SlideShare ist ein Scribd-Unternehmen logo
1 von 12
Downloaden Sie, um offline zu lesen
Brandon Turner                           Receptors & Channels                           May 10, 2012




                 Investigating the Roles of GABAergic Inhibitory Currents in Epilepsy



Introduction


       Epilepsy is one of the more common neurological disorders in humans and is

characterized by the occurrence of repeated seizures. Seizures themselves result from an area of

the brain exhibiting large amounts of depolarizing currents/over activity. For this reason, past

research has focused on an up-regulation of excitatory neurotransmission, such as AMPA

receptors, to be the cause of such over activity (Rogawski, 2011). However, treatment of seizures

with anti-convulsant that target excitability has proved to be less effective than desired. More

recent research has discussed the role of inhibitory currents, notably those mediated by the γ-

amino butyric acid (GABAA) type A receptor, which is responsible for the majority of the fast

inhibitory signaling in the brain. The GABAA receptor exists as a heteropentamer usually

comprised of two pairs of α and β subunits and a variable subunit (γ, δ, ρ, π, ε), each of which

may have several subtypes (α [1-6], β [1-3], etc). With such subunit variability and differences of

subunit expression levels in specific brain regions, the pharmacology of the GABAA receptor has

a high degree of variance. The receptor binds several allosteric potentiating ligands, such as

benzodiazepines (BZDs), which require the γ subunit, and neurosteroids which bind directly to

the α subunit.


       The involvement of such receptors in epilepsy is bound to be highly complex. Indeed,

many factors can contribute to epilepsy via GABAergic currents, such as environmental

intoxicants like RDX (Williams et al 2011), developmental defects involving time of onset of

seizures (Briggs SW and Galanopoulou AS, 2011), regulation of GABA transport both, synaptic
2


and extrasynaptic (Chiu et al 2005), neuronal migration (Bozzi et al 2012), or inherent genetic

factors (Gurba et al 2012, Macdonald et al 2009). It has also been shown that GABA subunit

composition changes during the course of seizures and epileptogenesis (Loup et al 2000).

Despite these problems, neurosteroids and benzodiazepines are known to be effective

anticonvulsants if administered with proper timing, yet some types of epilepsy, such as temporal

lobe epilepsy (TLE), remain resistant to such treatment. Clearly the role of GABAA in epilepsy is

not straightforward and requires additional investigation if a more conclusive understanding of

the disease and its treatment are to be found. In this review, I will focus on changes in

GABAergic inhibitory current due to developmental changes, GABA concentration variations in

multiple forms of epilepsy, and on receptor specific changes in temporal lobe epilepsy in the

hopes of directing future research to develop anticonvulsants targeting these receptors that more

accurately despite the inherent complexity of their role in epileptogenesis.


Developmental Malformations in GABAergic Systems


       Developmental miscues can result in a host of neurological disorders that include many

subtypes of epilepsy. Some of these include genetic defects that decrease the migration of

GABAergic neurons to their proper positions, including interneurons of the cortex and

hippocampus. Two of these factors, Dlx and Reelin, have been shown in knockout and mutation

studies to impair the migration of such inhibitory neurons to the cortex and proper layer in the

hippocampus, respectively (Bozzi et al 2012), and have been implicated several forms of

epilepsy. Many other factors have been shown to induce epilepsy in model animals as well, all

concerning cellular circuits and positioning in the brain. Notably, Velisek et al have recently

shown that haploinsufficiency of BRD2 (Bromodomain- containing gene 2) in mice contributes

to lack of GABAergic neurons in the neocortex and striatum and a deficiency in GABAergic
3


signaling in other regions of the brain associated with idiopathic generalized epilepsies. Clearly,

developmental factors and genetic regulation can play into developing epilepsy by removing

inhibition in different brain regions.


       Other changes in normal neuronal function, most notably seizures at an early age, can

induce epilepsy in adult animal models. Depending on age, GABAA currents can be either

depolarizing (early age) or hyperpolarizing. Recently, this has been shown to be linked to the

concentration of Cl- within the cell, which affects GABAA subunit compositions (Succol et al

2012). The authors used both electrophysiological and immunostaining to show that changes in

intracellular chloride concentrations affected EGABA depending on the expression levels of

KCC2, a potassium/chloride cotransporter. Consistent with this data, it has been shown that

disruption of KCC2 funciton can contribute to early life seizures and that these seizures

themselves can alter GABAA subunit composition as well (Reviewed by Briggs SW,

Galanopoulou AS, 2011). It may be that a rapid influx of chloride ions due to compensatory

inhibition during seizures may contribute to the changes in GABAA receptor subunit

composition, or that changes in chloride trafficking may result in a reversion to immature-like

expression and function of GABAA receptors, causing them to be depolarizing rather than

inhibitory. Further research into the temporal allocation of shifts in chloride gradients,

transporter activity, and subunit composition during the ictal period of seizures may provide

insight into epileptogenesis.


Changes in Extracellular and Synaptic GABA Concentrations


       Changes in GABA concentrations at the synapse due to disruptions in transport or

synthesis could contribute to altered levels of GABAA signaling, which could, in turn, contribute
4


to epileptic phenotypes. As reviewed by Pavlov et al, 2012, “Reversal of GABA transport in

certain subpopulations of neurons or individual cells in epileptic tissue cannot be ruled out. Some

interneurons in chronic epilepsy may become metabolically more active, express more GAD and

have elevated intracellular GABA concentrations.” This speaks the idea that overexpression of

GAD (Glutamate Decarboxylase), which converts Glutamate to GABA, could raise intracellular

GABA and reverse transportation, which is largely dependent on voltage and concentration

gradients (Pavlov et al, 2012). Consistent with this, impaired GABA uptake by GABA

transporter deficiency has been shown to cause tremors and ataxia in mice (Chiu et al 2005).

Despite the possible negative roles of GAT impairment in epilepsy, it has been shown that GAT

impairment functions to increase tonic inhibition in epileptic rats (Frahm et al, 2003) by

increasing available GABA to the extrasynaptic, high affinity and slow desensitizing receptors.

In this sense, GABA transporters could serve as a possible drug target for anticonvulsants should

allosteric up-regulation be possible.


