Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2007 Sep;5(3):195–201. doi: 10.2174/157015907781695919

Allosteric Modulators of GABAB Receptors: Mechanism of Action and Therapeutic Perspective

Jean-Philippe Pin 1,*, Laurent Prézeau 1
PMCID: PMC2656813  PMID: 19305802

Abstract

γ-aminobutyric acid (GABA) plays important roles in the central nervous system, acting as a neurotransmitter on both ionotropic ligand-gated Cl--channels, and metabotropic G-protein coupled receptors (GPCRs). These two types of receptors called GABAA (and C) and GABAB are the targets of major therapeutic drugs such as the anxiolytic benzodiazepines, and antispastic drug baclofen (lioresal®), respectively. Although the multiplicity of GABAA receptors offer a number of possibilities to discover new and more selective drugs, the molecular characterization of the GABAB receptor revealed a unique, though complex, heterodimeric GPCR. High throughput screening strategies carried out in pharmaceutical industries, helped identifying new compounds positively modulating the activity of the GABAB receptor. These molecules, almost devoid of apparent activity when applied alone, greatly enhance both the potency and efficacy of GABAB agonists. As such, in contrast to baclofen that constantly activates the receptor everywhere in the brain, these positive allosteric modulators induce a large increase in GABAB-mediated responses only WHERE and WHEN physiologically needed. Such compounds are then well adapted to help GABA to activate its GABAB receptors, like benzodiazepines favor GABAA receptor activation. In this review, the way of action of these molecules will be presented in light of our actual knowledge of the activation mechanism of the GABAB receptor. We will then show that, as expected, these molecules have more pronounced in vivo responses and less side effects than pure agonists, offering new potential therapeutic applications for this new class of GABAB ligands.

Key Words: Baclofen, anxiety, drug addiction, allosteric modulators, class C GPCRs.

INTRODUCTION

As one of the major neurotransmitters in the brain, γ -amino-butyric acid (GABA) plays critical roles in brain development and physiology. By activating GABAA receptors, which are Cl--gated channels, this neurotransmitter prevents neuronal depolarization, and as such controls the transmission of excitatory signals. In young animals, these GABAA receptors generate instead excitatory responses, and replace the glutamatergic system not yet fully established. Controlling GABAA receptor activity soon appeared as an interesting way for the treatment of brain dysfunction. This led to the discovery of benzodiazepines that allosterically enhance GABAA receptor activation by acting at a site distinct from the GABA binding site. These positive modulators act by increasing GABA affinity and potency, and by facilitating Cl--channel opening, and are widely used for the treatment of insomnia, anxiety and epilepsies.

GABA also acts on G protein-coupled GABAB receptors [6]. These receptors limit neurotransmitter release at many synapses, including most GABAergic and glutamatergic ones, by inhibiting at least Ca2+-channel opening. They are also located in post-synaptic elements where they activate G protein-regulated inward-rectifying K+-channels (GIRK channels) [42]. These receptors were pharmacologically identified in the early 80's, being selectively activated by baclofen (β p-chlorophenyl-GABA) [31], a molecule that is used for the treatment of spasticity in multiple sclerosis patients due to its muscle-relaxant properties [9]. The GABAB receptors are also responsible for most effects of the drug of abuse gamma-hydroxybutyrate (GHB) that acts as a GABAB partial agonist at high doses [34, 48]. GABAB agonists also demonstrated a number of beneficial effects both in animals and in humans [5]. Indeed, activation of GABAB receptors exerts analgesic/antinociceptive effects in animal models of chronic inflammation and neuropathy (see [5]), suppresses drug seeking behavior [15], and has some anxiolytic activity both in animal models and in human [17]. However, undesired side effects such as hypothermic and sedative effects, greatly limits the use of GABAB agonists in therapeutics [5]. Moreover, tolerance to baclofen chronic treatment is well established [44]. In addition to agonists, GABAB antagonists were also shown to have potential therapeutic effects, such as antidepressant activity [17], cognition improvement [22], and beneficial effects in rat models of absence epilepsy [53].

GABAB receptors have therefore been used as a target in high throughput screening strategies with the aim at identifying new ligands acting at this receptor. As already well documented for the related metabotropic glutamate (mGlu) receptors [26, 28], this strategy leads to the discovery of allosteric modulators acting at the GABAB receptor [76, 77]. In contrast to mGluRs for which both positive and negative (non-competitive antagonists) were identified, only positive allosteric modulators (PAMs) have been described so far for the GABAB receptor. These compounds display no or very partial agonist activity, but enhance both the potency and efficacy of GABAB agonists. As such, these molecules appear as a better alternative to GABAB agonists, allowing the specific enhancement of GABAB receptor activity when and where needed, and as such, are less prone to tolerance in contrast to the pure agonists that constantly activate the receptor in any region where it is expressed.

In the present chapter, we aimed at describing the mechanism of action of the identified allosteric modulators of the GABAB receptor. We will first describe our current knowledge of the functioning of this complex receptor (for reviews see [2, 65]). We will then highlight the potential new therapeutic possibilities offered by these molecules, as based on the recent preclinical studies reported in the literature.

STRUCTURE AND ACTIVATION MECHANISM OF THE GABAB RECEPTOR

The GABAB receptor is part of the class C of GPCRs that also includes the mGlu, the Ca2+-sensing, and the sweet and umami taste receptors among others [64]. These receptors are dimers, either homodimers linked by a disulphide bond (mGlu and Ca2+-sensing receptors), or heterodimers made of two similar, but distinct subunits (the GABAB and taste receptors). Indeed, the GABAB receptor was the first GPCR to be identified that requires two distinct subunits to function: the GABAB1 and GABAB2 subunits [33, 36, 79] (Fig. 1). Although the GABAB1 subunit was soon shown to bind all known GABAB ligands (both agonists and antagonists), this protein did not form a functional GABAB receptor when expressed alone [35]. Only when GABAB1 was co-expressed with the homologous GABAB2 subunit was a functional GABAB receptor observed, either in cell lines or in cultured neurons. The GABAB dimeric entity was confirmed in native tissue [36]. Indeed, both GABAB1 and GABAB2 mRNAs are co-localized in most brain regions. Second, both proteins are found in the same neurons, even in the same subcellular compartments as observed at the electron microscopic level. Moreover, co-immunoprecipitation of GABAB1 with a GABAB2 antibody could be demonstrated from brain membranes. Eventually, mice lacking either GABAB1 or GABAB2 share very similar phenotypes, and none of the known GABAB-mediated responses could be measured in either mice [66, 69]. Although unusual baclofen-mediated inhibition of GIRK channels could be observed in mice lacking GABAB2, it is still not known whether this represents a natural response, or is the consequence of the absence of the GABAB2 subunit. Taken together, these data demonstrate that the assembly between these two proteins is required to get a functional GABAB receptor in native tissues.

Fig. (1).

Fig. (1)

Expected structure of the heterodimeric GABAB receptor in its inactive (left) and active (right) states. This receptor is made of two homologous subunits, GABAB1 (in the front, in black) and GABAB2 (in the back, in light grey). Each subunit is made of two main domains, the extracellular Venus Flytrap domain (VFT) and the heptahelical domain (HD). GABA and other orthosteric GABAB ligands are known to bind in the GABAB1 VFT. No known ligand bind at the equivalent site in GABAB2. Only the GABAB2 HD appears to be responsible for G-protein coupling. These images were made using the coordinates of the dimer of mGlu1 VFTs in the inactive empty state (left) and those of the active Glu occupied state (right) [43], in association with a dimer of HD generated based on the proposed model of the dimer of rhodopsin [47].

