User:Samsara/GABA

From Wikipedia, the free encyclopedia

The GABAA receptor is one of the two ligand-gated ion channels responsible for mediating the effects of Gamma-Amino Butyric Acid (GABA), the major inhibitory neurotransmitter in the brain.

Structure and function[edit]

The receptor is a multimeric transmembrane receptor that consists of five subunits arranged around a central pore. The receptor sits in the membrane of its neuron at a synapse. The ligand GABA is the endogenous compound that causes this receptor to open; once bound to GABA, the protein receptor changes conformation within the membrane, opening the pore in order to allow chloride ions (Cl-) to pass down an electrochemical gradient. Because the [reversal potential] for chloride in most neurons is close to or more negative than the resting membrane potential, activation of GABAA receptors tends to stabilize the resting potential, and can make it more difficult for excitatory neurotransmitters to depolarize the neuron and generate an action potential. The net effect is typically inhibitory, reducing the activity of the neuron. The GABAA channel opens quickly and thus contributes to the early part of the inhibitory postsynaptic potential (IPSP) (Siegel et al., 1999; Chen et al., 2005).

Subunits[edit]

GABAA receptors are members of the large "Cys-loop" superfamily of evolutionarily related and structurally similar ligand-gated ion channels that also includes nicotinic acetylcholine receptors, glycine receptors, and the 5HT3 serotonin receptor. There are numerous subunit isoforms for the GABAA receptor, which determine the receptor’s agonist affinity, chance of opening, conductance, and other properties (Cossart et al., 2005). In man, there are six types of α subunits, three β's, three γ's, as well as a δ, an ε, a π, a θ, and three ρs (Martin and Dunn, 2002; Sieghart et al., Neurochem Int 1999;34:379–85). Five subunits can combine in different ways to form GABAA channels, but the most common type in the brain has two α's, two β's, and a γ (Martin and Dunn, 2002). The receptor binds two GABA molecules (Siegel et al., 1999; Colquhoun and Sivilotti, 2004), somewhere between an α and a β subunit (Martin and Dunn, 2002).

Agonists and antagonists[edit]

Other ligands (besides GABA) interact with the GABAA receptor to activate it (agonists), to inhibit its activation (antagonists) or to increase or decrease its response to an agonist (positive and negative allosteric modulators). Such other ligands include benzodiazepines (increase pore opening frequency; often the ingredient of sleep pills and anxiety medications), imidazopyridines (newer class of sleep medications), barbiturates (increase pore opening duration; used as sedatives), and certain steroids, called neuroactive steroids.

Among antagonists are picrotoxin (which blocks the channel pore) and bicuculline (which occupies the GABA site and prevents GABA from activating the receptor). The antagonist flumazenil is used medically to reverse the effects of the benzodiazepines.

A useful property of the many agonists and some antagonists is that they often have a greater interaction with GABAA receptors which contain specific subunits. This allows one to determine which GABAA receptor subunit combinations are prevalent in particular brain areas and provides a clue as to which subunit combinations may be responsible for behavioral effects of drugs acting at GABAA receptors. Among the behavioral effects of such drugs are relief of anxiety (anxiolysis), muscle relaxation, sedation, anticonvulsion, and anesthesia.

See also[edit]

References[edit]

  • Chen K., Lia H.Z., Yea N., Zhanga J., and Wang J.J. 2005. Role of GABAB receptors in GABA and baclofen-induced inhibition of adult rat cerebellar interpositus nucleus neurons in vitro. Brain Research Bulletin, 67(4), 310-318.
  • Colquhoun D. and Sivilotti L.G. 2004. Function and structure in glycine receptors and some of their relatives. Trends in Neurosciences, 27(6), 337-344.
  • Martin I.L., and Dunn S.M.J. 2002. "GABA Receptors". Tocris Cookson Ltd.
  • Siegel G.J., Agranoff B.W., Fisher S.K., Albers R.W., and Uhler M.D. 1999. Basic Neurochemistry: Molecular, Cellular and Medical Aspects, Sixth Edition. GABA Receptor Physiology and Pharmacology. American Society for Neurochemistry. Lippincott Williams and Wilkins.
  • Cossart R, Bernard C, Ben-Ari Y. 2005. Multiple facets of GABAergic neurons and synapses: multiple fates of GABA signalling in epilepsies. TRENDS in Neurosciences, 28(2), 108-115

External links[edit]


GABA ABC[edit]

GABA acts via three classes of receptors (GABAA, GABAB and GABAC receptors) in the vertebrate CNS that differ in their structure, function, and pharmacology. The vast majority of fast GABA responses that are inhibited by bicuculline and picrotoxin and enchanced by benzodiazepines or barbiturates, result from the direct activation of an anion channel, that is referred to as the GABAA receptor.


A second type of ionotropic GABA receptor (the GABAC receptor) is insensitive to bicuculline, benzodiazepines and barbiturates, for review see Sieghart and Sperk, 2002. While GABAA receptors are widely distributed in the CNS, the presence of GABAC receptors is largely restricted to retinal bipolar or horizontal cells across vertebrate species. GABAA receptors are heterooligomeric channel forming proteins, formed by different subunits that can belong to 8 distinct classes (α, β, γ, δ, ε, π, θ, and ρ)providing the potential for a huge number of receptor subtypes. In contrast, GABAC receptors are homooligomeric receptors composed of ρ subunits. Although the GABAC receptor terminology is still used frequently, these receptors are thought to be part of the GABAA receptor family.


Fast responding GABA receptors are members of the Cys-loop ligand-gated ion channel superfamily (reviewed by Barnard et al., 1998) comprising nicotinic acetylcholine, GABAA, GABAC, glycine and 5-HT3 receptors that possess a characteristic loop formed by a disulphide bond between two cysteine residues. GABA binding to the extracellular domain of GABAA and GABAC receptors triggers the opening of an intrinsic chloride-selective ionophore that drives the membrane potential towards the reversal potential for Cl¯ ions (about –80 mV) in neurons. As a consequence, the probability that action potentials will be generated by excitatory neurotransmission is decreased. But GABAA receptor activation can also induce depolarizing responses, particularly in embryonic neurons. This phenomenon is due to increased intracellular Cl¯ concentration in certain neurons that occurs prior to the expression of the KCC2 transporter, which is principally responsible for Cl¯ extrusion. In adult brain, the GABAA receptor can also mediate depolarizing responses under certain physiological and pathological conditions that involve intense receptor activation.

GABAA Receptors[edit]

GABAA Receptor Subunit Types[edit]

Initially two subunits of the GABAA receptor named α and β were purified (Sigel et al., 1983, Sigel and Bernard, 1984). Subsequently the cDNAs coding for these subunits have been cloned (Schofield et al., 1987). So far 20 related GABAA receptor subunits in mammals were identified 6α, 4β, 3γ, 1δ, 1ε, 1π, 1θ, and 3ρ (Barnard et al., 1998, Bonnert et al., 1999, Moragues et al., 2000). A mammalian counterpart of the avian γ4 subunit (Harvey et al., 1993) has not yet been isolated by cDNA cloning and so is not included here. However, the β4 subunit gene, likewise discovered in the chicken (Bateson et al., 1991), has more recently been shown to be present in humans (Levin et al., 1996). Sequence homology within same class of GABAA receptor subunits is up to 80% and between subunit classes up to 40%. Homology with other members of Cys-loop receptor superfamily is about 10-20%. The subunits of this family are distributed in two groups, one containing the subunits forming anion selective receptors (GABAA, GABAC and glycine receptors), and one containing subunits forming cation selective receptors (5-HT3 and nicotinic receptors). For more complete insights in phylogenetical relationships within Cys-loop receptor family see reviews Ortells and Lunt, (1995) and Hervers and Luddens (1998). Splice variants add to the subunit diversity. Two forms of the γ2 subunit are generated from one gene (Whiting et al., 1990, Kofuji et al., 1991). These receptors are differently expressed in the brain (Glencorse et al., 1992). Two forms are also known for both the β2 and the β4 subunits (Bateson et al., 1991, Harvey et al., 1994). In each case, the longer and shorter products were designated “L” and “S,” and differ by the presence of absence of short peptide in the long intracellular loop between TM3 and TM4. Splicing of exon-1 results in two alternative forms of the β3 subunit (Kirkness and Fraser, 1993). The α6 subunit is alternatively spliced in approximately 20% of its transcripts in rat brain, causing a deletion at the N-terminus of 10-amino acid residues (Korpi et al., 1994). Interestingly this deletion abolishes the functional receptor activity in all subunit combinations tested so far.


Regional Distribution of GABAA Receptor Subunits in the Brain[edit]

Subunit isoform distribution in brain has been studied by using “in situ” hybridization at the mRNA level (Laurie et al., 1992, Persohn et al., 1992, Wisden and Seeburg, 1992, Miralles et al., 1994, Bonnert et al., 1999, Sinkkonen et al., 2000) and by using immuno-histochemical studies at the protein level (Benke et al., 1991a, b, c, Zimprich, et al., 1991, Fritschy et al., 1992, Gutierrez et al., 1994, Fritschy et al., 1995, Sperk et al., 1997, Kultas-Ilinsky et al., 1998, Fritschy et al., 1998, Moragues et al., 2000, Pirker et al., 2000, Schwarzer et al., 2001, Moragues et al., 2002, Moragues et al., 2003, Plotl et al., 2003). The individual subunits exhibit a distinct but overlapping regional and cellular distribution. Subunits α1, β1, β2, β3 and γ2 are found throughout the brain, although differences in their distribution were observed.

The α1 subunit is the most abundant, subunits α2, α3, α4, α5, α6, γ1, and δ are more confined to certain brain areas. The α2 subunits are preferentially located in forebrain areas. The highest concentrations were found in olfactory bulb, striatum, nucleus accumbens, septum, dentate gyros, amygdale and hypothalamus. α2 subunits were less abundant in thalamus (except reticular nucleus), midbrain and brainstem areas. α3 subunits were strongly expressed the glomerular and external plexiform layers of the olfactory bulb, in the inner layers of the cerebral cortex, the reticular thalamic nucleus, and the zonal and superficial layers of the superior colliculus, the amygdala and cranial nerve nuclei. Subunit α4 was strongly expressed in the thalamus, dentate gyros, olfactory tubercle and basal ganglia (Benke et al., 1997). The α5 subunit immunoreactivity was strongest in Ammon’s horn, the olfactory bulb and hypothalamus, whereas the α6 subunit is exclusively expressed in granule cells of the cerebellum and the cochlear nucleus (Pirker et al., 2000). The β subunits are widely distributed. The β2 subunit is one of the most widely distributed subunits in the brain. β1 and β3 subunits are less abundant (Benke et al., 1994).

Among the γ subunits the γ2 is most widely distributed throughout the brain, whereas γ1 and γ3 are relatively rare (Somogyi et al., 1996). The subunit γ1 is the rarest subunit and exhibits a quite specific distribution in the brain. It is preferentially located in the central and medial amygdaloid nuclei, in pallidal areas, the substantia nigra pars reticulate and the inferior olive. In contrast, the γ3 subunit is expressed in most brain areas but with low abundancy. The δ subunit can be co-localized with the α4 subunit, e.g. in the thalamus, striatum outer layers of the cortex and in the dentate molecular layer and in the neonatal hippocampus (Sur et al., 1999, Bencsits et al., 1999, Didelon et al., 2000). In cerebellum it is co-distributed with α6 subunit (Pirker et al., 2000). The π subunit was detected in several peripheral human tissues as well as in the brain (hippocampus and temporal cortex) and was particularly abundant in the uterus (Hedblom and Kirkness, 1997) So far no study investigating the detailed regional distribution of the π subunit has been published. The θ subunit (Bonnert et al., 1999) seems to be expressed in various regions, including the hypothalamus, amygdala, hippocampus, substantia nirga, dorsal raphe and locus coeruleus (Sinkkonen et al., 2000, Moragues et al., 2002). θ subunits showed strikingly overlapping expression patterns with ε subunits throughout the brain, especially in the septum, preoptic areas, various hypothalamic nuclei, amygdala, and thalamus, as well as in monoaminergic groups (Moragues et al., 2002). As with the ε subunit, there were some discrepancies in the cDNA sequence obtained by different groups (Bonnert et al., 1999, Sinkkonen et al., 2000).

The ρ subunits seem to be preferentially expressed in the retina. Immunohistochemistry in the retina using an antibody recognizing all 3 ρ subunits revealed a staining pattern restricted to the terminals of bipolar cells in the inner plexiform layer which did not overlap with GABAA α or β subunits (Enz et al., 1996, Koulen et al., 1998, Fletcher et al., 1998, Koulen, 1999). mRNA encoding ρ subunits, however, is present also in the superior colliculus, dorsal lateral geniculate nucleus and cerebral Purkinje sells (Boue-Grabot et al., 1998, Wegelius et al., 1998). In addition, bicuculline-resistant and baclofen-independent GABA effects were reported in the cerebellum (Drew and Johnston, 1984, Drew and Johnston, 1992), superior colliculus (Arakawa et al., 1988, Clark et al., 2001), amygdala (Delaney and Sah, 1999), hippocampus (Martina et al., 1996, Cherubini et al., 1998, Didelon et al., 2002), dorsal geniculate cells (Zhu and Lo, 1999) and spinal cord (Park et al., 1999). This indicates that ρ subunits may be present in many CNS regions and are more prevalent than previously suspected.


Architecture of Recombinant GABAA Receptors[edit]

Architecture of Recombinant GABAA Receptor Subunits[edit]

All GABAA receptor subunits are composed of a large N-terminal extracellular domain, four transmembrane (TM) domains, and a large intracellular loop between TM3 and TM4 (Schofield et al., 1987). Subunits are up to 460 amino acids in length. The N-terminal extracellular domain is carrying several potential sites for N-linked glycosylation (Buller et al., 1994) and two conserved cysteine residues. Upon receptor assembly homologous parts of this domain form at subunit interfaces binding sites for agonists and ligands of the benzodiazepine binding site. Other binding sites as those for anesthetics, barbiturates, ethanol, furosemide, zinc and some other compounds located within transmembrane domains. Each subunit contributes to the channel lining that is largely formed by residues of TM2, and possibly of TM3 membrane-spanning segments (Xu and Akabas, 1996, Williams and Akabas, 1999, Goren et al., 2004).

The three-dimensional structure of a related receptor – the nicotinic acetylcholine receptor (Unwin, 1993, Miyazawa et al., 1999, Miyazawa et al., 2003) indicates that the transmembrane domains have an α-helical structure, the pore is shaped by an inner ring of α-helices, which curve radially to create a tapering path for the ions, and an outer ring of α-helices, which coil around each other and shield the inner ring from the lipids. The gate is a constricting hydrophobic girdle at the middle of the lipid bilayer, formed by weak interactions between neighboring inner helices.


How Many Subunits Make a GABAA Receptor?[edit]

As mentioned above the GABAA receptor subunits share amino acid sequence homology with the subunits of the nicotinic acetylcholine receptors. The muscle type of the nicotinic acetylcholine (nAChR) receptor occurs in Torpedo electric organ in such a high density that it is possible to prepare membranes containing a lattice of the receptors, from which a low-resolution three-dimensional structure of the molecule could be obtained by electron optical diffraction techniques (Toyoshima and Unwin, 1988, Unwin, 1993, Miyazawa et al., 2003, Unwin, 2003). Those studies clearly showed that the muscle type nicotinic receptor is pentameric, with the ion channel located in the center of a rosette. For the GABAA receptors, the situation is more complex as no such rich sources exist. Using purified GABAA receptors from pig brain cortex combined with image analysis in the electron microscope, dispersed single receptor molecules have been visualized and analyzed. This method indicated a pentamer (Nayeem et al., 1994). Furthermore, the negatively stained images indicated a central pore in the pentameric rosette.

Independent evidence to support a pentameric structure has been obtained in several ways. Hydrodynamic estimates of the size of GABAA receptors in solution, either native (Mamalaki et al., 1989) or α1β3γ2 recombinant receptors (Tretter et al., 1997) are consistent with the molecular weight of a pentamer. Further, the integral ratios of the subunits combined in several forms of functional recombinant receptors, as determined by diverse methods, fit best in each case with total of five subunits (Backus et al., 1993, Im et al., 1995, Chang et al., 1996, Tretter et al., 1997, Ferrar et al., 1999). A powerful way to gain insight into the arrangement of subunits in GABAA receptors and their stoichiometry is the use of a predefined alignment of subunits by producing linked subunit constructs with the aid of gene fusion (Im et al., 1995, Baumann et al., 2001, Baumann et al., 2002). Analysis of receptors formed by linked subunits also indicated a pentameric stoichiometry.


Assembly of GABAA Receptor Subunits[edit]

GABAA receptor assembly occurs within the endoplasmic reticulum (ER) (Czajkowski and Farb, 1989, Kittler et al., 2000, Moss and Smart, 2001, Kittler et al., 2002). Their distinct subunit compositions may provide distinct functional properties e.g. modulation by endogenous ligands such as neurosteroids (Twyman and Macdonald, 1992, Wohlfarth et al., 2002, Akk and Steinbach, 2003, Bianchi and Macdonald, 2003) or second messenger systems (Angelotti et al., 1993, Moss and Smart, 2001), subcellular localization (Connolly et al., 1996a), or long term differences in the regulation of types of receptor surface expression (Connolly et al., 1999a,b). Many neuron express multiple receptor subunit mRNAs simultaneously (Wisden and Seeburg, 1992, Sieghart and Sperk, 2002), suggesting that cellular mechanisms for differential receptor assembly may also exist. To achieve the correct arrangement of subunits around the pore, each subunit must form specific contacts, assembly signals, on interfaces contacting with neighbors subunits. The presence of such multiple assembly signals is capable of differential interaction with other subunits may permit construction of different GABAA receptors. Individual subunits may not be committed to a particular receptor subtype, but may function as universal building blocks in the generation of diverse receptor compositions.

In the α1 subunit residues (54–68) on (-) side were identified as important for assembly with β subunits (Taylor et al., 2000, Klausberger et al., 2000, Sarto et al., 2002a). An additional interaction site included the residues (80–100) on α1 subunit (+) interface, which are believed to be important for assembly with the γ2 subunit (Klausberger et al., 2001). Subsequently, single amino acid residues implicated in assembly were identified. Thus it was shown that a single amino acid residue Q67 (Taylor et al., 2000) is important for assembly of α1 with β3 but not with γ2 subunits. Conversion of a single amino acid in α1 to that of γ2 (R66A) was shown to be sufficient to alter the assembly profile of the α1 subunit to that of the γ2 subunit. It was also shown that presence of this residue is required for the assembly of α1β2 but not α1β1 or α1β3 (Bollan et al., 2003a,b). Two tryptophan residues α1W69 and α2W94, on the rat α1 subunit were found to be critical for the assembly of the GABAA receptor pentamer (Srinivasan et al., 1999).

In β2 and β3 subunits it was found that region (52–66) on (-) interface (Taylor et al., 1999, Klausberger et al., 2000, Sarto et al., 2002a) is important for assembly with α1 subunits. An additional region (76–89) located on β3 subunit (+) interface is important for assembly with α1 subunit was identified later (Ehya et al., 2003). In the γ subunits amino acid sequences γ2 (67–81) (Sarto et al., 2002a), γ3 (70–84) (Sarto et al., 2002b) located at (-) interface and γ2(83–90) and γ2 (91–104) located at (+) interface (Klausberger et al., 2000) were identified as sites important for assembly with α1 and β3 subunits. There is also a report that a singe amino acid γ2W82 residing on (-) interface upon mutation to cysteine failed to express with α1 and β2 subunits (Teissere and Czajkowski, 2001). Regions α1 (54–68), β2 (52–66), and γ2 (67–81), are located in homologous regions of (-) sides of the different subunits (Sarto et al., 2002b). It has been reported that dimer or trimer assembly intermediates of GABAA receptor subunits can form binding sites for [3H]muscimol and [3H]Ro15-1788 (Klausberger et al., 2001).

Interestingly, these assembly signals or intersubunit contact points at the α, β and γ subunits (Taylor et al., 2000, Klausberger et al., 2000, Klausberger et al., 2001, Sarto et al., 2002a,b) overlap with the GABA (Boileau et al., 1999b) and benzodiazepine binding sites (Buhr et al., 1997, Boileau et al., 1999a, Teissere et al., 2001, Sigel, 2002) formed at subunit interfaces between the α/β and α/γ subunits.


Subunit Composition of Recombinant GABAA Receptors[edit]

With the application of molecular biology approaches, in the late 1980s and 1990s, it soon became clear that a family of GABAA receptor subtypes composed from different subunits exists within the brain. If all these subunits could randomly co-assemble with each other, more than 151,887 GABAA receptors subtypes with distinct subunit composition, arrangement would be formed (Burt and Kamatchi, 1991). Not all subunits can assemble efficiently with each other and form functional receptors.


Homo-oligomeric Recombinant GABAA Receptors[edit]

Recombinant expression studies have indicated that at least some of the GABAA receptor subunits can form homo-oligomers. The extent of formation of these homo-oligomers, however, varies dramatically. Whereas some are robustly formed in all recombinant expression systems, others are formed with low efficiency only (Blair et al., 1988). Xenopus oocytes or HEK-293 cells have been used mainly as host cells.

A robust expression of GABA-activated homo-oligomeric chloride channels was observed with ρ subunits (Cutting et al., 1991, Shimada et al., 1992, Kusama et al., 1993a, b, Wang et al., 1994, Shingai et al., 1996). To smaller extent expression of homo-oligomeric receptors was observed with β1 or β3 subunits (Connolly et al., 1996a, b, Krishek et al., 1996b, Wooltorton et al., 1997b) and γ2L subunits (Martinez-Torres and Miledi, 2004).

