2007;11:3316.

    158. Gram L, Larsson OM, Johnsen AH, Schousboe A. Effects of valproate, vigabatrinand aminooxyacetic acid on release of endogenous and exogenous GABA fromcultured neurons. Epilepsy Research 1988;2:8795.

    159. Petroff OA, Behar KL, Mattson RH, Rothman DL. Human brain gamma-ami-nobutyric acid levels and seizure control following initiation of vigabatrintherapy. Journal of Neurochemistry 1996;67:2399404.

    160. Stuchlik A, Kubova H, Mares P. Single systemic dose of vigabatrin induces earlyproconvulsant and later anticonvulsant effect in rats. Neuroscience Letters2001;312:3740.

    161. Mares P, Slamberova R. Biphasic action of vigabatrin on cortical epileptic after-discharges in rats. Naunyn-Schmiedebergs Archives of Pharmacology2004;369:30511.

    162. Panayiotopoulos CP, Agathonikou A, Sharoqi IA, Parker AP. Vigabatrin aggra-vates absences and absence status. Neurology 1997;49:1467.

    163. Iannetti P, Spalice A, Perla FM, Conicella E, Raucci U, Bizzarri B. Visual eldconstriction in children with epilepsy on vigabatrin treatment. Pediatrics2000;106:83842.

    164. Hammoudi DS, Lee SS, Madison A, Mirabella G, Buncic JR, Logan WJ, Snead OC,Westall CA. Reduced visual function associated with infantile spasms inchildren on vigabatrin therapy. Investigative Ophthalmology & Visual Science2005;46:51420.

    165. Wild JM, Ahn HS, Baulac M, Bursztyn J, Chiron C, Gandolfo E, Safran AB,Schiefer U, Perucca E. Vigabatrin and epilepsy: lessons learned. Epilepsia2007;48:131827.

    166. Lukasiuk K, Pitkanen A. GABA(A)-mediated toxicity of hippocampal neurons invitro. Journal of Neurochemistry 2000;74:244554.

    167. Lorsignol A, Taupignon A, Dufy B. Short applications of gamma-aminobutyricacid increase intracellular calcium concentrations in single identied ratlactotrophs. Neuroendocrinology 1994;60:38999.

    168. Kahraman S, Zup SL, McCarthy MM, Fiskum G. GABAergic mechanism ofpropofol toxicity in immature neurons. Journal of Neurosurgical Anesthesiology2008;20:23340.

    169. Staley KJ, Soldo BL, Proctor WR. Ionic mechanisms of neuronal excitation byinhibitory GABAA receptors. Science 1995;269:97781.

    170. Brumback AC, Staley KJ. Thermodynamic regulation of NKCC1-mediated Cl-cotransport underlies plasticity of GABA(A) signaling in neonatal neurons.Journal of Neuroscience 2008;28:130112.

    171. Kriegstein AR, Owens DF. GABA may act as a self-limiting trophic factor atdeveloping synapses. Sciences STKE 2001;2001:E1.

    172. Dzhala VI, Talos DM, Sdrulla DA, Brumback AC, Mathews GC, Benke TA, DelpireE, Jensen FE, Staley KJ. NKCC1 transporter facilitates seizures in the developingbrain. Nature Medical 2005;11:120513.

    173. Dzhala VI, Staley KJ. Excitatory actions of endogenously released GABAcontribute to initiation of ictal epileptiform activity in the developing hippo-campus. Journal of Neuroscience 2003;23:18406.

    174. Zhang SJ, Jackson MB. GABAA receptor activation and the excitability of nerveterminals in the rat posterior pituitary. Journal of Physiology 1995;483(Pt3):58395.

    175. Bormann J, Hamill OP, Sakmann B. Mechanism of anion permeation throughchannels gated by glycine and gamma-aminobutyric acid in mouse culturedspinal neurones. Journal of Physiology 1987;385:24386.

    176. Zeng XJ, Tietz EI. Role of bicarbonate ion in mediating decreased synapticconductance in benzodiazepine tolerant hippocampal CA1 pyramidal neu-rons. Brain Research 2000;868:20214.

    177. Cohen I, Navarro V, Clemenceau S, Baulac M, Miles R. On the origin of interictalactivity in human temporal lobe epilepsy in vitro. Science 2002;298:141821.

    178. Shorvon SD. Status epilepticus. Its clinical features and treatment in childrenand adults. New York: Cambridge University Press; 1994.

    179. Tassinari CA, Dravet C, Roger J, Cano JP, Gastaut H. Tonic status epilepticusprecipitated by intravenous benzodiazepine in ve patients with Lennox-Gastaut syndrome. Epilepsia 1972;13:42135.

    180. Dzhala VI, Brumback AC, Staley KJ. Bumetanide enhances phenobarbitalefcacy in a neonatal seizure model. Annals of Neurology 2008;63:22235.

    181. Glykys J, Dzhala VI, Kuchibhotla KV, Feng G, Kuner T, Augustine G, Bacskai BJ,Staley KJ. Differences in cortical versus subcortical GABAergic signaling:

  • a candidate mechanism of electroclinical uncoupling of neonatal seizures.Neuron 2009;63:65772.