When expressed alone, the GABAB1 subunit is mostly retained in the endoplasmic reticulum (ER), both in transfected cell lines and in neurons [16]. This is due to the presence of an intracellular retention signal (RXR) located in its intracellular tail that constitutes a binding site for the coat protein-I complex (COPI) [8, 10, 54, 61]. COPI is known to target back to the ER proteins that reached the cis-Golgi, therefore preventing their trafficking through the Golgi and their targeting to the cell surface. COPI binding to the RXR motif of GABAB1 is prevented by GABAB2 thanks to a direct interaction between the intracellular tails of these two subunits through a coiled-coil interaction [8, 10, 54, 61]. Such a system is assumed to control the trafficking to the cell surface of correctly assembled GABAB heterodimers.

Each GABAB receptor subunit is made of two main domains: a large extracellular domain structurally similar to bacterial periplasmic amino-acid binding proteins often called a Venus Flytrap domain (VFT) [24], linked to a 7 transmembrane domain (the heptahelical domain (HD)) typical of all GPCRs (Fig. 1). Besides these common features, most class C GPCRs, except the GABAB receptor subunits, possess a cystein-rich domain that interconnects, both physically and functionally, the VFT and the HD in mGlu receptors [68]. A third domain composed of two short consensus repeats, also known as Sushi domains, is found at the N-terminus of the GABAB1a splice variant but in the GABAB1b variant, [30, 35]. These Sushi domains are responsible for the specific targeting of the GABAB1a receptor in nerve terminals of glutamatergic neurons [78].

The VFT domain of class C GPCRs contains the binding site for agonists and competitive antagonists (the orthosteric ligands). In the case of the GABAB receptor, GABA and all other orthosteric ligands bind to the GABAB1 VFT only, notably by interacting with Ser246 and Glu465 (nomenclature based on the rat GABAB1a sequence) [24, 39, 52]. Indeed, mutational and evolution analyses of the GABAB2 VFT suggest that no natural ligand binds in this domain [39].

Although GABA binds in the GABAB1 VFT, it is now well demonstrated that the GABAB2 HD is responsible for G-protein activation (Fig. 1). Indeed, a mutated receptor dimer with two GABAB2 HDs is functional, whereas a mutated receptor with two GABAB1 HDs does not [23]. Moreover, mutations in either the second or third intracellular loop of GABAB2 suppress G-protein activation whereas the equivalent mutations in GABAB1 do not [21, 29, 67]. Finally, a recent study identified an Arg residue at the bottom of TM3 conserved in most class C GPCRs that plays a critical role in G-protein activation [4]. This Arg may possibly play a role similar to that of the conserved D/ERY motif of class A GPCRs. Of interest, this Arg is found in the GABAB2, but not in the GABAB1, further highlighting the pivotal role played by GABAB2 in G-protein activation.

How can agonist binding in GABAB1 VFT activate the GABAB2 HD? Much information to answer that question came from the solved crystal structure of the mGlu1 VFT dimer with and without bound agonist or antagonist [43, 73]. These structures revealed that agonist binding in the VFT stabilizes a closed conformation that is also associated with a major change in the relative orientation of the two VFTs in the dimer (Fig. 1). This relative movement is expected to induce a relative movement of the HDs, a proposal that is consistent with FRET studies [72]. Of interest, although both HDs in a mGlu homodimer are identical, this process leads to the active state of only one of them [32], likely because a single G-protein can interact at a time with such dimeric entities [19]. This model perfectly fits with all mutational analysis of GABAB receptor functioning. Indeed, the closure of the GABAB1 VFT has been shown to be responsible for GABAB receptor activation [40], and such a closure activates GABAB2 HD whether it is part of the associated subunit (like in the wild-type heterodimer) or linked to the GABAB1 VFT [23]. Moreover, point mutations introduced into either the GABAB1 VFT or the GABAB2 HD were found to increase constitutive activity of this receptor, consistent with these two domains playing a critical role in receptor activation [56].

In summary, the GABAB receptor is a complex allosteric protein made or four main domains working "de concert" to allow GABA binding in the VFT of one subunit (GABAB1) to activate the HD of the associated subunit (GABAB2), likely through relative movement between these domains (Fig. 1). As we will see now, such a complex structure offers a number of possibilities to modulate GABAB receptor function.

ALLOSTERIC MODULATORS OF THE GABABRECEPTOR: PROPERTIES AND MECHANISM OF ACTION

Early studies following the molecular characterization of the GABAB receptor heterodimer indicated that Ca2+ ions act as enhancers of this receptor [80]. Indeed, few hundred micromolar of Ca2+ increased the potency of GABA in stimulating GTPγ S binding or G-protein activation measured in second messenger assays [25]. This effect is observed both with the recombinant and the native receptor [25], even in post-morten human tissues [58], and results from a direct increase in GABA affinity. Of interest, this effect of Ca2+ was not observed with baclofen, suggesting that the chlorophenyl group of baclofen prevents the action of Ca2+ ions, pointing to the possibility that Ca2+ directly binds within the GABA binding site in the GABAB1 VFT. This was further validated using site directed and 3D modeling studies [25]. According to the expected physiological Ca2+ concentration range, the GABAB receptor is expected to be always potentiated under physiological condition. Only under pathological conditions, when the extracellular Ca2+ concentration reaches values as low as few micromolar, can this effect disappear. Whatever, these data revealed that it is possible to positively modulate GABAB receptor function with small molecules.

Few years before this observation, a number of allosteric modulators of the other class C GPCRs, and especially mGlu receptors, were identified, including both negative and positive allosteric modulators [26, 28] (see this issue). The negative modulators first identified for mGlu1 and mGlu5, were found to inhibit in a non-competitive manner the activity of the receptors, and to display in most cases inverse agonist activity [12, 62]. In contrast, PAMs were found to have no, or weak agonist activity when applied alone, but to greatly enhance both the potency and the efficacy of agonists [41, 57]. Both types of modulators were found to bind in a cavity within the HD, contacting residues of TM3, TM5 TM6 and TM7, therefore at a site clearly distinct from the glutamate binding site located in the VFT. Residues that constitute this binding site differ between receptor subtypes, such that most modulators identified so far, either positive or negative, were found to be highly subtype selective, in contrast to the orthosteric ligands that usually do not discriminate between mGlu receptors from the same group [26, 28].

Taken together, these data indicated that compounds interacting in the HD of class C GPCRs could allosterically modulate their activity, and these compounds had three main advantages: 1) original chemical structures, different from that of the orthosteric ligands, usually poly-cyclic with a good bioavailability, more prone to chemical modifications, and in agreement with the Lipinski's rules for drug-likelyness; 2) much higher selectivity among related sequences; and 3) a good respect of the biological activity of the receptors, especially for the PAMs that facilitate agonist action, and therefore enhance receptor activation when and where needed physiologically.