Interestingly, channels formed by murine or rat β1 (Sigel et al., 1989, Krishek et al., 1996b) or β3 subunits (Wooltorton et al., 1997b) were open in the absence of GABA, but could be inhibited with channel blocker picrotoxin. This effect seems to be species dependent, because human or bovine β1 subunits seem to be able to form homo-oligomeric channels closed in the absence of GABA (Pritchett et al., 1988, Krishek et al., 1996b, Sanna et al., 1995).


Hetero-oligomeric Recombinant GABAA Receptors Composed of Two Different Subunits[edit]

The efficiency of receptor formation of two different subunits depends on the subunit combination. Whereas different αβ subunit combinations are expressed efficiently and form GABA-activated channels in all systems investigated, conflicting results were obtained with αγ or βγ subunit combinations (Verdoorn et al., 1990, Sigel et al., 1990, Knoflach et al., 1992, Angelotti et al., 1993). The efficiency of formation of pentameric α1γ2 or β3γ2 receptors heterologously expressed in HEK-293 cells seems to be low (Tretter et al., 1997). For the cells co-expressing β3 and γ2L subunits, γ2L could be detected on the surface of only about 15% of cells, indicating that most of the receptors formed in these cells were homo-oligomeric β3 receptors (Taylor et al., 1999, Bollan et al., 2003a, b). β1γ2S, β2γ2Sand β3γ2S receptors also formed in HEK-293 cells to a comparable extent and exhibit pharmacological properties distinct from that of homo-oligomeric β1-3 receptor (Taylor et al., 1999, Hamon et al., 2003). It has been observed that α1γ2 or β2γ2L subunits combinations were retained within the endoplasmatic reticulum (Connolly et al., 1996a, b). It is thus possible that receptors composed of these subunit combinations can only be formed under certain experimental conditions, such as in the presence of suitable chaperons, at high subunit concentrations due to high synthesis rates (conditions that are present in some recombinant receptor systems). No information is available on the possible formation of GABAA receptors composed of αδ, βδ or γδ subunits. No functional channels, however, were formed on co-transfection of α1ε or β1ε (Whiting et al., 1997) or of α1π or β1π (Hedblom et al., 1997) subunit combinations.

In contrast, different ρ subunits can combine with each other and might also co-assemble to functional receptors in vivo (Enz and Cutting, 1998). Although in one study it was demonstrated that ρ subunits are unable to assemble with α1, β1 or γ2 subunits (Enz and Cutting, 1998), other studies indicated that ρ subunits can assemble with γ2 subunits and possibly also with glycine receptor subunits, and also form functional receptors found in certain cell types of the retina (Pan et al., 1997, Qian and Ripps, 1999, Pan et al., 2000).


Hetero-oligomeric Recombinant GABAA Receptors Composed of Three and More Different Subunits[edit]

Recombinant receptors subtypes composed of an α, a β and a γ subunit mainly have been studied so far (Sieghart, 1995, Herves and Luddens, 1998, Sieghart and Sperk, 2002). Assuming the stoichiometry of 2:2:1 these receptors can have at least one of three general compositions: 2α/2β/γ; 2α/β/2γ; α/2β/2γ. Here, a notation is introduced in which the numeral represents the number of molecules of a given subunit class (α, β, etc.) present in one receptor molecule and not the isoform identity within that class. Based on abundance of co-expression, it is assumed that α1β2γ2 represents the most abundant GABAA receptor in adult mammalian brain (Herves and Luddens, 1998, Sieghart and Sperk, 2002). Additional cases such as 3α/β/γ, α/3β/γ and α/β/3γ are theoretically possible, but immunoprecipitation (Tretter et al., 1997), measurements of electrophysiological properties (Backus et al., 1993, Chang et al., 1996) and fluorescence energy transfer (Ferrar et al., 1999) have excluded (at least in those cases) the presence of three identical subunit isoforms in one receptor molecule.

Additional studies have indicated that receptors containing two different α subunit isoforms, in combination with a β and a γ subunit can assemble and exhibit properties that are distinct from those of receptors containing only a single type of α subunit (Verdoorn et al., 1990, Sigel et al., 1990, Polenzani et al., 1991, Verdoorn, 1994, Sigel and Baur, 2000, Hansen et al., 2001).

Similarly, it has been demonstrated that receptors containing two different types of β subunits together with one of α and γ subunit are able to assemble and to exhibit properties different form receptors that contain only a single β subunit subtype (Fisher and Macdonald, 1997). Finally, it has been demonstrated that recombinant receptors composed of α1, β1, the long splice variant of γ2L, and δ (α1β2γ2Lδ) or α1, β3, γ3, and π (α1β3π and α1β3γ3π) subunits can also be formed and exhibit properties distinct from those of α1β1γ2L or α1β1δ receptors (Saxena and MacDonald, 1994, Hansen et al., 2001, Hevers et al., 2000) or from those of α1β3 and α1β3γ3 receptors (Neelands and Macdonald, 1999) respectively. Although experiments investigating the expression of five different subunits have been performed in Xenopus oocytes, the results obtained were difficult to interpret (Sigel et al., 1990). This is not surprising because from the five different subunits simultaneously expressed in the oocytes a variety of different receptor subtypes composed of 3, 4, or 5 different subunits could have been formed, that all could have contributed to the chloride current measured in these cells. This problem could be solved by linking multi-subunits by gene fusion. This methodology has already been applied successfully to GABAA receptors (Baumann et al., 2001, Baumann et al., 2002, Baumann et al., 2003).


Functional Architecture of GABAA Receptors[edit]

The N-terminal domains of GABAA receptor subunits are implicated in receptor assembly and in formation of agonist and benzodiazepine binding sites. Two agonist binding sites are harbored by α(-)/β(+) subunit interfaces (Boileau et al., 1999b, Teissere and Czajkowski, 2001). It was found recently that even though agonist sites are located at similar interfaces formed by identical (-) sides of α and (+) sides of β subunits, they have dissimilar properties: site 2 has an approximately threefold higher affinity for GABA than site 1, whereas muscimol and bicuculline show some preference for site 1 (Baumann et al., 2003). The benzodiazepine binding site is located at α(+) and γ(-) subunit interface. Interestingly, domains involved in the formation of the GABA and benzodiazepine binding sites are homologous (reviewed in Sigel, 2002). A large body of evidence has been collected suggesting that anesthetics, barbiturates, alcohols and number of other drugs share some overlapping structural determinants for their actions on the GABAA receptor. All these allosteric sites are located within the gating domain and it was observed that compounds acting at these sites were capable of direct activation of channel in the absence of GABA (Belelli et al., 1999, Akk and Steinbach, 2000).

On the cytoplasmatic side at the entry to channel pore is located binding site for channel blockers. This chemically inhomogeneous group of compounds is comprised from substances acting as physical plugs. Prototypic compounds of this class are picrotoxinin (Inoue and Akaike, 1988, Yoon et al., 1993), the bicyclic caged compound TBPS (Squires et al., 1983, Supavilai and Karobath, 1983). Properties of this binding site were found to be similar among different αβγ receptors (Bell-Horner et al., 2000), however, some unusual subunit combinations, like receptors formed by β subunits (Sigel et al., 1989, Krishek et al., 1996b, Wooltorton et al., 1997b) showed increased sensitivity to channel blockers. In following the binding sites for the receptor agonist GABA, the benzodiazepine binding pocket, binding sites located within transmembrane domains of subunits and binding site of channel blockers are discussed in more detail.


Agonist Binding Sites of the GABAA Receptor[edit]

Ligands Acting at Agonist Sites of GABAA Receptors[edit]

The endogenous activator of GABAA receptors is GABA. Various compounds of different type and intrinsic activity are also recognized by the agonist binding site of the GABAA receptor. Binding of agonist is coupled to the opening of the channel, the so-called channel gating. Partial agonists differ from agonists in respect of channel opening efficacy. Binding of a competitive antagonist is stabilizing the closed state of the receptor channel. Competitive antagonists are viewed as classic competitive inhibitors of GABAA receptor (Macdonald and Olsen, 1994), but there are indications that they can induce conformational changes (Ueno et al., 1997, Bianchi and Macdonald, 2001, Wagner and Czajkowski, 2001).


Architecture of the Agonist Binding Sites[edit]

Residues implicated in agonist binding are assigned to at least six different non-contiguous extracellular N-terminal regions of the α and β subunits. These regions have been designated loops A–F in the homologous nicotinic acetylcholine receptor (Corringer et al., 2000, Le Novere et al., 2002a). In GABAA receptors, the agonist binding site is formed by (-) side of α and (+) side of β subunit. Residues in different loops likely have different functional roles. Some residues may directly contact ligand, some may be important for maintaining the structural integrity of the binding site, and others may mediate local conformational movements within the site.

The following residues are thought to take part in the formation of the agonist site. On the α1 subunit, residues identified include F64 (Sigel et al., 1992, Smith and Olsen, 1994), R66, S68 (Boileau et al., 1999) (on loop D, K116, R119, and I120 (Westh-Hansen et al., 1997, Westh-Hansen et al., 1999, Hartvig et al., 2000) (on loop E) and V178, V180, D183 (Newell and Czajkowski, 2003) (on loop F). Complementary residues in the β2 subunit include Y97, L99 (Boileau et al., 2002) (on loop A), Y157, T160 (Amin and Weiss, 1993) (on loop B), T202, S204, Y205, R207, and S209 (Amin and Weiss, 1993, Wagner and Czajkowski, 2001) (on loop C) have been identified.

The spatial arrangement of these residues was unclear until direct crystallographic evidence was obtained on protein involved in synaptic transmission of the snail Lymnaea stagnalis has helped to visualize residues forming the binding site after homology modeling. This water-soluble protein is called acetylcholine binding protein (AChBP). AChBP subunit is 210 residues long, forms a stable homopentamer (Brejc et al., 2001, Smit et al., 2003) and shares 24% sequence homology with the N-terminal part of human α7 nicotinic receptor subunit and about 15% with subunits of GABAA receptor family.


The Benzodiazepine Binding Site[edit]

Ligands of the Benzodiazepine Binding Site[edit]

In 1957, scientists at a drug company (Hoffmann-La Roche) by accident discovered that a new compound, chlordiazepoxide reduced fear in animals. This compound was a benzodiazepine and its discovery ushered a new era in treatment of anxiety and related disorders. Since then, the number of compounds with the structure of benzodiazepine template reached more than 3000. Among the pharmacological agents that allosterically modulate GABAA receptors, the benzodiazepines have gained major clinical relevance (Mohler et al., 1996a,b, Sieghart, 2003). Present evidence suggests that GABAA receptors are the only effector sites of benzodiazepines in the central nervous system. Ligands of the benzodiazepine binding site have been subdivided into three classes according to their intrinsic activity: positive allosteric modulators, negative allosteric modulatorsand antagonists. The names “agonist”, “inverse agonist” and “antagonist” are also used for these compounds. Classical benzodiazepines upon binding to GABAA receptor exert their positive allosteric effect by increasing the affinity of GABA for its binding sites without affecting maximum response. This results in increased probability of channel opening (Rogers et al., 1994). Antagonists of the benzodiazepine site do not affect GABA-elicited responses. However, they prevent positive allosteric modulators from binding and thus, from allosteric modulation of receptor function. Negative allosteric modulators have opposite effects to positive allosteric modulators, decreasing affinity for GABA. An additional, “peripheral” recognition site of benzodiazepines, structurally and functionally unrelated to GABAA receptors, is located at 18 kDa protein found in the mitochondrial membrane (for review see Papadopoulos et al., 2001).


Architecture of the Benzodiazepine Binding Site in Recombinant αβγ Receptors[edit]

The benzodiazepine binding site has been shown to be located at the interface between the α- and γ-subunits, with residues from each subunit contributing to the binding site (Casalotti et al., 1986, Deng et al., 1986, Pritchett et al., 1989, Smith and Olsen, 1995, Sigel and Buhr, 1997, Sigel, 2002). Photoaffinity labeling of the receptor by benzodiazepines [3H]flunitrazepam and [3H]Ro15–4513 has been performed (Mohler et al., 1980, Fuchs et al., 1988, Stephenson et al., 1990, McKernan et al., 1995, Davies et al., 1996, Smith and Olsen, 2000, Sawyer et al., 2002). The residues H101 (rat numbering) (McKernan et al., 1995, Duncalfe and Dunn, 1996, Duncalfe et al., 1996, Smith and Olsen, 2000) and P97 (Smith and Olsen, 2000) have been shown to be the major sites of incorporation of [3H]Flunitrazepam into the α1 subunits. [3H]Ro15-4513 can also be photoincorporated into α subunits of the GABAA receptor (Sieghart et al., 1987). The amino acid(s) photolabeled by [3H]Ro15-4513 are contained within a subunit fragment extending from residue 104 to the C terminus of the α1 subunit (Duncalfe and Dunn, 1996), possibly within amino acids 247–289 spanning the end of the TM1 to the beginning of the TM3 (Davies and Dunn, 1998). The results from a recent study suggest that [3H]Ro15-4513 is photoincorporated into α1Y209 and in homologous positions in the α2 and α3 subunits (Sawyer et al., 2002).

Extensive mutagenesis experiments have also identified other α1 residues implicated in benzodiazepine binding. The significance of α1H101 has initially been demonstrated in studies in which this residue has been substituted with arginine, the native residue at the homologous position in α4 and α6 subunits (Wieland et al., 1992). Substitution of this histidine by arginine resulted in about 500-800 fold decrease in affinity of classical benzodiazepines (Wieland et al., 1992). Extensive mutational analysis of α1H101 residue has also revealed its implication in allosteric coupling between GABA and benzodiazepine binding sites (Davies et al., 1998b).

The following residues in the α1 subunit were shown to either affect benzodiazepine sensitivity in functional assays or benzodiazepine affinity in binding studies, Y159 (Amin et al., 1997) and Y209 (Amin et al., 1997, Buhr et al., 1997b), T162 (Wieland and Luddens, 1994), G200 (Pritchett and Seeburg, 1991, Schaerer et al., 1998, Wingrove et al., 2002), T206 (Buhr et al., 1997b), V211 (Casula et al., 2001) and I215 (Strakhova et al., 2000). In the γ2 subunit M57 (Buhr and Sigel, 1997) and Y59 (Kucken et al., 2000) were found to be essential determinants for conferring high-affinity for classical and atypical benzodiazepines. The F77 residue was absolutely crucial for maintaining ability of benzodiazepine binding site to recognize its classical ligands (Buhr et al., 1997a, Wingrove et al., 1997, Sigel et al., 1998). It should be noted that this residue is homologous to F64 in α subunit, which has been previously shown to be a key determinant of the GABA binding site, initially suggesting a conservation of motifs between different ligand binding sites on the GABAA receptor (Sigel et al., 1992, Smith and Olsen, 1994, Buhr et al., 1997a, Sigel et al., 1998). Residue M130 is required for high affinity binding of benzodiazepine binding site ligands (Buhr and Sigel, 1997, Wingrove et al., 1997, Sigel et al., 1998). And finally, threonine residue at position 142 was implicated in the efficacy of benzodiazepine binding site ligands (Mihic et al., 1994). Recently using analogy to a GABA binding pocket residues A79 and T81 which are clustered on a β-strand around F77 were found to line up the part of binding pocket (Kucken et al., 2000, Teissere and Czajkowski, 2001).


Benzodiazepine Binding Sites May Be Composed of Different α and γ Subunit Isoforms[edit]

Different α and γ subunit isoforms can assemble to form a benzodiazepine binding site thereby imposing different pharmacological properties. Benzodiazepine receptors have been classified pharmacologically into those which recognize the classical, 5-phenyl-1,4-benzodiazepines (for example diazepam and flunitrazepam) referred to as ‘diazepam-sensitive’ receptor and those which do not recognize these ligands referred to as ‘diazepam-insensitive’ receptor (Malminiemi and Korpi, 1989, Hadingham et al., 1996, Knoflach et al., 1996). As described above, the residue at position 101 of α1 (and homologous positions in other α subunits) has been shown to determine the affinity for diazepam. α1, α2, α3 or α5 have a histidine in this position and display high affinity for diazepam, while α4 or α6 have arginine at the homologous position do not bind diazepam (Wieland et al., 1992, Dunn et al., 1999, Kelly et al., 2002). α1, α2, α3 and α5 subunit containing receptors can be further subdivided by their affinity to CL218872 with the higher affinity α1 subunit containing receptors have a higher affinity and are referred to BZI type receptors and the α2, α2 or α5 subunit containing receptors to BZII type receptors (Pritchett and Seeburg, 1991, Yang et al., 1995). Selectivity for CL218872 and zolpidem of α1 subunits over α6 is conferred mainly by α6T161 (Wieland and Luddens, 1994, Renard et al., 1999), α1G201 (Pritchett and Seeburg, 1991, Schaerer et al., 1998), α1S205 (Wieland and Luddens, 1994, Renard et al., 1999) and α1V211 (Casula et al., 2001).

It is noticeable that the subunits conferring the higher affinity to zolpidem have the smaller amino acid residues at both positions 201 and 211, suggesting a steric role for these residues in benzodiazepine selectivity. It was proposed that a similar mechanism also underlies more than 1000-fold decrease in affinity for diazepam, flunitrazepam, zolpidem and CL218872 at α6-containing receptors compared to those having α1 (Luddens et al., 1990, Hadingham et al., 1996, Casula et al., 2001, Wingrove et al., 2002). Another residue conferring insensitivity of α6 subunit containing receptors to β-carboline β-CCE is α6N204. Introduction of a point mutation α6N204S or α6N204I, found in homologous position of α1 or α4 subunits restored affinity (Derry et al., 2004).

From two studies of Stephenson et al. (1990), Puia et al. (1991) and McKernan et al. (1995) it was clear that the type of γ subunit also contributes significantly to the properties of the benzodiazepine binding site. GABAA receptors containing a γ1 subunit have a >5000-fold lower affinity for the antagonist Ro15-1788 than do those containing a γ2 or γ3 subunit, whereas γ1- and γ3-containing receptors have a about 100-fold reduced affinity for zolpidem and 10–30-fold lower affinity for flunitrazepam than do receptors containing γ2 (Hadingham et al., 1995, Benke et al., 1996, Wingrove et al., 1997). Two amino acid residues are determining selectivity of zolpidem for γ2 subunit containing receptors (Wingrove et al., 1997, Buhr et al., 1997a, Buhr and Sigel, 1997). These are γ2F77 and γ2M130; γ1 subunit has an isoleucine (γ1I79) at position homologous to γ2F77 and additionally γ1 and γ3 subunits contain a leucine in position homologous to γ2M130 (γ1L132 and γ3L133). Introduction of point mutations γ1I79F, γ1L132M in γ1 subunit and γ3L133M in γ3 restored zolpidem affinity (Wingrove et al., 1997, Buhr and Sigel, 1997).


Relative Position of Ligands in the Benzodiazepine Binding Site[edit]

Many attempts have been made to characterize interactions of benzodiazepine binding site ligands in their binding pocket and to superimpose positive and negative allosteric modulators and antagonists (Borea et al., 1987, Villar et al., 1989, Schove et al., 1994, Zhang et al., 1995b, Huang et al., 1998, Huang et al., 1999, He et al., 2000, Marder et al., 2001, Verli et al., 2002). All these studies have investigated quantitative-structure affinity/activity relationships using chemically related compounds and have inferred type of interacting points e.g. lipophilic, aromatic, H-donor, H-acceptor and distances between these interaction centers. Such studies, have been found to be useful to estimate binding affinities and mode of action of newly designed ligands of the benzodiazepine binding site, however have very limited use for positioning of a ligand in the binding pocket. As detailed above, a soluble remote homologue of the N-terminal extracellular domain of nicotinic acetylcholine receptors (nAChR), the acetylcholine binding protein (AChBP), has been recently crystallized (Brejc et al., 2001). This crystal structure, featuring a novel fold of modified immunoglobulin-like topology, was used to construct homology models of the N-terminal domain of other superfamily members. A few models of nAChRs (Fruchart-Gaillard et al., 2002, Le Novere et al., 2002b, Schapira et al., 2002) and, also of GABAA receptors (Cromer et al., 2002, Trudell, 2002, Ernst et al., 2003) have been published.

These models of the extracellular domain of GABAA receptors provide a tool for the visualization of existing data (Holden and Czajkowski, 2002, Kash et al., 2003, Kucken et al., 2003a, Kash et al., 2004) and planning of rational mutagenesis studies. However, authors indicate many uncertainties in these models (Ernst et al., 2003). Their limited accuracy results from the low sequence homology between GABAA receptor subunits and AChBP template. Computational docking in models of the benzodiazepine site is presently hampered (Ernst et al., 2003). In spite of these problems possible positioning of the imidazobenzodiazepine Ro15-4513 has been suggested (Sawyer et al., 2002). [3H]Flunitrazepam primarily labeled residue H101 (rat numbering) (McKernan et al., 1995, Duncalfe et al., 1996, Smith and Olsen, 2000) and P97 (Smith and Olsen, 2000) and it has been assumed that in the binding pocket flunitrazepam molecule is pointed in the direction of these amino acids (reactive group in this case presumably not diffusing away from the primary site of radioactivity incorporation). In case of [3H]Ro15-4513 was shown to label α1Y209 and homologous positions of the α2 and α3 subunits (Sawyer et al., 2002). It is, however, difficult to infer the precise geometrical orientation from these labeling studies.