       Much of the regulation of extrasynaptic GABA concentrations is dependent on the

function of GABA transporters in astrocytes. GAT-3, which is primarily found in astrocytes,

contributes substantially to extracellular GABA levels and can induce increased tonic inhibition

in the presensce of glutamate (Heja et al 2009). In vivo studies have also shown that uptake of

glutamate in hippocampal astrocytes is necessary for GABA release by GAT-2,3 and can convert

excess amounts of glutamate excitatory signaling to inhibition in an epileptic model (Heja et al

2012). Due to increased variability of GABAA subunits during seizures, increased tonic GABA

can initiate a negative feedback loop and prevent the spread of excess excitation to other brain

regions without the need of specifically designed drugs for the variant receptors. However, this

may not serve as an effective treatment for epilepsy, as past studies have demonstrated in vitro
5


that increased extra-cellular GABA concentrations are not enough to mitigate the hyper-

excitability of hippocampal neurons in an epileptic model (Yeh et al 2005).


       Since extracellular GABA levels and tonic inhibition seem to be inefficient at preventing

seizures, investigation of vesicular GABA transporters (VGAT) may provide insight into

mechanisms for preventing seizures. It has recently been shown that epileptic neurons, as

induced by excess exposure to glutamate, express a truncated form of VGAT that accumulates at

non-synaptic sites (Gomes et al 2011). However, removal of VGAT from vesicles contributes to

increased synaptic GABA concentrations, suggesting that VGAT truncation may serve as a

mechanism to increase inhibition in their model of temporal lobe epilepsy rather than furthering

the progression of the disease. Given that synaptic GABAA receptors desensitize more rapidly

than extrasynaptic receptors, it would seem that enhancing GABA at the synapse may also prove

to be ineffective at alleviating epileptogenesis. Since the level of GABA transport appears to be

providing negative feedback in epileptic models, their dysfunction could contribute to

epileptogenesis. However, due to their regulation by both voltage and concentration gradients,

modifying these transporters would prove difficult at the least, suggesting that changes in

receptor expression and subunit concentration could provide more insight into understanding

epileptogenesis and drug design.


GABA Receptor Expression and Composition Prior to and Following Seizures


       Temporal lobe epilepsy (TLE) is among the most common forms of epilepsy in adults

and is mainly thought to arise from alterations in excitation/inhibition in the hippocampus (Joshi

et al 2011). Those with the disease normally display a loss of both CA1 and CA3 pyramidal

neurons and changes in receptor expression in dentate gyrus cells (DGCs). These changes have
6


been shown by S. Joshi et al to lower neurosteroid sensitivity, which they attribute to changes in

seizure susceptibility of women dependent on the menstrual cycle (known to cause alterations in

the levels of endogenous neurosteroids in the brain). Indeed, recent studies have shown using

radiolabeled ligand binding that rats with temporal lobe epilepsy show decreased binding in the

cerebral cortex and further show that this corresponds to a decrease in expression of α1, γ, δ,

GABAB receptors, and GAD, the enzyme that converts glutamate to GABA (Mathew et al

2012). Past studies have also shown that changes in CA1 and CA3 cells include a marked

decrease in α1 subunit expression and an increase in α2/3 expression (Loup et al 2000). Also,

Rajasekaran et al had recently shown that the low affinity of neurosteroids for epileptic GABAA

receptors is likely due to lower incorporation of the δ subunit, which is down-regulated in TLE.

How intracellular mechanisms contribute to subunit regulation is currently not well understood,

but recent studies have shown that δ subunit incorporation is largely dependent on intracellular

[Cl-]. Increased intracellular [Cl] in the presence of a KCC2 knockdown decreased the amount of

δ subunit incorporated into receptors, as shown by immunostaining, present at the membrane, as

well as increasing the incorporation of α3 subunits and decreasing the incorporation of α1

subunits (Succol et al 2012). The authors focused on this shift of subunit incorporation primarily

to elucidate the shift of GABAA from depolarizing to hyperpolarizing during neuronal

development, which they showed to be mediated by KCC2 up-regulation. However, this could

also point to a method of altered subunit expression in epileptic animals, but whether the KCC2

ion transporter is involved is not known.


       In addition to differential regulation of GABAA subunits during epilepsy, there is also a

loss of synaptic GABAA receptors that may be due to changes in gephyrin and collybistin

scaffolding. Past studies have shown that gephyrin preferentially binds to α2 and α3 subunits,
7


while collybistin binds preferentially to α2 only. They assert that the subunit and the scaffolding

proteins form a trimeric complex via co-immunoprecipitation and that disruption of this

interaction may lead to epilepsy, as seen in a genetic mutation causing a change in the SH3

domain of collybistin which is known to cause mental disability and seizures (Saiepour et al

2010). More recent studies have also shown that gephyrin is able to bind to α1 subunits as well,

also using immunoprecipitation (Mukherjee et al 2011). Since α1-3 containing receptors are the

primary receptors found at synaptic sites, changes in gephyrin binding, expression, or

localization may contribute to loss of these receptors at the synapse and change the

pharmacological properties of these sites during epilepsy. Congruent with this idea, other studies

have shown that induction of status epilepticus alters gephyrin and neuroligin-2 expresssion, but

these did not have an effect on GABAA clustering . Mechanisms underlying the lack of

correlation between the two remain unexplained, but other studies have shown that such a

relationship does exist. Forstera et al had recently demonstrated that splicing of gephyrin exons

is altered in brain regions undergoing TLE or cellular stress which corresponded to a loss of α2

subunit containing receptors at the synapse. It is possible that a decrease of gephyrin binding and

the reported up-regulation of α2 GABAA subunits could contribute to an unnatural clustering of

these receptors at extra-synaptic sites, which normally contain α4 α6 subunits, specifically.

However, Forstera et al report that no concurrent mutations in gephyrin are concurrent with

temporal lobe epilepsy in humans, which may indicate that collybistin-gephyrin clustering, along

with their association with GABAA receptors is complex and requires additional study. Whether

these changes in protein scaffolding are reflect potential feedback mechanisms to stop

epileptogenesis or a method in which seizures are generated will require additional research and
8


holds promise in understanding how spatial localization of inhibitory receptors may contribute to

TLE.


       Most studies have alluded to the involvement of hippocampal neurons in the pathogenesis

of TLE, but, due to the aberrant shuffling of subunits and inherent neuro-plasticity that

accompanies seizures many forms of TLE have remained resistant to anti-convulsants. As

previously described, GABAA receptors in the hippocampus display lowered sensitivity to both

benzodiazepines and neurosteroids, possibly due to increases in α4 containing receptors, which

are insensitive to BZDs, and a decrease in δ containing receptors, which have a high sensitivity

to neurosteroids (Joshi et al 2011). A recent study has shown that other brain regions may be

involved in the propagation of excitatory signaling from the hippocampus and are involved in the

development of seizures. The thalamus, particularly the Thalamic Parafascicular Nucleus, has

been shown to be directly involved in the development of seizures (Langlois et al 2010).