    182. Kahle KT, Barnett SM, Sassower KC, Staley KJ. Decreased seizure activity in ahuman neonate treated with bumetanide, an inhibitor of the Na(+)-K(+)-2Cl() cotransporter NKCC1. Journal of Child Neurology 2009;24:5726.

    183. Wang DD, Kriegstein AR. Blocking early GABA depolarization with bumeta-nide results in permanent alterations in cortical circuits and sensorimotorgating decits. Cerebral Cortex 2010.

    184. Kapur J, Macdonald RL. Rapid seizure-induced reduction of benzodiazepineand Zn2+ sensitivity of hippocampal dentate granule cell GABAA receptors.Journal of Neuroscience 1997;17:753240.

    185. Goodkin HP, Sun C, Yeh JL, Mangan PS, Kapur J. GABA(A) receptor internaliza-tion during seizures. Epilepsia 2007;48(Suppl 5):10913.

    186. Lauren HB, Lopez-Picon FR, Korpi ER, Holopainen IE. Kainic acid-induced statusepilepticus alters GABA receptor subunit mRNA and protein expression in thedeveloping rat hippocampus. Journal of Neurochemistry 2005;94: 138494.

    187. Feng HJ, Mathews GC, Kao C, Macdonald RL. Alterations of GABA A-receptorfunction and allosteric modulation during development of status epilepticus.Journal of Neurophysiology 2008;99:128593.

    188. Alldredge BK, Gelb AM, Isaacs SM, Corry MD, Allen F, Ulrich S, Gottwald MD,ONeil N, Neuhaus JM, Segal MR, Lowenstein DH. A comparison of lorazepam,diazepam, and placebo for the treatment of out-of-hospital status epilepticus.New England Journal of Medicine 2001;345:6317.

    189. Treiman DM, Meyers PD, Walton NY, Collins JF, Colling C, Rowan AJ, HandforthA, Faught E, Calabrese VP, Uthman BM, Ramsay RE, Mamdani MB. A compari-son of four treatments for generalized convulsive status epilepticus. VeteransAffairs Status Epilepticus Cooperative Study Group. New England Journal ofMedicine 1998;339(12):7928.

    190. Brooks-Kayal AR, Shumate MD, Jin H, Rikhter TY, Coulter DA. Selective changesin single cell GABA(A) receptor subunit expression and function in temporallobe epilepsy. Nature Medicine 1998;4:116672.

    191. Raol YH, Zhang G, Lund IV, Porter BE, Maronski MA, Brooks-Kayal AR.Increased GABA(A)-receptor alpha1-subunit expression in hippocampaldentate gyrus after early-life status epilepticus. Epilepsia 2006;47:166573.

    192. Raol YH, Lund IV, Bandyopadhyay S, Zhang G, Roberts DS, Wolfe JH, Russek SJ,Brooks-Kayal AR. Enhancing GABA(A) receptor alpha 1 subunit levels inhippocampal dentate gyrus inhibits epilepsy development in an animal modelof temporal lobe epilepsy. Journal of Neuroscience 2006;26:113426.

    193. Raol YH, Zhang G, Budreck EC, Brooks-Kayal AR. Long-term effects of diaze-pam and phenobarbital treatment during development on GABA receptors,transporters and glutamic acid decarboxylase. Neuroscience 2005;132:399407.

    194. Kang I, Miller LG. Decreased GABAA receptor subunit mRNA concentrationsfollowing chronic lorazepam administration. British Journal of Pharmacology1991;103:12857.

    195. Chen S, Huang X, Zeng XJ, Sieghart W, Tietz EI. Benzodiazepine-mediatedregulation of alpha1, alpha2, beta1-3 and gamma2 GABA(A) receptor subunitproteins in the rat brain hippocampus and cortex. Neuroscience 1999;93:3344.

    196. Tseng YT, Wellman SE, Ho IK. In situ hybridization evidence of differentialmodulation by pentobarbital of GABAA receptor alpha 1- and beta 3-subunitmRNAs. Journal of Neurochemistry 1994;63:3019.

    197. Klein RL, Harris RA. Regulation of GABAA receptor structure and function bychronic drug treatments in vivo and with stably transfected cells. JapaneseJournal of Pharmacology 1996;70:115.

    198. Unwin N. Rened structure of the nicotinic acetylcholine receptor at 4Aresolution. Journal of Molecular Biology 2005;346:96789.

    199. Olsen RW, Tobin AJ. Molecular biology of GABAA receptors. FASEB Journal1990;4:146980.

    200. Macdonald RL, Kang JQ, Gallagher MJ. GABAA receptor subunit mutations andgenetic epilepsies, 2012.

    L.J. Greeneld Jr. / Seizure 22 (2013) 589600600

    Molecular mechanisms of antiseizure drug activity at GABAA receptorsGABAARs and epilepsyBenzodiazepinesBarbituratesLoreclezoleGanaxoloneGABAAR effects of other ASDsTopiramateFelbamateEzogabineLosigamoneStiripentolGABA-enhancing agentsGabapentin and pregabalinValproic acidTiagabineVigabatrin

    Excitatory GABAA currentsGABAAR subunit changes and ASD efficacy in epilepsy and status epilepticusReferences