These observations lead a number of pharmaceutical companies to search for new GABAB modulators using high throughput functional assays. So far, only 2 PAMs have been reported in the literature (2,6-Di-tert.-butyl-4-(3-hydroxy-2,2-dimethyl-propyl)-phenol (CGP7930) and N,N'-Dicyclopentyl-2-methylsulfanyl-5-nitro-pyrimidine-4,6-diamine (GS39783) and some of their derivatives) [76, 77] (Fig. 2), and some others have been reported in patents [49-51]. Aryl-alkylamine (such as fendiline), amino acids like phenylalanine, leucine and isoleucine as well as dipeptides have also been shown to enhance GABAB receptor activity in brain slices [14, 37, 38]. However, others reported that fendiline inhibits, rather than mimick, the effect of CGP7930 [58], and Urwyller and colleagues show that the effect of Aryl-alkylamine and amino acids are rather indirect, and do not result from a direct PAM action on the receptor itself [74].

Fig. (2).

Fig. (2)

Structure of the two PAMs identified for the GABAB receptor.

CGP7930 and GS39783 were found to enhance agonist potency as well as efficacy on recombinant GABAB receptors in various assays (Fig. 3), on both human and rat receptors [20, 76, 77]. GS39783 was also shown to be active on fish and chicken receptors, but not on the Drosophila one [20], and CGP7930 enhances GABA affinity on the bullfrog receptor [1], demonstrating a good conservation of the allosteric site in vertebrates. In the GTPγ S binding assay CGP7930 and GS39783 increase GABA potency by 5-10 fold, and increase the maximal effect from 1.5 to up to 2 fold, with potencies ranging from 3 to 5 µM, depending on the agonist concentration. The same positive allosteric effect was also observed when coupling of the GABAB receptor to GIRK channels was measured in Xenopus oocytes [76, 77], or when the coupling of the receptor to phospholipase C was made possible with recombinant chimeric Gqi/o proteins [3, 76]. Very similar enhancing effects were observed with all three well known GABAB receptor agonists, GABA, baclofen and APPA. Of interest, the PAMs largely increased the efficacy of partial agonists like CGP47656 to make it a full agonist, with a similar maximal effect as that of GABA. Moreover, among 7 competitive antagonists, two (CGP35348 and 2-OH-saclofen) became partial agonists [75].

Fig. (3).

Fig. (3)

The PAMs increase both the potency and efficacy of GABA on the GABAB receptor. Data were obtained from membranes prepared from HEK 293 cells expressing GABAB1, GABAB2 and the Gαo proteins. GTPγS binding was measured in the presence of the indicated concentration of GABA with (open triangles) or without (closed squares) 100 µM CGP7930. This figure is adapted from [3].

CGP7930 and GS39783 increased agonist affinity as measured with [3H]-APPA or through the displacement of radio-labeled antagonists [75-77]. However, the increase in affinity (2 fold) is lower than the measured increase in potency. In agreement with the allosteric potentiator further stabilizing the closed state of the GB1 VFT, a decrease in both the ON and OFF binding rates of agonists was observed, as well as a slight decrease in the affinity of most antagonists [63]. Only the affinities of the antagonists CGP35348 and 2-OH-saclofen that became partial agonists in the presence of the PAMs, were increased [75].

The two identified GABAB PAMs show no or only slight agonist activity when applied alone in most assays, both in recombinant systems, and in native preparations [20, 59, 76, 77]. However, partial agonist activity of CGP7930 could be observed when IP production was measured in HEK293 cells co-expressing the GABAB receptor and the chimeric G-protein Gqi9 [3]. This was not the consequence of endogenous agonists in the preparation since the competitive antagonists could not fully inhibit the effect of CGP7930. Although this likely results from the over-expression of the receptor and/or its coupling to non natural G-proteins, these data show that CGP7930 acts by stabilizing the receptor in its active state, an effect that is greatly favored in the presence of agonist. In agreement with this proposal, point mutations in the GABAB2 subunit were found to convert GS39783 from a pure PAM into a partial agonist, even though these mutations did not appear to generate a constitutively active receptor [20].

To identify the mode of action of CGP7930, Binet and colleagues studied its effect on various combinations of wild-type and chimeric GABAB subunits, and took advantage of the agonist activity of this molecule in their assay [3]. This study revealed that the GABAB2 HD was required and sufficient for CGP7930 action. Indeed, CGP7930 was found to activate GABAB2 subunit expressed alone, as well as a truncated version of this subunit corresponding to the HD only. Dupuis and colleagues make use of the absence of effect of GS39783 on the Drosophila GABAB receptor to identify its mechanism of action using chimeric drosophila/rat subunits [20]. They also bring further evidence for GS39783 acting in the HD of GABAB2. These authors also tried to identify the residues within the GABAB2 HD that interact with GS39783. Although no such residues were identified, mutations in TM6 were found to convert the modulator into an agonist, suggesting that the mutated residues are involved in stabilizing the GABAB2 HD into its inactive conformation. Residues of the extracellular side of TM7 were also found to decrease GS39783 efficacy, but not its potency, suggesting that these residues are involved in the allosteric coupling between the HD and the VFT in the GABAB receptor. It is quite surprising that among the large number of mutants generated, none affected GS39783 potency. More work is therefore needed to better understand the mode of action of GABAB PAMs at the atomic level.

In summary, GABAB receptor activation is due to the closure of the GABAB1 VFT, that likely results in a relative movement of one subunit compare to the other. This new conformation of the heterodimer stabilizes the active conformation of the GABAB2 HD that promote the GDP-GTP exchange in the associated G-protein (Fig. 4). As such there are two possibilities to enhance receptor activity. By further stabilizing the close state of GABAB1 VFT, as likely does Ca2+, or stabilizing the active conformation of GABAB2 HD as do CGP7930 and GS39783 (Fig. 4). In the absence of agonist, these later compounds may still bind in the GABAB2 HD, but may not lead to the relative movement between the subunits, preventing them from being agonists (Fig. 4).

Fig. (4).

Fig. (4)

Schematic view of the mechanism of action of GABAB PAMs as based on the proposed activation mechanism of this heterodimeric receptor. By stabilizing the closed state of the GABAB1 VFT (dark grey), Ca2+ increases GABA affinity and potency. By stabilizing the active conformation of the GABAB2 HD, small molecule PAMs increase both the potency and efficacy of agonists. The absence of agonist activity of these molecule may be due to their difficulty in promoting the relative movement between the subunits, a change that is proposed to play a critical role in receptor activation.

IN VIVO EFFECT OF POSITIVE ALLOSTERIC MODULATORS (PAMS)

Action of GABAB PAMs on Native Receptors

Soon after their identification and characterization on recombinant GABAB receptors, the PAMs were shown to be effective on native receptors. This is nicely illustrated with the increase in agonist affinity and the potentiation of baclofen stimulated GTPγS binding by both CGP7930 and GS39783 in rat cortical membranes [75-77], as well as in human fontal cortex membranes [58]. Measurement of either the inhibition or stimulation of cAMP formation in native brain membranes and in vivo also confirmed the PAM activity of these two compounds at the native GABAB receptor [27, 59]. When examined in brain slices, these compounds potentiated GABAB receptor action on synaptic transmission. GS39783 suppresses the paired pulse inhibition of population spikes recorded on hippocampal CA1 pyramidal cells, an effect that likely results from the potentiation of the action of ambiant GABA at pre-synaptic GABAB receptors located on GABAergic terminals [77]. The other GABAB enhancer CGP7930 enhances baclofen-induced depression of dopaminergic neurons in the ventral tegmental area [14] and the GABAergic synaptic transmission in the CA1 area of the hippocampus [13]. Surprisingly, no significant effect on excitatory synaptic transmission in hippocampal CA1 network was observed [13] with CGP7930. It is proposed that this may result from a differential effect of this enhancer on the autoreceptors located in GABAergic terminals, and the heteroreceptors located in glutamatergic terminals. Although GABAB1a and GABAB1b splice variants have been shown to be differentially distributed in these two types of terminals [78], CGP7930 was found to be equally active on both recombinant receptors. Further studies are therefore required to clarify this issue.

