The GABAB receptor (also denoted as GABA_B receptor) is a metabotropic G protein-coupled receptor (GPCR) of class C that mediates the slow and prolonged inhibitory neurotransmission elicited by gamma-aminobutyric acid (GABA), the principal inhibitory neurotransmitter in the central nervous system (CNS).[1] Unlike ionotropic GABA_A receptors, which produce fast synaptic inhibition, GABAB receptors couple to Gαi/o proteins to inhibit adenylyl cyclase, activate G-protein-coupled inwardly rectifying potassium (GIRK) channels, and suppress voltage-gated calcium channels, thereby reducing neuronal excitability and neurotransmitter release.[1] These receptors function as obligatory heterodimers composed of two distinct subunits, GABAB1 (GB1) and GABAB2 (GB2), each featuring an extracellular Venus flytrap domain for ligand binding, a seven-transmembrane helical domain, and intracellular C-terminal tails that regulate trafficking and signaling.[2]GABAB receptors are widely distributed throughout the CNS, including key regions such as the cerebral cortex, hippocampus, thalamus, cerebellum, and spinal cord, where they operate as presynaptic autoreceptors on GABAergic terminals or heteroreceptors on glutamatergic terminals to modulate synaptic transmission, as well as postsynaptic receptors to generate inhibitory postsynaptic potentials.[1] Their activation plays critical roles in regulating diverse physiological processes, including synaptic plasticity, pain sensation, reward pathways, feeding behavior, and motor control, with dysregulation implicated in neurological and psychiatric disorders such as epilepsy, anxiety, depression, addiction, and spasticity.[1] Splice variants of the GB1 subunit (e.g., GB1a and GB1b) further diversify receptor localization and function, with GB1a predominantly in axonal terminals and GB1b in somatodendritic compartments.[2]The GABAB receptor was first identified in the 1980s through pharmacological studies distinguishing it from ionotropic GABA receptors.[3]Pharmacologically, the orthosteric binding site for GABA resides in the Venus flytrap domain of the GB1 subunit, while allosteric modulation occurs at the GB2 subunit's transmembrane domain, enabling the development of agonists like baclofen (the only clinically approved GABAB agonist as of 2025, used for spasticity and muscle rigidity) and positive allosteric modulators such as GS39783 and CGP7930, which enhance GABA affinity with fewer side effects than orthosteric ligands.[1] Antagonists like CGP55845 and saclofen have been instrumental in dissecting receptor functions, and emerging research explores GABAB-targeted therapies for epilepsy, alcohol use disorder, pain, and obesity, leveraging the receptor's role in inhibiting binge eating and fat intake.[1] Structural insights from cryo-electron microscopy have illuminated ligand-bound states, facilitating drug design to address unmet needs in CNS disorders.[4]
Introduction
Definition and classification
The GABAB receptor is a metabotropic G-protein-coupled receptor (GPCR) primarily activated by gamma-aminobutyric acid (GABA), the main inhibitory neurotransmitter in the vertebrate nervous system.[5] Unlike ionotropic GABAA receptors, which function as ligand-gated ion channels to mediate rapid synaptic inhibition, GABAB receptors elicit slower, modulatory responses through G-protein-mediated activation of intracellular second messenger pathways.[2] This distinction arises from their GPCR architecture, enabling prolonged regulation of neuronal excitability rather than direct ion flux.[6]GABAB receptors are classified within class C of the GPCR superfamily, a group characterized by large extracellular domains and roles in neurotransmission, sensory perception, and ion homeostasis.[7] They form a distinct subfamily alongside metabotropic glutamate receptors and calcium-sensing receptors, setting them apart from the rhodopsin-like class A GPCRs that include many other neurotransmitter receptors.[2] The existence of these receptors was first identified in 1981 through radioligand binding studies using the selective agonist baclofen on rat brain membranes.[8]Functional GABAB receptors require heterodimeric assembly of principal GABAB1 and auxiliary GABAB2 subunits to traffic to the cell surface and bind ligands effectively.[6] They are expressed throughout the central nervous system, including cortex, hippocampus, and cerebellum, as well as in peripheral tissues such as the gastrointestinal tract and immune cells.[2] In synaptic transmission, GABAB receptors mediate presynaptic inhibition by suppressing calcium influx and neurotransmitter release at excitatory and inhibitory terminals, while postsynaptic activation leads to potassium channel opening and neuronal hyperpolarization.[5]GABAB receptors demonstrate strong evolutionary conservation, with orthologs present in all vertebrates and functional homologs in select invertebrates such as mollusks and arthropods, indicating an ancient role in inhibitory signaling.[6] Recent studies have identified GABAB receptor homologs in cnidarians, such as Nematostella vectensis, where they regulate neurogenesis.[9] This conservation underscores their fundamental contribution to neural modulation across phyla, from basic reflex arcs in invertebrates to complex cognitive processes in mammals.[6]