Specifically, they show in vivo that inhibition of NMDA receptors and/or the presence of

GABAA antagonists injected into the TPN was sufficient to prevent behavioral effects following

the creation of an epileptic center in the hippocampus. In agreement with these findings, similar

data was published using muscimol to activate GABAA receptors in the mediodorsal region of

the thalamus and similar results were observed, most notably being the attenuation of seizure

duration (Sloan et al 2011). Although changes in receptor signaling and expression may be

present in the thalamus as well, it provides a novel target for preventing TLE behavioral effects

that circumvents the complex problem of receptor subunit regulation, shuffling, or changes in

ligand concentrations. Until the generation of seizures is fully understood, the thalamus may be

the best option for treating patients with TLE and provides a novel target for specific drugs that

could serve as anti-convulsants for TLE patients.
9


Conclusion


       Comprehending epilepsy from the molecular to the physiological level is a difficult gap

to bridge, but the numerous studies already done have shown many changes at both levels, both

before and after the occurrence of seizures. The myriad of developmental factors that could

contribute to the onset of epilepsy are not well understood beyond their number, yet the

numerous in ways in which they affect the nervous system, as per neuronal development and

migration or receptor/transporter mutations that cause aberrant signaling in mature neurons. This

significantly narrows the mechanisms in which developmental disorders can contribute to

epileptogenesis, but correcting neuronal migration in the brain is unlikely, while finding methods

that can target malformed regions and restore them to their normal inhibitory function by anti-

convulsant drugs is much more likely. GABA transporters, however, seem to serve a role as

increasing GABAergic current in epileptic areas of the brain and their selective modulation by

allosteric drugs would also prove arduous due to their voltage sensitivity and dependence on

intra/extra cellular GABA concentrations. It is possible that an increase in GAD activity may in

neurons or astrocytes may be able to shunt the concentration of GABA to favor increased

exportation, but it has been shown that increased ambient GABA at the synapse or in extra-

synaptic regions have been ineffective at preventing seizures. A more novel target would be

selective modulation of GABAA receptors to increase tonic and phasic inhibition, but the regions

affected by seizure generation display irregular GABAA subunit composition and pharmacology,

further complicating the effect of anti-convulsants. Also, TLE is markedly insensitive to anti-

convulsant drugs, perhaps due to this or the inability of such drugs to act in the epileptic centers.

Yet new research suggests that inhibiting the progression of seizures to other areas of the brain

via increased inhibition or decreased excitation in the thalamus may be an effective way of
10


mitigating seizure genesis at a behavioral level. Although working in the hippocampus to

discover the origin of TLE genesis, preventing the spread of seizures from here by increased

inhibition in the thalamus may be the most effective direction to work with until the exact

mechanism of the marked increase in excitability of hippocampal neurons is discovered and

more specific drugs can be developed.
11


                                          References


Bozzi Y, Casarosa S and Caleo M. Epilepsy as a neurodevelopmental disorder (2012). Front.
       Psych. 3(19): DOI: 10/3389/fpsyt.2012.00019.
Briggs SW and Galanopoulou AS. Altered GABA signaling in early life epilepsies (2011).
       Neural Plasticity. 2011: DOI: 10.1155/2011/527605
Chiu CS, Brickley S, Jensen K. Southwell A, Mckinney S, Cull-Candy S, Mody I and Lester
      HA. GABA transporter deficiency causes tremor, ataxia, nervousness, and increased
      GABA-induced tonic conductance in cerebellum (2005). Journ. Neurosci. 25(12): 3234 –
      3245.
Forstera B, Belaidi AA, Juttner R, Bernert C, Tsokos M, Lehmann TN, Horn P, Dehnicke C,
       Schwarz G and Meir JC. Irregular RNA splicing curtails postsynaptic gephyrin in the
       cornu ammonis of patients with epilepsy. Brain. 133:3778-3794.
Frahm C, Stief F, Zuschratter W, Draguh A. Unaltered control of extracellular GABA-
      concentration through GAT-1 in the hippocampus of rats after pilocarpine-induced status
      epilepticus (2003). Epilepsy Res. 52: 243-252
Gomes JR, Lobo AC, Melo CV, Inacio AR, Takano J, Iwata N, Saido TC, de Almeida LP,
      Wieloch T and Duarte CB. Cleavage of the vesicular GABA transporter under
      excitotoxic conditions is followed by accumulation of the truncated transporter in
      nonsynaptic sites (2011). 31(12): 4622 – 4635.
Gurba KN, Hernandez CC, Hu N and Macdonald RL. The GABRB3 mutation, G32R, associated
      with childhood absence epilepsy alters α1β3γ2L GABAA receptor expression and channel
      gating (2012). Journ. Biol. Chem. 287, 12083-12097.
Heja L, Barabas P, Nyitrai G, Kekesi KA, Lasztoczi B, Toke O, Tarkanyi G, Madsen K,
       Schousboe A, Dobolyi A, Palkovits M and Kardos J. Glutamate uptake triggers
       transporter-mediated GABA release from astrocytes (2009). PLoS ONE. 4(9): e7153.
       DOI:10.1371/jounal.pone.0007153.
Heja L, Nyitrai G, Kekesi O, Dobolyi A, Szabo P, Fiath R, Ulbert I, Pal-Szenthe B, Palkovits M
       and Kardos J. Astrocytes convert network excitation to tonic inhibition of neurons
       (2012). BMC Biology. 10(26): http://www.biomedcentral.com/1741-7007/10/26.
Jackson J, Chugh D, Nilsson P, Wood J, Carlstrom K, Lindvall O and Ekdahl CT. Altered
       synaptic properties during integration of adult-born hippocampal neurons following a
       seiaure insult (2012). PLoS One. 7(4). E35557.
Joshi S, Rajasekaran K, Kapur J. GABAergic transmission in temporal lobe epilepsy: The role of
        neurosteroids (2011). Exp Neurol. Doi:10.1016/j.expneurol.2011.10.028
Langlois M, Polack PO, Bernard H, David O, Charpier S, Depaulis A, and Deransart C.
       Involvement of the Thalamic Parafascicular Nucleus in Mesial Temporal Lobe Epilepsy
       (2010). Journ. Neurosci. 300(49):16523-16535
12