Most importantly, both CGP7930 and GS39783 were found to pass the blood brain barrier when injected i.p. (or even when given orally in the case of GS39783) allowing the examination of their behavioral effects in vivo. Indeed, GS39783 decreased cAMP formation in vivo in the striatum only when co-administered orally with a threshold concentration of baclofen [27]. In vivo efficacy of CGP7930 was also illustrated by its marked enhancement of the sedative and hypnotic effect of both baclofen and GHB in DBA mice [11]. Due to the original mechanism of action of these PAMs, it was therefore of interest to examine whether such compounds have different effects than the GABAB agonist baclofen.

Differential Effects of PAMs and Agonists

Although baclofen is being used in the treatment of spasticity for multiple sclerosis patients, its myorelaxant, sedative, cognitive and hypothermic effects limit its use in a number of other pathologies. In contrast to baclofen and other GABAB agonists that activate constantly and everywhere the receptor, PAMs are expected to enhance receptor activity only WHEN and WHERE needed physiologically (when and where GABA is produced to act on the GABAB receptor) (Fig. 5). As such, differential effects of PAMs and agonists were expected. Indeed, GS39783 given alone did not display sedative, cognitive, myorelaxant activities [18]. However, sedative effects were reported for CGP7930 at high doses [46]. No effect of GS39783 on body temperature was also observed [18]. This document the general idea that PAMs could be a better alternative to baclofen for the treatment of pathologies in which such side effects are not desired. Of interest, as described in more details bellow, the PAMs display more pronounced anxiolytic effects than GABAB agonists and keep most of the known positive actions of baclofen (Table 1).

Fig. (5).

Fig. (5)

Major difference in the effect of agonists and PAMs acting at the GABAB receptor. Scheme illustrate biological responses resulting from the physiological activity of the GABAB receptor. In plain thick line is the response mediated under control condition. In the presence of a pure agonist, the receptor is always activated, with a decline resulting from the desensitization of the system and tolerance to the drug (dashed thick line). In contrast the PAM does not activate the system unless GABA is released close to the receptor. As such, the PAM enhances the response mediated by physiologically released GABA, enhancing the GABA mediated response, WHEN and WHERE needed (dashed thin line).

Table 1.

Comparison of the Effect and Properties of GABAB Agonists and PAMs

Agonists PAMs Ref.
tolerance yes Not after 3 weeks [44,55]
Body temperature decrease No effect [18]
sedation increase No effect [18]but see [46]
myorelaxation yes No effect [9, 18]
cognition decrease No effect [18]
Anxiety variable decrease [5, 17, 18, 55]
Cocaine self-admin decrease decrease [15, 45, 70, 71]
Alcohol intake decrease decrease [15, 46, 60]

GABAB PAMs as Potential New Anxiolytics

The GABA system is well known to be involved in anxiety, as illustrated by the effect of benzodiazepines. However, the involvement of the GABAB receptor remained elusive for a long time due to the difficulty in assessing the effect of baclofen because of its above mentioned side effects. Anxiolytic effects of baclofen were however observed in some specific tests in rats, and also in humans [5, 17]. The generation of knockout mice deleted of either the GABAB1 or the GABAB2 gene confirmed a role of GABAB receptor in anxiety [17], as illustrated in several tests such as the light-dark box, the elevated plus maze or the elevated zero maze [18, 55]. In these same tests, the GABAB PAM GS39783 show strong anxiolytic activity, in contrast to baclofen [18, 55]. GS39783 was also efficient in reducing stress-induced hyperthermia [18], a test that could not be performed with baclofen due to its hypothermic action. Of most interest, the anxiolytic effect of GS39783 could still be observed after three weeks of treatment, demonstrating an absence of tolerence [55]. Moreover, no synergy with alcohol was observed [17]. As such, GABAB PAMs appear as a new class of anxiolytics that lack the side effects of the commonly used benzodiazepines.

GABAB PAMs for the Treatment of Drug Addiction

The GABAB receptor is known for its role in modulating the reinforcing effect of abused drugs such as cocaine, heroin, alcohol, amphetamine and nicotine [15]. In rats, baclofen decreases self-administration of such drugs, and preclinical studies further indicated the potential of baclofen for the treatment of cocaine, alcohol and nicotine dependence. In support of these effects, baclofen attenuates the activation of limbic regions resulting from cocaine-associated cues as revealed by neuroimaging in humans [7]. However, the use of baclofen as a therapeutic strategy for these indications is limited due to its side effects. The effect of GABAB PAMs on drug dependence and reinforcement has therefore been studied recently as a potential alternative to baclofen.

Both CGP7930 and GS39783 were found to inhibit cocaine self-administration in rats responding to different schedule of reinforcement [71]. Moreover, GS39783 inhibits the reward-facilitating effect of acute cocaine administration, as assessed by the reward threshold in intracranial self-stimulation paradigm [70]. The positive action of GS39783 on cocaine addiction is further supported by the inhibition of most biochemical and behavioral effects of acute and chronic cocaine treatment [45]. These include increased locomotor activity, up regulation of cAMP-response-element-binding protein (CREB) and phosphorylation of dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARP32) in the nucleus accumbens and dorsal striatum [45].

The GABAB PAMs were also shown to have beneficial effects in alcohol consumption in rats. Like baclofen, CGP7930 or GS39783 reduced ethanol drinking behavior in two types of inbred alcohol-preferring rats [46, 60]. Both acquisition and maintenance of alcohol dependence were largely inhibited by PAMs, similarly to baclofen.

These first data reveal that GABAB PAMs represent a novel therapeutic strategy for the treatment of drug addiction, a strategy that will certainly benefit from the anxiolytic activity of these molecules.

CONCLUSION

Although drugs activating the GABAB receptor were found to have a number of possible therapeutic actions, these were limited because of tolerance and undesired side effects which include sedation, myorelaxing activity and hypothermia. By only enhancing the activity of GABAB receptors when and where needed, the GABAB PAMs respect the physiological activity of the receptor (Fig. 5). Not surprisingly, PAMs were found to have different behavioral effects than the pure agonist baclofen. These molecules lack the undesired side effects of baclofen, can be used in long-term treatment without tolerance, display a more pronounced anxiolytic activity, and show similar positive effects as baclofen in drug addiction. These observations make these modulators excellent alternatives to baclofen for a number of therapeutic applications.

These recent findings on the GABAB receptor nicely illustrate the power of allosteric enhancers compared to agonists. After the benzodiazepines acting as PAMs at the GABAA receptors, these data represent certainly the second best example of such a class of compounds. A search for similar molecules acting at other receptors is now open.