Observations relevant for the interaction of ligands of the benzodiazepine binding site with the γ subunit were made in two different studies. In the first affinities of ligands based on two templates – imidazobenzodiazepines (Ro15-1788-like) and 5-phenyl-1,4-benzodiazepine (diazepam- and flunitrazepam-like) to wild-type and mutant receptors were determined (Sigel et al., 1998). The authors concluded, that the extra hydroxyl group in tyrosine introduced in the mutant γ2F77Y interferes with the phenyl moiety of benzodiazepine and therefore γ2F77 should be close to the phenyl substituent in 5-phenyl-1,4-benzodiazepines (Sigel et al., 1998). Another study, where the size of moiety occupying the 3’-imidazo substituent (ester group in Ro15-1788/Ro15-4513) was varied together with the volume of the residue introduced in the γ2A79 position suggested that that Ro 15-4513 spans the binding site between α1Y209 and γ2A79, with the azide substituent facing the α1 subunit and the 3’-imidazo substituent facing the γ2 subunit. Computational docking of Ro 15-4513 and Ro15-1788 into the benzodiazepine binding site performed in the same study position the 3’-imidazo substituent near γ2A79 and γ2T81 residues (Kucken et al., 2003).


Allosteric Sites within Transmembrane Domains[edit]

Compounds Acting at Allosteric Sites within Transmembrane Domains[edit]

Modulation of GABAA receptor function by most volatile, intravenous, general anesthetics (Belelli et al., 1997, Belelli et al., 1999, Krasowski et al., 2001a,b, Korpi et al., 2002, Siegwart et al., 2002, Bali and Akabas, 2004), alcohols (Wick et al., 1998, Ueno et al., 1999, Mascia et al., 2000, Ueno et al., 2000), anticonvulsants (Vaught, and Wauquier, 1991, Wafford et al., 1994) and allosteric antagonists (Korpi et al., 1995, Thompson et al., 1999a, Fisher, 2002) is mediated via allosteric binding sites located within transmembrane domains of α and β subunits. At high concentration some compounds like propofol, barbiturates, loreclezole and etomidate can directly gate ion channel of the GABAA receptor in the absence of its agonist, GABA (Sanna et al., 1996, Akk and Steinbach, 2000, Steinbach and Akk, 2001). At low concentrations they modulate GABA-induced openings (Belelli et al., 1999), and, depending on the type of compound, this potentiation of GABA-gated currents appears to alter receptor deactivation and/or desensitization (Mozrzymas et al., 1999, Li and Pearce, 2000, Bai et al., 2001).


Allosteric Binding Sites Located within α and β Subunits[edit]

A set of residues located in the transmembrane domains 1-4 of GABAA receptor α and β subunits confer potency of various clinically used compounds. Residues implicated in formation of these binding sites are located within homologous domains of α and β subunits. It is worth noting, that the same transmembrane regions have been described as an integral part of the channel gating domain of the GABAA receptor (Xu and Akabas, 1996, Horenstein et al., 2001) and other ligand-gated ion channels (Le Novere et al., 2002a, Unwin, 2003, Miyazawa et al., 2003).

Residue α1G223F of TM1 segment of α subunits affects receptor gating induced by pentobarbital and propofol (Engblom et al., 2002), another residue α2L232F was implicated in halothane action (Jenkins et al., 2001). It was found that a single amino acid in α6 subunit, α6Ι230 confers sensitivity to furosemide (Jackel et al., 1998, Thompson et al., 1999a). In the TM2 and TM3 segments residues α1S270 and α1A291 are forming part of binding site for ethanol (Ueno et al., 1999, Krasowski and Harrison, 2000, Ueno et al., 2000), halothane, isoflurane and propofol (Mihic et al., 1997, Krasowski et al., 1997, Krasowski et al., 1998a, Koltchine et al., 1999, Jenkins et al., 2001, Nishikawa et al., 2002, Nishikawa and Harrison, 2003).

Complementing residues of this allosteric site were identified on the α1 subunit TM4 segment. Introduction of a tryptophane mutation in residues α1Y411, α1T414 and α1Y415 was reducing ability of isoflurane, halothane and chloroform to modulate channel function (Jenkins et al., 2002).

A number of residues located on transmembrane domains 1-4 of β subunits are implicated in the formation of recognition sites for compounds discussed above. Residues β2G219F, β2N265 and β2M286, which are homologous to α1G232, α1S270 and α1A291 confer sensitivity to inhaled, general anesthetics and anticonvulsants (Wafford et al., 1994, Thompson et al., 1999, Carlson et al., 2000, Krasowski and Harrison, 2000, Serafiniet al., 2000, Krasowski et al., 2001a,b, Siegwart et al., 2002, Thompson et al., 2002, Chang et al., 2003, Bali and Akabas, 2004).

Findings concerning the β2N265 residue were recently confirmed in studies of genetically modified mice. Thus, mice carrying β2N265S knock-in mutation were lacking the sedative effects produced by etomidate (Reynolds et al., 2003) whereas the β3N265S mutation rendered mice insensitive to anesthetic effects of propofol and etomidate, with small reduction in potency of volatile anesthetics (Jurd et al., 2003). However, there are some subtle differences concerning presence of additional sites – for loreclezole and zinc. Selectivity of loreclezole for β23 over β1 subunit containing receptors is determined by TM3 residue β2N289/β3N290. Introduction of a single serine to asparagine mutation in β1 subunit (β1S289N) was enough to confer loreclezole sensitivity of otherwise loreclezole-insensitive GABAA receptors (Wingrove et al., 1994).

Residues conferring sensitivity of GABAA receptors to Zn2+ were identified in the TM2 domain. Mutations of residues β2H267 and β2G270 located close to the entrance of the channel pore were found to reduce inhibition by zinc about 650-fold (Wooltorton et al., 1997a,b, Horenstein and Akabas, 1998, Hosie et al., 2003).


Channel Blockers and their Binding Site[edit]

Channel blockers antagonize GABA-elicited currents in a non-competitive fashion (Dillon et al., 1993, Nagata et al., 1994, Nagata and Narahashi, 1994, Ikeda et al., 2001, Huang et al., 2001) and act as convulsants in vivo. Picrotoxinin, U-93631, TBPS and some insecticides are thought to bind at a single binding site located within the channel pore (Xu et al., 1995, Perret et al., 1999, Dibas and Dillon, 2000, Jursky et al., 2000, Buhr et al., 2001).

The binding site of channel blockers is formed by residues located on TM2 segments of both α and β subunits (Figure 1.4.4.2.). Following residues conferring sensitivity to picrotoxinin and TBPS were identified. Residues α1V257 and α1T261 were found using the cysteine accessibility method (Xu et al., 1995). Residue α1V257 was also labelled using site-directed cysteine probes by Perret et al., (1999). Using α12 chimeric receptors were β2A252 and β2L253 identified residues (Jursky et al., 2000). An additional residue contributing to this binding site β2T246 located on the linker between TM1 and TM2 segments affects the potency of the convulsant compound pentylenetetrazole (Dibas and Dillon, 2000).


Modulation of GABAA Receptor Function via Unidentified Allosteric Sites[edit]

The majority of compounds acting at the GABAA receptor discussed in the previous chapters exert their actions via known recognition sites. However, for a number of compounds which are able to allosterically potentiate actions of GABA or directly act on GABAA receptors structural determinants of their recognition by the receptor have not been identified yet. Neurosteroids is one class of such compounds. They can be divided into two functional groups – uncharged, that can act as positive allosteric modulators (Gee and Lan, 1991, Akk and Steinbach, 2003, Stell et al., 2003) and charged – negative allosteric modulators of receptor function (Zaman et al., 1992, Park-Chung et al., 1999, Akk et al., 2001). Enhancement of submaximal GABAA receptor currents occurs through increases in both channel open frequency and open duration (Puia et al., 1990, Twyman and Macdonald, 1992, Akk and Steinbach, 2003, Bianchi and Macdonald, 2003). Charged neurosteroids inhibit GABA-gated channel openings by enhancing receptor desensitization and stabilizing desensitized states (Zhu and Vicini, 1997, Shen et al., 2000).

Another group are the γ-butyrolactones and related compounds interacting with the GABAA receptor, but not at the benzodiazepine or barbiturate sites (Klunk et al., 1982, Mathews et al., 1996). Displacement studies with [S]TBPS suggested an interaction between the γ-butyrolactones and the picrotoxinin site (Holland et al., 1990a,b,c), however when the picrotoxinin binding site was disrupted by a point mutation potentiation of GABA responses was maintained (Holland et al., 1993, Holland et al., 1995, Williams et al., 1997).

A number of fatty and unsaturated acids were found to modulate GABAA receptor function. Arachidonic, eicosatetraenoicpentayonic and oleic acids were found to inhibit currents elicited by GABA and muscimol in brain preparations and recombinant GABAA receptors in dose-dependent manner (Schwartz et al., 1988, Schwartz and Yu, 1992, Saxena, 2000). Thyroid hormones such as L-triiodothyronine (T3) and L-thyroxine are also reported to interact with GABAA receptors (Chapell et al., 1998), and it has been suggested that the α1-subunit imparts T3 sensitivity (Chapell et al., 1998). The antihelminthic compound ivermectin (Pong and Wang, 1982, Krusek and Zemkova, 1994), the anxiolytic anticonvulsant compounds chlormethiazole and trichloroethanol (Moody and Skolnick, 1989, Hales and Lambert, 1992, Peoples and Weight, 1994), polyamines such as spermine and spermidine (Gilad et al., 1992), and antidepressants such as amoxapine and mianserin (Squires and Saederup, 1988) have been reported to interact with GABAA receptors but the exact site of action of these drugs and their subunit requirements are not known.


Pharmacology Mediated by GABAA Receptors in vivo[edit]

Different isoforms of the GABAA receptor differ in their channel kinetics, affinity for GABA, rate of desensitization, subcellular positioning and pharmacology. In the absence of selective pharmacological tools the in vivo function of defined receptor isoform cannot be investigated. Therefore, alternative approaches were used to address this problem. Specific subunit isoforms were either deleted or mutated to alter its properties. Such a deletion or alternation of a subunit isoform would be expected to affect all receptors containing the corresponding subunit isoform. Knockout of individual GABAA receptor subunits may lead to compensatory upregulation of other subunits. An alternative strategy, which avoids compensatory changes, is the knock-in approach. In this approach, a subunit isoform is mutated such as to alter its pharmacological properties. Transgenic mice were subsequently screened for deficit in the behavioral responses to defined drugs. Thus, allowing conclusions on the in vivo contribution of the GABAA receptors containing defined subunit isoform.

This strategy helped to understand the in vivo pharmacology of GABAA receptors containing α1, α2, α3 and α5 subunits. The point mutation α1H101R (or equivalent position in other subunits), which renders the mutated subunit isoform insensitive to classical benzodiazepines, has separately been introduced into the different subunit isoforms (Rudolph and Mohler, 2004). A similar approach has been applied to β2 and β3 subunit with introduction of β2N265S and β3N265S mutations, which render receptors containing mutated subunit isoforms insensitive to general anesthetics (Reynolds et al., 2003, Jurd et al., 2003).


Pharmacological Properties Mediated by the α Subunits[edit]

GABAA receptors containing the α1 subunit are the most abundant and expressed in all brain areas. Several studies were undertaken to elucidate functions mediated by receptors containing α1 subunit, using both the knock-out and knock-in approaches. Mice with a deleted gene coding for the α1 subunit developed normally and it was found that ablation of this particular subunit was compensated by overexpression of α2 and α3 subunits enough to sustain function of GABAergic inhibitory system (Sur et al., 2001, Kralic et al., 2002a,b, Goldstein et al., 2002). This deletion caused developmental changes (Vicini et al., 2001) and reduced sensitivity of mutant mice to the locomotor-stimulating effects of ethanol (Kralic et al., 2003).

In mice carrying α1 subunit containing receptors in which α1H101R mutation had been introduced diazepam lost its ability to mediate sedation (Rudolph et al., 1999, Crestani et al., 2000a,b, Low et al., 2000, McKernan et al., 2000). Additionally α1-containing receptors were found to mediate the amnestic and anticonvulsant activity of diazepam (Rudolph et al., 1999, Crestani et al., 2000a). Mice carrying the α2H101R point mutation lost the anxiolytic effect of diazepam (Low et al., 2000). This lack of response was specific for ligands of the benzodiazepine site, since α2H101R mice retained the ability to display an anxiolytic-like response to sodium pentobarbital. Thus, the anxiolytic action of diazepam is selectively mediated by the enhancement of GABAergic transmission in a population of neurons expressing the α2 subunit containing GABAA receptors (Low et al., 2000). Additionally α2 subunit containing GABAA receptors were found to mediate the muscle relaxant effect (Crestani et al., 2001). The analysis of mice carrying the α3H126R mutation indicated that the anxiolytic effect of benzodiazepine drugs is not mediated by α3-receptors (Low et al., 2000). However, α3 subunit containing receptors seem to be implicated in muscle relaxant effect of diazepam, but only at high doses (Crestani et al., 2001).

The native α4 subunit containing receptors in the brain are associated with actions of the neurosteroids (Mihalek et al., 1999, Spigelman, 2002, Spigelman, 2003, Stell et al., 2003), implicated in actions of alcohol (Mihalek et al., 2001, Sundstrom-Poromaa et al., 2002, Wallner et al., 2003) and formation of alcohol-dependence (Mahmoudi et al., 1997, Follesa et al., 2003). In steroid-withdrawal models of premenstrual syndrome and postpartum or postmenopausal dysphoria, particularly the increased anxiety and incidence of seizures was also attributed to α4 subunit containing receptors (Smith et al., 1998, Follesa et al., 2000, Gulinello et al., 2001, Hsu and Smith, 2003, Gulinello et al., 2003a,b).

Two transgenic models have been generated to study contribution of α5 subunit. In one the entire subunit has been deleted (Collinson et al., 2002), and in the second the α5H105R point mutation has been introduced (Crestani et al., 2002). Both of these genetically modified mice showed an improved performance in animal models of learning and memory (Collinson et al., 2002, Crestani et al., 2002), suggesting that a selective inhibitor of α5 subunit containing receptors could have use as a cognitive enhancer, for instance in mild cognitive impaired elderly, or Alzheimer’s disease patients.

Studies on knockout mice that lack the α6 subunit reported no change in the response of these mice to pentobarbital, general anesthetics or ethanol, compared with wild-type mice (Homanics et al., 1997a), but the knockout mice were more sensitive to the motor-impairing action of diazepam (although in a limited dose range only) than their wild-type counterparts (Korpi et al., 1999). In addition, a selective post-translational loss of the δ subunit was apparent in cerebellar granule cells, which indicates that the δ subunit is co-assembled with the α6 subunit (Jones et al., 1997). The absence of the α6 subunit triggered various additional changes in the cerebellum, which included a reduction in the affinity of the GABAA receptor for muscimol (Homanics et al., 1997a), an increase in the number of receptors containing the β3 subunit compared with wild-type (Nusser et al., 1999b) and, interestingly, a compensatory upregulation of a K+ channel (TASK-1) in granule cells (Brickley et al., 2001).


Pharmacological Properties Mediated by the β Subunits[edit]

GABAA receptors that contain the β2 and β3 subunits are a prevalent receptor population present in most brain areas (Fritschy and Mohler, 1995). It was observed that expression of β subunits is altered in patients with temporal lobe epilepsy (Brooks-Kayal et al., 1998, Loup et al., 2001, Pirker et al., 2003) and also in various experimental models of epilepsy (Tsunashima et al., 1997, Schwarzer et al., 1997). Deletion of the gene encoding the β3-subunit results in mice that possess only half of the normal density of GABAA receptors in the brain (Krasowski et al., 1998b). Most of these mice die in the neonatal period; however, a few survive and grow to normal body size (Homanics et al., 1997b), although these mice display various neurological impairments including hyperresponsiveness to sensory stimuli (Ugarte et al., 2000), strong motor impairment and epileptic seizures (DeLorey et al., 1998), which might be due to the lack of β3-containing receptors as ‘desynchronizers’ of neuronal activity (Huntsman et al., 1999, Ramadan et al., 2003).

The sedative and anesthetic effects of anesthetics were also found to be mediated via GABAA receptors composed from different β subunit isoforms (Quinlan et al., 1998, Laposky et al., 2001, Wong et al., 2001). Mice carrying β2N265S mutation were lacking the sedative effects produced by etomidate (Reynolds et al., 2003). In another study β3N265S mutation rendered mice insensitive to anesthetics propofol and etomidate, suggesting that it has a key role in mediating the hypnotic and immobilizing responses in vivo. Volatile anesthetics showed only small reduction in their effects and appear to act via a broader spectrum of molecular targets (Jurd et al., 2003).


Pharmacological Properties Mediated by the γ Subunits[edit]

Mice deficient in both the γ2S and γ2L subunits are entirely devoid of a response to benzodiazepines as shown behaviorally and in cultured dorsal root ganglion cells (Gunther et al., 1995). Most homozygous γ2 knockout mice die perinatally. This is due, at least in part, to the requirement of the γ2 subunit for synaptic clustering of GABAA receptors, although not for receptor assembly (Essrich et al., 1998). In animals that survive for up to two weeks, diazepam failed to induce sedation and to impair the righting reflex. This failure reflects the requirement of the γ2 subunit for the formation of the benzodiazepine site of GABAA receptors (Gunther et al., 1995, Sigel, 2002). By contrast, mice heterozygous for the γ2 subunit knockout mutation develop and behave normally. The synaptic clustering of GABAA receptors is only partly reduced (~15–30%, depending on the brain region); the unclustered receptors consist of α and β subunits. When exposed to certain fear-inducing stimuli, these animals show a striking disease phenotype with a high anxiety response to natural and learned aversive stimuli, as well as a cognitive bias for threat cues (Crestani et al., 1999).


Pharmacological Properties Mediated by the δ Subunit[edit]

Disruption of the gene encoding the δ subunit produced mice with an epileptic phenotype (Spigelman et al., 2002), changes in expression of α4 and γ2 subunits in the forebrain (Peng et al., 2002, Korpi et al., 2002) and cerebellar granule cells (Tretter et al., 2001). δ subunit knockout mice displayed an attenuation of the sleep time following the administration of the neurosteroids alphaxalone and pregnenolone and ethanol, whereas the response to propofol, etomidate, ketamine and midazolam was indistinguishable from that observed in wild-type mice (Quinlan et al., 2000). Behavioral responses of δ deficient mice to neurosteroids and ethanol were also greatly altered, suggesting important role of this subunit type in endogenous functional modulation of δ subunit containing GABAA receptors (Mihalek et al., 2001). This behavioral changes may be attributed to reduced sensitivity to neurosteroids in hippocampus (Spigelman et al., 2003), thalamic relay neurons (Porcello et al., 2003) and cerebellum (Vicini et al., 2002).