Loup F, Wieser HG, Yonekawa Y, Aguzzi A and Fritschy JM. Selective alterations in GABA A
      subtypes in human temporal lobe epilepsy (2000). Journ. Neurosci. 20(14): 5401 – 5419.
Mathew J, Balakrishnan S, Antony S, Abraham PM and Paulose CS. Decreased GABA receptor
      in the cerebral cortex of epileptic rats: effect of Bacopa monnieri and Bacoside-A (2012).
      Journal of Biomedical Science. 19:25. http://www.jbiomedsci.com/content/19/1/25.
Mukherjee J, Kretschmannova K, Gouzer G, Maric HM, Ramsden S, Tretter V, Harvey K,
      Davies PA, Triller A, Schindelin H and Moss SJ. The residence time of GABAARs at
      inhibitory synapses is determined by direct binding of the receptor α1 subunit to gephyrin
      (2011). Journ. Neurosci. 32(41):14677-14687.
Pavlov I & Walker MC. Tonic GABAA receptor-mediated signaling in temporal lobe epilepsy
       (2012). Neuropharm. Doi:10.1016/j.neuropharm.2012.04.003
Rajasekaran K, Joshi S, Sun C, Mtchedlishvilli Z and Kapur J. Receptors with low affinity for
       neurosteroids and GABA contribute to tonic inhibition of granule cells in epileptic
       animals (2010). Neurobio. Disease. 40: 490-501.
Rogawski MA. Revisiting AMPA receptors as an antiepileptic drug target (2011). Epilep. Curr.
     11(2): 56-63.
Saiepour L, Fuchs C, Patrizi A, Sassoe-Pognetto M, Harvey RJ and Harvey K. Complex role of
       collybistin and gephyrin in GABAA receptor clustering (2010). Journ. Biol. Chem.
       285(38): 29623-29631.
Sloan DM, Zhang DX, and Bertram EH III. Increased GABAergic inhibition in the midline
       thalamus affects signaling and seizure spread in the hippocampus-prefrontal cortex
       pathway (2011). Epilepsia. 52(3):523-530.
Succol F, Fiumelli H, Benfanati F, Cancedda L and Barberis A. Intracellular chloride
       concentration influences the GABAA receptor subunit composition (2012). Nature
       Comm. 3:738. DOI: 10.1038/ncomms1744.
Velisek L, Shang E, Velskova J, Chachau T, Macchiarulo S, Maglakelidze G, Wolgemuth DJ
       and Greenberg DA. GABAergic neuron defecit as an idiopathic generalized epilepsy
       mechanism: The role of BRD2 haploinsufficiency in juvenile myoclonic epilepsy (2011).
       PLoS One. 6(8): E23656.
Williams LR, Aroniadou-Anderjaska V, Qashu F, Finne H, Pidoplichko V, Bannon DI and Braga
       MFM. RDX binds to the GABAA receptor-convulsant site and blocks GABAA receptor-
       mediated currents in the amygdale: A mechanism for RDX-induced seizures. Envir.
       Health. Persp. 119(3): 357 – 363.
Yeh JH, Jeng CJ, Chen YW, Lin HM, Wu YS and Tang CY. Selective enhancement of tonic
      inhibition by increasing ambient GABA is insufficient to suppress excitotoxicity in
      hippocampal neurons (2005). Biochem. Biophys. Res. Comm. 338: 1417-1425.

Weitere ähnliche Inhalte

Was ist angesagt?

Marinus GNX SFN poster 16Nov16
Marinus GNX SFN poster 16Nov16Marinus GNX SFN poster 16Nov16
Marinus GNX SFN poster 16Nov16
John Gruner
 
Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.
Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.
Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.
Beth Murphy
 

Was ist angesagt? (16)

Glutamate receptors
Glutamate receptorsGlutamate receptors
Glutamate receptors
 
Nrn ad posters- nature
Nrn ad posters- natureNrn ad posters- nature
Nrn ad posters- nature
 
Cohen, Akiva
Cohen, AkivaCohen, Akiva
Cohen, Akiva
 
Decreases Expression of PGC-1α in the Alzheimer Disease Brain Impaire Mitocho...
Decreases Expression of PGC-1α in the Alzheimer Disease Brain Impaire Mitocho...Decreases Expression of PGC-1α in the Alzheimer Disease Brain Impaire Mitocho...
Decreases Expression of PGC-1α in the Alzheimer Disease Brain Impaire Mitocho...
 
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
The Role of DNA Methylation as an Epigenetic Mechanism of the Neuroadaptation...
 
74th ICREA Colloquium "Autoimmunity meets neurodegeneration: different pathwa...
74th ICREA Colloquium "Autoimmunity meets neurodegeneration: different pathwa...74th ICREA Colloquium "Autoimmunity meets neurodegeneration: different pathwa...
74th ICREA Colloquium "Autoimmunity meets neurodegeneration: different pathwa...
 
Marinus GNX SFN poster 16Nov16
Marinus GNX SFN poster 16Nov16Marinus GNX SFN poster 16Nov16
Marinus GNX SFN poster 16Nov16
 
Nmda receptors and glun3 subunits: a review
Nmda receptors and glun3 subunits: a reviewNmda receptors and glun3 subunits: a review
Nmda receptors and glun3 subunits: a review
 
Neurosteroids and neuropeptides
Neurosteroids and neuropeptidesNeurosteroids and neuropeptides
Neurosteroids and neuropeptides
 
Epileptogenesis
EpileptogenesisEpileptogenesis
Epileptogenesis
 
Final poster (002)
Final poster (002)Final poster (002)
Final poster (002)
 
Neuroactive steroids
Neuroactive steroidsNeuroactive steroids
Neuroactive steroids
 
Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.
Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.
Murphy et al, AJP Cell Physiol 296_ C746–C756, 2009.
 
Nicholas Young, Sphingosine 1-phosphate Receptor Subtype Influence over Gliob...
Nicholas Young, Sphingosine 1-phosphate Receptor Subtype Influence over Gliob...Nicholas Young, Sphingosine 1-phosphate Receptor Subtype Influence over Gliob...
Nicholas Young, Sphingosine 1-phosphate Receptor Subtype Influence over Gliob...
 