REFERENCES

  • 1.Asay MJ, Boyd SK. Characterization of the binding of [3H] CGP54626 to GABAB receptors in the male bullfrog (Rana catesbeiana) Brain Res. 2006;1094:76–85. doi: 10.1016/j.brainres.2006.03.008. [DOI] [PubMed] [Google Scholar]
  • 2.Bettler B, Tiao JY. Molecular diversity, trafficking and subcellular localization of GABAB receptors. Pharmacol. Ther. 2006;110:533–543. doi: 10.1016/j.pharmthera.2006.03.006. [DOI] [PubMed] [Google Scholar]
  • 3.Binet V, Brajon C, Le Corre L, Acher F, Pin JP, Prézeau L. The heptahelical domain of GABAB2 is activated directly by CGP7930, a positive allosteric modulator of the GABAB receptor. J. Biol. Chem. 2004;279:29085–29091. doi: 10.1074/jbc.M400930200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Binet V, Duthey B, Lecaillon J, Vol C, Quoyer J, Labesse G, Pin J-P, Prézeau L. Common structural requirements for heptahelical domain function in class A and class C G protein-coupled receptors. J. Biol. Chem. 2007;282:12154–12163. doi: 10.1074/jbc.M611071200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bowery NG. GABAB receptor: a site of therapeutic benefit. Curr. Opin. Pharmacol. 2006;6:37–43. doi: 10.1016/j.coph.2005.10.002. [DOI] [PubMed] [Google Scholar]
  • 6.Bowery NG, Bettler B, Froestl W, Gallagher JP, Marshall F, Raiteri M, Bonner TI, Enna SJ. International union of pharmacology.XXXIII. mammalian gamma- aminobutyric acid(B) receptors structure and function. Pharmacol. Rev. 2002;54:247–264. doi: 10.1124/pr.54.2.247. [DOI] [PubMed] [Google Scholar]
  • 7.Brebner K, Childress AR, Roberts DC. A potential role for GABA(B) agonists in the treatment of psychostimulant addiction. Alcohol Alcohol. 2002;37:478–484. doi: 10.1093/alcalc/37.5.478. [DOI] [PubMed] [Google Scholar]
  • 8.Brock C, Boudier L, Maurel D, Blahos J, Pin J-P. Assembly-dependent surface targeting of the heterodimeric GABAB receptor is controlled by COPI, but not 14-3-3. Mol. Biol. Cell. 2005;16:5572–5578. doi: 10.1091/mbc.E05-05-0400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Brogden RN, Speight TM, Avery GS. Baclofen a preliminary report of its pharmacological properties and therapeutic efficacy in spasticity. Drugs. 1974;8:1–14. doi: 10.2165/00003495-197408010-00001. [DOI] [PubMed] [Google Scholar]
  • 10.Calver AR, Robbins MJ, Cosio C, Rice SQ, Babbs AJ, Hirst WD, Boyfield I, Wood MD, Russell RB, Price GW, Couve A, Moss SJ, Pangalos MN. The C-terminal domains of the gabab receptor subunits mediate intracellular trafficking but are not required for receptor signaling. J. Neurosci. 2001;21:1203–1210. doi: 10.1523/JNEUROSCI.21-04-01203.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Carai MA, Colombo G, Froestl W, Gessa GL. In vivo effectiveness of CGP7930, a positive allosteric modulator of the GABAB receptor. Eur. J. Pharmacol. 2004;504:213–216. doi: 10.1016/j.ejphar.2004.10.008. [DOI] [PubMed] [Google Scholar]
  • 12.Carroll FY, Stolle A, Beart PM, Voerste A, Brabet I, Mauler F, Joly C, Antonicek H, Bockaert J, Müller T, Pin JP, Prézeau L. BAY36-7620: a potent non-competitive mGlu1 receptor antagonist with inverse agonist activity. Mol. Pharmacol. 2001;59:965–973. [PMC free article] [PubMed] [Google Scholar]
  • 13.Chen Y, Menendez-Roche N, Sher E. Differential modulation by the GABAB receptor allosteric potentiator 2,6-di-tert-butyl-4-(3-hydroxy-2,2-dimethylpropyl)-phenol (CGP7930) of synaptic transmission in the rat hippocampal CA1 area. J. Pharmacol. Exp. Ther. 2006;317:1170–1177. doi: 10.1124/jpet.105.099176. [DOI] [PubMed] [Google Scholar]
  • 14.Chen Y, Phillips K, Minton G, Sher E. GABA(B) receptor modulators potentiate baclofen-induced depression of dopamine neuron activity in the rat ventral tegmental area. Br. J. Pharmacol. 2005;144:926–932. doi: 10.1038/sj.bjp.0706100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cousins MS, Roberts DC, de Wit H. GABA(B) receptor agonists for the treatment of drug addiction a review of recent findings. Drug Alcohol Depend. 2002;65:209–220. doi: 10.1016/s0376-8716(01)00163-6. [DOI] [PubMed] [Google Scholar]
  • 16.Couve A, Filippov AK, Connolly CN, Bettler B, Brown DA, Moss SJ. Intracellular retention of recombinant GABAB receptors. J. Biol. Chem. 1998;273:26361–26367. doi: 10.1074/jbc.273.41.26361. [DOI] [PubMed] [Google Scholar]
  • 17.Cryan JF, Kaupmann K. Don't worry 'B' happy!: a role for GABA(B) receptors in anxiety and depression. Trends Pharmacol. Sci. 2005;26:36–43. doi: 10.1016/j.tips.2004.11.004. [DOI] [PubMed] [Google Scholar]
  • 18.Cryan JF, Kelly PH, Chaperon F, Gentsch C, Mombereau C, Lingenhoehl K, Froestl W, Bettler B, Kaupmann K, Spooren WP. Behavioral characterization of the novel GABAB receptor-positive modulator GS39783 N,N'-dicyclopentyl-2-methylsulfanyl-5-nitro-pyrimidine-4,6-diamine anxiolytic-like activity without side effects associated with baclofen or benzodiazepines. J. Pharmacol. Exp. Ther. 2004;310:952–963. doi: 10.1124/jpet.104.066753. [DOI] [PubMed] [Google Scholar]
  • 19.Damian M, Mesnier D, Martin A, Pin J-P, Banères J-L. Asymmetric conformational changes in a GPCR dimer controlled by G-proteins. EMBO J. 2006;25:5693–5702. doi: 10.1038/sj.emboj.7601449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dupuis DS, Relkovic D, Lhuillier L, Mosbacher J, Kaupmann K. Point mutations in the transmembrane region of GABAB2 facilitate activation by the positive modulator N,N'-dicyclopentyl-2-methylsulfanyl-5-nitro-pyrimidine-4,6-diamine (GS39783) in the absence of the GABAB1 subunit. Mol. Pharmacol. 2006;70:2027–2036. doi: 10.1124/mol.106.028183. [DOI] [PubMed] [Google Scholar]