Historical discovery
The GABAB receptor was first identified in 1981 by Norman Bowery and colleagues, who used the radioligand [^3H]baclofen to demonstrate a bicuculline-insensitive binding site for GABA in rat brain membranes, distinct from the ionotropic GABAA receptor. This discovery arose from observations that baclofen, a GABA analog, elicited inhibitory effects in the central nervous system that were not blocked by bicuculline, prompting the pharmacological characterization of a novel GABA receptor subtype termed GABAB. The binding assays revealed high-affinity sites with a distribution enriched in synaptic regions, laying the foundation for recognizing GABAB as a mediator of presynaptic inhibition and postsynaptic hyperpolarization.In the early 1980s, subsequent studies confirmed the metabotropic nature of the GABAB receptor through functional assays linking its activation to second messenger systems, rather than direct ion channel gating. For instance, baclofen application was shown to inhibit adenylate cyclase activity and modulate potassium conductances via G-protein coupling in neuronal preparations, with effects sensitive to pertussis toxin, indicating involvement of Gi/o proteins. These experiments, including electrophysiological recordings in hippocampal slices and biochemical measurements of cyclic AMP levels, established GABAB as a G-protein-coupled receptor (GPCR), contrasting with the fast, chloride-mediated responses of GABAA receptors. This period also saw the development of selective antagonists like phaclofen, further delineating GABAB pharmacology.The molecular era began in 1997 with the expression cloning of the GABAB1 subunit by Kaupmann et al., who isolated a cDNA from rat brain that encoded a protein with sequence homology to metabotropic glutamate receptors, classifying it within the class C GPCR family. When expressed alone, GABAB1 bound GABA and baclofen but failed to couple effectively to G-proteins or elicit functional responses, highlighting the need for additional components. This breakthrough enabled detailed studies of receptor isoforms, such as GABAB1a and GABAB1b, differing in their N-terminal splice variants and expression patterns.[10]In 1998, the GABAB2 subunit was identified through parallel cloning efforts, revealing that functional GABAB receptors require heterodimerization of GABAB1 and GABAB2 subunits for proper trafficking to the cell surface and G-protein activation.[11] Coexpression studies demonstrated that GABAB2 provides the domain for efficient Gi/o coupling, while GABAB1 retains the orthosteric binding site for agonists, resolving earlier puzzles about the receptor's poor monomeric function. This heterodimeric architecture was confirmed via co-immunoprecipitation and functional assays measuring inhibition of calcium currents.Nomenclature was standardized by the International Union of Pharmacology (IUPHAR) in the early 2000s, designating the genes as GABBR1 (encoding GABAB1) and GABBR2 (encoding GABAB2), with the receptor officially termed GABAB to reflect its metabotropic properties. A key milestone in 2001 involved detailed mapping of the intracellular loops, confirming that specific motifs in the GABAB2 C-terminus are essential for selective Gi/o coupling and downstream signaling, such as activation of GIRK channels. These advancements solidified the receptor's role in inhibitory neurotransmission and spurred targeted drug development.[12][13]
Molecular biology
Structure