Epileptogenesis and drugs with mechaism
Epileptogenesis and drugs with mechaismEpileptogenesis and drugs with mechaism
Epileptogenesis and drugs with mechaism
 
Research Proposal
Research ProposalResearch Proposal
Research Proposal
 

Ähnlich wie GABAergic Currents In Epilepsy

Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014
Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014
Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014
Elizabeth Lucas
 
1-s2.0-S0197018615000613-main
1-s2.0-S0197018615000613-main1-s2.0-S0197018615000613-main
1-s2.0-S0197018615000613-main
Natalia K?eczek
 
Epilepsy Poster titiu
Epilepsy Poster titiuEpilepsy Poster titiu
Epilepsy Poster titiu
Tamana Yousof
 

Ähnlich wie GABAergic Currents In Epilepsy (20)

Gaba
GabaGaba
Gaba
 
GABA.pptx
GABA.pptxGABA.pptx
GABA.pptx
 
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...
 
Management of seizures
Management of seizuresManagement of seizures
Management of seizures
 
ppt on neuro transmitters
ppt on neuro transmittersppt on neuro transmitters
ppt on neuro transmitters
 
Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014
Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014
Dougherty Bartley Lucas Hablitz Dobrunz Cowell 2014
 
1-s2.0-S0197018615000613-main
1-s2.0-S0197018615000613-main1-s2.0-S0197018615000613-main
1-s2.0-S0197018615000613-main
 
Newer antiepileptic drugs
Newer antiepileptic drugsNewer antiepileptic drugs
Newer antiepileptic drugs
 
Issues in brainmapping...EEG in dementias and hereditary encephalopathies
Issues in brainmapping...EEG in dementias and hereditary encephalopathiesIssues in brainmapping...EEG in dementias and hereditary encephalopathies
Issues in brainmapping...EEG in dementias and hereditary encephalopathies
 
Seminar 5
Seminar 5 Seminar 5
Seminar 5
 
Dr. kristen park kcnq2 Cure professional track - learn more at kcnq2cure.org
Dr. kristen park kcnq2 Cure professional track - learn more at kcnq2cure.orgDr. kristen park kcnq2 Cure professional track - learn more at kcnq2cure.org
Dr. kristen park kcnq2 Cure professional track - learn more at kcnq2cure.org
 
GABA (Gamma aminobutyric acid)
GABA (Gamma aminobutyric acid)GABA (Gamma aminobutyric acid)
GABA (Gamma aminobutyric acid)
 
Basic neuroanatomy & neurophysiology
Basic neuroanatomy & neurophysiologyBasic neuroanatomy & neurophysiology
Basic neuroanatomy & neurophysiology
 
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...
Developmental Inhibitory Changes in the Primary Somatosensory Cortex of the S...
 
RECENT ADVANCES IN ALZHEIMER'S DISEASE
RECENT ADVANCES IN ALZHEIMER'S DISEASERECENT ADVANCES IN ALZHEIMER'S DISEASE
RECENT ADVANCES IN ALZHEIMER'S DISEASE
 
Status epilepticus sign and symptoms, etiology ,pathogenesis , diagnostic te...
Status epilepticus  sign and symptoms, etiology ,pathogenesis , diagnostic te...Status epilepticus  sign and symptoms, etiology ,pathogenesis , diagnostic te...
Status epilepticus sign and symptoms, etiology ,pathogenesis , diagnostic te...
 
Era of Glutamate
Era of GlutamateEra of Glutamate
Era of Glutamate
 
Bu32451456
Bu32451456Bu32451456
Bu32451456
 
Epilepsy Poster titiu
Epilepsy Poster titiuEpilepsy Poster titiu
Epilepsy Poster titiu
 