  • 21.Duthey B, Caudron S, Perroy J, Bettler B, Fagni L, Pin J-P, Prézeau L. A single subunit (GB2) is required for G-protein activation by the heterodimeric GABAB receptor. J. Biol. Chem. 2002;277:3236–3241. doi: 10.1074/jbc.M108900200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Froestl W, Gallagher M, Jenkins H, Madrid A, Melcher T, Teichman S, Mondadori CG, Pearlman R. SGS742 the first GABA(B) receptor antagonist in clinical trials. Biochem. Pharmacol. 2004;68:1479–1487. doi: 10.1016/j.bcp.2004.07.030. [DOI] [PubMed] [Google Scholar]
  • 23.Galvez T, Duthey B, Kniazeff J, Blahos J, Rovelli G, Bettler B, Prézeau L, Pin J-P. Allosteric interactions between GB1 and GB2 subunits are required for optimal GABAB receptor function. EMBO J. 2001;20:2152–2159. doi: 10.1093/emboj/20.9.2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Galvez T, Prézeau L, Milioti G, Franek M, Joly C, Froestl W, Bettler B, Bertrand H-O, Blahos J, Pin J-P. Mapping the agonist binding site of GABAB type 1 subunit sheds light on the activation process of GABAB receptors. J. Biol. Chem. 2000;275:41166–41174. doi: 10.1074/jbc.M007848200. [DOI] [PubMed] [Google Scholar]
  • 25.Galvez T, Urwyler S, Prézeau L, Mosbacher J, Joly C, Malitschek B, Heid J, Brabet I, Froestl W, Bettler B, Kaupmann K, Pin J-P. Ca2+-requirement for high affinity γ-aminobutyric acid (GABA) binding at GABAB receptors involvement of serine 269 of the GABABR1 subunit. Mol. Pharmacol. 2000;57:419–426. doi: 10.1124/mol.57.3.419. [DOI] [PubMed] [Google Scholar]
  • 26.Gasparini F, Kuhn R, Pin J-P. Allosteric modulators of group I metabotropic glutamate receptors: novel subtype-selective ligands and therapeutic perspectives. Curr. Opin. Pharmacol. 2002;2:43–49. doi: 10.1016/s1471-4892(01)00119-9. [DOI] [PubMed] [Google Scholar]
  • 27.Gjoni T, Desrayaud S, Imobersteg S, Urwyler S. The positive allosteric modulator GS39783 enhances GABA(B) receptor-mediated inhibition of cyclic AMP formation in rat striatum in vivo. J. Neurochem. 2006;96:1416–1422. doi: 10.1111/j.1471-4159.2006.03660.x. [DOI] [PubMed] [Google Scholar]
  • 28.Goudet C, Binet V, Prezeau L, Pin J-P. Allosteric modulators of class-C G-Protein coupled receptors open new possibilities for therapeutic application. Drug Discov. Today Ther. Strat. 2004;1:125–133. [Google Scholar]
  • 29.Havlickova M, Prezeau L, Duthey B, Bettler B, Pin J-P, Blahos J. The intracellular loops of the GB2 subunit are crucial for G-protein coupling of the heteromeric γ-aminobutyrate B receptor. Mol. Pharmacol. 2002;62:343–350. doi: 10.1124/mol.62.2.343. [DOI] [PubMed] [Google Scholar]
  • 30.Hawrot E, Xiao Y, Shi Q-I, Norman D, Kirkitadze M, Barlow PN. Demonstration of a tandem pair of complement protein modules in GABA(B) receptor1a. FEBS Lett. 1998;432:103–108. doi: 10.1016/s0014-5793(98)00794-7. [DOI] [PubMed] [Google Scholar]
  • 31.Hill DR, Bowery NG. 3H-Baclofen and 3H GABA bind to bicuculline-insensitive GABAB sites in rat brain. Nature. 1981;290:149–152. doi: 10.1038/290149a0. [DOI] [PubMed] [Google Scholar]
  • 32.Hlavackova V, Goudet C, Kniazeff J, Zikova A, Maurel D, Vol C, Trojanova J, Prézeau L, Pin J-P, Blahos J. Evidence for a single heptahelical domain being turned on upon activation of a dimeric GPCR. EMBO J. 2005;24:499–509. doi: 10.1038/sj.emboj.7600557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jones KA, Borowsky B, Tamm JA, Craig DA, Durkin MM, Dai M, Yao W-J, Johnson M, Gunwaldsen C, Huang L-Y, Tang C, Shen Q, Salon JA, Morse K, Laz T, Smith KE, Nagarathnam D, Noble SA, Branchek TA, Gerald C. GABA B receptors function as a heteromeric assembly of the subunits GABA B R1 and GABA B R2. Nature. 1998;396:674–679. doi: 10.1038/25348. [DOI] [PubMed] [Google Scholar]
  • 34.Kaupmann K, Cryan JF, Wellendorph P, Mombereau C, Sansig G, Klebs K, Schmutz M, Froestl W, van der Putten H, Mosbacher J, Brauner-Osborne H, Waldmeier P, Bettler B. Specific gamma-hydroxybutyrate-binding sites but loss of pharmacological effects of gamma-hydroxybutyrate in GABA(B)(1)-deficient mice. Eur. J. Neurosci. 2003;18:2722–2730. doi: 10.1111/j.1460-9568.2003.03013.x. [DOI] [PubMed] [Google Scholar]
  • 35.Kaupmann K, Huggel K, Heid J, Flor PJ, Bischoff S, Mickel SJ, McMaster G, Angst C, Bittiger H, Froestl W, Bettler B. Expression cloning of GABAB receptors uncovers similarity to metabotropic glutamate receptors. Nature. 1997;386:239–246. doi: 10.1038/386239a0. [DOI] [PubMed] [Google Scholar]
  • 36.Kaupmann K, Malitschek B, Schuler V, Heid J, Froestl W, Beck P, Mosbacher J, Bischoff S, Kulik A, Shigemoto R, Karschin A, Bettler B. GABA B-receptor subtypes assemble into functional heteromeric complexes. Nature. 1998;396:683–687. doi: 10.1038/25360. [DOI] [PubMed] [Google Scholar]
  • 37.Kerr DI, Ong J. Potentiation of metabotropic GABA(B) receptors by L-amino acids and dipeptides in rat neocortex. Eur. J. Pharmacol. 2003;468:103–108. doi: 10.1016/s0014-2999(03)01675-3. [DOI] [PubMed] [Google Scholar]
  • 38.Kerr DI, Ong J, Puspawati NM, Prager RH. Arylalkylamines are a novel class of positive allosteric modulators at GABA(B) receptors in rat neocortex. Eur. J. Pharmacol. 2002;451:69–77. doi: 10.1016/s0014-2999(02)02195-7. [DOI] [PubMed] [Google Scholar]
  • 39.Kniazeff J, Galvez T, Labesse G, Pin J-P. No ligand binding in the GB2 subunit of the GABAB receptor is required for activation and allosteric interaction between the subunits. J. Neurosci. 2002;22:7352–7361. doi: 10.1523/JNEUROSCI.22-17-07352.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kniazeff J, Saintot P-P, Goudet C, Liu J, Charnet A, Guillon G, Pin J-P. Locking the dimeric GABAB G-protein coupled receptor in its active state. J. Neurosci. 2004;24:370–377. doi: 10.1523/JNEUROSCI.3141-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Knoflach F, Mutel V, Jolidon S, Kew JN, Malherbe P, Vieira E, Wichmann J, Kemp JA. Positive allosteric modulators of metabotropic glutamate 1 receptor: Characterization, mechanism of action, and binding site. Proc. Natl. Acad. Sci. USA. 2001;98:13402–13407. doi: 10.1073/pnas.231358298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kulik A, Vida I, Fukazawa Y, Guetg N, Kasugai Y, Marker CL, Rigato F, Bettler B, Wickman K, Frotscher M, Shigemoto R. Compartment-dependent colocalization of Kir3.2-containing K+ channels and GABAB receptors in hippocampal pyramidal cells. J. Neurosci. 2006;26:4289–4297. doi: 10.1523/JNEUROSCI.4178-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kunishima N, Shimada Y, Tsuji Y, Sato T, Yamamoto M, Kumasaka T, Nakanishi S, Jingami H, Morikawa K. Structural basis of glutamate recognition by a dimeric metabotropic glutamate receptor. Nature. 2000;407:971–977. doi: 10.1038/35039564. [DOI] [PubMed] [Google Scholar]