The GABAB receptor is an obligate heterodimer composed of two distinct subunits, GABAB1 and GABAB2, which assemble to form a functional G protein-coupled receptor (GPCR). The GABAB1 subunit features an extracellular Venus flytrap (VFT) domain responsible for GABA binding, while the GABAB2 subunit contains a heptahelical transmembrane domain that couples to G proteins for signal transduction.[1][12]The GABAB1 subunit exists in two primary isoforms: GABAB1a, which has a longer N-terminal region with two sushi domains facilitating synaptic targeting, and GABAB1b, characterized by a shorter N-terminus that promotes extrasynaptic localization. Both isoforms heterodimerize with GABAB2 to form active receptors, but their distinct N-terminal extensions influence subcellular distribution without altering core binding or coupling properties.[14][15]Key structural elements stabilize the receptor's architecture, including disulfide bridges within the VFT domain of GABAB1 that maintain the ligand-binding pocket's integrity and an intracellular coiled-coil domain at the C-termini of both subunits that mediates heterodimerization and ensures proper trafficking to the cell surface. These features are essential for the receptor's stability and assembly, with the coiled-coil interaction preventing homodimerization and promoting the obligatory heterodimeric configuration.[16][17]Cryo-electron microscopy (cryo-EM) studies from 2020 and 2021 have elucidated the receptor's conformational dynamics, revealing distinct inactive and active states. In the inactive conformation, the VFT domains are open, and transmembrane helices maintain a loosely packed arrangement; agonist binding induces VFT closure in GABAB1, propagating allosteric changes to the transmembrane domains of GABAB2 for G protein engagement, with identified phospholipid binding pockets modulating stability and allosteric sites influencing activation. These structures highlight an asymmetric activation mechanism where GABAB2's transmembrane domain undergoes principal rearrangements.[18][7][19]A 2025 study demonstrated that mechanical forces, such as traction and shear stress, can induce conformational changes in the GABAB receptor independent of GABA binding, activating downstream signaling through G protein coupling and suggesting a mechanosensitive role in cellular contexts.[20]
Genetics and expression
The GABAB receptor is composed of two principal subunits encoded by distinct genes: GABBR1 and GABBR2. The GABBR1 gene is located on chromosome 6p22.1, spanning approximately 78 kb and consisting of 24 exons.[21] The GABBR2 gene resides on chromosome 9q22.33, covering about 421 kb with 22 exons.[22] These genes produce the GABAB1 and GABAB2 subunits, respectively, which must heterodimerize to form functional receptors.Alternative splicing of the GABBR1 gene generates multiple isoforms, most notably GABAB1a and GABAB1b, which arise from alternative promoter usage and differ in their N-terminal regions, influencing subcellular targeting and tissue distribution.[23] For instance, GABAB1a predominates in axonal compartments, while GABAB1b is more prevalent in somatodendritic regions. In contrast, the GABBR2 gene exhibits limited alternative splicing, with no major isoforms reported that significantly alter function.[24]Expression of GABBR1 and GABBR2 mRNAs is highest in the central nervous system, particularly in the cerebral cortex, hippocampus, thalamus, and cerebellum, where they support inhibitory neurotransmission.[14] Moderate expression occurs in peripheral nervous tissues, such as dorsal root ganglia and autonomic nerves, while levels are low in non-neuronal peripheral organs like liver and kidney.[2] Within neurons, the receptors localize presynaptically to modulate neurotransmitter release and postsynaptically to induce hyperpolarization, with isoform-specific patterns enhancing this distribution. GABAB receptor expression is developmentally upregulated, increasing progressively from embryonic stages to adulthood in regions like the hippocampus and cortex to refine excitatory-inhibitory balance.[25]Transcriptional regulation involves neuron-restrictive silencer factor (NRSF/REST), which represses GABBR1 in non-neuronal cells, ensuring restricted expression in neural tissues.[26]
Function and signaling
Mechanisms of action
The GABAB receptor is a G-protein-coupled receptor that preferentially couples to the Gi/o family of heterotrimeric G-proteins upon activation by GABA. This coupling leads to the dissociation of the G-protein into αi/o and βγ subunits, initiating downstream signaling cascades that mediate inhibitory effects in the central nervous system.[2]The Gαi/o subunit primarily inhibits adenylyl cyclase isoforms I, III, V, and VI, resulting in reduced cyclic AMP (cAMP) levels and subsequent modulation of protein kinase A activity, which dampens neuronal excitability. In parallel, the free Gβγ subunits directly interact with effector proteins: they activate G-protein-gated inwardly rectifying potassium (GIRK) channels, promoting K⁺ efflux and membrane hyperpolarization, while inhibiting presynaptic N-type (Caᵥ2.2) and P/Q-type (Caᵥ2.1) voltage-gated Ca²⁺ channels, thereby reducing Ca²⁺ influx and neurotransmitter release. These βγ-mediated effects occur via membrane-delimited pathways, as demonstrated by rapid occlusion of inhibition upon direct Gβγ scavenging or strong depolarizing pulses that relieve channel blockade.