Glutamate and GABA
Glutamate and GABAGlutamate and GABA
Glutamate and GABA
 

GABAergic Currents In Epilepsy

  • 1. Brandon Turner Receptors & Channels May 10, 2012 Investigating the Roles of GABAergic Inhibitory Currents in Epilepsy Introduction Epilepsy is one of the more common neurological disorders in humans and is characterized by the occurrence of repeated seizures. Seizures themselves result from an area of the brain exhibiting large amounts of depolarizing currents/over activity. For this reason, past research has focused on an up-regulation of excitatory neurotransmission, such as AMPA receptors, to be the cause of such over activity (Rogawski, 2011). However, treatment of seizures with anti-convulsant that target excitability has proved to be less effective than desired. More recent research has discussed the role of inhibitory currents, notably those mediated by the γ- amino butyric acid (GABAA) type A receptor, which is responsible for the majority of the fast inhibitory signaling in the brain. The GABAA receptor exists as a heteropentamer usually comprised of two pairs of α and β subunits and a variable subunit (γ, δ, ρ, π, ε), each of which may have several subtypes (α [1-6], β [1-3], etc). With such subunit variability and differences of subunit expression levels in specific brain regions, the pharmacology of the GABAA receptor has a high degree of variance. The receptor binds several allosteric potentiating ligands, such as benzodiazepines (BZDs), which require the γ subunit, and neurosteroids which bind directly to the α subunit. The involvement of such receptors in epilepsy is bound to be highly complex. Indeed, many factors can contribute to epilepsy via GABAergic currents, such as environmental intoxicants like RDX (Williams et al 2011), developmental defects involving time of onset of seizures (Briggs SW and Galanopoulou AS, 2011), regulation of GABA transport both, synaptic
  • 2. 2 and extrasynaptic (Chiu et al 2005), neuronal migration (Bozzi et al 2012), or inherent genetic factors (Gurba et al 2012, Macdonald et al 2009). It has also been shown that GABA subunit composition changes during the course of seizures and epileptogenesis (Loup et al 2000). Despite these problems, neurosteroids and benzodiazepines are known to be effective anticonvulsants if administered with proper timing, yet some types of epilepsy, such as temporal lobe epilepsy (TLE), remain resistant to such treatment. Clearly the role of GABAA in epilepsy is not straightforward and requires additional investigation if a more conclusive understanding of the disease and its treatment are to be found. In this review, I will focus on changes in GABAergic inhibitory current due to developmental changes, GABA concentration variations in multiple forms of epilepsy, and on receptor specific changes in temporal lobe epilepsy in the hopes of directing future research to develop anticonvulsants targeting these receptors that more accurately despite the inherent complexity of their role in epileptogenesis. Developmental Malformations in GABAergic Systems Developmental miscues can result in a host of neurological disorders that include many subtypes of epilepsy. Some of these include genetic defects that decrease the migration of GABAergic neurons to their proper positions, including interneurons of the cortex and hippocampus. Two of these factors, Dlx and Reelin, have been shown in knockout and mutation studies to impair the migration of such inhibitory neurons to the cortex and proper layer in the hippocampus, respectively (Bozzi et al 2012), and have been implicated several forms of epilepsy. Many other factors have been shown to induce epilepsy in model animals as well, all concerning cellular circuits and positioning in the brain. Notably, Velisek et al have recently shown that haploinsufficiency of BRD2 (Bromodomain- containing gene 2) in mice contributes to lack of GABAergic neurons in the neocortex and striatum and a deficiency in GABAergic
  • 3. 3 signaling in other regions of the brain associated with idiopathic generalized epilepsies. Clearly, developmental factors and genetic regulation can play into developing epilepsy by removing inhibition in different brain regions. Other changes in normal neuronal function, most notably seizures at an early age, can induce epilepsy in adult animal models. Depending on age, GABAA currents can be either depolarizing (early age) or hyperpolarizing. Recently, this has been shown to be linked to the concentration of Cl- within the cell, which affects GABAA subunit compositions (Succol et al 2012). The authors used both electrophysiological and immunostaining to show that changes in intracellular chloride concentrations affected EGABA depending on the expression levels of KCC2, a potassium/chloride cotransporter. Consistent with this data, it has been shown that disruption of KCC2 funciton can contribute to early life seizures and that these seizures themselves can alter GABAA subunit composition as well (Reviewed by Briggs SW, Galanopoulou AS, 2011). It may be that a rapid influx of chloride ions due to compensatory inhibition during seizures may contribute to the changes in GABAA receptor subunit composition, or that changes in chloride trafficking may result in a reversion to immature-like expression and function of GABAA receptors, causing them to be depolarizing rather than inhibitory. Further research into the temporal allocation of shifts in chloride gradients, transporter activity, and subunit composition during the ictal period of seizures may provide insight into epileptogenesis. Changes in Extracellular and Synaptic GABA Concentrations Changes in GABA concentrations at the synapse due to disruptions in transport or synthesis could contribute to altered levels of GABAA signaling, which could, in turn, contribute
  • 4. 4 to epileptic phenotypes. As reviewed by Pavlov et al, 2012, “Reversal of GABA transport in certain subpopulations of neurons or individual cells in epileptic tissue cannot be ruled out. Some interneurons in chronic epilepsy may become metabolically more active, express more GAD and have elevated intracellular GABA concentrations.” This speaks the idea that overexpression of GAD (Glutamate Decarboxylase), which converts Glutamate to GABA, could raise intracellular GABA and reverse transportation, which is largely dependent on voltage and concentration gradients (Pavlov et al, 2012). Consistent with this, impaired GABA uptake by GABA transporter deficiency has been shown to cause tremors and ataxia in mice (Chiu et al 2005). Despite the possible negative roles of GAT impairment in epilepsy, it has been shown that GAT impairment functions to increase tonic inhibition in epileptic rats (Frahm et al, 2003) by increasing available GABA to the extrasynaptic, high affinity and slow desensitizing receptors. In this sense, GABA transporters could serve as a possible drug target for anticonvulsants should allosteric up-regulation be possible. Much of the regulation of extrasynaptic GABA concentrations is dependent on the function of GABA transporters in astrocytes. GAT-3, which is primarily found in astrocytes, contributes substantially to extracellular GABA levels and can induce increased tonic inhibition in the presensce of glutamate (Heja et al 2009). In vivo studies have also shown that uptake of glutamate in hippocampal astrocytes is necessary for GABA release by GAT-2,3 and can convert excess amounts of glutamate excitatory signaling to inhibition in an epileptic model (Heja et al 2012). Due to increased variability of GABAA subunits during seizures, increased tonic GABA can initiate a negative feedback loop and prevent the spread of excess excitation to other brain regions without the need of specifically designed drugs for the variant receptors. However, this may not serve as an effective treatment for epilepsy, as past studies have demonstrated in vitro
  • 5. 5 that increased extra-cellular GABA concentrations are not enough to mitigate the hyper- excitability of hippocampal neurons in an epileptic model (Yeh et al 2005). Since extracellular GABA levels and tonic inhibition seem to be inefficient at preventing seizures, investigation of vesicular GABA transporters (VGAT) may provide insight into mechanisms for preventing seizures. It has recently been shown that epileptic neurons, as induced by excess exposure to glutamate, express a truncated form of VGAT that accumulates at non-synaptic sites (Gomes et al 2011). However, removal of VGAT from vesicles contributes to increased synaptic GABA concentrations, suggesting that VGAT truncation may serve as a mechanism to increase inhibition in their model of temporal lobe epilepsy rather than furthering the progression of the disease. Given that synaptic GABAA receptors desensitize more rapidly than extrasynaptic receptors, it would seem that enhancing GABA at the synapse may also prove to be ineffective at alleviating epileptogenesis. Since the level of GABA transport appears to be providing negative feedback in epileptic models, their dysfunction could contribute to epileptogenesis. However, due to their regulation by both voltage and concentration gradients, modifying these transporters would prove difficult at the least, suggesting that changes in receptor expression and subunit concentration could provide more insight into understanding epileptogenesis and drug design. GABA Receptor Expression and Composition Prior to and Following Seizures Temporal lobe epilepsy (TLE) is among the most common forms of epilepsy in adults and is mainly thought to arise from alterations in excitation/inhibition in the hippocampus (Joshi et al 2011). Those with the disease normally display a loss of both CA1 and CA3 pyramidal neurons and changes in receptor expression in dentate gyrus cells (DGCs). These changes have
  • 6. 6 been shown by S. Joshi et al to lower neurosteroid sensitivity, which they attribute to changes in seizure susceptibility of women dependent on the menstrual cycle (known to cause alterations in the levels of endogenous neurosteroids in the brain). Indeed, recent studies have shown using radiolabeled ligand binding that rats with temporal lobe epilepsy show decreased binding in the cerebral cortex and further show that this corresponds to a decrease in expression of α1, γ, δ, GABAB receptors, and GAD, the enzyme that converts glutamate to GABA (Mathew et al 2012). Past studies have also shown that changes in CA1 and CA3 cells include a marked decrease in α1 subunit expression and an increase in α2/3 expression (Loup et al 2000). Also, Rajasekaran et al had recently shown that the low affinity of neurosteroids for epileptic GABAA receptors is likely due to lower incorporation of the δ subunit, which is down-regulated in TLE. How intracellular mechanisms contribute to subunit regulation is currently not well understood, but recent studies have shown that δ subunit incorporation is largely dependent on intracellular [Cl-]. Increased intracellular [Cl] in the presence of a KCC2 knockdown decreased the amount of δ subunit incorporated into receptors, as shown by immunostaining, present at the membrane, as well as increasing the incorporation of α3 subunits and decreasing the incorporation of α1 subunits (Succol et al 2012). The authors focused on this shift of subunit incorporation primarily to elucidate the shift of GABAA from depolarizing to hyperpolarizing during neuronal development, which they showed to be mediated by KCC2 up-regulation. However, this could also point to a method of altered subunit expression in epileptic animals, but whether the KCC2 ion transporter is involved is not known. In addition to differential regulation of GABAA subunits during epilepsy, there is also a loss of synaptic GABAA receptors that may be due to changes in gephyrin and collybistin scaffolding. Past studies have shown that gephyrin preferentially binds to α2 and α3 subunits,
  • 7. 7 while collybistin binds preferentially to α2 only. They assert that the subunit and the scaffolding proteins form a trimeric complex via co-immunoprecipitation and that disruption of this interaction may lead to epilepsy, as seen in a genetic mutation causing a change in the SH3 domain of collybistin which is known to cause mental disability and seizures (Saiepour et al 2010). More recent studies have also shown that gephyrin is able to bind to α1 subunits as well, also using immunoprecipitation (Mukherjee et al 2011). Since α1-3 containing receptors are the primary receptors found at synaptic sites, changes in gephyrin binding, expression, or localization may contribute to loss of these receptors at the synapse and change the pharmacological properties of these sites during epilepsy. Congruent with this idea, other studies have shown that induction of status epilepticus alters gephyrin and neuroligin-2 expresssion, but these did not have an effect on GABAA clustering . Mechanisms underlying the lack of correlation between the two remain unexplained, but other studies have shown that such a relationship does exist. Forstera et al had recently demonstrated that splicing of gephyrin exons is altered in brain regions undergoing TLE or cellular stress which corresponded to a loss of α2 subunit containing receptors at the synapse. It is possible that a decrease of gephyrin binding and the reported up-regulation of α2 GABAA subunits could contribute to an unnatural clustering of these receptors at extra-synaptic sites, which normally contain α4 α6 subunits, specifically. However, Forstera et al report that no concurrent mutations in gephyrin are concurrent with temporal lobe epilepsy in humans, which may indicate that collybistin-gephyrin clustering, along with their association with GABAA receptors is complex and requires additional study. Whether these changes in protein scaffolding are reflect potential feedback mechanisms to stop epileptogenesis or a method in which seizures are generated will require additional research and
  • 8. 8 holds promise in understanding how spatial localization of inhibitory receptors may contribute to TLE. Most studies have alluded to the involvement of hippocampal neurons in the pathogenesis of TLE, but, due to the aberrant shuffling of subunits and inherent neuro-plasticity that accompanies seizures many forms of TLE have remained resistant to anti-convulsants. As previously described, GABAA receptors in the hippocampus display lowered sensitivity to both benzodiazepines and neurosteroids, possibly due to increases in α4 containing receptors, which are insensitive to BZDs, and a decrease in δ containing receptors, which have a high sensitivity to neurosteroids (Joshi et al 2011). A recent study has shown that other brain regions may be involved in the propagation of excitatory signaling from the hippocampus and are involved in the development of seizures. The thalamus, particularly the Thalamic Parafascicular Nucleus, has been shown to be directly involved in the development of seizures (Langlois et al 2010). Specifically, they show in vivo that inhibition of NMDA receptors and/or the presence of GABAA antagonists injected into the TPN was sufficient to prevent behavioral effects following the creation of an epileptic center in the hippocampus. In agreement with these findings, similar data was published using muscimol to activate GABAA receptors in the mediodorsal region of the thalamus and similar results were observed, most notably being the attenuation of seizure duration (Sloan et al 2011). Although changes in receptor signaling and expression may be present in the thalamus as well, it provides a novel target for preventing TLE behavioral effects that circumvents the complex problem of receptor subunit regulation, shuffling, or changes in ligand concentrations. Until the generation of seizures is fully understood, the thalamus may be the best option for treating patients with TLE and provides a novel target for specific drugs that could serve as anti-convulsants for TLE patients.
  • 9. 9 Conclusion Comprehending epilepsy from the molecular to the physiological level is a difficult gap to bridge, but the numerous studies already done have shown many changes at both levels, both before and after the occurrence of seizures. The myriad of developmental factors that could contribute to the onset of epilepsy are not well understood beyond their number, yet the numerous in ways in which they affect the nervous system, as per neuronal development and migration or receptor/transporter mutations that cause aberrant signaling in mature neurons. This significantly narrows the mechanisms in which developmental disorders can contribute to epileptogenesis, but correcting neuronal migration in the brain is unlikely, while finding methods that can target malformed regions and restore them to their normal inhibitory function by anti- convulsant drugs is much more likely. GABA transporters, however, seem to serve a role as increasing GABAergic current in epileptic areas of the brain and their selective modulation by allosteric drugs would also prove arduous due to their voltage sensitivity and dependence on intra/extra cellular GABA concentrations. It is possible that an increase in GAD activity may in neurons or astrocytes may be able to shunt the concentration of GABA to favor increased exportation, but it has been shown that increased ambient GABA at the synapse or in extra- synaptic regions have been ineffective at preventing seizures. A more novel target would be selective modulation of GABAA receptors to increase tonic and phasic inhibition, but the regions affected by seizure generation display irregular GABAA subunit composition and pharmacology, further complicating the effect of anti-convulsants. Also, TLE is markedly insensitive to anti- convulsant drugs, perhaps due to this or the inability of such drugs to act in the epileptic centers. Yet new research suggests that inhibiting the progression of seizures to other areas of the brain via increased inhibition or decreased excitation in the thalamus may be an effective way of
  • 10. 10 mitigating seizure genesis at a behavioral level. Although working in the hippocampus to discover the origin of TLE genesis, preventing the spread of seizures from here by increased inhibition in the thalamus may be the most effective direction to work with until the exact mechanism of the marked increase in excitability of hippocampal neurons is discovered and more specific drugs can be developed.
  • 11. 11 References Bozzi Y, Casarosa S and Caleo M. Epilepsy as a neurodevelopmental disorder (2012). Front. Psych. 3(19): DOI: 10/3389/fpsyt.2012.00019. Briggs SW and Galanopoulou AS. Altered GABA signaling in early life epilepsies (2011). Neural Plasticity. 2011: DOI: 10.1155/2011/527605 Chiu CS, Brickley S, Jensen K. Southwell A, Mckinney S, Cull-Candy S, Mody I and Lester HA. GABA transporter deficiency causes tremor, ataxia, nervousness, and increased GABA-induced tonic conductance in cerebellum (2005). Journ. Neurosci. 25(12): 3234 – 3245. Forstera B, Belaidi AA, Juttner R, Bernert C, Tsokos M, Lehmann TN, Horn P, Dehnicke C, Schwarz G and Meir JC. Irregular RNA splicing curtails postsynaptic gephyrin in the cornu ammonis of patients with epilepsy. Brain. 133:3778-3794. Frahm C, Stief F, Zuschratter W, Draguh A. Unaltered control of extracellular GABA- concentration through GAT-1 in the hippocampus of rats after pilocarpine-induced status epilepticus (2003). Epilepsy Res. 52: 243-252 Gomes JR, Lobo AC, Melo CV, Inacio AR, Takano J, Iwata N, Saido TC, de Almeida LP, Wieloch T and Duarte CB. Cleavage of the vesicular GABA transporter under excitotoxic conditions is followed by accumulation of the truncated transporter in nonsynaptic sites (2011). 31(12): 4622 – 4635. Gurba KN, Hernandez CC, Hu N and Macdonald RL. The GABRB3 mutation, G32R, associated with childhood absence epilepsy alters α1β3γ2L GABAA receptor expression and channel gating (2012). Journ. Biol. Chem. 287, 12083-12097. Heja L, Barabas P, Nyitrai G, Kekesi KA, Lasztoczi B, Toke O, Tarkanyi G, Madsen K, Schousboe A, Dobolyi A, Palkovits M and Kardos J. Glutamate uptake triggers transporter-mediated GABA release from astrocytes (2009). PLoS ONE. 4(9): e7153. DOI:10.1371/jounal.pone.0007153. Heja L, Nyitrai G, Kekesi O, Dobolyi A, Szabo P, Fiath R, Ulbert I, Pal-Szenthe B, Palkovits M and Kardos J. Astrocytes convert network excitation to tonic inhibition of neurons (2012). BMC Biology. 10(26): http://www.biomedcentral.com/1741-7007/10/26. Jackson J, Chugh D, Nilsson P, Wood J, Carlstrom K, Lindvall O and Ekdahl CT. Altered synaptic properties during integration of adult-born hippocampal neurons following a seiaure insult (2012). PLoS One. 7(4). E35557. Joshi S, Rajasekaran K, Kapur J. GABAergic transmission in temporal lobe epilepsy: The role of neurosteroids (2011). Exp Neurol. Doi:10.1016/j.expneurol.2011.10.028 Langlois M, Polack PO, Bernard H, David O, Charpier S, Depaulis A, and Deransart C. Involvement of the Thalamic Parafascicular Nucleus in Mesial Temporal Lobe Epilepsy (2010). Journ. Neurosci. 300(49):16523-16535
  • 12. 12 Loup F, Wieser HG, Yonekawa Y, Aguzzi A and Fritschy JM. Selective alterations in GABA A subtypes in human temporal lobe epilepsy (2000). Journ. Neurosci. 20(14): 5401 – 5419. Mathew J, Balakrishnan S, Antony S, Abraham PM and Paulose CS. Decreased GABA receptor in the cerebral cortex of epileptic rats: effect of Bacopa monnieri and Bacoside-A (2012). Journal of Biomedical Science. 19:25. http://www.jbiomedsci.com/content/19/1/25. Mukherjee J, Kretschmannova K, Gouzer G, Maric HM, Ramsden S, Tretter V, Harvey K, Davies PA, Triller A, Schindelin H and Moss SJ. The residence time of GABAARs at inhibitory synapses is determined by direct binding of the receptor α1 subunit to gephyrin (2011). Journ. Neurosci. 32(41):14677-14687. Pavlov I & Walker MC. Tonic GABAA receptor-mediated signaling in temporal lobe epilepsy (2012). Neuropharm. Doi:10.1016/j.neuropharm.2012.04.003 Rajasekaran K, Joshi S, Sun C, Mtchedlishvilli Z and Kapur J. Receptors with low affinity for neurosteroids and GABA contribute to tonic inhibition of granule cells in epileptic animals (2010). Neurobio. Disease. 40: 490-501. Rogawski MA. Revisiting AMPA receptors as an antiepileptic drug target (2011). Epilep. Curr. 11(2): 56-63. Saiepour L, Fuchs C, Patrizi A, Sassoe-Pognetto M, Harvey RJ and Harvey K. Complex role of collybistin and gephyrin in GABAA receptor clustering (2010). Journ. Biol. Chem. 285(38): 29623-29631. Sloan DM, Zhang DX, and Bertram EH III. Increased GABAergic inhibition in the midline thalamus affects signaling and seizure spread in the hippocampus-prefrontal cortex pathway (2011). Epilepsia. 52(3):523-530. Succol F, Fiumelli H, Benfanati F, Cancedda L and Barberis A. Intracellular chloride concentration influences the GABAA receptor subunit composition (2012). Nature Comm. 3:738. DOI: 10.1038/ncomms1744. Velisek L, Shang E, Velskova J, Chachau T, Macchiarulo S, Maglakelidze G, Wolgemuth DJ and Greenberg DA. GABAergic neuron defecit as an idiopathic generalized epilepsy mechanism: The role of BRD2 haploinsufficiency in juvenile myoclonic epilepsy (2011). PLoS One. 6(8): E23656. Williams LR, Aroniadou-Anderjaska V, Qashu F, Finne H, Pidoplichko V, Bannon DI and Braga MFM. RDX binds to the GABAA receptor-convulsant site and blocks GABAA receptor- mediated currents in the amygdale: A mechanism for RDX-induced seizures. Envir. Health. Persp. 119(3): 357 – 363. Yeh JH, Jeng CJ, Chen YW, Lin HM, Wu YS and Tang CY. Selective enhancement of tonic inhibition by increasing ambient GABA is insufficient to suppress excitotoxicity in hippocampal neurons (2005). Biochem. Biophys. Res. Comm. 338: 1417-1425.