  • 44.Lehmann A, Mattsson JP, Edlund A, Johansson T, Ekstrand AJ. Effects of repeated administration of baclofen to rats on GABAB receptor binding sites and subunit expression in the brain. Neurochem. Res. 2003;28:387–393. doi: 10.1023/a:1022353923578. [DOI] [PubMed] [Google Scholar]
  • 45.Lhuillier L, Mombereau C, Cryan JF, Kaupmann K. GABA(B) receptor-positive modulation decreases selective molecular and behavioral effects of cocaine. Neuropsychopharmacology. 2006; 32:388–398. doi: 10.1038/sj.npp.1301102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Liang JH, Chen F, Krstew E, Cowen MS, Carroll FY, Crawford D, Beart PM, Lawrence AJ. The GABA(B) receptor allosteric modulator CGP7930, like baclofen, reduces operant self-administration of ethanol in alcohol-preferring rats. Neuropharmacology. 2006;50:632–639. doi: 10.1016/j.neuropharm.2005.11.011. [DOI] [PubMed] [Google Scholar]
  • 47.Liang Y, Fotiadis D, Filipek S, Saperstein DA, Palczewski K, Engel A. Organization of the G protein-coupled receptors rhodopsin and opsin in native membranes. J. Biol. Chem. 2003;278:21655–21662. doi: 10.1074/jbc.M302536200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lingenhoehl K, Brom R, Heid J, Beck P, Froestl W, Kaupmann K, Bettler B, Mosbacher J. Gamma-hydroxybutyrate is a weak agonist at recombinant GABA(B) receptors. Neuropharmacology. 1999;38:1667–1673. doi: 10.1016/s0028-3908(99)00131-8. [DOI] [PubMed] [Google Scholar]
  • 49.Malherbe P, Masciadri R, Norcross R, Ratni H, Thomas A. Quinoline as allosteric enhancers of the GABA-B receptor. WO 2006/048146 A1. 2006.
  • 50.Malherbe P, Masciadri R, Norcross R, Ratni H, Thomas A. Thieno-pyridine derivatives as GABA-B allosteric enhancers. WO 2006/063732 A1. 2006 [Google Scholar]
  • 51.Malherbe P, Masciadri R, Prinssen E, Spooren W, Thomas A. 4-(sulfanyl-pyrimidin-4-ylmethyl)-morpholine derivatives and related compounds as GABA receptor ligands for the treatment of anxiety, depression and epilepsy. WO 2005/094828 A1. 2005.
  • 52.Malitschek B, Schweizer C, Keir M, Heid J, Froestl W, Mosbacher J, Kuhn R, Henley J, Joly C, Pin J-P, Kaupmann K, Bettler B. The N-terminal domain of γ-aminobutyric acid B receptors is sufficient to specify agonist and antagonist binding. Mol. Pharmacol. 1999;56:448–454. doi: 10.1124/mol.56.2.448. [DOI] [PubMed] [Google Scholar]
  • 53.Manning JP, Richards DA, Bowery NG. Pharmacology of absence epilepsy. Trends Pharmacol. Sci. 2003;24:542–549. doi: 10.1016/j.tips.2003.08.006. [DOI] [PubMed] [Google Scholar]
  • 54.Margeta-Mitrovic M, Jan YN, Jan LY. A trafficking checkpoint controls GABA(B) receptor heterodimerization. Neuron. 2000;27:97–106. doi: 10.1016/s0896-6273(00)00012-x. [DOI] [PubMed] [Google Scholar]
  • 55.Mombereau C, Kaupmann K, Froestl W, Sansig G, van der Putten H, Cryan JF. Genetic and pharmacological evidence of a role for GABA(B) receptors in the modulation of anxiety- and antidepressant-like behavior. Neuropsychopharmacology. 2004;29:1050–1062. doi: 10.1038/sj.npp.1300413. [DOI] [PubMed] [Google Scholar]
  • 56.Mukherjee RS, McBride EW, Beinborn M, Dunlap K, Kopin AS. Point mutations in either subunit of the GABAB receptor confer constitutive activity to the heterodimer. Mol. Pharmacol. 2006;70:1406–1413. doi: 10.1124/mol.106.024463. [DOI] [PubMed] [Google Scholar]
  • 57.O'Brien JA, Lemaire W, Chen T-B, Chang RSL, Jacobson MA, Ha SN, Lindsley CW, Schaffhauser HJ, Sur C, Pettibone DJ, Conn PJ, Williams Jr DL. A family of highly selective allosteric modulators of the metabotropic glutamate receptor subtype 5. Mol. Pharmacol. 2003;64:731–740. doi: 10.1124/mol.64.3.731. [DOI] [PubMed] [Google Scholar]
  • 58.Olianas MC, Ambu R, Garau L, Onali P. Allosteric modulation of GABA(B) receptor function in human frontal cortex. Neurochem. Int. 2005;46:149–158. doi: 10.1016/j.neuint.2004.08.002. [DOI] [PubMed] [Google Scholar]
  • 59.Onali P, Mascia FM, Olianas MC. Positive regulation of GABA(B) receptors dually coupled to cyclic AMP by the allosteric agent CGP7930. Eur. J. Pharmacol. 2003;471:77–84. doi: 10.1016/s0014-2999(03)01823-5. [DOI] [PubMed] [Google Scholar]
  • 60.Orru A, Lai P, Lobina C, Maccioni P, Piras P, Scanu L, Froestl W, Gessa GL, Carai MA, Colombo G. Reducing effect of the positive allosteric modulators of the GABA(B) receptor, CGP7930 and GS39783, on alcohol intake in alcohol-preferring rats. Eur. J. Pharmacol. 2005;525:105–111. doi: 10.1016/j.ejphar.2005.10.005. [DOI] [PubMed] [Google Scholar]
  • 61.Pagano A, Rovelli G, Mosbacher J, Lohmann T, Duthey B, Stauffer D, Ristig D, Schuler V, Meigel I, Lampert C, Stein T, Prézeau L, Blahos J, Pin J-P, Froestl W, Kuhn R, Heid J, Kaupmann K, Bettler B. C-terminal interaction is essential for surface trafficking but not for heteromeric assembly of GABAB receptors. J. Neurosci. 2001;21:1189–1202. doi: 10.1523/JNEUROSCI.21-04-01189.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pagano A, Rüegg D, Litschig S, Stoehr N, Stierlin C, Heinrich M, Floersheim P, Prézeau L, Carroll F, Pin J-P, Cambria A, Vranesic I, Flor PJ, Gasparini F, Kuhn R. The non-competitive antagonists 2-Methyl-6-(phenylethynyl)pyridine and 7-Hydroxyiminocyclopropan[b]chro-men-1a-carboxylic acid ethyl ester Interact with overlapping binding pockets in the transmembrane region of group I metabotropic glutamate receptors. J. Biol. Chem. 2000;275:33750–33758. doi: 10.1074/jbc.M006230200. [DOI] [PubMed] [Google Scholar]