[2][27][28]Postsynaptically, GABAB receptor activation via Gβγ subunits enhances GIRK conductance, shifting the membrane potential toward the potassium equilibrium potential (E_K). This hyperpolarization can be described by the Nernst equation for potassium:\Delta V = \frac{RT}{F} \ln \left( \frac{[\mathrm{K}^+]_{\mathrm{out}}}{[\mathrm{K}^+]_{\mathrm{in}}} \right)where increased GIRK-mediated K⁺ permeability drives V_m closer to E_K (typically around -90 mV), generating slow inhibitory postsynaptic potentials (IPSPs). Presynaptically, the receptor functions as an autoreceptor, where βγ-mediated Ca²⁺ channel inhibition reduces GABA release probability, providing feedback inhibition at GABAergic synapses. In contrast to these canonical inhibitory roles, a recent study in medial habenula terminals revealed that GABAB activation recruits release-ready vesicles in an activity-dependent manner, enhancing phasic neurotransmitter release through Ca²⁺-dependent mechanisms involving CAPS2, thereby modulating inhibition in this specific circuit.[28][2][29]
Physiological roles
The GABAB receptor plays a key role in synaptic transmission by mediating slow inhibitory postsynaptic potentials (IPSPs) in various brain regions, including the hippocampus and cortex, where it regulates neuronal excitability and prevents excessive firing.[30] Activation of these receptors on presynaptic terminals inhibits the release of excitatory neurotransmitters like glutamate, contributing to the fine-tuning of network activity during information processing.[31] In the hippocampus, this mechanism supports the balance between excitation and inhibition essential for learning and memory formation.[32]In the spinal cord, GABAB receptors contribute to pain modulation through presynaptic inhibition of glutamate release from primary afferent fibers, thereby reducing the transmission of nociceptive signals to second-order neurons in the dorsal horn.[33] This inhibitory action helps maintain sensory gating under normal conditions, limiting hypersensitivity to mechanical or thermal stimuli.[34]GABAB receptors in the ventral tegmental area (VTA) modulate dopamine release from dopaminergic neurons, influencing reward processing and susceptibility to addictive substances.[35] By inhibiting GABAergic inputs to these neurons, the receptors facilitate dopamine signaling in reward pathways, while their activation can enhance sensitivity to ethanol and gamma-hydroxybutyrate through GIRK channel-mediated hyperpolarization.[36]During central nervous system development, GABAB receptors promote axon guidance and synaptogenesis by regulating neuronal migration and neurite outgrowth in early cortical circuits.[37] Their activation triggers brain-derived neurotrophic factor (BDNF) release, which supports the maturation of inhibitory synapses and network refinement.[38]Recent findings highlight additional physiological roles, including integration in the gut-brain axis where GABAB receptors in gastrointestinal tissues influence feeding behaviors via bidirectional signaling with central circuits.[39] In non-neuronal contexts, these receptors can be mechanically activated by shear stress independently of GABA, modulating cellular responses in vascular or astrocytic environments.[20] Furthermore, in cerebrospinal fluid-contacting neurons of the spinal cord, GABAB receptor activation enhances neural stem cell potential, promoting proliferation and recovery mechanisms in healthy tissue maintenance.[40]
Pharmacology
Agonists
The primary endogenous agonist of the GABAB receptor is γ-aminobutyric acid (GABA), which binds to the orthosteric site with an EC50 of approximately 100 μM in functional assays measuring G protein-coupled responses.[41]Baclofen, a synthetic analog of GABA featuring a 4-chlorophenyl substitution at the β-position, serves as a selective orthosteric agonist with an EC50 of around 1 μM, enabling potent activation of receptor signaling pathways.[42]Among synthetic agonists, γ-hydroxybutyric acid (GHB) acts as a partial agonist at the GABAB receptor, exhibiting dual activity by also engaging high-affinity GHB receptors at low doses (typically <1 mM), which contributes to its sedative effects.[43]Phenibut, structurally similar to baclofen with a phenyl group instead of chlorine, functions as a GABAB agonist alongside weak interactions at GABAA receptors, promoting anxiolytic and nootropic properties. Lesogaberan (AZD3355), a fluorinated phosphinic acid derivative, is a peripherally restricted agonist with high potency (EC50 ≈ 8.6 nM in recombinant systems) and limited central nervous system penetration, developed primarily for gastroesophageal reflux disease by reducing transient lower esophageal sphincter relaxations.[44]The orthosteric binding site resides in the Venus flytrap domain of the GABAB1 subunit, where agonist binding induces conformational changes that require heterodimerization with GABAB2 for effective G protein coupling and downstream signaling.[43] Selectivity challenges include partial agonism observed with certain ligands like GHB at higher concentrations, which limits maximal efficacy compared to full agonists such as baclofen.[43]
Antagonists
Orthosteric antagonists of the GABAB receptor bind competitively to the orthosteric site in the Venus flytrap domain (VFTD) of the GABAB1 subunit, thereby preventing the binding of GABA or synthetic agonists and inhibiting the conformational changes necessary for receptor activation and G-protein coupling.[6] This blockade disrupts downstream signaling, including inhibition of adenylyl cyclase and modulation of ion channels, allowing researchers to isolate GABAB-mediated effects in experimental settings.[5]The first selective GABAB antagonist identified was phaclofen, a phosphonate analog of baclofen, which competitively antagonizes GABAB responses with moderate potency (IC50 in the micromolar range) but limited brain penetration.[45] Subsequent developments included saclofen, a more potent competitive antagonist (IC50 ~10-50 μM) that improved upon phaclofen's selectivity and efficacy in peripheral and central assays.[46] For systemic applications, CGP 35348 emerged as a brain-penetrant antagonist (IC50 ~34 μM), enabling in vivo studies of GABAB function without invasive delivery.[47] Among these, CGP 55845 stands out for its high potency and selectivity for GABAB receptors over ionotropic GABA receptors (IC50 ~5 nM), making it a preferred tool for precise pharmacological blockade.[48]These antagonists exhibit strong selectivity for metabotropic GABAB receptors compared to GABAA or GABAC receptors, though early compounds like phaclofen showed some off-target effects at higher concentrations; later iterations like CGP 55845 achieved near-exclusive GABAB specificity.[5] In research, they are widely employed to dissect GABAB roles in neuronal circuits, such as blocking presynaptic inhibition in brain slices to reveal contributions to synaptic transmission and plasticity.[49] Despite their utility in preclinical models, no GABAB antagonists have advanced to major clinical use, primarily due to challenges in achieving therapeutic windows without disrupting physiological inhibition.[50] Recent applications include the use of CGP 35348 in 2024 rodent models of cocaine addiction, where intra-accumbens administration enhanced drug-seeking behaviors and altered gene expression related to reward pathways, highlighting GABAB's suppressive role in addiction.[51]
Allosteric modulators
Allosteric modulators of the GABAB receptor bind to sites distinct from the orthosteric GABA-binding pocket, thereby enhancing or inhibiting receptor activity in a manner dependent on the endogenous ligand. These compounds offer advantages over orthosteric ligands by providing spatiotemporal control over signaling, as they only modulate responses when GABA is present, and they exhibit reduced desensitization compared to direct agonists.[52]Positive allosteric modulators (PAMs) stabilize the active conformation of the receptor, increasing the potency and efficacy of GABA without directly activating the receptor. CGP7930, one of the first identified PAMs, binds within the transmembrane domain of the GABAB2 subunit (specifically involving transmembrane helices 3, 5, 6, and 7), enhancing GABA potency 5- to 10-fold and efficacy up to 2-fold, with an EC50 of approximately 5 μM in recombinant and native systems.[52][53] Similarly, GS39783 acts at the same transmembrane site on GABAB2, producing comparable 5- to 10-fold increases in GABA potency and up to 2-fold efficacy enhancements across species including rat and human, without intrinsic agonist activity.[52] ADX71441 represents a more advanced, brain-penetrant PAM with high potency and selectivity for GABAB receptors, demonstrating dose-dependent attenuation of alcohol self-administration in rodents at doses as low as 1-3 mg/kg, while avoiding sedative side effects at therapeutically relevant levels; although preclinical studies showed promise, its development was discontinued in 2019.[54][55]Negative allosteric modulators (NAMs) are less common and counteract GABA-induced activation by stabilizing the inactive receptor state. Examples include CLH304a, which inhibits native GABAB receptor activity in cerebellar neurons by binding to the heptahelical domain of the GABAB2 subunit, and COR758, which blocks G protein activation, subunit rearrangements, and downstream signaling (such as cAMP inhibition and ERK phosphorylation) via a novel intrahelical site in the GABAB1 subunit, without competing at the orthosteric site.[56][57]Recent advances have focused on developing subtype-selective PAMs to overcome limitations of orthosteric ligands, with novel chemical scaffolds identified for improved pharmacokinetics and pharmacodynamics; for instance, candidates like ASP8062, which has progressed to clinical trials (completed phase II as of 2025) for alcohol use disorder, demonstrating reductions in craving and consumption. A 2024 review highlights novel chemical scaffolds for improved pharmacokinetics, while the 2025 Guide to Pharmacology update underscores new insights into synaptic regulation. Cryo-EM structures from 2021 onward have elucidated allosteric binding sites, including the 2021 GABAB-Gi complex revealing flexible G protein coupling and transmembrane pocket details that inform modulator design, with ongoing studies (up to 2025) refining activation mechanisms and enabling targeted chemistry for enhanced selectivity.[58][7][58][59][60]
Clinical aspects
Disease associations
Dysfunction of the GABAB receptor has been implicated in various neurological and psychiatric disorders, primarily through loss-of-function mutations or altered expression that disrupt inhibitory signaling in the central nervous system. In epilepsy, loss-of-function mutations in the GABBR1 and GABBR2 genes reduce GABAB receptor-mediated inhibition, leading to hyperexcitability and epileptic phenotypes such as epileptic encephalopathy. For instance, de novo monoallelic variants in GABBR1 have been linked to neurodevelopmental disorders including epilepsy, with functional studies showing impaired receptor trafficking and signaling.[61][62] Similarly, variants in GABBR2 are associated with epileptic encephalopathy and milder intellectual disability syndromes, where reduced receptor function diminishes presynaptic inhibition of glutamate release.[63]In addiction, particularly alcohol dependence, GABAB receptor signaling modulates reward pathways and withdrawal symptoms, with evidence suggesting altered receptor sensitivity contributes to dependence vulnerability. Although direct upregulation of GABAB receptors in chronic alcohol dependence remains under investigation, preclinical models show that GABAB agonists like baclofen reduce alcohol intake by enhancing inhibitory tone in the mesolimbic system, implying baseline hypoactivity in dependent states. Additionally, gamma-hydroxybutyric acid (GHB), a GABAB receptor agonist, poses significant overdose risks due to dose-dependent respiratory depression and neuroinhibition via excessive GABAB activation, which can lead to coma or death in recreational misuse.[64][65][66]Chronic pain disorders, such as neuropathic pain, involve GABAB receptor deficits that impair presynaptic inhibition of nociceptive transmission, resulting in central hypersensitivity. Downregulation or impaired function of presynaptic GABAB receptors in the spinal cord reduces GABA-mediated suppression of glutamate release from primary afferents, exacerbating pain signaling in models of nerve injury. This presynaptic deficit contributes to mechanical allodynia and hyperalgesia, highlighting GABAB receptors as key regulators of spinal inhibitory circuits in chronic pain states.[67]Recent evidence from 2025 links GABAB hypoactivity to schizophrenia, where genetic variants in GABBR1, including single nucleotide polymorphisms (SNPs), correlate with disrupted inhibitory neurotransmission and psychotic symptoms. Functional analyses of these variants reveal reduced receptor expression or signaling efficacy in prefrontal cortex regions, contributing to excitatory-inhibitory imbalance and cognitive deficits characteristic of the disorder. A comprehensive review of GABAB genetics underscores how such hypoactivity exacerbates schizophrenia etiology alongside other psychiatric conditions.[24][68]Other disorders associated with GABAB dysfunction include autism spectrum disorder (ASD), where downregulation of GABAB receptors leads to synaptic excitatory-inhibitory imbalance, manifesting as social deficits and repetitive behaviors. In spinal cord injury, impaired GABAB receptor signaling hinders the neural stem cell potential of cerebrospinal fluid-contacting neurons, reducing proliferation and repair responses post-injury, as evidenced by 2025 studies showing GABAB activation enhances recovery via PI3K/Akt pathways.[69][70]In brain-gut disorders like irritable bowel syndrome (IBS), dysregulation of GABAB receptors within gut-brain GABA networks contributes to visceral hypersensitivity and motility issues, with 2024 findings indicating altered GABAergic signaling in the enteric nervous system amplifies stress-induced symptoms. Decreased GABAB-mediated inhibition in vagal and spinal pathways disrupts the bidirectional communication, linking central hypoactivity to peripheral gut dysfunction in IBS pathogenesis.[71][72]
Therapeutic applications
Baclofen, a selective GABAB receptoragonist, was approved by the FDA in 1977 for the treatment of spasticity associated with multiple sclerosis, spinal cord injuries, and other neurological conditions, where it reduces muscle tone by inhibiting excitatory neurotransmitter release in the spinal cord.[73] Clinical use has also extended off-label to alcohol use disorder, where doses of 30–80 mg/day have demonstrated efficacy in increasing abstinence rates and reducing craving through modulation of reward pathways in preclinical and open-label studies.[65] However, its application remains limited by central nervous system side effects, prompting exploration of more targeted GABAB therapeutics.Arbaclofen, the R-enantiomer of baclofen, has been investigated as a GABAB agonist for neurodevelopmental disorders, particularly autism spectrum disorder (ASD) and fragile X syndrome (FXS). An open-label phase 2 trial in children and adolescents with ASD showed improvements in social avoidance and irritability at doses up to 0.3 mg/kg/day, attributed to restoration of excitatory-inhibitory balance.[74] Despite these findings, a phase 3 trial in FXS patients failed to meet primary endpoints for behavioral outcomes in 2015, leading to discontinued development by Roche; however, recent 2025 data from Allos Pharma reported positive results in an ASD trial, supporting further evaluation of its safety and efficacy in youth aged 5–17.[75] Lesogaberan (AZD3355), another GABAB agonist, was developed for gastroesophageal reflux disease (GERD) by targeting transient lower esophageal sphincter relaxations; phase 2 trials in PPI-refractory patients showed a 25% reduction in reflux episodes at 65 mg twice daily but only marginal superiority over placebo, resulting in halted development in 2012 due to insufficient efficacy and side effects like paresthesia.[76][77]Positive allosteric modulators (PAMs) of GABAB receptors offer a promising approach to enhance endogenous GABA signaling with potentially fewer side effects than orthosteric agonists. ADX71441, a potent and selective GABAB PAM, has shown preclinical efficacy in models of anxiety and epilepsy by potentiating receptor activation without direct agonism, with early-phase trials indicating good tolerability for conditions like spasticity and pain.[78] In schizophrenia, GABAB PAMs in recent preclinical studies have ameliorated cognitive and negative symptoms by modulating prefrontal cortex excitability, positioning them as adjunctive therapies to antipsychotics.[79] Recent advances highlight PAMs' advantages, including higher selectivity and reduced sedation risk compared to agonists.[79]Key challenges in GABAB-targeted therapies include sedation and dizziness from broad receptor activation, as seen with baclofen, and the development of tolerance with chronic use, which diminishes efficacy in spasticity and addiction treatment.[73] Strategies to address these involve biased agonists that preferentially activate G-protein signaling over β-arrestin pathways, potentially separating therapeutic benefits from adverse effects, though clinical translation remains preclinical.[80]Emerging research in 2025 has identified GABA-independent mechanical activation of GABAB receptors via integrins in response to traction forces and shear stress, opening avenues for therapies in tissue repair by promoting cellular signaling in wound healing and fibrosis models without ligand dependence.[20] Indirect modulation through orexin-GABAB interactions holds potential for narcolepsy, where GABAB agonism reduces cataplexy and promotes slow-wave sleep in murine models by inhibiting orexin neuron activity presynaptically.[81] Looking ahead, gene therapy approaches targeting rare GABAB receptor mutations linked to epilepsy are in early conceptual stages, with recent preclinical advances using AAV vectors to restore receptor function in animal models, informed by GABAA-related precedents and GPCR structural insights.[82][83]