References[edit]

  • Akk, G., Steinbach, J. H. (2003) Low doses of ethanol and a neuroactive steroid positively interact to modulate rat GABAA receptor function. J Physiol 546: 641-646
  • Akk, G., Bracamontes, J., Steinbach, J. H. (2001) Pregnenolone sulfate block of GABAA receptors: mechanism and involvement of a residue in the M2 region of the alpha subunit. J Physiol 532: 673-684
  • Akk, G., Steinbach, J. H. (2000) Activation and block of recombinant GABAA receptors by pentobarbitone: a single-channel study. Br J Pharmacol 130: 249-258
  • Allman, K., Page, K. M., Curtis, C. A., Hulme, E. C. (2000) Scanning mutagenesis identifies amino acid side chains in transmembrane domain 5 of the M(1) muscarinic receptor that participate in binding the acetyl methyl group of acetylcholine. Mol Pharmacol 58: 175-184
  • Amin, J. (1999) A single hydrophobic residue confers barbiturate sensitivity to gamma-aminobutyric acid type C receptor. Mol Pharmacol 55: 411-423
  • Amin, J., Brooks-Kayal, A., and Weiss, D. S. (1997) Two tyrosine residues on the asubunit are crucial for benzodiazepine binding and allosteric modulation of γ-aminobutyric acidA receptors. Mol Pharmacol 51: 833–841
  • Amin, J., Weiss, D. S. (1993) GABAA receptor needs two homologous domains of the β-subunit for activation by GABA but not by pentobarbital. Nature 366: 565–569
  • Angelotti, T. P., and Macdonald, R. L. (1993) Assembly of GABAA receptor subunits: α1β1 and α1β1γ2S subunits produce unique ion channels with dissimilar single-channel properties. J Neurosci 13: 1429–1440
  • Arakawa, T., Okada, Y. (1988) Excitatory and inhibitory action of GABA on synaptic transmission in slices of guinea pig superior colliculus. Eur J Pharmacol 158: 217-224
  • Backus, K. H., Arigoni, M., Drescher, U., Scheurer, L., Malherbe, P., Mohler, H. and Benson, J. A. (1993) Stoichiometry of a recombinant GABAA-receptor deduced from mutation-induced rectification. Neuroreport 5: 285–288
  • Bali, M., Akabas, M. H. (2004) Defining the Propofol Binding Site Location on the GABAA Receptor. Mol Pharmacol 65: 68-76
  • Barnard, E. A., Skolnick, P., Olsen, R. W., Mohler, H., Sieghart, W., Biggio, G., Braestrup, C., Bateson, A. N., and Langer, S. Z. (1998) International Union of Pharmacology. XV. Subtypes of gamma-aminobutyric acidA Receptors: Classification on the Basis of Subunit Structure and Receptor Function. Pharmacol Rev 50: 291-314
  • Bateson, A. N., Lasham, A. and Darlison, M. G. (1991) Gamma-aminobutyric acid-A receptor heterogeneity is increased by alternative splicing of a novel beta-subunit gene transcript. J Neurochem 56: 1437–1440
  • Baumann, S. W., Baur, R., Sigel, E. (2003) Individual Properties of the Two Functional Agonist Sites in GABAA Receptors. J Neurosci 23: 11158-11166
  • Baumann, S. W., Baur, R., Sigel, E. (2002) Forced subunit assembly in alpha1beta2gamma2 GABAA receptors. Insight into the absolute arrangement. J Biol Chem 277: 46020-46025
  • Baumann, S. W., Baur, R., Sigel, E. (2001) Subunit arrangement of gamma-aminobutyric acid type A receptors. J Biol Chem 276: 36275-36280.
  • Bein, H. J., (1972) Pharmacological differentiations of muscle relaxants, in Spasticity: A Topical Survey (Birkmayer W. ed) pp 76–89, Hans Huber, Vienna
  • Bell-Horner, C. L., Dibas, M., Huang, R. Q., Drewe, J. A., Dillon, G. H. (2000) Influence of subunit configuration on the interaction of picrotoxin-site ligands with recombinant GABAA receptors. Brain Res Mol Brain Res 76: 47-55
  • Belelli, I., Pistis, I., Peter, J. A., and Lambert, J. J. (1999) General anaesthetic action at transmitter-gated inhibitory amino acid receptors. Trends Pharmacol Sci 20: 496-502
  • Belelli, D., Lambert, J. J., Peters, J. A., Wafford, K., and Whiting, P. J. (1997) The interaction of the general anesthetic etomidate with the γ-aminobutyric acid type A receptor is influenced by a single amino acid. Proc Natl Acad Sci USA 94: 11031–11036
  • Ben-Ari, Y., Khazipov, R., Leinekugel, X., Caillard, O., and Galarsa, J.-L. (1997) GABAA, NMDA and AMPA receptors: A developmentally regulated “menage a trois”. Trends Neurosci 20: 523–529
  • Bencsits, E., Ebert, V., Tretter, V., Sieghart, W. (1999) A significant part of native gamma-aminobutyric AcidA receptors containing alpha4 subunits do not contain gamma or delta subunits. J Biol Chem 274: 19613-19616
  • Benke, D., Michel, C., Mohler, H. (1997) GABAA receptors containing the alpha4-subunit: prevalence, distribution, pharmacology, and subunit architecture in situ. J Neurochem 69: 806-814
  • Benke, D., Honer, M., Michel, C., Mohler, H. (1996) GABAA receptor subtypes differentiated by their gamma-subunit variants: prevalence, pharmacology and subunit architecture. Neuropharmacology 35: 1413-1423
  • Benke, D., Fritschy, J. M., Trzeciak, A., Bannerworth, W. and Mohler, H. (1994) Distribution, prevalence and drug-binding profile of GABAA-receptor subtypes differing in β-subunit isoform. J Biol Chem 269: 27100–27107
  • Benke, D., Mertens, S., Trzeciak, A., Gillessen, D., Mohler, H. (1991a) Identification and immunohistochemical mapping of GABAA receptor subtypes containing the delta-subunit in rat brain. FEBS Lett 283: 145-149
  • Benke, D., Mertens, S., Trzeciak, A., Gillessen, D., Mohler, H. (1991b) GABAA receptors display association of gamma 2-subunit with alpha 1- and beta 2/3-subunits. J Biol Chem 266: 4478-4483
  • Benke, D., Cicin-Sain, A., Mertens, S., Mohler, H. (1991c) Immunochemical identification of the alpha 1- and alpha 3-subunits of the GABAA-receptor in rat brain. J Recept Res 11: 407-424
  • Bera, A. K., Chatav, M., Akabas, M. H. (2002) GABAA receptor M2-M3 loop secondary structure and changes in accessibility during channel gating. J Biol Chem 277: 43002-43010
  • Bertocci, B., Miggiano, V., Da Prada, M., Dembic, Z., Lahm, H. W., Malherbe, P. (1991) Human catechol-O-methyltransferase: cloning and expression of the membrane-associated form. Proc Natl Acad Sci USA 88: 1416-1420
  • Bianchi, M. T., Macdonald, R. L. (2003) Neurosteroids shift partial agonist activation of GABAA receptor channels from low- to high-efficacy gating patterns. J Neurosci 23: 10934-10943
  • Bianchi, M. T., Macdonald, R. L. (2001) Agonist Trapping by GABAA Receptor Channels. J Neurosci 21: 9083-9091
  • Blair, L. A., Levitan, E. S., Marshall, J., Dionne, V. E., Barnard, E. A. (1988) Single subunits of the GABAA receptor form ion channels with properties of the native receptor. Science 242: 577-579.
  • Boue-Grabot, E., Roudbaraki, M., Bascles, L., Tramu, G., Bloch, B., Garret, M. (1998) Expression of GABA receptor rho subunits in rat brain. J Neurochem 70: 899-907
  • Bouchet, M. J., Jacques, P., Ilien, B., Goeldner, M., Hirth, C. (1992) m-Sulfonate benzene diazonium chloride: a powerful affinity label for the gamma-aminobutyric acid binding site from rat brain. J Neurochem 59: 1405-1413
  • Boileau, A. J., Baur, R,, Sharkey, L, M,, Sigel, E,, Czajkowski, C. (2002) The relative amount of cRNA coding for gamma2 subunits affects stimulation by benzodiazepines in GABAA receptors expressed in Xenopus oocytes. Neuropharmacology 43: 695-700
  • Boileau, A. J., Newell, J. G., Czajkowski, C. (2002) GABAA receptor beta2 Tyr97 and Leu99 line the GABA-binding site. Insights into mechanisms of agonist and antagonist actions. J Biol Chem 277: 2931-2937
  • Boileau, A. J., Czajkowski, C. (1999a) Identification of transduction elements for benzodiazepine modulation of the GABAA receptor: three residues are required for allosteric coupling. J Neurosci 19: 10213-10220
  • Boileau, A. J., Evers, A. R., Davis, A. F., Czajkowski, C. (1999b) Mapping the agonist binding site of the GABAA receptor: evidence for a beta-strand. J Neurosci 19: 4847-4854
  • Bollan, K., Robertson, L. A., Tang, H., Connolly, C. N. (2003a) Multiple assembly signals in gamma-aminobutyric acid (type A) receptor subunits combine to drive receptor construction and composition. Biochem Soc Trans 31: 875-879
  • Bollan, K., King, D., Robertson, L. A., Brown, K., Taylor, P. M., Moss, S. J., Connolly, C. N. (2003b) GABAA receptor composition is determined by distinct assembly signals within alpha and beta subunits. J Biol Chem 278: 4747-4755
  • Bonnert, T. P., McKernan, R. M., Farrar, S., le Bourdelles, B., Heavens, R. P., Smith, D. W., Hewson, L., Rigby, M. R., Sirinathsinghji, D. J., Brown, N., Wafford, K. A., Whiting, P. J. (1999) Theta, a novel gamma-aminobutyric acid type A receptor subunit. Proc Natl Acad Sci USA 96: 9891-9896
  • Borea, P. A., Gilli, G., Bertolasi, V., and Ferretti, V. (1987) Stereochemical features controlling binding and intrinsic activity properties of benzodiazepine-receptor ligands. Mol Pharmacol 31: 334–344
  • Bormann, J., and Feigenspan, A. (1995) GABAC receptors. Trends Neurosci 18: 515–519
  • Bowery, N. G., Bettler, B., Froestl, W., Gallagher, J. P., Marshall, F., Raiteri, M., Bonner, T. I., and Enna, S. J. (2002) International Union of Pharmacology. XXXIII. Mammalian γ-Aminobutyric AcidB Receptors: Structure and Function. Pharmacol Rev 54: 247-264
  • Bowery, N. G., Enna, S. J. (2000) gamma-aminobutyric acid(B) receptors: first of the functional metabotropic heterodimers. J Pharmacol Exp Ther 292: 2-7
  • Bowery, N. G., Hill, D. R., Hudson, A. L., Doble A., Middlemiss, D. N., Shaw J., and Turnbull, M. (1980) (β)-Baclofen decreases neurotransmitter release in the mammalian CNS by an action at a novel GABA receptor. Nature 283: 92–94
  • Brejc, K., van Dijk, W. J., Klaassen, R. V., Schuurmans, M., van Der Oost, J., Smit, A. B., Sixma, T. K. (2001) Crystal structure of an ACh-binding protein reveals the ligand-binding domain of nicotinic receptors. Nature 411: 269-276
  • Brickley, S. G., Revilla, V., Cull-Candy, S. G., Wisden, W., Farrant, M. (2001) Adaptive regulation of neuronal excitability by a voltage-independent potassium conductance. Nature 409: 88-92
  • Brooks-Kayal, A. R., Shumate, M. D., Jin, H., Rikhter, T. Y., Coulter, D. A. (1998) Selective changes in single cell GABAA receptor subunit expression and function in temporal lobe epilepsy. Nat Med 4: 1166-1172, erratum: Nat Med (1999) 5: 590
  • Buhr, A., Wagner, C., Fuchs, K., Sieghart, W., Sigel, E. (2001) Two novel residues in M2 of the gamma-aminobutyric acid type A receptor affecting gating by GABA and picrotoxin affinity. J Biol Chem 276: 7775-7781
  • Buhr, A., Sigel, E. (1997) A point mutation in the gamma2 subunit of gamma-aminobutyric acid type A receptors results in altered benzodiazepine binding site specificity. Proc Natl Acad Sci 94: 8824-8829
  • Buhr, A., Baur, R., Sigel, E. (1997a) Subtle changes in residue 77 of the gamma subunit of alpha1beta2gamma2 GABAA receptors drastically alter the affinity for ligands of the benzodiazepine binding site. J Biol Chem 272: 11799-11804
  • Buhr, A., Schaerer, M. T., Baur, R., Sigel, E. (1997b) Residues at positions 206 and 209 of the alpha1 subunit of gamma-aminobutyric AcidA receptors influence affinities for benzodiazepine binding site ligands. Mol Pharmacol 52: 676-682
  • Buller, A. L., Hastings, G. A., Kirkness, E. F., Fraser, C. M. (1994) Site-directed mutagenesis of N-linked glycosylation sites on the gamma-aminobutyric acid type A receptor alpha 1 subunit. Mol Pharmacol 46: 858-865
  • Burt, D. R., and Kamatchi, G. L. (1991) GABAA receptor subtypes: From pharmacology to molecular biology. FASEB J 5: 2916–2923
  • Carlson, B. X., Engblom, A. C., Kristiansen, U., Schousboe, A., Olsen, R. W. (2000) A single glycine residue at the entrance to the first membrane-spanning domain of the gamma-aminobutyric acid type A receptor beta(2) subunit affects allosteric sensitivity to GABA and anesthetics. Mol Pharmacol 57: 474-484
  • Casalotti, S. O., Stephenson, A., Barnard, E. A. (1986) Separate subunits for agonist and benzodiazepine binding in the γ-aminobutyric acidA receptor oligomer. J Biol Chem 261: 15013–15016
  • Casula, M. A., Bromidge, F. A., Pillai, G. V., Wingrove, P. B., Martin, K., Maubach, K., Seabrook, G. R., Whiting, P. J., Hadingham, K. L. (2001) Identification of amino acid residues responsible for the alpha5 subunit binding selectivity of L-655,708, a benzodiazepine binding site ligand at the GABAA receptor. J Neurochem 77: 445-451
  • Cestari, I. N., Min, K. T., Kulli, J. C., Yang, J. (2000) Identification of an amino acid defining the distinct properties of murine beta1 and beta3 subunit-containing GABAA receptors. J Neurochem 74: 827-838
  • Chang, C. S., Olcese, R., Olsen, R. W. (2003) A single M1 residue in the beta2 subunit alters channel gating of GABAA receptor in anesthetic modulation and direct activation. J Biol Chem 278: 42821-42828
  • Chang, Y., Wang, R., Barot, S., and Weiss, D. S. (1996) Stoichiometry of a recombinant GABAA receptor. J Neurosci 16: 5415–5424
  • Chapell, R., Martin, J., Machu, T. K., Leidenheimer, N. J. (1998) Direct channel-gating and modulatory effects of triiodothyronine on recombinant GABAA receptors. Eur J Pharmacol 349: 115–121
  • Chen, C., Okayama, H. (1987) High-efficiency transformation of mammalian cells by plasmid DNA. Mol Cell Biol 7: 2745-2752
  • Cheng, Y., and Prusoff, W. H. (1973) Relationship between the inhibition constant (Ki) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol 22: 3099–3108
  • Cherubini, E., Martina, M., Sciancalepore, M., Strata, F. (1998) GABA excites immature CA3 pyramidal cells through bicuculline-sensitive and -insensitive chloride-dependent receptors. Perspect Dev Neurobiol 5: 289-304
  • Cherubini, E., Giairsa, J. L., and Ben-Ari, Y. (1991) GABA, an excitatory transmitter in early postnatal life. Trends Neurosci 14: 515–519
  • Clark, S. E., Garret, M., Platt, B. (2001) Postnatal alterations of GABA receptor profiles in the rat superior colliculus. Neuroscience 104: 441-454
  • Collinson, N., Kuenzi, F. M., Jarolimek, W., Maubach, K. A., Cothliff, R., Sur, C., Smith, A., Out, F. M., Howell, O., Atack, J. R., McKernan, R. M., Seabrook, G. R., Dawson, G. R., Whiting, P. J., Rosahl, T. W. (2002) Enhanced learning and memory and altered GABAergic synaptic transmission in mice lacking the alpha 5 subunit of the GABAA receptor. J Neurosci 22: 5572-5580
  • Connolly, C. N., Uren, J. M., Thomas, P., Gorrie, G. H., Gibson, A., Smart, T. G., and Moss, S. J. (1999a) Subcellular Localization and Endocytosis of Homomeric γ2 Subunit Splice Variants of γ-Aminobutyric Acid Type A Receptors. Mol Cel Neurosci 13: 259–271
  • Connolly, C. N., Kittler, J. T., Thomas, P., Uren, J. M., Brandon, N. J., Smart, T. G., Moss, S. J. (1999b) Cell surface stability of gamma-aminobutyric acid type A receptors. Dependence on protein kinase C activity and subunit composition. J Biol Chem 274: 36565-36572
  • Connolly, C. N., Wooltorton, J. R., Smart, T. G., Moss, S. J. (1996a) Subcellular localization of gamma-aminobutyric acid type A receptors is determined by receptor beta subunits. Proc Natl Acad Sci USA 93: 9899-9904
  • Connolly, C. N., Krishek, B. J., McDonald, B. J., Smart, T. G., Moss, S. J. (1996b) Assembly and cell surface expression of heteromeric and homomeric gamma-aminobutyric acid type A receptors. J Biol Chem 271: 89-96
  • Corringer, P. J., Le Novere, N., Changeux, J. P. (2000) Nicotinic receptors at the amino acid level. Annu Rev Pharmacol Toxicol 40: 431-458
  • Crestani, F., Keist, R., Fritschy, J. M., Benke, D., Vogt, K., Prut, L., Bluthmann, H., Mohler, H., Rudolph, U. (2002) Trace fear conditioning involves hippocampal alpha5 GABAA receptors. Proc Natl Acad Sci USA 99: 8980-8985
  • Crestani, F., Low, K., Keist, R., Mandelli, M., Mohler, H., Rudolph, U. (2001) Molecular targets for the myorelaxant action of diazepam. Mol Pharmacol 59: 442-445
  • Crestani, F., Martin, J. R., Mohler, H., and Rudolph, U. (2000a) Mechanism of action of the hypnotic zolpidem in vivo. Br J Pharmacol 131: 1251–1256
  • Crestani, F., Martin, J. R., Mohler, H., and Rudolph, U. (2000b) Resolving differences in GABAA receptor mutant mouse studies. Nature Neurosci 3: 1059
  • Crestani, F., Lorez, M., Baer, K., Essrich, C., Benke, D., Laurent, J. P., Belzung, C., Fritschy, J M., Luscher, B., Mohler, H. (1999) Decreased GABAA-receptor clustering results in enhanced anxiety and a bias for threat cues. Nat Neurosci 2: 833-839
  • Cromer, B. A., Morton, C. J., Parker, M. W. (2002) Anxiety over GABAA receptor structure relieved by AChBP. Trends Biochem Sci 27: 280–287
  • Cutting, G. R., Lu, L., O’Hara, B. F., Kasch, L. M., Montrose-Rafizadeh, C., Donovan, D. M., Shimada, S., Antonarakis, S. E., Guggino, W. B., Uhl, G. R., and Kazazian, H. H. Jr. (1991) Cloning of the γ-aminobutyric acid (GABA) rho1 cDNA: A GABA receptor subunit highly expressed in the retina. Proc Natl Acad Sci USA 88: 2673–2677
  • Czajkowski, C., Farb, D. H. (1989) Identification of an intracellular pool of gamma-aminobutyric acidA/benzodiazepine receptors en route to the cell surface of brain neurons in culture. Mol Pharmacol 35: 183-188
  • Dalziel, J. E., Cox, G. B., Gage, P. W., Birnir, B. (1999) Mutant human alpha(1)beta(1)(T262Q) GABAA receptors are directly activated but not modulated by pentobarbital. Eur J Pharmacol 385: 283-286
  • Davies, M., Newell, J. G., Dunn, S. M. (2001) Mutagenesis of the GABAA receptor alpha1 subunit reveals a domain that affects sensitivity to GABA and benzodiazepine-site ligands. J Neurochem 79: 55-62
  • Davies, M., Dunn, S. M. (1998) Identification of a unique domain in bovine brain GABAA receptors that is photoaffinity labelled by [3H]Ro15-4513. Biochem Biophys Res Commun 246: 650-653
  • Davies, M., Bateson, A. N., Dunn, S. M. (1998b) Structural requirements for ligand interactions at the benzodiazepine recognition site of the GABAA receptor. J Neurochem 70: 2188-2194
  • Davies, M., Martin, I. L., Bateson, A. N., Hadingham, K. L., Whiting, P. J., Dunn, S. M. (1996) Identification of domains in human recombinant GABAA receptors that are photoaffinity labelled by [3H]flunitrazepam and [3H]Ro15-4513. Neuropharmacology 35: 1199-1208
  • Delaney, A. J., Sah, P. (1999) GABA receptors inhibited by benzodiazepines mediate fast inhibitory transmission in the central amygdala. J Neurosci 19: 9698-9704
  • DeLorey, T. M., Handforth, A., Anagnostaras, S. G., Homanics, G. E., Minassian, B. A., Asatourian, A., Fanselow, M. S., Delgado-Escueta, A., Ellison, G. D., Olsen, R. W. (1998) Mice lacking the beta3 subunit of the GABAA receptor have the epilepsy phenotype and many of the behavioral characteristics of Angelman syndrome. J Neurosci 18: 8505-8514
  • Deng, L., Ransom, R. W., Olsen, R. W. (1986) [3H]Muscimol photolabels the γ-aminobutyric acid receptor binding site on a peptide subunit distinct from that labelled with benzodiazepine. Biochem Biophys Res Commun 138: 1308–1314
  • Dibas, M. I., Dillon, G. H. (2000) The central nervous system convulsant pentylenetetrazole stimulates gamma-aminobutyric acid (GABA)-activated current in picrotoxin-resistant GABAA receptors in HEK293 cells. Neurosci Lett 285: 193-196
  • Derry, J. M., Dunn, S. M., Davies, M. (2004) Identification of a residue in the gamma-aminobutyric acid type A receptor alpha subunit that differentially affects diazepam-sensitive and -insensitive benzodiazepine site binding. J Neurochem 88: 1431-1438
  • Didelon, F., Sciancalepore, M., Savic', N., Mladinic', M., Bradbury, A., Cherubini, E. (2002) gamma-Aminobutyric acidA rho receptor subunits in the developing rat hippocampus. J Neurosci Res 67: 739-744
  • Didelon, F., Mladinic', M., Cherubini, E., Bradbury, A. (2000) Early expression of GABAA receptor delta subunit in the neonatal rat hippocampus. J Neurosci Res 62: 638-643
  • Dillon, G. H., Im, H. K., Hamilton, B. J., Carter, D. B., Gammill, R. B., Judge, T. M., Im, W. B. (1993) U-93631 causes rapid decay of gamma-aminobutyric acid-induced chloride currents in recombinant rat gamma-aminobutyric acid type A receptors. Mol Pharmacol 44: 860-865
  • Drew, C. A., Johnston, G. A. (1992) Bicuculline- and baclofen-insensitive gamma-aminobutyric acid binding to rat cerebellar membranes. J Neurochem 58: 1087-1092
  • Drew, C. A., Johnston, G. A. R., and Weatherby, R. P. (1984) Bicuculline-insensitive GABA receptors: Studies on the binding of (β)-baclofen to rat cerebellar membranes. Neurosci Lett 52: 317–321
  • Duncalfe, L. L., Carpenter, M. R., Smillie, L. B., Martin, I. L., Dunn, S. M. (1996) The major site of photoaffinity labeling of the gamma-aminobutyric acid type A receptor by [3H]flunitrazepam is histidine 102 of the alpha subunit. J Biol Chem 271: 9209-9214

Duncalfe, L. L., Dunn, S. M. (1996) Mapping of GABAA receptor sites that are photoaffinity-labelled by [3H]flunitrazepam and [3H]Ro 15-4513. Eur J Pharmacol 298: 313-319

  • Dunn, S. M., Davies, M., Muntoni, A. L., Lambert, J. J. (1999) Mutagenesis of the rat alpha1 subunit of the gamma-aminobutyric acidA receptor reveals the importance of residue 101 in determining the allosteric effects of benzodiazepine site ligands. Mol Pharmacol 56: 768-774
  • Ehya, N., Sarto, I., Wabnegger, L., Sieghart, W. (2003) Identification of an amino acid sequence within GABAA receptor beta3 subunits that is important for receptor assembly. J Neurochem 84: 127-135
  • Engblom, A. C., Carlson, B. X., Olsen, R. W., Schousboe, A., Kristiansen, U. (2002) Point mutation in the first transmembrane region of the beta 2 subunit of the gamma-aminobutyric acid type A receptor alters desensitization kinetics of gamma-aminobutyric acid and anesthetic-induced channel gating. J Biol Chem 277: 17438-17447
  • Enna, S. J. (2000) GABAB receptor signaling pathways, in Pharmacology of GABA and Glycine Neurotransmission (Mohler H. ed) pp 329–342, Springer-Verlag, Berlin
  • Enna, S. J. (1997) GABAB receptor agonists and antagonists: pharmacological properties and therapeutic possibilities. Exp Opin Invest Drugs 6: 1319–1325
  • Enna, S. J. and Bowery, N. G. (1997) in The GABA Receptors, 2nd ed, Humana Press, Totowa, NJ
  • Enz, R., and Cutting, G. R. (1998) Molecular composition of GABAC receptors. Vision Res 38: 1431–1441
  • Enz, R., Brandstatter, J. H., Hartveit, E., Wassle, H., and Bormann, J. (1995) Expression of GABA receptor ρ1 and ρ2 subunits in the retina and brain of the rat. Eur J Neurosci 7: 1495–1501
  • Ernst, M., Brauchart, D., Boresch, S., Sieghart, W. (2003) Comparative modeling of GABAA receptors: limits, insights, future developments. Neuroscience 119: 933-943
  • Essrich, C., Lorez, M., Benson, J. A., Fritschy, J. M., Luscher, B. (1998) Postsynaptic clustering of major GABAA receptor subtypes requires the gamma 2 subunit and gephyrin. Nat Neurosci 1: 563-571

Farrar, S. J., Whiting, P. J., Bonnert, T. P., and McKernan, R. M. (1999) Stoichiometry of a ligand-gated ion channel determined by fluorescence energy transfer. J Biol Chem 274: 10100–10104

  • Faure-Halley, C., Graham, D., Arbilla, S., and Langer, S. Z. (1993) Expression and properties of recombinant α1β2γ2 and α5β2γ2 forms of the rat GABAA receptor. Eur J Pharmacol 246: 283–287
  • Feigenspan, A., Wassle, H., and Bormann, J. (1993) Pharmacology of the GABA receptor C1─ channel in rat retina bipolar cells. Nature 361: 159–162
  • Fisher, J. L. (2002) Amiloride inhibition of gamma-aminobutyric acidA receptors depends upon the alpha subunit subtype. Mol Pharmacol 61: 1322-1328
  • Fisher, J. L., Macdonald, R. L. (1997) Functional properties of recombinant GABAA receptors composed of single or multiple beta subunit subtypes. Neuropharmacology 36: 1601-1610
  • Fletcher, E. L., Koulen, P., Wassle, H. (1998) GABAA and GABAC receptors on mammalian rod bipolar cells. J Comp Neurol 396: 351-365
  • Foucaud, B., Laube, B., Schemm, R., Kreimeyer, A., Goeldner, M., Betz, H. (2003) Structural model of the N-methyl-D-aspartate receptor glycine site probed by site-directed chemical coupling. J Biol Chem 278: 24011-24017
  • Foucaud, B., Perret, P., Grutter, T., Goeldner, M. (2001) Cysteine mutants as chemical sensors for ligand-receptor interactions. Trends Pharmacol Sci 22: 170-173
  • Follesa, P., Mancuso, L., Biggio, F., Mostallino, M. C., Manca, A., Mascia, M. P., Busonero, F., Talani, G., Sanna, E., Biggio, G. (2003) Gamma-hydroxybutyric acid and diazepam antagonize a rapid increase in GABAA receptors alpha(4) subunit mRNA abundance induced by ethanol withdrawal in cerebellar granule cells. Mol Pharmacol 63: 896-907
  • Follesa, P., Serra, M., Cagetti, E., Pisu, M. G., Porta, S., Floris, S., Massa, F., Sanna, E., Biggio, G. (2000) Allopregnanolone synthesis in cerebellar granule cells: roles in regulation of GABAA receptor expression and function during progesterone treatment and withdrawal. Mol Pharmacol 57:1262-1270
  • Fritschy, J. M., Weinmann, O., Wenzel, A., Benke, D. (1998) Synapse-specific localization of NMDA and GABAA receptor subunits revealed by antigen-retrieval immunohistochemistry. J Comp Neurol 390: 194-210
  • Fritschy, J. M., Mohler, H. (1995) GABAA-receptor heterogeneity in the adult rat brain: differential regional and cellular distribution of seven major subunits. J Comp Neurol 359: 154-194
  • Fritschy, J. M., Benke, D., Mertens, S., Oertel, W. H., Bachi, T., and Mohler, H. (1992) Five subtypes of type A γ-aminobutyric acid receptors identified in neurons by double and triple immunofluorescence staining with subunit-specific antibodies. Proc Nat

Acad Sci USA 89: 6726–6730

  • Fuchs, K., Mohler, H., Sieghart, W. (1988) Various proteins from rat brain, specifically and irreversibly labeled by [3H]flunitrazepam, are distinct alpha-subunits of the GABA-benzodiazepine receptor complex. Neurosci Lett 90: 314-319
  • Fruchart-Gaillard, C., Gilquin, B., Antil-Delbeke, S., Le Novere, N., Tamiya, T., Corringer, P. J., Changeux, J. P., Menez, A., Servent, D. (2002) Experimentally based model of a complex between a snake toxin and the alpha 7 nicotinic receptor. Proc Natl Acad Sci USA 99: 3216–3221
  • Gee, K. W., Lan, N. C. (1991) γ-Aminobutyric acidA receptor complexes in rat cortex and spinal cord show differential responses to steroid modulation. Mol Pharmacol 40: 995–999
  • Gilad, G. M., Gilad, V. H., Wyatt, R. J. (1992) Polyamines modulate the binding of GABAA-benzodiazepine receptor ligands in membranes from the rat forebrain. Neuropharmacology 31: 895–898
  • Glencorse, T. A., Bateson, A. N., and Darlison, M. G. (1992) Differential localization of two alternatively spliced GABAA receptor γ2-subunit mRNAs in the chick brain. Eur J Neurosci 4: 271–277
  • Goldstein, P. A., Elsen, F. P., Ying, S. W., Ferguson, C., Homanics, G. E., Harrison, N. L. (2002) Prolongation of hippocampal miniature inhibitory postsynaptic currents in mice lacking the GABAA receptor alpha1 subunit. J Neurophysiol 88: 3208-3217
  • Goren, E. N., Reeves, D. C., Akabas, M. H. (2004) Loose protein packing around the extracellular half of the GABAA receptor beta 1 subunit M2 channel-lining segment. J Biol Chem 279: 11198-11205
  • Gulinello, M., Orman, R., Smith, S. S. (2003a) Sex differences in anxiety, sensorimotor gating and expression of the alpha4 subunit of the GABAA receptor in the amygdala after progesterone withdrawal. Eur J Neurosci 17: 641-648
  • Gulinello, M., Gong, Q. H., Smith, S. S. (2003b) Progesterone withdrawal increases the anxiolytic actions of gaboxadol: role of alpha4betadelta GABAA receptors. Neuroreport 14: 43-46
  • Gulinello, M., Gong, Q. H., Li, X., Smith, S. S. (2001) Short-term exposure to a neuroactive steroid increases alpha4 GABAA receptor subunit levels in association with increased anxiety in the female rat. Brain Res 910: 55-66
  • Gunther, U., Benson, J. A., Benke, D., Fritschy, J.-M., Reyes, G., Knoflach, F., Crestani, F., Aguzzi, A., Arigoni, M., Lang, Y., Bluethmann, H., Mohler, H., and Luscher, B. (1995) Benzodiazepine-insensitive mice generated by targeted disruption of the γ2 subunit gene of γ-aminobutyric acid type A receptors. Proc Natl Acad Sci USA 92: 7749–7753
  • Gutierrez, A., Khan, Z. U., De Blas, A. L. (1994) Immunocytochemical localization of gamma2 short and gamma2 long subunits of the GABAA receptor in the rat brain. J Neurosci 4: 7168-7179
  • Hadingham, K. L., Garrett, E. M., Wafford, K. A., Bain, C., Heavens, R. P., Sirinathsinghji, D. S., and Whiting, P. J. (1996) Cloning of cDNA encoding the human γ-aminobutyric acidA receptor α6 subunit and characterization of the pharmacology of α6-containing receptors. Mol Pharmacol 49: 253–259
  • Hadingham, K. L., Garrett, E. M., Wafford, K. A., Bain, C., Heavens, R. P., Sirinathsinghji, D. J., Whiting, P. J. (1996) Cloning of cDNAs encoding the human γ-aminobutyric acid type A receptor α6 subunit and characterization of the pharmacology of α6-containing receptors. Mol Pharmacol 49: 253–259
  • Hadingham, K. L., Wafford, K. A., Thompson, S. A., Palmer, K. J., and Whiting, P. J. (1995) Expression and pharmacology of human GABAA receptors containing γ3 subunits. Eur J Pharmacol 291: 301–309
  • Hales, T. G., Lambert, J. J. (1992) Modulation of GABAA and glycine receptors by chlormethiozole. Eur J Pharmacol 210: 239–246
  • Hamon, A., Morel, A., Hue, B., Verleye, M., Gillardin, J. M. (2003) The modulatory effects of the anxiolytic etifoxine on GABAA receptors are mediated by the beta subunit. Neuropharmacology 45: 293-303
  • Hansen, S. L., Ebert, B., Fjalland, B., Kristiansen, U. (2001) Effects of GABAA receptor partial agonists in primary cultures of cerebellar granule neurons and cerebral cortical neurons reflect different receptor subunit compositions. Br J Pharmacol 133: 539-549
  • Hartvig, L., Lukensmejer, B., Liljefors, T., Dekermendjian, K. (2000) Two conserved arginines in the extracellular N-terminal domain of the GABAA receptor α5 subunit are crucial for receptor function. J Neurochem 75: 1746–1753
  • Harvey, R. J., Chinchetru, M. A., and Darlison, M. G. D. (1994) Alternative splicing of a 51-nucleotide exon that encodes a putative protein kinase C phosphorylation site generates two forms of the chicken γ-aminobutyric acidA receptor β2 subunit. J Neurochem 62: 10–16
  • Harvey, R. J., Kim, H. C., and Darlison, M. G. (1993) Molecular cloning reveals the existence of a fourth gamma subunit of the vertebrate brain GABAA receptor. FEBS Lett 331: 211–216
  • He, X., Huang, Q., Ma, C., Yu, S., McKernan, R., Cook, J. M. (2000) Pharmacophore/receptor models for GABAA/BzR alpha2beta3gamma2, alpha3beta3gamma2 and alpha4beta3gamma2 recombinant subtypes. Included volume analysis and comparison to alpha1beta3gamma2, alpha5beta3gamma2, and alpha6beta3gamma2 subtypes. Drug Des Discov 17: 131-171

Hedblom, E., and Kirkness, E. F. (1997) A novel class of GABAA receptor subunit in tissues of the reproductive system. J Biol Chem 272: 15346–15350

  • Herb, A., Wisden, W., Luddens, H., Puia, G., Vicini, S., and Seeburg, P. H. (1992) The third γ-subunit of the γ-aminobutyric acid type A receptor family. Proc Natl Acad Sci USA 89: 1433–1437
  • Hevers, W., Korpi, E. R., Luddens, H. (2000) Assembly of functional alpha6beta3gamma2delta GABAA receptors in vitro. Neuroreport 11: 4103-4106
  • Hervers, W., Luddens, H. (1998) The diversity of GABAA receptors. Mol Neurobiol 18: 35-86
  • Hill, D. R. and Bowery, N. G. (1981) [3H]-baclofen and [3H]-GABA bind to bicuculline insensitive GABAB sites in rat brain. Nature 290: 149–152
  • Holden, J. H., Czajkowski, C. (2002) Different residues in the GABAA receptor alpha1T60-alpha1K70 region mediate GABA and SR-95531 actions. J Biol Chem 277: 18785-18792
  • Holland, K. D., Mathews, G. C., Bolos-Sy, A. M., Tucker, J. B., Reddy, P. A., Covey, D. F., Ferrendelli, J. A., and Rothman, S. M. (1995) Dual modulation of the γ-aminobutyric acid type A receptor/ionophore by alkyl-substituted γ-butyrolactones. Mol Pharmacol 47: 1217–1223
  • Holland, K. D., Bouley, M. G., Covey, D. F., and Ferrendelli, J. A. (1993) Alkylsubstituted γ-butyrolactones act at a distinct site allosterically linked to the TBPS/picrotoxinin site on the GABAA receptor complex. Brain Res 615:170–174
  • Holland, K. D., Ferrendelli. J. A., Covey. D. F.. and Rothman S. M. (1990a) Physiological regulation of the picrotoxin receptor by γ-butyrolactones and γ-thiobutyrolactones in cultured hippocampal neurons. J Neurosci 10: 1719-1727
  • Holland, K. D., McKeon, A. C., Covey, D. F., and Ferrendelli, J. A. (1990b) Binding interactions of convulsant and anticonvulsant γ-butyrolactones and γ-thiobutyrolactones with the picrotoxin receptor. J Pharmac Exp Ther 254: 578-583
  • Holland, K. D., Yoon, K.-W., Ferrendelli, J. A., Covey, D. F., and Rothman S. M. (1990c) γ-Butyrolactone antagonism of the picrotoxin receptor: comparison of a pure antagonist and a mixed antagonist/inverse agonist. Mol Pharmacol 39: 79-84
  • Homanics, G. E., Harrison, N. L., Quinlan, J. J., Krasowski, M. D., Rick, C. E., de Blas, A. L., Mehta, A. K., Kist, F., Mihalek, R. M., Aul, J. J., Firestone, L. L. (1999) Normal electrophysiological and behavioral responses to ethanol in mice lacking the long splice variant of the gamma2 subunit of the gamma-aminobutyrate type A receptor. Neuropharmacology 38: 253-265
  • Homanics, G. E., Ferguson, C., Quinlan, J. J., Daggett, J., Snyder, K., Lagenaur, C., Mi, Z. P., Wang, X. H., Grayson, D. R., Firestone, L. L. (1997a) Gene knockout of the alpha6 subunit of the gamma-aminobutyric acid type A receptor: lack of effect on responses to ethanol, pentobarbital, and general anesthetics. Mol Pharmacol 51: 588-596
  • Homanics, G. E., DeLorey, T. M., Firestone, L. L., Quinlan, J. J., Handforth, A., Harrison, N. L., Krasowski, M. D., Rick, C. E., Korpi, E. R., Makela, R., Brilliant, M. H., Hagiwara, N., Ferguson, C., Snyder, K., Olsen, R.W. (1997b) Mice devoid of gamma-aminobutyrate type A receptor beta3 subunit have epilepsy, cleft palate, and hypersensitive behavior. Proc Natl Acad Sci USA 94: 4143-4148
  • Hosie, A. M., Dunne, E. L., Harvey, R. J., Smart, T. G. (2003) Zinc-mediated inhibition of GABAA receptors: discrete binding sites underlie subtype specificity. Nat Neurosci 6: 362-369
  • Horenstein, J., Wagner, D. A., Czajkowski, C., Akabas, M. H. (2001) Protein mobility and GABA-induced conformational changes in GABAA receptor pore-lining M2 segment. Nat Neurosci 4: 477-485
  • Horenstein, J., and Akabas, M.H. (1998) Location of a high affinity Zn2+ binding site in the channel of α1β1 γ-aminobutyric acidA receptors. Mol Pharmacol 53: 870–877
  • Hsu, F. C., Smith, S. S. (2003) Progesterone withdrawal reduces paired-pulse inhibition in rat hippocampus: dependence on GABAA receptor alpha4 subunit upregulation. J Neurophysiol 89: 186-198
  • Huang, R. Q., Bell-Horner, C. L., Dibas, M. I., Covey, D. F., Drewe, J. A., Dillon, G. H. (2001) Pentylenetetrazole-induced inhibition of recombinant gamma-aminobutyric acid type A GABAA) receptors: mechanism and site of action. J Pharmacol Exp Ther 298: 986-995
  • Huang, Q., Cox, E. D., Gan, T., Ma, C., Bennett, D. W., McKernan, R. M., Cook, J. M. (1999) Studies of molecular pharmacophore/receptor models for GABAA/benzodiazepine receptor subtypes: binding affinities of substituted beta-carbolines at recombinant alpha x beta 3 gamma 2 subtypes and quantitative structure-activity relationship studies via a comparative molecular field analysis. Drug Des Discov 16: 55-76
  • Huang, Q., Liu, R., Zhang, P., He, X., McKernan, R., Gan, T., Bennett, D. W., Cook, J. M. (1998) Predictive models for GABAA/benzodiazepine receptor subtypes: studies of quantitative structure-activity relationships for imidazobenzodiazepines at five recombinant GABAA/benzodiazepine receptor subtypes [alphaxbeta3gamma2 (x = 1-3, 5, and 6)] via comparative molecular field analysis. J Med Chem 41: 4130-4142
  • Huntsman, M. M., Porcello, D. M., Homanics, G. E., DeLorey, T. M., Huguenard, J. R. (1999) Reciprocal inhibitory connections and network synchrony in the mammalian thalamus. Science 283: 541-543
  • Im, W. B., Pregenzer, J. F., Binder, J. A., Dillon, G. H., and Alberts, G. L. (1995) Chloride channel expression with the tandem construct of α6-β2 GABAA receptor subunit requires a monomeric subunit of α6 or γ2. J Biol Chem 270: 26063–26066
  • Inoue, M. and Akaike, N. (1988) Blockade of γ-aminobutyric acid-gated chloride current in frog sensory neurons by picrotoxin. Neurosci Res 5: 380–394
  • Jackel, C., Kleinz, R., Makela, R., Hevers, W., Jezequel, S., Korpi, E. R., and Luddens. H. (1998) The main determinant of furosemide inhibition on GABAA receptors is located close to the first transmembrane domain. Eur J Pharmacol 357: 251–256
  • Jenkins, A., Andreasen, A., Trudell, J. R., Harrison, N. L. (2002) Tryptophan scanning mutagenesis in TM4 of the GABAA receptor alpha1 subunit: implications for modulation by inhaled anesthetics and ion channel structure. Neuropharmacology 43: 669-678
  • Jenkins, A., Greenblatt, E. P., Faulkner, H. J., Bertaccini, E., Light, A., Lin, A., Andreasen, A., Viner, A., Trudell, J. R., Harrison, N. L. (2001) Evidence for a common binding cavity for three general anesthetics within the GABAA receptor. J Neurosci 21: RC136
  • Johnston, G. A. R. (1996) GABAC receptors: relatively simple transmitter-gated ion channels? Trends Pharmacol Sci 17: 319–323
  • Jones, A., Korpi, E. R., McKernan, R. M., Pelz, R., Nusser, Z., Ma.kela, R., Mellor, J. R., Pollard, S., Bahn, S., Stephenson, F. A., Randall, A. D., Sieghart, W., Somogyi, P., Smith, A. J. H., and Wisden, W. (1997) Ligand-gated ion channel subunit partnerships: GABAA receptor α6 subunit gene inactivation inhibits δ subunit expression. J Neurosci 17: 1350– 1362
  • Jurd, R., Arras, M., Lambert, S., Drexler, B., Siegwart, R., Crestani, F., Zaugg, M., Vogt, K. E., Ledermann, B., Antkowiak, B., Rudolph, U. (2003) General anesthetic actions in vivo strongly attenuated by a point mutation in the GABAA receptor beta3 subunit. FASEB J 17: 250-252
  • Jursky, F., Fuchs, K., Buhr, A., Tretter, V., Sigel, E., Sieghart, W. (2000) Identification of amino acid residues of GABAA receptor subunits contributing to the formation and affinity of the tert-butylbicyclophosphorothionate binding site. J Neurochem 74: 1310-1316
  • Karlin, A., Akabas, M. H. (1998) Substituted-cysteine accessibility method. Methods Enzymol 293: 123-145
  • Kash, T. L., Dizon, M. J., Trudell, J. R., Harrison, N. L. (2004) Charged residues in the GABA-A receptor beta-2 subunit are crucial for efficient receptor activation. J Biol Chem 279: 4887-4893
  • Kash, T. L., Jenkins, A., Kelley, J. C., Trudell, J. R., Harrison, N. L. (2003) Coupling of agonist binding to channel gating in the GABAA receptor. Nature 421: 272-275
  • Kaupmann, K., Malitschek, B., Schuler, V., Heid, J., Froestl, W., Beck, P., Mosbacher, J., Bischoff, S., Kulik, A., Shigemoto, R., Karschin, A., and Betterli, B. (1998a) GABAB-receptor subtypes assemble into functional heteromeric complexes. Nature 396: 683–687
  • Kaupmann, K., Schuler, V., Mosbacher, J., Bischoff, S., Bittiger, H., Heid, J., Froestl, W., Leonhard, S., Pfaff, T., Karschin, A. and Bettler, B. (1998b) Human gamma-aminobutyric acid type B receptors are differentially expressed and regulate inwardly rectifying K+ channels. Proc Natl Acad Sci USA 95: 14991–14996
  • Kaupmann, K., Huggel, K., Heid, J., Flor, P. J., Bischoff, S., Mickel, S. J., McMaster, G., Angst, C., Bittiger, H., Froestl, W., and Bettler, B. (1997) Expression cloning of GABAB receptors uncovers similarity to metabotropic glutamate receptors. Nature 386: 239–246
  • Keberle, H., and Faigle, J. W., (1972) Synthesis and structure-activity relationship of the gamma-aminobutyric acid derivatives, in Spasticity: A Topical Survey (Birkmayer W. ed) pp 90–100, Hans Huber, Vienna
  • Kelly, M. D., Smith, A., Banks, G., Wingrove, P., Whiting, P. W., Atack, J., Seabrook, G. R., Maubach, K. A. (2002) Role of the histidine residue at position 105 in the human alpha5 containing GABAA receptor on the affinity and efficacy of benzodiazepine site ligands. Br J Pharmacol 135: 248-256
  • Kirkness, E. F., and Fraser, C. M. (1993) A strong promotor element is located between alternative exons of a gene encoding the human γ-aminobutyric acid-type A receptor β2 subunit (GABRB3). J Biol Chem 268: 4420–4428
  • Kittler, J. T., Moss, S. J. (2003) Modulation of GABAA receptor activity by phosphorylation and receptor trafficking: implications for the efficacy of synaptic inhibition. Curr Opin Neurobiol 13: 341-347
  • Kittler, J. T., McAinsh, K., Moss, S. J. (2002) Mechanisms of GABAA receptor assembly and trafficking: implications for the modulation of inhibitory neurotransmission. Mol Neurobiol 26: 251-268
  • Kittler, J. T., Wang, J., Connolly, C. N., Vicini, S., Smart, T. G., Moss, S. J. (2000) Analysis of GABAA receptor assembly in mammalian cell lines and hippocampal neurons using gamma 2 subunit green fluorescent protein chimeras. Mol Cell Neurosci, 16: 440-452
  • Klausberger, T., Sarto, I., Ehya, N., Fuchs, K., Furtmuller, R., Mayer, B., Huck, S., Sieghart, W. (2001) Alternate use of distinct intersubunit contacts controls GABAA receptor assembly and stoichiometry. J Neurosci 21: 9124-9133
  • Klausberger, T., Ehya, N., Fuchs, K., Fuchs, T., Ebert, V., Sarto, I., Sieghart, W. (2001)

Detection and binding properties of GABAA receptor assembly intermediates. J Biol Chem 276: 16024-16032

  • Klausberger, T., Fuchs, K., Mayer, B., Ehya, N., Sieghart, W. (2000) GABAA receptor assembly. Identification and structure of gamma(2) sequences forming the intersubunit contacts with alpha(1) and beta(3) subunits. J Biol Chem 275: 8921-8928
  • Klunk, W. E., A. McKeon, D. F. Covey, and J. A. Ferrendelli. (1982) α-Substituted γ-butyrolactones: new class of anticonvulsant drugs. Science (Washington DC) 217:1040–1042
  • Knoflach, F., Benke, D., Wang, Y., Scheurer, L., Luddens, H., Hamilton, B. J., Carter, D. B., Mohler, H., and Benson, J. A. (1996) Pharmacological modulation of the diazepaminsensitive recombinant γ-aminobutyric acid A receptors α4β2γ2 and α6β2γ2. Mol Pharmacol 50: 1243–1261
  • Knoflach, F., Bakus, K. H., Giller, T., Malherbe, P., Pflimlin, P., Mohler, H., Trube, B. (1992) Pharmacological and electrophysiological properties of recombinant GABAA receptors comprising the α3, β1 and γ2 subunits. Eur J Neurosci 4: 1-9
  • Knoflach, F., Rhyner, T., Villa, M., Kellenberger, S., Drescher, U., Malherbe, P., Sigel, E., Mohler, H. (1991) The gamma 3-subunit of the GABAA-receptor confers sensitivity to benzodiazepine receptor ligands. FEBS Lett 293: 191-194
  • Kofuji, P., Wang, J. B., Moss, S. J., Huganir, R. L., and Burt, D. R. (1991) Generation of two forms of the gamma-aminobutyric acid-A receptor gamma-2-subunit in mice by alternative splicing. J Neurochem 56: 713–715
  • Koltchine, V. V., Finn, S. E., Jenkins, A., Nikolaeva, N., Lin, A., Harrison, N. L. (1999) Agonist gating and isoflurane potentiation in the human gamma-aminobutyric acid type A receptor determined by the volume of a second transmembrane domain residue. Mol Pharmacol 56: 1087-1093
  • Korpi, E. R., Grunder, G., Luddens, H. (2002) Drug interactions at GABAA receptors. Prog Neurobiol 67: 113-159
  • Korpi, E. R., Mihalek, R. M., Sinkkonen, S. T., Hauer, B., Hevers, W., Homanics, G. E., Sieghart, W., Luddens, H. (2002) Altered receptor subtypes in the forebrain of GABAA receptor delta subunit-deficient mice: recruitment of gamma 2 subunits. Neuroscience 109: 733-743


  • Korpi, E. R., Koikkalainen, P., Vekovischeva, O. Y., Makela, R., Kleinz, R., Uusi-Oukari, M., Wisden, W. (1999) Cerebellar granule-cell-specific GABAA receptors attenuate benzodiazepine-induced ataxia: evidence from alpha 6-subunit-deficient mice. Eur J Neurosci 11: 233-240
  • Korpi, E.R., Kuner, T., Seeburg, P. H., and Luddens, H. (1995) Selective antagonist for the cerebellar granule cell - specific γ-aminobutyric acid type A receptor. Mol Pharmacol 47: 283–289
  • Korpi, E. R., Kuner, T., Kristo, P., Kohcer, M., Herb, A., Luddens, H., and Seeburg, P. H. T. (1994) Small N-terminal deletion by splicing in cerebellar α6 - subunit abolishes GABAA receptor function. J Neurochem 63: 1167–1170
  • Koulen, P. (1999) Postnatal development of GABAA receptor beta1, beta2/3, and gamma2 immunoreactivity in the rat retina. J Neurosci Res 57: 185-194
  • Koulen, P., Brandstatter, J.H., Enz, R., Bormann, J., and Wassle, H. (1998) Synaptic clustering of GABAC receptor ρ-subunits in the rat retina. Eur J Neurosci 10: 115–127
  • Kralic, J. E., Wheeler, M., Renzi, K., Ferguson, C., O'Buckley, T. K., Grobin, A. C., Morrow, A. L., Homanics, G. E.. (2003) Deletion of GABAA receptor alpha 1 subunit-containing receptors alters responses to ethanol and other anesthetics. J Pharmacol Exp Ther 305: 600-607
  • Kralic, J. E., Korpi, E. R., O'Buckley, T. K., Homanics, G. E., Morrow, A. L. (2002a) Molecular and pharmacological characterization of GABAA receptor alpha1 subunit knockout mice. J Pharmacol Exp Ther 302: 1037-1045
  • Kralic, J. E., O'Buckley, T. K., Khisti, R. T., Hodge, C. W., Homanics, G. E., Morrow, A. L. (2002b) GABAA receptor alpha-1 subunit deletion alters receptor subtype assembly, pharmacological and behavioral responses to benzodiazepines and zolpidem. Neuropharmacology 43: 685-694
  • Krasowski, M. D., Jenkins, A., Flood, P., Kung, A. Y., Hopfinger, A. J., Harrison, N. L. (2001a) General anesthetic potencies of a series of propofol analogs correlate with potency for potentiation of gamma-aminobutyric acid (GABA) current at the GABAA receptor but not with lipid solubility. J Pharmacol Exp Ther 297: 338-351
  • Krasowski, M. D., Nishikawa, K., Nikolaeva, N., Lin, A., Harrison, N. L. (2001b) Methionine 286 in transmembrane domain 3 of the GABAA receptor beta subunit controls a binding cavity for propofol and other alkylphenol general anesthetics. Neuropharmacology 41: 952-964
  • Krasowski, M. D., Harrison, N. L. (2000) The actions of ether, alcohol and alkane general anaesthetics on GABAA and glycine receptors and the effects of TM2 and TM3 mutations. Br J Pharmacol 129: 731-443
  • Krasowski, M. D., Koltchine, V. V., Rick, C. E., Ye, Q., Finn, S. E., Harrison, N. L. (1998a) Propofol and other intravenous anesthetics have sites of action on the gamma-aminobutyric acid type A receptor distinct from that for isoflurane. Mol Pharmacol 53: 530-538
  • Krasowski, M. D., Rick, C. E., Harrison, N. L., Firestone, L. L., Homanics, G. E. (1998b) A deficit of functional GABAA receptors in neurons of beta 3 subunit knockout mice. Neurosci Lett 240: 81-84
  • Krasowski, M. D., O'Shea, S. M., Rick, C. E., Whiting, P. J., Hadingham, K. L., Czajkowski, C, Harrison, N. L. (1997) Alpha subunit isoform influences GABAA receptor modulation by propofol. Neuropharmacology 36: 941-949
  • Kravitz, E. A. (1963) Gamma-aminobutyric acid and other blocking compounds in crustacea. III. Their relative concentrations in separated motor and inhibitory axons. J Neurophysiol 49: 831-850
  • Kreimeyer, A., Laube, B., Sturgess, M., Goeldner, M., Foucaud, B. (1999a) Evaluation and biological properties of reactive ligands for the mapping of the glycine site on the N-methyl-D-aspartate (NMDA) receptor. J Med Chem 42: 4394-4404
  • Kreimeyer, A., Laube, B., Sturgess, M., Goeldner, M., Foucaud, B. (1999b) Reactive affinity probes for the mapping of the glycine-binding site of the NMDA receptor NR1 subunit. J Recept Signal Transduct Res 19: 547-557
  • Krishek, B.J., Moss, S. J., and Smart, T. G. (1996b) Homomeric β1 γ−aminobutyric acid A receptor-ion channels: evaluation of pharmacological and physiological properties. Mol Pharmacol 49: 494–504
  • Krnjevic, K., and Schwartz, S. (1967) The action of γ-aminobutyric acid on cortical neurons. Expl Brain Res 3: 320-336
  • Krusek, J., Zemkova, H. (1994) Effect of ivermectin on γ-aminobutyric acid-induced chloride currents in mouse hippocampal embryonic neurones. Eur J Pharmacol 259: 121–128
  • Kucken, A. M., Teissere, J. A., Seffinga-Clark, J., Wagner, D. A., Czajkowski, C. (2003) Structural requirements for imidazobenzodiazepine binding to GABAA receptors. Mol Pharmacol 63: 289-296
  • Kucken, A. M., Wagner, D. A., Ward, P. R., Teissere, J. A., Boileau, A. J., Czajkowski, C. (2000) Identification of benzodiazepine binding site residues in the gamma2 subunit of the gamma-aminobutyric acidA receptor. Mol Pharmacol 57: 932-939
  • Kuffler, S. W., and Edwards, C. (1958) Mechanism of gamma-aminobutyric acid (GABA) action and its relation to synaptic inhibition. J Neurophysiol 21: 589-610
  • Kusama, T., Spivak, C. E., Whiting, P., Dawson, V. L., Schaeffer, J. C., and Uhl, G. R. (1993a) Pharmacology of GABAρ1 and GABAα/β receptors expressed in Xenopus oocytes and Cos cells. Br J Pharmacol 109: 200–206
  • Kusama, T., Wang, T. L., Guggino, W. B., Cutting, G. R., and Uhl, G. R. (1993b) GABA rho2 receptor pharmacological profile: GABA recognition site similarities to rho1. Eur J Pharmacol 245: 83–84
  • Kultas-Ilinsky, K., Leontiev, V., Whiting, P. J. (1998) Expression of 10 GABAA receptor subunit messenger RNAs in the motor-related thalamic nuclei and basal ganglia of Macaca mulatta studied with in situ hybridization histochemistry. Neuroscience 85: 179-204
  • Lamsa, K., Taira, T. (2003) Use-dependent shift from inhibitory to excitatory GABAA receptor action in SP-O interneurons in the rat hippocampal CA3 area. J Neurophysiol 90:1983-1995
  • Laposky, A. D., Homanics, G. E., Basile, A., Mendelson, W. B. (2001) Deletion of the GABAA receptor beta 3 subunit eliminates the hypnotic actions of oleamide in mice. Neuroreport 12: 4143-4147
  • Laurie, D. J., Seeburg, P. H., and Wisden, W. (1992) The distribution of 13 GABAA receptor subunit mRNAs in the rat brain. II. Olfactory bulb and cerebellum. J Neurosci 12: 1063–1076
  • Le Novere, N., Corringer, P. J., Changeux, J. P. (2002a) The diversity of subunit composition in nAChRs: evolutionary origins, physiologic and pharmacologic consequences. J Neurobiol 53: 447-456
  • Le Novere, N., Grutter, T., Changeux, J. P. (2002b) Models of the extracellular domain of the nicotinic receptors and of agonist- and Ca2+-binding sites. Proc Natl Acad Sci USA 99: 3210–3215
  • Levin, M. L., Chatterjee, A., Pragliola, A., Worley, K. C., Wehnert, M., Zhuchenko, O., Smith, R. F., Lee, C. C., and Herman, G. E. (1996) A comparative transcription map of the murine bare patches (Bpa) and striated (Str) critical regions and human Xq28. Genome Res 6: 465–477
  • Li, X., Pearce, R. A. (2000) Effects of halothane on GABAA receptor kinetics: evidence for slowed agonist unbinding. J Neurosci 20: 899-907
  • Li, X., Czajkowski, C., Pearce, R. A. (2000) Rapid and direct modulation of GABAA receptors by halothane. Anesthesiology 92: 1366-1375
  • Lolait, S. J., O'Carroll, A. M., Kusano, K., Muller, J. M., Brownstein, M. J., Mahan, L. C. (1989) Cloning and expression of a novel rat GABAA receptor. FEBS Lett 246: 145-148
  • Loup, F., Wieser, H. G., Yonekawa, Y., Aguzzi, A., Fritschy, J. M. (2000) Selective alterations in GABAA receptor subtypes in human temporal lobe epilepsy. J Neurosci 20: 5401-5419
  • Low, K., Crestani, F., Keist, R., Benke, D., Brunig, I., Benson, J. A., Fritschy, J. M., Rulicke, T., Bluethmann, H., Mohler, H., and Rudolph, U. (2000) Molecular and neuronal substrate for the selective attenuation of anxiety. Science (Wash DC) 290: 131–134
  • Luddens, H., Seeburg, P. H., and Korpi, E. R. (1994) Impact of α and γ variants on ligand-binding properties of γ-aminobutyric acid type A receptors. Mol Pharmacol 45: 810–814
  • Luddens, H., Pritchett, D. B., Kohler, M., Killisch, L., Keinanan, K., Monyer, H., Sprengel, R., and Seeburg, P. H. (1990) Cerebellar GABAA receptor selective for a behavioural alcohol antagonist. Nature 346: 648–651
  • Lukasiewicz, P. D. (1996) GABAC receptors in the vertebrate retina. Mol Neurobiol 12: 181–194
  • Macdonald, R. L., and Olsen, R. W. (1994) GABAA receptor channels. Annu Rev Neurosci 17: 569–602
  • Mahmoudi, M., Kang, M. H., Tillakaratne, N., Tobin, A. J., Olsen, R. W. (1997) Chronic intermittent ethanol treatment in rats increases GABAA receptor alpha4-subunit expression: possible relevance to alcohol dependence. J Neurochem 68: 2485-2492
  • Malherbe, P., Sigel, E., Baur, R., Persohn, E., Richards, J. G., Mohler, H. (1990a) Functional characteristics and sites of gene expression of the alpha1, beta1, gamma2-isoform of the rat GABAA receptor. J Neurosci 10: 2330-2337
  • Malherbe, P., Sigel, E., Baur, R., Persohn, E., Richards, J. G., Mohler, H. (1990b) Functional expression and sites of gene transcription of a novel alpha subunit of the GABAA receptor in rat brain. FEBS Lett 260: 261-265
  • Malminiemi, O., Korpi, E. R., (1989) Diazepam-insensitive [3H]Ro15-4513 binding in intact cultured cerebellar granule cells. Eur J Pharmacol 169: 53– 60
  • Mamalaki, C., Barnard, E. A., and Stephenson, F. A. (1989) Molecular size of the γ-aminobutyric acidA receptor purified from mammalian cerebral cortex. J Neurochem 52: 125–134
  • Marder, M., Estiu, G., Blanch, L. B., Viola, H., Wasowski, C., Medina, J. H., Paladini, A. C. (2001) Molecular modeling and QSAR analysis of the interaction of flavone derivatives with the benzodiazepine binding site of the GABAA receptor complex. Bioorg Med Chem 9: 323-335
  • Marshall, F. H., Jones, K. A., Kaupmann, K., and Bettler, B. (1999a) GABAB receptors−the first 7TM heterodimers. Trends Pharmacol Sci 20: 396–399
  • Marshall, F. H., White, J., Main, M., Green, A., and Wise, A. (1999b) GABAB receptors function as heterodimers. Biochem Soc Trans 27: 530–535
  • Martina, M., Mozrzymas, J. W., Strata, F., Cherubini, E. (1996) Zinc modulation of bicuculline-sensitive and -insensitive GABA receptors in the developing rat hippocampus. Eur J Neurosci 8: 2168-2176
  • Martinez-Torres, A., Miledi, R. (2004) Expression of functional receptors by the human gamma-aminobutyric acid A gamma 2 subunit. Proc Natl Acad Sci USA 101: 3220-3223
  • Mascia, M. P., Trudell, J. R., Harris, R. A. (2000) Specific binding sites for alcohols and anesthetics on ligand-gated ion channels. Proc Natl Acad Sci USA 97: 9305-9310
  • Mathews, G. C., Bolos-Sy, A. M., Covey, D. F., Rothman S. M., and Ferrendelli, J. A. (1996) Physiological comparison of α-ethyl-α-methyl-γ-thiobutyrolactone with benzodiazepine and barbiturate modulators of GABAA receptors. Neuropharmacology 35:123–136
  • McKernan, R. M., Rosahl, T. W., Reynolds, D. S., Sur, C., Wafford, K. A., Atack, J. R., Farrar S., Myers, J., Cook, G., Ferris. P., Garrett, L., Bristow, L., Marshall, G., Macaulay, A., Brown, N., Howell, O., Moore, K. W., Carling, R. W., Street, L. J., Castro, J. L., Ragan, C. I., Dawson, G. R., Whiting, P. J. (2000) Sedative but not anxiolytic properties of benzodiazepines are mediated by the GABAA receptor α1 subtype. Nature Neurosci 3: 587–592
  • McKernan, R. M., Wafford, K., Quirk, K., Hadingham, K. L., Harley, E. A., Ragan, C. I., and Whiting, P. J. (1995) The pharmacology of the benzodiazepine site of the GABA-A receptor is dependent upon the type of γ-subunit present. J Recept Signal Transduct Res 15: 173–183
  • Mihalek, R. M., Bowers, B. J., Wehner, J. M., Kralic, J. E., VanDoren, M. J., Morrow, A. L., Homanics, G. E. (2001) GABAA-receptor delta subunit knockout mice have multiple defects in behavioral responses to ethanol. Alcohol Clin Exp Res 25: 1708-1718
  • Mihalek, R. M., Banerjee, P. K., Korpi, E. R., Quinlan, J. J., Firestone, L. L., Mi, Z. P., Lagenaur, C., Tretter, V., Sieghart, W., Anagnostaras, G., Sage, J. R., Fanselow, M. S., Guidotti, A., Spigelman, I., Li, Z., DeLorey, T. M., Olsen, R. W., Homanics, G. E. (1999) Attenuated sensitivity to neuroactive steroids in γ-aminobutyrate type A receptor delta subunit knock-out mice. Proc Natl Acad Sci USA 96: 12905–12910
  • Mihic, S. J., Ye, Q., Wick, M. J., Koltchine, V. V., Krasowski, M. D., Finn, S. E., Mascia, M. P., Valenzuela, C. F., Hanson, K. K., Greenblatt, E. P., Harris, R. A., Harrison, N. L. (1997) Sites of alcohol and volatile anaesthetic action on GABAA and glycine receptors. Nature 389: 385-389
  • Mihic, S. J., Whiting, P. J., Klein, R. L., Wafford, K. A., and Harris, R. A. (1994) A single amino acid of the human gamma-aminobutyric acid type A receptor γ2 subunit determines benzodiazepine efficacy. J Biol Chem 269: 32768–32773
  • Miralles, C. P., Gutierrez, A., Khan, Z. U., Vitorica, J., De Blas, A. L. (1994) Differential expression of the short and long forms of the gamma 2 subunit of the GABAA/benzodiazepine receptors. Brain Res Mol Brain Res 24: 129-139
  • Miyazawa, A., Fujiyoshi, Y., Unwin, N. (2003) Structure and gating mechanism of the acetylcholine receptor pore. Nature 424: 949-955
  • Miyazawa, A., Fujiyoshi, Y., Stowell, M., and Unwin, N. (1999) Nicotinic acetylcholine receptor at 4.6 Å resolution: transverse tunnels in the channel wall. J Mol Biol 288: 765–786
  • Mohler, H., Battersby, M. K., and Richards, J. G. (1980) Benzodiazepine receptor protein identified and visualized in brain tissue by a photoaffinity label. Proc Natl Acad Sci USA 77: 1666–1670
  • Mohler, H., Fritschy, J. M., Benke, D., Rudolph, U., and Luscher, B. (1996a) GABAA receptor subtypes: Pharmacological significance and mutational analysis in vivo, in GABA: Receptors, Transporters and Metabolism (Tanaka C. and Bowery N. G., eds) pp 157–171, Bilkhauser, Basel
  • Mohler, H., Fritschy, J. M., Luscher, B., Rudolph, U., and Benson, J. (1996b) The GABAA receptors: From subunits to diverse functions, in Ion Channels (Narahashi, T. ed) vol. 4., pp 89–113, Plenum Press, New York
  • Mohler, H., and Richards, J. G. (1981) Agonist and antagonist of benzodiazepine receptor: interaction in vitro. Nature 294: 763–765
  • Moody, E. J., Skolnick, P. (1989) Chlormethiozole: neurochemical actions at the γ-aminobutyric acid receptor complex. Eur J Pharmacol 164: 153–158
  • Moragues, N., Ciofi, P., Lafon, P., Tramu, G., Garret, M. (2003) GABAA receptor epsilon subunit expression in identified peptidergic neurons of the rat hypothalamus. Brain Res 967 : 285-289
  • Moragues, N., Ciofi, P., Tramu, G., Garret, M. (2002) Localisation of GABAA receptor epsilon-subunit in cholinergic and aminergic neurones and evidence for co-distribution with the theta-subunit in rat brain. Neuroscience 111: 657-669
  • Moragues, N., Ciofi, P., Lafon, P., Odessa, M. F., Tramu, G., Garret, M. (2000) cDNA cloning and expression of a gamma-aminobutyric acid A receptor epsilon-subunit in rat brain. Eur J Neurosci 12: 4318-4330
  • Moss, S. J., Smart, T. G. (2001) Constructing inhibitory synapses. Nat Rev Neurosci, 2: 240-250
  • Mozrzymas, J. W., Barberis, A., Michalak, K., Cherubini, E. (1999) Chlorpromazine inhibits miniature GABAergic currents by reducing the binding and by increasing the unbinding rate of GABAA receptors. J Neurosci 19: 2474-2488
  • Nagata, K., Hamilton, B. J., Carter, D. B., Narahashi, T. (1994) Selective effects of dieldrin on the GABAA receptor-channel subunits expressed in human embryonic kidney cells. Brain Res 645: 19-26
  • Nagata, K., and Narahashi, T. (1994) Dual action of the cyclodien insecticide dieldrin on the g-aminobutyric acid receptor-chloride channel complex of rat dorsal root ganglion neurons. J Pharmacol Exp Ther 269: 164-171
  • Nayeem, N., Green, T. P., Martin, I. L., and Barnard, E. A. (1994) Quaternary structure of the native GABAA receptor determined by electron microscope image analysis. J Neurochem 62: 815–818
  • Neelands, T. R., and Macdonald, R. L. (1999) Incorporation of the π subunit into functional γ-aminobutyric acidA receptors. Mol Pharmacol 56: 598–610
  • Newell, J. G., Czajkowski, C. (2003) The GABAA receptor alpha1 subunit Pro174-Asp191 segment is involved in GABA binding and channel gating. J Biol Chem 278: 13166-13172
  • Nishikawa, K., Harrison, N. L. (2003) The actions of sevoflurane and desflurane on the gamma-aminobutyric acid receptor type A: effects of TM2 mutations in the alpha and beta subunits. Anesthesiology 99: 678-684
  • Nishikawa, K., Jenkins, A., Paraskevakis, I., Harrison, N. L. (2002) Volatile anesthetic actions on the GABAA receptors: contrasting effects of alpha 1(S270) and beta 2(N265) point utations. Neuropharmacology 42: 337-345
  • Nusser, Z., Ahmad, Z., Tretter, V., Fuchs, K., Wisden, W., Sieghart, W., Somogyi, P. (1999b) Alterations in the expression of GABAA receptor subunits in cerebellar granule cells after the disruption of the alpha6 subunit gene. Eur J Neurosci 11: 1685-1697
  • O'Shea, S. M., Harrison, N. L. (2000) Arg-274 and Leu-277 of the gamma-aminobutyric acid type A receptor alpha 2 subunit define agonist efficacy and potency. J Biol Chem 275: 22764-22778
  • Ortells, M. O. and Lunt, G. G. (1995) Evolutionary history of the ligand-gated ion-channel superfamily of receptors. Trends Neurosci 18: 121-126
  • Pan, Z. H., Zhang, D., Zhang, X., Lipton, S. A. (2000) Evidence for coassembly of mutant GABAC rho1 with GABAA gamma2S, glycine alpha1 and glycine alpha2 receptor subunits in vitro. Eur J Neurosci 12: 3137-3145
  • Pan, Z. H., Zhang, X., Zhang, Z., and Lipton, S. A. (1997) Evidence for co-assembly of GABA ρ1 with GABAA or glycine subunits in vitro. Soc Neurosci Abstr 7: 6
  • Papadopoulos, V., Amri, H., Li, H., Yao, Z., Brown, R. C., Vidic, B., Culty, M. (2001) Structure, function and regulation of the mitochondrial peripheral-type benzodiazepine receptor. Therapie 56: 549-556
  • Park-Chung, M., Malayev, A., Purdy, R. H., Gibbs, T. T., Farb, D. H. (1999) Sulfated and unsulfated steroids modulate γ-aminobutyric acidA receptor function through distinct sites. Brain Res 830: 72–87
  • Park, J. S., Higashi, H., Nagata, K., Yoshimura, M. (1999) Bicuculline-resistant, Cl¯ dependent GABA response in the rat spinal dorsal horn. Neurosci Res 33: 261-268
  • Peng, Z., Hauer, B., Mihalek, R. M., Homanics, G. E., Sieghart, W., Olsen, R. W., Houser, C. R. (2002) GABAA receptor changes in delta subunit-deficient mice: altered expression of alpha4 and gamma2 subunits in the forebrain. J Comp Neurol 446: 179-197
  • Peoples, R. W., Weight, F. F. (1994) Trichloroethanol potentiation of γ-aminobutyric acid-activated chloride current in mouse hippocampal neurones. Br J Pharmacol 113: 555–563
  • Perret, P., Sarda, X., Wolff, M., Wu, T. T., Bushey, D., Goeldner, M. (1999) Interaction of non-competitive blockers within the gamma-aminobutyric acid type A chloride channel using chemically reactive probes as chemical sensors for cysteine mutants. J Biol Chem 274: 25350-25354
  • Persohn, E., Malherbe, P., Richards, J. G. (1992) Comparative molecular neuroanatomy of cloned GABAA receptor subunits in the rat CNS. J Comp Neurol 326: 193-216
  • Pirker, S., Schwarzer, C., Czech, T., Baumgartner, C., Pockberger, H., Maier, H., Hauer, B., Sieghart, W., Furtinger, S., Sperk, G. (2003) Increased expression of GABAA receptor beta-subunits in the hippocampus of patients with temporal lobe epilepsy. J Neuropathol Exp Neurol 62: 820-834
  • Pirker, S., Schwarzer, C., Wieselthaler, A., Sieghart, W., Sperk, G. (2000) GABAA receptors: immunocytochemical distribution of 13 subunits in the adult rat brain. Neuroscience 101: 815-850
  • Poltl, A., Hauer, B., Fuchs, K., Tretter, V., Sieghart, W. (2003) Subunit composition and quantitative importance of GABAA receptor subtypes in the cerebellum of mouse and rat. J Neurochem 87: 1444-1455
  • Polenzani, L., Woodward, R. M., and Miledi, R. (1991) Expression of mammalian γ-aminobutyric acid receptors with distinct pharmacology in Xenopus oocytes. Proc Natl Acad Sci USA 88: 4318–4322
  • Pong, S.-S., Wang, C. C. (1982) Avermectin B modulation of γ-aminobutyric acid receptors in rat brain membranes. J Neurochem 38: 375–379
  • Papadopoulos, V., Amri, H., Li, H., Yao, Z., Brown, R. C., Vidic, B., Culty, M. Structure, function and regulation of the mitochondrial peripheral-type benzodiazepine receptor. Therapie 56: 549-556
  • Porcello, D. M., Huntsman, M. M., Mihalek, R. M., Homanics, G. E., Huguenard, J. R. (2003) Intact Fast Synaptic GABAergic Inhibition and Altered Neurosteroid Modulation of Thalamic Relay Neurons in Mice Lacking the {delta} Subunit. J Neurophysiol 89: 1378-1386
  • Pritchett, D. B., Seeburg, P. H. (1991) gamma-Aminobutyric acid type A receptor point mutation increases the affinity of compounds for the benzodiazepine site. Proc Natl Acad Sci USA 88: 1421-1425
  • Pritchett, D. B., and Seeburg, P. H. (1990) γ-aminobutyric acidA receptor α5 – subunit creates novel type II benzodiazepine receptor pharmacology. J Neurochem 54: 1802–1804
  • Pritchett, D. B., Sontheimer, H., Shivers, B. D., Ymer, S., Kettenmann, H., Schofield, P.R., Seeburg, P. H. (1989) Importance of a novel GABAA receptor subunit for benzodiazepine pharmacology. Nature 338: 582–585
  • Pritchett, D. B., Sontheimer, H., Gorman, C. M., Kettenmann, H., Seeburg, P. H., Schofield, P. R. (1988) Transient expression shows ligand gating and allosteric potentiation of GABAA receptor subunits. Science 242: 1306-1308
  • Puia, G., Vicini, S., Seeburg, P. H., and Costa, E. (1991) Influence of recombinant γ-aminobutyric acid-A receptor subunit composition on the action of allosteric modulators of γ-aminobutyric acid-gated Cl─ currents. Mol Pharmacol 39: 691–696
  • Puia, G., Santi, M. R., Vicini, S., Pritchett, D. B., Purdy, R. H., Paul, S. M., Seeburg, P. H., Costa, E. (1990) Neurosteroids act on recombinant human GABAA receptor. Neuron 4: 759–765
  • Qian, H., Ripps, H. (1999) Response kinetics and pharmacological properties of heteromeric receptors formed by co-assembly of GABA rho- and gamma 2-subunits. Proc R Soc Lond B Biol Sci 266: 2419-2425
  • Quian, H., and Dowling, J. E. (1993) Novel GABA responses from rod-driven retinal horizontal cells. Nature 361: 162–164
  • Quinlan, J. J., Firestone, L. L., Homanics, G. E. (2000) Mice lacking the long splice variant of the gamma 2 subunit of the GABAA receptor are more sensitive to benzodiazepines. Pharmacol Biochem Behav 66: 371-374
  • Quinlan, J. J., Homanics, G. E., Firestone, L. L. (1998) Anesthesia sensitivity in mice that lack the beta3 subunit of the gamma-aminobutyric acid type A receptor. Anesthesiology 88: 775-780
  • Ramadan, E., Fu, Z., Losi, G., Homanics, G. E., Neale, J. H., Vicini, S. (2003) GABAA receptor beta3 subunit deletion decreases alpha2/3 subunits and IPSC duration. J Neurophysiol 89: 128-134
  • Renard, S., Olivier, A., Granger, P., Avenet, P., Graham, D., Sevrin, M., George, P., Besnard, F. (1999) Structural elements of the gamma-aminobutyric acid type A receptor conferring subtype selectivity for benzodiazepine site ligands. J Biol Chem 274: 13370-13374
  • Reynolds, D. S., Rosahl, T. W., Cirone, J., O'Meara, G. F., Haythornthwaite, A., Newman, R. J., Myers, J., Sur, C., Howell, O., Rutter, A. R., Atack, J., Macaulay, A. J., Hadingham, K. L., Hutson, P. H., Belelli, D., Lambert, J. J., Dawson, G. R., McKernan, R., Whiting, P. J., Wafford, K. A. (2003) Sedation and anesthesia mediated by distinct GABAA receptor isoforms. J Neurosci 23: 8608-8617
  • Rogers, C. J., Twyman, R. E., and MacDonald, R. L. (1994) Benzodiazepine and betacarboline regulation of single GABAA-receptor channels of mouse spinal neurons in culture. J Physiol 475: 69–82
  • Rudolph, U., Mohler, H. (2004) Analysis of GABAA receptor function and dissection of the pharmacology of benzodiazepines and general anesthetics through mouse genetics. Annu Rev Pharmacol Toxicol 44: 475-498
  • Rudolph, U., Crestani, F., Benke, D., Brunig, I., Benson, J. A., Fritschy, J. M., Martin, J. R., Bluethmann, H., Mohler, H. (1999) Benzodiazepine actions mediated by specific gamma-aminobutyric acidA receptor subtypes. Nature 401: 796-800, erratum: Nature 2000 404: 629
  • Sander, C., and Schneider, R. (1991) Database of Homology-Derived Structures and the Structural Meaning of Sequence Alignment. Proteins 9: 56-68
  • Sanna, E., Murgia, A., Casula, A., Usala, M., Maciocco, E., Tuligi, G., Biggio, G. (1996) Direct activation of GABAA receptors by loreclezole, an anticonvulsant drug with selectivity for the beta-subunit. Neuropharmacology 35: 1753-1760
  • Sanna, E., Garau, F., Harris, R. A. (1995) Novel properties of homomeric β1 γ-aminobutyric acid type A receptors: actions of the anesthetics propofol and pentobarbital. Mol Pharmacol 47: 213–217
  • Sarto, I., Wabnegger, L., Dogl, E., Sieghart, W. (2002a) Homologous sites of GABAA receptor alpha(1), beta(3) and gamma(2) subunits are important for assembly. Neuropharmacology 43: 482-491
  • Sarto, I., Klausberger, T., Ehya, N., Mayer, B., Fuchs, K., Sieghart, W. (2002b) A novel site on gamma 3 subunits important for assembly of GABAA receptors. J Biol Chem 277: 30656-30664
  • Sawyer, G. W., Chiara, D. C., Olsen, R. W., Cohen, J. B. (2002) Identification of the bovine gamma-aminobutyric acid type A receptor alpha subunit residues photolabeled by the imidazobenzodiazepine [3H]Ro15-4513. J Biol Chem 277: 50036-50045
  • Saxena, N. C. (2000) Inhibition of GABAA receptor (GABAR) currents by arachidonic acid in HEK 293 cells stably transfected with alpha1beta2gamma2 GABAR subunits. Pflugers Arch 440: 380-392
  • Saxena, N. C., and MacDonald, R. L. (1994) Assembly of GABAA receptor subunits: role of the δ-subunit. J Neurosci 14: 7077–7086
  • Schaerer, M. T., Buhr, A., Baur, R., Sigel, E. (1998) Amino acid residue 200 on the alpha1 subunit of GABAA receptors affects the interaction with selected benzodiazepine binding site ligands. Eur J Pharmacol 354: 283-287
  • Schapira, M., Abagyan, R., Totrov, M. (2002) Structural model of nicotinic acetylcholine receptor isotypes bound to acetylcholine and nicotine. BMC Struct Biol 2: 1
  • Schofield, P. R., Darlison, M. G., Fujita, N., Burt, D. R., Stephenson, F. A., Rodriguez, H., Rhee, L. M., Ramachandran, J., Reale, V., Glencorse, T. A., Seeburg, P. H., and Barnard, E. A. (1987) Sequence and functional expression of the GABAA receptor shows a ligand-gated receptor superfamily. Nature 328: 221–227
  • Schove, L. T., Perez, J. J., and Loew, G. H. (1994) Molecular determinants of recognition and activation at the cerebellar benzodiazepine receptor site. Bioorg Med Chem 2: 1029–1049
  • Schwarzer, C., Berresheim, U., Pirker, S., Wieselthaler, A., Fuchs, K., Sieghart, W., Sperk, G. (2001) Distribution of the major gamma-aminobutyric acidA receptor subunits in the basal ganglia and associated limbic brain areas of the adult rat. J Comp Neurol 433: 526-549
  • Sperk, G., Schwarzer, C., Tsunashima, K., Fuchs, K., Sieghart, W. (1997) GABAA receptor subunits in the rat hippocampus I: immunocytochemical distribution of 13 subunits. Neuroscience 80: 987-1000
  • Schwarzer, C., Tsunashima, K., Wanzenbock, C., Fuchs, K., Sieghart, W., Sperk, G. (1997) GABAA receptor subunits in the rat hippocampus II: altered distribution in kainic acid-induced temporal lobe epilepsy. Neuroscience 80: 1001-1017
  • Schwartz, R. D., Yu, X. (1992) Inhibition of GABA-gated chloride channel function by arachidonic acid. Brain Res 585: 405-410
  • Schwartz, R. D., Skolnick, P., Paul, S. M. (1988) Regulation of gamma-aminobutyric acid/ barbiturate receptor-gated chloride ion flux in brain vesicles by phospholipase A2: possible role of oxygen radicals. J Neurochem 50: 565-571
  • Serafini, R., Bracamontes, J., Steinbach, J. H. (2000) Structural domains of the human GABAA receptor beta3 subunit involved in the actions of pentobarbital. J Physiol 524: 649-676
  • Shen, W., Mennerick, S., Covey, D. F., Zorumski, C. F. (2000) Pregnenolone sulfate modulates inhibitory synaptic transmission by enhancing GABAA receptor desensitization. J Neurosci 20: 3571-3579
  • Shimada, S., Cutting, G. R., and Uhl, G. R. (1992) γ-Aminobutyric acid A or C receptor? γ-aminobutyric acid ρ1 receptor RNA induces bicuculline-, barbiturate-, and benzodiazepine-insensitive γ-aminobutyric acid responses in Xenopus oocytes. Mol Pharmacol 41: 683–687
  • Shingai, R., Yanagi, K., Fukushima, T., Sakata, K., and Ogurusu, T. (1996) Functional expression of rat GABA receptor ρ3 subunit. Neurosci Res 26: 387–390
  • Sieghart, W. (2003) Benzodiazepines, Benzodiazepine receptor, and Endogenous Ligands in Handbook of anxiety and depression (Kasper S., Boer, J., Sitsen, J. M. eds) pp 415–442, Marcel Dekker, New York, Basel
  • Sieghart, W., Sperk, G. (2002) Subunit composition, distribution and function of GABAA receptor subtypes. Curr Top Med Chem 2: 795-816
  • Sieghart, W. (1995) Structure and pharmacology of gamma-aminobutyric acidA receptor subtypes. Pharmacol Rev 47: 181–234
  • Sieghart, W., Eichinger, A., Richards, J. G., Mohler, H. (1987) Photoaffinity labeling of benzodiazepine receptor proteins with the partial inverse agonist [3H]Ro15-4513: a biochemical and autoradiographic study. J Neurochem 48: 46-52
  • Sigel, E. (2002) Mapping of the benzodiazepine recognition site on GABAA receptors. Curr Top Med Chem 2: 833-839
  • Sigel, E., Baur, R. (2000) Electrophysiological evidence for the coexistence of alpha1 and alpha6 subunits in a single functional GABAA receptor. J Neurochem 74: 2590-2596
  • Sigel, E., Buhr, A., Baur, R. (1999) Role of the conserved lysine residue in the middle of the predicted extracellular loop between M2 and M3 in the GABAA receptor. J Neurochem 73: 1758-1764.
  • Sigel, E., Schaerer, M. T., Buhr, A., Baur, R. (1998) The benzodiazepine binding pocket of recombinant alpha1beta2gamma2 gamma-aminobutyric acidA receptors: relative orientation of ligands and amino acid side chains. Mol Pharmacol 54: 1097-1105.
  • Sigel, E., Buhr, A. (1997) The benzodiazepine binding site of GABAA receptors. Trends Pharmacol Sci 18: 425-429
  • Sigel, E., Baur, R., Kellenberger, S., Malherbe, P. (1992) Point mutations affecting antagonist affinity and agonist dependent gating of GABAA receptor channels. EMBO J 11: 2017-2023
  • Sigel, E., Baur, R., Trube, G., Mohler, H., Malherbe, P. (1990) The effect of subunit composition of rat brain GABAA receptors on channel function. Neuron 5: 703-711.
  • Sigel, E. (1990) Use of Xenopus oocytes for the functional expression of plasma membrane proteins. J Membr Biol 117: 201-221
  • Sigel, E., Baur, R., Malherbe, P., Mohler, H. (1989) The rat beta1 subunit of the GABAA receptor forms a picrotoxin-sensitive anion channel open in the absence of GABA. FEBS Lett 257: 377-379
  • Sigel, E., Baur, R. (1988) Allosteric modulation by benzodiazepine receptor ligands of the GABAA receptor channel expressed in Xenopus oocytes. J Neurosci 8: 289-295
  • Sigel, E. (1987) Properties of single sodium channels translated by Xenopus oocytes after injection with messenger ribonucleic acid. J Physiol 386: 73-90
  • Sigel, E., and Barnard, E. A. (1984) A γ-aminobutyric acid/benzodiapzepine receptor complex from bovine cerebral cortex: Improved purification with preservation of regulatory sites and their regulations. J Biol Chem 259: 7129–7223
  • Sigel, E., Stephenson, F. A., Mamalaki, C., Barnard, E. A. (1983) A γ-aminobutyric acid/benzodiapzepine receptor complex from bovine cerebral cortex. J Biol Chem 258: 6965–6971
  • Siegwart, R., Jurd, R., Rudolph, U. (2002) Molecular determinants for the action of general anesthetics at recombinant alpha(2)beta(3)gamma(2)gamma-aminobutyric acidA receptors. J Neurochem 80: 140-148
  • Sinkkonen, S. T., Hanna, M, C., Kirkness, E. F., Korpi, E. R. (2000)GABAA receptor epsilon and theta subunits display unusual structural variation between species and are enriched in the rat locus ceruleus. J Neurosci 20: 3588-3595
  • Sivilotti, L., and Nistri, A. (1991) GABA receptor mechanisms in the central nervous system. Prog Neurobiol 36: 35–92
  • Smit, A. B., Brejc, K., Syed, N., Sixma, T. K. (2003) Structure and function of AChBP, homologue of the ligand-binding domain of the nicotinic acetylcholine receptor. Ann N Y Acad Sci 998: 81-92
  • Smith, G. B., Olsen, R. W. (2000) Deduction of amino acid residues in the GABAA receptor alpha subunits photoaffinity labeled with the benzodiazepine flunitrazepam. Neuropharmacology 39: 55-64
  • Smith, S. S., Gong, Q. H., Li, X., Moran, M. H., Bitran, D., Frye, C. A., Hsu, F. C. (1998) Withdrawal from 3alpha-OH-5alpha-pregnan-20-One using a pseudopregnancy model alters the kinetics of hippocampal GABAA-gated current and increases the GABAA receptor alpha4 subunit in association with increased anxiety. J Neurosci 18: 5275-5284
  • Smith, G. B., and Olsen, R. W. (1995) Functional domains of GABAA receptors. Trends Pharmacol Sci 16: 162–168
  • Smith, G. B., Olsen, R. W. (1994) Identification of a [3H]muscimol photoaffinity substrate in the bovine gamma-aminobutyric acidA receptor alpha subunit. J Biol Chem 269: 20380-20387
  • Somogyi, P., Fritschy, J.M., Benke, D., Roberts, J.D.B., Sieghart, W., (1996) The γ2 subunit of the GABAA receptor is concentrated in synaptic junctions containing the α1 and β2/3 subunits in hippocampus, cerebellum, and globus pallidus. Neuropharmacol 35: 1425-1444
  • Sperk, G., Schwarzer, C., Tsunashima, K., Fuchs, K., Sieghart, W. (1997) GABAA receptor subunits in the rat hippocampus I: immunocytochemical distribution of 13 subunits. Neuroscience 80: 987-1000
  • Spigelman, I., Li, Z., Liang, J., Cagetti, E., Samzadeh, S., Mihalek, R. M., Homanics, G. E., Olsen, R. W. (2003) Reduced inhibition and sensitivity to neurosteroids in hippocampus of mice lacking the GABAA receptor delta subunit. J Neurophysiol 90: 903-910
  • Spigelman, I., Li, Z., Banerjee, P. K., Mihalek, R. M., Homanics, G. E., Olsen, R. W. (2002) Behavior and physiology of mice lacking the GABAA-receptor delta subunit. Epilepsia 43: 3-8
  • Squires, R. F., Saederup, E. (1988) Antidepressants and metabolites that block GABAA receptors coupled to [35S] t-butylbicyclophosphorothionate binding sites in rat brain. Brain Res 441: 15–22
  • Squires, R. F., Saederup, E. (1987) GABAA receptor blockers reverse the inhibitory effect of GABA on brain-specific [35S]TBPS binding. Brain Res 414: 357-364
  • Squires, R. F., Casida, J. E., Richardson, M., Saederup, E. (1983) [35S] t-butylbicyclophosphoro-thionate binds with high affinity to brain-specific sites coupled to gamma-aminobutyric acid-A and ion recognition sites. Mol Pharmacol 23: 326-336
  • Srinivasan, S., Nichols, C. J., Lawless, G. M., Olsen, R. W., Tobin, A. J. (1999) Two invariant tryptophans on the alpha1 subunit define domains necessary for GABAA receptor assembly. J Biol Chem 274: 26633-26638
  • Stell, B. M., Brickley, S. G., Tang, C. Y., Farrant, M., Mody, I. (2003) Neuroactive steroids reduce neuronal excitability by selectively enhancing tonic inhibition mediated by delta subunit-containing GABAA receptors. Proc Natl Acad Sci USA 100: 14439-14444
  • Stephenson, F. A., Duggan, M. J., and Pollard, S. (1990) The γ2 subunit is an integral component of the γ-aminobutyric acidA receptor but the alpha1 polypeptide is the principal site of the agonist benzodiazepine photoaffinity labeling reaction. J Biol Chem 265: 21160–21165
  • Strakhova, M. I., Harvey, S. C., Cook, C. M., Cook, J. M., Skolnick, P. (2000) A single amino acid residue on the alpha(5) subunit (Ile215) is essential for ligand selectivity at alpha(5)beta(3)gamma(2) gamma-aminobutyric acidA receptors. Mol Pharmacol 58: 1434-1440
  • Sullivan, D. A., Cohen, J. B. (2000) Mapping the agonist binding site of the nicotinic acetylcholine receptor. Orientation requirements for activation by covalent agonist. J Biol Chem 275: 12651-12660
  • Sundstrom-Poromaa, I., Smith, D. H., Gong, Q. H., Sabado, T. N., Li, X., Light, A., Wiedmann, M., Williams, K., Smith, S. S. (2002) Hormonally regulated alpha(4)beta(2)delta GABAA receptors are a target for alcohol. Nat Neurosci 5: 721-722
  • Supavilai, P., Karobath, M. (1983) Differential modulation of [35S]TBPS binding by the occupancy of benzodiazepine receptors with its ligands. Eur J Pharmacol 91: 145-146
  • Sur, C., Wafford, K. A., Reynolds, D. S., Hadingham, K. L., Bromidge, F., Macaulay, A., Collinson, N., O’Meara, G., Howell, O., Newman, R., Myers, J., Atack, J. R., Dawson, G. R., McKernan, R. M., Whiting, P. J., Rosahl, T. W. (2001). Loss of the major GABAA receptor subtype in the brain is not lethal in mice. J Neurosci 21: 3409–3418
  • Sur, C., Farrar, S. J., Kerby, J., Whiting, P. J., Atack, J. R., McKernan, R. M. (1999) Preferential coassembly of alpha4 and delta subunits of the gamma-aminobutyric acidA receptor in rat thalamus. Mol Pharmacol 56: 110-115
  • Taketo, M., and Yoshioka, T. (2000) Developmental change of GABAA receptor-mediated current in rat hippocampus. Neuroscience 96: 507–514
  • Takeuchi, A., and Onodera, K. (1972) Effect of bicuculline on the GABA receptor of the crayfish neuromuscular junction. Nature New Biol 236: 55-56
  • Takeuchi, A., and Takeuchi, N. (1969) A study of the action of picrotoxin on the inhibitory neuromuscular junction of the crayfish. J Physiol 205: 377-391
  • Takeuchi, A., and Takeuchi, N. (1967) Anion permeability of the inhibitory post-synaptic membrane of the crayfish neuromuscular junction. J Physiol 191: 575-590
  • Taylor, P. M., Connolly, C. N., Kittler, J. T., Gorrie, G. H., Hosie, A., Smart, T. G., Moss, S. J. (2000) Identification of residues within GABAA receptor alpha subunits that mediate specific assembly with receptor beta subunits. J Neurosci 20: 1297-1306
  • Taylor, P. M., Thomas, P., Gorrie, G. H., Connolly, C. N., Smart, T. G., Moss, S. J. (1999) Identification of amino acid residues within GABAA receptor beta subunits that mediate both homomeric and heteromeric receptor expression. J Neurosci 19: 6360-6371
  • Teissere, J. A., Czajkowski, C. A (2001) (beta)-strand in the (gamma)2 subunit lines the benzodiazepine binding site of the GABAA receptor: structural rearrangements detected during channel gating. J Neurosci 21: 4977-4986
  • Thompson, S. A., Wingrove, P. B., Connelly, L., Whiting, P. J., Wafford, K. A. (2002) Tracazolate reveals a novel type of allosteric interaction with recombinant gamma-aminobutyric acidA receptors. Mol Pharmacol 61: 861-869
  • Thompson, S. A., Arden, S. A., Marshall, G., Wingrove, P. B., Whiting, P. J., Wafford, K. A. (1999) Residues in transmembrane domains I and II determine γ-aminobutyric acid type AA receptor subtype-selective antagonism by furosemide. Mol Pharmacol 55: 993–999
  • Tomiko, S. A., Taraskevich, P. S., and Douglas, W. W. (1983) GABA acts directly on cells of pituitary pars intermedia. Nature 301: 706–707
  • Toyoshima, C., and Unwin, N. (1988) Ion channel of acetylcholine receptor reconstructed from images of postsynaptic membranes. Nature 336: 247–250
  • Tretter, V., Hauer, B., Nusser, Z., Mihalek, R. M., Hoger, H., Homanics, G. E., Somogyi, P., Sieghart, W. (2001) Targeted disruption of the GABAA receptor delta subunit gene leads to an up-regulation of gamma 2 subunit-containing receptors in cerebellar granule cells. J Biol Chem 276: 10532-10538
  • Tretter, V., Ehya, N., Fuchs, K., and Sieghart, W. (1997) Stoichiometry and assembly of a recombinant GABAA receptor subtype. J Neurosci 17: 2728–2737
  • Trudell, J. (2002) Unique assignment of inter-subunit association in GABAA alpha1beta3gamma2 receptors determined by molecular modeling. Biochim Biophys Acta 1565: 91–96
  • Tsunashima, K., Schwarzer, C., Kirchmair, E., Sieghart, W., Sperk, G. (1997) GABAA receptor subunits in the rat hippocampus III: altered messenger RNA expression in kainic acid-induced epilepsy. Neuroscience 80: 1019-1032
  • Twyman, R. E., Macdonald, R. L. (1992) Neurosteroid regulation of GABAA receptor single-channel kinetic properties of mouse spinal cord neurons in culture. J Physiol 456: 215–245
  • Ueno, S., Lin, A., Nikolaeva, N., Trudell, J. R., Mihic, S. J., Harris, R. A., Harrison, N. L. (2000) Tryptophan scanning mutagenesis in TM2 of the GABAA receptor alpha subunit: effects on channel gating and regulation by ethanol. Br J Pharmacol 131: 296-302
  • Ueno, S., Wick, M. J., Ye, Q., Harrison, N. L., Harris, R. A. (1999) Subunit mutations affect ethanol actions on GABAA receptors expressed in Xenopus oocytes. Br J Pharmacol 127: 377-382
  • Ueno, S., Bracamontes, J., Zorumski, C., Weiss, D. S., Steinbach, J. H. (1997) Bicuculline and gabazine are allosteric inhibitors of channel opening of the GABAA receptor. J Neurosci 17: 625-634
  • Ugarte, S. D., Homanics, G. E., Firestone, L. L., Hammond, D. L. (2000) Sensory thresholds and the antinociceptive effects of GABA receptor agonists in mice lacking the beta3 subunit of the GABAA receptor. Neuroscience 95: 795-806
  • Unwin, N. (2003) Structure and action of the nicotinic acetylcholine receptor explored by electron microscopy. FEBS Lett 555: 91-95
  • Unwin, N. (1993) Neurotransmitter action: Opening of ligand-gated ion channels. Cell 72(Suppl): 31–41
  • Vaught, K. A., and Wauquier, A. (1991) Evidance for a unique interaction of loreclezole with the GABA receptor complex. Drug Dev Res 23: 181-189
  • Verdoorn, T. A. (1994) Formation of heteromeric γ-aminobutyric acid type A receptors containing two different α subunits. Mol Pharmacol 45: 475–480
  • Verdoorn, T. A., Draguhn, A., Ymer, S., Seeburg, P. H., Sakmann, B. (1990) Functional properties of recombinant rat GABAA receptors depend upon subunit composition. Neuron 4: 919-928
  • Verli, H., Albuquerque, M. G., Bicca de Alencastro, R., Barreiro, E. J. (2002) Local intersection volume: a new 3D descriptor applied to develop a 3D-QSAR pharmacophore model for benzodiazepine receptor ligands. Eur J Med Chem 37: 219-229
  • Vicini, S., Losi, G., Homanics, G. E. (2002) GABAA receptor delta subunit deletion prevents neurosteroid modulation of inhibitory synaptic currents in cerebellar neurons. Neuropharmacology 43: 646-650
  • Vicini, S., Ferguson, C., Prybylowski, K., Kralic, J., Morrow, A. L., Homanics, G. E. (2001) GABAA receptor alpha1 subunit deletion prevents developmental changes of inhibitory synaptic currents in cerebellar neurons. J Neurosci 21: 3009-3016
  • Villar, H. O., Uyeno, E. T., Toll, L., Polgar, W., Davies, M. F., and Loew, G. H. (1989) Molecular determinants of benzodiazepine receptor affinities and anticonvulsant activities. Mol Pharmacol 36: 589–600
  • Wafford, K. A., Bain, C. J., Quirk, K., McKernan, R. M., Wingrove, P. B., Whiting, P. J., and Kemp, J. A. (1994) A novel allosteric modulatory site on the GABAA receptor β subunit. Neuron 12: 775–782
  • Wagner, D. A., Czajkowski, C. (2001) Structure and dynamics of the GABA binding pocket: A narrowing cleft that constricts during activation. J Neurosci 21: 67-74
  • Wallner, M., Hanchar, H. J., Olsen, R. W. (2003) Ethanol enhances alpha 4 beta 3 delta and alpha 6 beta 3 delta gamma-aminobutyric acid type A receptors at low concentrations known to affect humans. Proc Natl Acad Sci USA 100: 15218-15223
  • Walters, R. J., Hadley, S. H., Morris, K. D. W., and Amin, J. (2000) Benzodiazepines act upon GABAA receptors via two distinct and separable mechanisms. Nat Neurosci 3: 1274-1281
  • Wang, T. L., Guggino, W. B., and Cutting, G. R. (1994) A novel γ-aminobutyric acid receptor subunit (rho2) cloned from human retina forms bicuculline-insensitive homooligomeric receptors in Xenopus oocytes. J Neurosci 14: 6524–6531
  • Wegelius, K., Pasternack, M., Hiltunen, J. O., River, C., Kaila, K., Saarma, M., Reeben, M. (1998) Distribution of GABA receptor rho subunit transcripts in the rat brain. Eur J. Neurosci 10: 350-357
  • Westh-Hansen, S. E., Witt, M. R., Dekermendjian, K., Liljefors, T., Rasmussen, P. B., Nielsen, M. (1999) Arginine residue 120 of the human GABAA receptor alpha 1, subunit is essential for GABA binding and chloride ion current gating. Neuroreport 10: 2417-2421
  • Westh-Hansen, S. E., Rasmussen, P. B., Hastrup, S., Nabekura, J., Noguchi, K., Akaike, N., Witt, M. R., Nielsen, M. (1997) Decreased agonist sensitivity of human GABAA receptors by an amino acid variant, isoleucine to valine, in the α1 subunit. Eur J Pharmacol 329: 253–257
  • Whiting, P. J., McAllister, G., Vassilatis, D., Bonnert, T. P., Heavens, R. P., Smith, D. W., Hewson, L., O’Donnell, R., Rigby, M. R., Sirinathsinghji, D. J. S., Marshall, G., Thompson, S. A., and Wafford, K. A. (1997) Neuronally restricted RNA splicing regulates the expression of a novel GABAA receptor subunit conferring atypical functional properties. J Neurosci 17: 5027–5037
  • Whiting, P., McKernan, R. M., and Iversen, L. L. (1990) Another mechanism for creating diversity in gamma-aminobutyrate type A receptors: RNA splicing directs expression of two forms of gamma 2 subunit, one of which contains a protein kinase C phosphorylation site. Proc Natl Acad Sci USA 87: 9966–9970
  • Wick, M. J., Mihic, S. J., Ueno, S., Mascia, M. P., Trudell, J. R., Brozowski, S. J., Ye, Q., Harrison, N. L., Harris, R. A. (1998) Mutations of gamma-aminobutyric acid and glycine receptors change alcohol cutoff: evidence for an alcohol receptor? Proc Natl Acad Sci USA 95: 6504-6509
  • Wieland, H. A., and Luddens, H. (1994) Four amino acid exchanges convert a diazepam-insensitive, inverse agonist-preferring GABAA receptor into a diazepam-preferring GABAA receptor. J Med Chem 37: 4576–4580
  • Wieland, H. A., Luddens, H., and Seeburg, P. H. (1992) A single histidine in GABAA receptors is essential for benzodiazepine agonist binding. J Biol Chem 267: 1426–1429
  • Williams, K. L., Tucker, J. B., White, G., Weiss, D. S., Ferrendelli, J. A., Covey, D. F., Krause, J. E., Rothman, S. M. (1997) Lactone modulation of the gamma-aminobutyric acid A receptor: evidence for a positive modulatory site. Mol Pharmacol 52: 114-119
  • Wingrove, P. B., Safo, P., Wheat, L., Thompson, S. A., Wafford, K. A., Whiting, P. J. (2002) Mechanism of alpha-subunit selectivity of benzodiazepine pharmacology at gamma- aminobutyric acid type A receptors. Eur J Pharmacol 437: 31-39
  • Wingrove, P. B., Thompson, S. A., Wafford, K. A., and Whiting, P. J. (1997) Key amino acids in the γ subunit of the γ-aminobutyric acidA receptor that determine ligand binding and modulation at the benzodiazepine site. Mol Pharmacol 52: 874–881
  • Wingrove, P. B., Wafford, K. A., Bain, C., Whiting, P. J. (1994) The modulatory action of loreclezole at the γ-aminobutyric acid type A receptor is determined by a single amino acid in the β2 and β3 subunit. Proc Natl Acad Sci USA 91: 4569 –573
  • Wisden, W., Herb, A., Wieland, H., Keinanen, K., Luddens, H., and Seeburg, P. (1991) Cloning, pharmacological characteristics and expression pattern of the rat GABAA receptor α4 subunit. FEBS Lett 289: 227–230
  • Wisden, W., and Seeburg, P. H. (1992) GABAA receptor channels: From subunits to functional entities. Curr Opin Neurobiol 2: 263–269
  • Wohlfarth, K. M., Bianchi, M. T., Macdonald, R. L. (2002) Enhanced neurosteroid potentiation of ternary GABAA receptors containing the delta subunit. J Neurosci 22: 1541-1549
  • Wong, S. M., Cheng, G., Homanics, G. E., Kendig, J. J. (2001) Enflurane actions on spinal cords from mice that lack the beta3 subunit of the GABAA receptor. Anesthesiology 95: 154-164
  • Wooltorton, J.R., McDonald, B.J., Moss, S.J., and Smart, T.G. (1997a) Identification of a Zn2+ binding site on the murine GABAA receptor complex: dependence on the second transmembrane domain of β subunits. J Physiol 505: 633–640
  • Wooltorton, J. R., Moss, S. J., Smart, T. G. (1997b) Pharmacological and physiological characterization of murine homomeric beta3 GABAA receptors. Eur J Neurosci 9: 2225-2235
  • Xu, M. and Akabas, M. H. (1996) Identification of channel-lining residues in the M2 membrane-spanning segment of the GABAA receptor alpha1 subunit. J Gen Physiol 107:195–205
  • Xu, M., Covey, D. F., Akabas, M. H. (1995) Interaction of picrotoxin with GABAA receptor channel-lining residues probed in cysteine mutants. Biophys J 69: 1858-1867
  • Yang, W., Drewe, J. A., and Lan, N. C. (1995) Cloning and characterization of the human GABAA receptor α4 subunit: Identification of a unique diazepam-insensitive binding site. Eur J Pharmacol 209: 319–325
  • Ymer, S., Draguhn, A., Wisden, W., Werner, P., Keinanen, K., Schofield, P. R., Sprengel, R., Pritchett, D. B., Seeburg, P. H. (1990) Structural and functional characterization of the γ1-subunit of GABAA benzodiazepine receptors. EMBO J 9: 3261–3267
  • Yoon, K. W., Covey, D. F., Rothman, S. M. (1993) Multiple mechanisms of picrotoxin block of GABA-induced currents in rat hippocampal neurons. J Physiol 464: 423-439
  • Zaman, S. H., Shingai, R., Harvey, R. J., Darlinson, M. G., and Barnard, E. A. (1992) Effects of subunit types of the recombinant GABAA receptor on the response to a neurosteroid. European Journal of Pharmacology 225: 321–330
  • Zhang, D., Pan, Z. H., Awobuluyi, M., Lipton, S. A. (2001) Structure and function of GABA(C) receptors: a comparison of native versus recombinant receptors. Trends Pharmacol Sci 22: 121-132
  • Zhang, W., Koehler, K. F., Zhang, P., and Cook, J. M. (1995b) Development of a comprehensive pharmacophore model for the benzodiazepine receptor. Drug Design Dis 12: 192–248
  • Zhu, J. J., Lo, F. S. (1999) Three GABA receptor-mediated postsynaptic potentials in interneurons in the rat lateral geniculate nucleus. J Neurosci 19: 5721-5730
  • Zhu, W. J., Vicini, S. (1997) Neurosteroid prolongs GABAA channel deactivation by altering kinetics of desensitized states. J Neurosci 17: 4022-4031
  • Zimprich, F., Zezula, J., Sieghart, W., Lassmann, H. (1991) Immunohistochemical localization of the alpha1, alpha2 and alpha3 subunit of the GABAA receptor in the rat brain. Neurosci Lett 127: 125-128

External links[edit]