  • 63.Parmentier M-L, Prézeau L, Bockaert J, Pin J-P. A model for the functioning of family 3 GPCRs. Trends Pharmacol. Sci. 2002;23:268–274. doi: 10.1016/s0165-6147(02)02016-3. [DOI] [PubMed] [Google Scholar]
  • 64.Pin J-P, Galvez T, Prézeau L. Evolution, structure and activation mechanism of family 3/C G-protein coupled receptors. Pharmacol. Ther. 2003;98:325–354. doi: 10.1016/s0163-7258(03)00038-x. [DOI] [PubMed] [Google Scholar]
  • 65.Pin JP, Kniazeff J, Binet V, Liu J, Maurel D, Galvez T, Duthey B, Havlickova M, Blahos J, Prezeau L, Rondard P. Activation mechanism of the heterodimeric GABAB receptor. Biochem. Pharmacol. 2004;68:1565–1572. doi: 10.1016/j.bcp.2004.06.035. [DOI] [PubMed] [Google Scholar]
  • 66.Prosser HM, Gill CH, Hirst WD, Grau E, Robbins M, Calver A, Soffin EM, Farmer CE, Lanneau C, Gray J, Schenck E, Warmerdam BS, Clapham C, Reavill C, Rogers DC, Stean T, Upton N, Humphreys K, Randall A, Geppert M, Davies CH, Pangalos MN. Epileptogenesis and enhanced prepulse inhibition in GABAB1-deficient mice. Mol. Cell Neurosci. 2001;17:1059–1070. doi: 10.1006/mcne.2001.0995. [DOI] [PubMed] [Google Scholar]
  • 67.Robbins MJ, Calver AR, Filippov AK, Hirst WD, Russell RB, Wood MD, Nasir S, Couve A, Brown DA, Moss SJ, Pangalos MN. GABAB2 is essential for G-protein coupling of the GABAB receptor heterodimer. J. Neurosci. 2001;21:8043–8052. doi: 10.1523/JNEUROSCI.21-20-08043.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rondard P, Liu J, Huang S, Malhaire F, Vol C, Pinault A, Labesse G, Pin J-P. Coupling of agonist binding to effector domain activation in metabotropic glutamate-like receptors. J. Biol. Chem. 2006;281:24653–24661. doi: 10.1074/jbc.M602277200. [DOI] [PubMed] [Google Scholar]
  • 69.Schuler V, Luscher C, Blanchet C, Klix N, Sansig G, Klebs K, Schmutz M, Heid J, Gentry C, Urban L, Fox A, Spooren W, Jaton A, Vigouret J, Pozza M, Kelly PH, Mosbacher J, Froestl W, Kaslin E, Korn R, Bischoff S, Kaupmann K, van der Putten H, Bettler B. Epilepsy, hyperalgesia, impaired memory, and loss of pre- and postsynaptic GABAB responses in mice lacking GABAB1. Neuron. 2001;31:47–58. doi: 10.1016/s0896-6273(01)00345-2. [DOI] [PubMed] [Google Scholar]
  • 70.Slattery DA, Markou A, Froestl W, Cryan JF. The GABAB receptor-positive modulator GS39783 and the GABAB receptor agonist baclofen attenuate the reward-facilitating effects of cocaine: intracranial self-stimulation studies in the rat. Neuropsychopharmacology. 2005;30:2065–2072. doi: 10.1038/sj.npp.1300734. [DOI] [PubMed] [Google Scholar]
  • 71.Smith MA, Yancey DL, Morgan D, Liu Y, Froestl W, Roberts DC. Effects of positive allosteric modulators of the GABAB receptor on cocaine self-administration in rats. Psychopharmacology (Berl.) 2004;173:105–111. doi: 10.1007/s00213-003-1706-5. [DOI] [PubMed] [Google Scholar]
  • 72.Tateyama M, Abe H, Nakata H, Saito O, Kubo Y. Ligand-induced rearrangement of the dimeric metabotropic glutamate receptor 1alpha. Nat. Struct. Mol. Biol. 2004;11:637–642. doi: 10.1038/nsmb770. [DOI] [PubMed] [Google Scholar]
  • 73.Tsuchiya D, Kunishima N, Kamiya N, Jingami H, Morikawa K. Structural views of the ligand-binding cores of a metabotropic glutamate receptor complexed with an antagonist and both glutamate and Gd3+ Proc. Natl. Acad. Sci. USA. 2002;99:2660–2665. doi: 10.1073/pnas.052708599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Urwyler S, Gjoni T, Kaupmann K, Pozza MF, Mosbacher J. Selected amino acids, dipeptides and arylalkylamine derivatives do not act as allosteric modulators at GABAB receptors. Eur. J. Pharmacol. 2004;483:147–153. doi: 10.1016/j.ejphar.2003.10.024. [DOI] [PubMed] [Google Scholar]
  • 75.Urwyler S, Gjoni T, Koljatic J, Dupuis DS. Mechanisms of allosteric modulation at GABAB receptors by CGP7930 and GS39783: effects on affinities and efficacies of orthosteric ligands with distinct intrinsic properties. Neuropharmacology. 2005;48:343–353. doi: 10.1016/j.neuropharm.2004.10.013. [DOI] [PubMed] [Google Scholar]
  • 76.Urwyler S, Mosbacher J, Lingenhoehl K, Heid J, Hofstetter K, Froestl W, Bettler B, Kaupmann K. Positive allosteric modulation of native and recombinant GABAB receptors by 2,6-Di-tert.butyl-4-(3-hydrxy-2-dimethyl-propyl)-phenol (CGP7930) and its aldehyde analogue CGP13501. . Mol. Pharmacol . 2001;60:963–971. [PubMed] [Google Scholar]
  • 77.Urwyler S, Pozza MF, Lingenhoehl K, Mosbacher J, Lampert C, Froestl W, Koller M, Kaupmann K. N,N'-Dicyclopentyl-2-methylsulfanyl-5-nitro-pyrimidine-4,6-diamine (GS39783) and structurally related compounds: novel allosteric enhancers of gamma-aminobutyric acid B receptor function. J. Pharmacol. Exp. Ther. 2003;307:322–330. doi: 10.1124/jpet.103.053074. [DOI] [PubMed] [Google Scholar]
  • 78.Vigot R, Barbieri S, Brauner-Osborne H, Turecek R, Shigemoto R, Zhang YP, Lujan R, Jacobson LH, Biermann B, Fritschy JM, Vacher CM, Muller M, Sansig G, Guetg N, Cryan JF, Kaupmann K, Gassmann M, Oertner TG, Bettler B. Differential compartmentalization and distinct functions of GABAB receptor variants. Neuron. 2006;50:589–601. doi: 10.1016/j.neuron.2006.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.White JH, Wise A, Main MJ, Green A, Fraser NJ, Disney GH, Barnes AA, Emson P, Foord SM, Marshall FH. Heterodimerization is required for the formation of a functional GABAB receptor. Nature. 1998;396:679–682. doi: 10.1038/25354. [DOI] [PubMed] [Google Scholar]
  • 80.Wise A, Green A, Main MJ, Wilson R, Fraser N, Marshall FH. Calcium sensing properties of the GABA(B) receptor. Neuropharmacology. 1999;38:1647–1656. doi: 10.1016/s0028-3908(99)00119-7. [DOI] [PubMed] [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES