Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.191
Filter
1.
Nat Commun ; 15(1): 7795, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242530

ABSTRACT

ρ-type γ-aminobutyric acid-A (GABAA) receptors are widely distributed in the retina and brain, and are potential drug targets for the treatment of visual, sleep and cognitive disorders. Endogenous neuroactive steroids including ß-estradiol and pregnenolone sulfate negatively modulate the function of ρ1 GABAA receptors, but their inhibitory mechanisms are not clear. By combining five cryo-EM structures with electrophysiology and molecular dynamics simulations, we characterize binding sites and negative modulation mechanisms of ß-estradiol and pregnenolone sulfate at the human ρ1 GABAA receptor. ß-estradiol binds in a pocket at the interface between extracellular and transmembrane domains, apparently specific to the ρ subfamily, and disturbs allosteric conformational transitions linking GABA binding to pore opening. In contrast, pregnenolone sulfate binds inside the pore to block ion permeation, with a preference for activated structures. These results illuminate contrasting mechanisms of ρ1 inhibition by two different neuroactive steroids, with potential implications for subtype-specific gating and pharmacological design.


Subject(s)
Cryoelectron Microscopy , Estradiol , Molecular Dynamics Simulation , Pregnenolone , Receptors, GABA-A , Humans , Binding Sites , Estradiol/metabolism , Estradiol/pharmacology , HEK293 Cells , Pregnenolone/metabolism , Pregnenolone/pharmacology , Pregnenolone/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry
2.
J Med Chem ; 67(14): 12349-12365, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39013072

ABSTRACT

Multitarget strategies are essential in addressing complex diseases, yet developing multitarget-directed ligands (MTDLs) is particularly challenging when aiming to engage multiple therapeutic targets across different tissues. Here, we present a molecular transformer strategy, enhancing traditional MTDLs. By utilizing esterase-driven hydrolysis, this approach mimics the adaptive nature of transformers for enabling molecules to modify their pharmacological effects in response to the biological milieu. By virtual screening and biological evaluation, we identified KGP-25, a novel compound initially targeting the voltage-gated sodium channel 1.8 (Nav1.8) in the peripheral nervous system (PNS) for analgesia, and later the γ-aminobutyric acid subtype A receptor (GABAA) in the central nervous system (CNS) for general anesthesia. Our findings confirm KGP-25's dual efficacy in cellular and animal models, effectively reducing opioid-related side effects. This study validates the molecular transformer approach in drug design and highlights its potential to overcome the limitations of conventional MTDLs, paving new avenues in innovative therapeutic strategies.


Subject(s)
Analgesics , Esterases , Ligands , Analgesics/pharmacology , Analgesics/chemistry , Animals , Humans , Esterases/metabolism , Anesthetics/pharmacology , Anesthetics/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Drug Design , Mice , Male
3.
Chemistry ; 30(50): e202401921, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38875450

ABSTRACT

From both pharmaceutical and structural perspectives, the large family of prostaglandins represent a truly remarkable class of natural products. Prostaglandin A2 is a tissue hormone naturally found in human seminal plasma and in the sea whip Plexaura homomalla with yet poorly understood biological or therapeutic effects. Herein, a novel strategy for the stereoselective construction of both naturally occurring prostaglandin A2 epimers and first insights into their functional effects on the major inhibitory neurotransmitter γ-aminobutyric acid (GABA) type A receptors (GABAAR) are provided. The synthesis of both epimers was achieved in only 11 steps starting from commercially available 2,5-dimethoxy-tetrahydrofuran employing an organocatalytic domino-aldol reaction, a Mizoroki-Heck reaction, a Wittig reaction as well as an oxidation-decarboxylation sequence. The (15R)-epimer significantly reduced GABA-induced currents through GABAA receptors while its (15S)-epimer did not show any significant effect. These data suggest that (15R)-PGA2 might serve as a novel scaffold for the development of selective GABAA receptor modulators.


Subject(s)
Receptors, GABA-A , Stereoisomerism , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Humans , Furans/chemistry , Furans/chemical synthesis , Dinoprost/chemistry , Dinoprost/chemical synthesis , Oxidation-Reduction
4.
Nat Commun ; 15(1): 5244, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898000

ABSTRACT

Methaqualone, a quinazolinone marketed commercially as Quaalude, is a central nervous system depressant that was used clinically as a sedative-hypnotic, then became a notorious recreational drug in the 1960s-80s. Due to its high abuse potential, medical use of methaqualone was eventually prohibited, yet it persists as a globally abused substance. Methaqualone principally targets GABAA receptors, which are the major inhibitory neurotransmitter-gated ion channels in the brain. The restricted status and limited accessibility of methaqualone have contributed to its pharmacology being understudied. Here, we use cryo-EM to localize the GABAA receptor binding sites of methaqualone and its more potent derivative, PPTQ, to the same intersubunit transmembrane sites targeted by the general anesthetics propofol and etomidate. Both methaqualone and PPTQ insert more deeply into subunit interfaces than the previously-characterized modulators. Binding of quinazolinones to this site results in widening of the extracellular half of the ion-conducting pore, following a trend among positive allosteric modulators in destabilizing the hydrophobic activation gate in the pore as a mechanism for receptor potentiation. These insights shed light on the underexplored pharmacology of quinazolinones and further elucidate the molecular mechanisms of allosteric GABAA receptor modulation through transmembrane binding sites.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-A , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Binding Sites , Humans , Animals , Etomidate/pharmacology , Etomidate/analogs & derivatives , Propofol/pharmacology , Propofol/chemistry , Quinazolinones/pharmacology , Quinazolinones/chemistry , Allosteric Regulation/drug effects , HEK293 Cells , Hypnotics and Sedatives/pharmacology
5.
Bioorg Med Chem Lett ; 110: 129854, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38914347

ABSTRACT

C9-methylated quazepam 1 was prepared, and its physicochemical properties were investigated. The atropisomers of 1 were isolated as (a1R, a2S) and (a1S, a2R) isomers. Their absolute configurations were determined based on ECD spectra in comparison with those calculated using the time-dependent density functional theory. Preliminary examination of affinity for the GABAA receptor revealed that the (a1R, a2S) isomer of 1 possessed higher activity than its antipode (a1S, a2R) isomer. The active configuration of C9-methylated quazepam 1 is the same as that of 1,4-benzodiazepin-2-ones.


Subject(s)
Receptors, GABA-A , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Stereoisomerism , Structure-Activity Relationship , Molecular Structure , Humans , Benzodiazepinones/chemistry , Benzodiazepinones/pharmacology , Benzodiazepinones/chemical synthesis , Density Functional Theory
6.
ChemMedChem ; 19(15): e202400135, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38687623

ABSTRACT

Tetrahydrolinalool (THL) is an acyclic monoterpene alcohol, produced during linalol metabolism and also a constituent of essential oils. As described in the literature, many monoterpenes present anticonvulsant properties, and thus we became interested in evaluating the anticonvulsant activity of Tetrahydrolinalool using in mice model as well as in silico approaches. Our results demonstrated that THL increased latency to seizure onset and also reduced the mortality, in picrotoxin induced seizure tests. The results may be related to GABAergic regulation, which was also suggested in seizure testing induced by 3-mercapto-propionic acid. In the strychnine-induced seizure testing, none of the groups pretreated with THL modulated the parameters indicative of anticonvulsant effect. The electrophysiological results revealed that THL treatment reduces seizures induced by pentylenetetrazole. The in silico molecular docking studies showed that the interaction between THL and a GABAA receptor model formed a stable complex, in comparison to the crystaligraphic structure of diazepam, a structurally related ligand. In conclusion, all the evidences showed that THL presents effective anticonvulsant activity related to the GABAergic pathway, being a candidate for treatment of epileptic syndromes.


Subject(s)
Acyclic Monoterpenes , Anticonvulsants , Molecular Docking Simulation , Monoterpenes , Pentylenetetrazole , Seizures , Anticonvulsants/pharmacology , Anticonvulsants/chemistry , Anticonvulsants/chemical synthesis , Animals , Mice , Seizures/drug therapy , Monoterpenes/pharmacology , Monoterpenes/chemistry , Monoterpenes/chemical synthesis , Acyclic Monoterpenes/pharmacology , Acyclic Monoterpenes/chemistry , Acyclic Monoterpenes/chemical synthesis , Male , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Structure-Activity Relationship , Behavior, Animal/drug effects , Picrotoxin/pharmacology
7.
Biophys J ; 123(14): 2085-2096, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38400541

ABSTRACT

GABAA receptors (GABAARs) are neurotransmitter-gated ion channels critical for inhibitory synaptic transmission as well as the molecular target for benzodiazepines (BZDs), one of the most widely prescribed class of psychotropic drugs today. Despite structural insight into the conformations underlying functional channel states, the detailed molecular interactions involved in conformational transitions and the physical basis for their modulation by BZDs are not fully understood. We previously identified that alanine substitution at the central residue in the α1 subunit M2-M3 linker (V279A) enhances the efficiency of linkage between the BZD site and the pore gate. Here, we expand on this work by investigating the effect of alanine substitutions at the analogous positions in the M2-M3 linkers of ß2 (I275A) and γ2 (V290A) subunits, which together with α1 comprise typical heteromeric α1ß2γ2 synaptic GABAARs. We find that these mutations confer subunit-specific effects on the intrinsic pore closed-open equilibrium and its modulation by the BZD diazepam (DZ). The mutations α1(V279A) or γ2(V290A) bias the channel toward a closed conformation, whereas ß2(I275A) biases the channel toward an open conformation to the extent that the channel becomes leaky and opens spontaneously in the absence of agonist. In contrast, only α1(V279A) enhances the efficiency of DZ-to-pore linkage, whereas mutations in the other two subunits have no effect. These observations show that the central residue in the M2-M3 linkers of distinct subunits in synaptic α1ß2γ2 GABAARs contribute asymmetrically to the intrinsic closed-open equilibrium and its modulation by DZ.


Subject(s)
Diazepam , Ion Channel Gating , Protein Subunits , Receptors, GABA-A , Receptors, GABA-A/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Diazepam/pharmacology , Diazepam/chemistry , Ion Channel Gating/drug effects , Protein Subunits/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Humans , Animals , Mutation , HEK293 Cells
8.
Mol Inform ; 43(2): e202300156, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37964718

ABSTRACT

Numerous studies reported an association between GABAA R subunit genes and epilepsy, eating disorders, autism spectrum disorders, neurodevelopmental disorders, and bipolar disorders. This study was aimed to find some potential positive allosteric modulators and was performed by combining the in silico approach with further in vitro evaluation of its real activity. We started from the GABAA R-diazepam complexes and assembled a lipid embedded protein ensemble to refine it via molecular dynamics (MD) simulation. Then we focused on the interaction of α1ß2γ2 with some Z-drugs (non-benzodiazepine compounds) using an Induced Fit Docking (IFD) into the relaxed binding site to generate a pharmacophore model. The pharmacophore model was validated with a reference set and applied to decrease the pre-filtered Enamine database before the main docking procedure. Finally, we succeeded in identifying a set of compounds, which met all features of the docking model. The aqueous solubility and stability of these compounds in mouse plasma were assessed. Then they were tested for the biological activity using the rat Purkinje neurons and CHO cells with heterologously expressed human α1ß2γ2 GABAA receptors. Whole-cell patch clamp recordings were used to reveal the GABA induced currents. Our study represents a convenient and tunable model for the discovery of novel positive allosteric modulators of GABAA receptors. A High-throughput virtual screening of the largest available database of chemical compounds resulted in the selection of 23 compounds. Further electrophysiological tests allowed us to determine a set of 3 the most outstanding active compounds. Considering the structural features of leader compounds, the study can develop into the MedChem project soon.


Subject(s)
Receptors, GABA-A , gamma-Aminobutyric Acid , Animals , Rats , Mice , Humans , Cricetinae , Cricetulus , Workflow , Allosteric Regulation , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/pharmacology
9.
Nature ; 622(7981): 195-201, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37730991

ABSTRACT

Type A γ-aminobutyric acid receptors (GABAARs) are the principal inhibitory receptors in the brain and the target of a wide range of clinical agents, including anaesthetics, sedatives, hypnotics and antidepressants1-3. However, our understanding of GABAAR pharmacology has been hindered by the vast number of pentameric assemblies that can be derived from 19 different subunits4 and the lack of structural knowledge of clinically relevant receptors. Here, we isolate native murine GABAAR assemblies containing the widely expressed α1 subunit and elucidate their structures in complex with drugs used to treat insomnia (zolpidem (ZOL) and flurazepam) and postpartum depression (the neurosteroid allopregnanolone (APG)). Using cryo-electron microscopy (cryo-EM) analysis and single-molecule photobleaching experiments, we uncover three major structural populations in the brain: the canonical α1ß2γ2 receptor containing two α1 subunits, and two assemblies containing one α1 and either an α2 or α3 subunit, in which the single α1-containing receptors feature a more compact arrangement between the transmembrane and extracellular domains. Interestingly, APG is bound at the transmembrane α/ß subunit interface, even when not added to the sample, revealing an important role for endogenous neurosteroids in modulating native GABAARs. Together with structurally engaged lipids, neurosteroids produce global conformational changes throughout the receptor that modify the ion channel pore and the binding sites for GABA and insomnia medications. Our data reveal the major α1-containing GABAAR assemblies, bound with endogenous neurosteroid, thus defining a structural landscape from which subtype-specific drugs can be developed.


Subject(s)
Cryoelectron Microscopy , Neurosteroids , Receptors, GABA-A , gamma-Aminobutyric Acid , Animals , Mice , Binding Sites/drug effects , Depression, Postpartum/drug therapy , Flurazepam/pharmacology , gamma-Aminobutyric Acid/metabolism , Hypnotics and Sedatives/pharmacology , Ion Channel Gating/drug effects , Neurosteroids/metabolism , Neurosteroids/pharmacology , Photobleaching , Pregnanolone/pharmacology , Protein Conformation/drug effects , Protein Subunits/chemistry , Protein Subunits/drug effects , Protein Subunits/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, GABA-A/ultrastructure , Sleep Initiation and Maintenance Disorders/drug therapy , Zolpidem/pharmacology
10.
J Phys Chem B ; 127(28): 6306-6315, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37432029

ABSTRACT

General anesthetics are indispensable in modern medicine because they induce a reversible loss of consciousness and sensation in humans. On the other hand, their molecular mechanisms of action have not yet been elucidated. Several studies have identified the main targets of some general anesthetics. The structures of γ-aminobutyric acid A (GABAA) receptors with the intravenous anesthetics such as propofol and etomidate have recently been determined. Although these anesthetic binding structures provide essential insights into the mechanism of action of anesthetics, the detailed molecular mechanism of how the anesthetic binding affects the Cl- permeability of GABAA receptors remains to be elucidated. In this study, we performed coarse-grained molecular dynamics simulations for GABAA receptors and analyzed the resulting simulation trajectories to investigate the effects of anesthetic binding on the motion of GABAA receptors. The results showed large structural fluctuations in GABAA receptors, correlations of motion between the amino acid residues, large amplitude motion, and autocorrelated slow motion, which were obtained by advanced statistical analyses. In addition, a comparison of the resulting trajectories in the presence or absence of the anesthetic molecules revealed a characteristic pore motion related to the gate-opening motion of GABAA receptors.


Subject(s)
Anesthetics, General , Propofol , Humans , Receptors, GABA-A/chemistry , Molecular Dynamics Simulation , Anesthetics, Intravenous/pharmacology , Propofol/pharmacology , Propofol/chemistry , Anesthetics, General/pharmacology , gamma-Aminobutyric Acid
11.
Cell Biol Toxicol ; 39(6): 2793-2819, 2023 12.
Article in English | MEDLINE | ID: mdl-37093397

ABSTRACT

GABAA receptors, members of the pentameric ligand-gated ion channel superfamily, are widely expressed in the central nervous system and mediate a broad range of pharmaco-toxicological effects including bidirectional changes to seizure threshold. Thus, detection of GABAA receptor-mediated seizure liabilities is a big, partly unmet need in early preclinical drug development. This is in part due to the plethora of allosteric binding sites that are present on different subtypes of GABAA receptors and the critical lack of screening methods that detect interactions with any of these sites. To improve in silico screening methods, we assembled an inventory of allosteric binding sites based on structural data. Pharmacophore models representing several of the binding sites were constructed. These models from the NeuroDeRisk IL Profiler were used for in silico screening of a compiled collection of drugs with known GABAA receptor interactions to generate testable hypotheses. Amoxapine was one of the hits identified and subjected to an array of in vitro assays to examine molecular and cellular effects on neuronal excitability and in vivo locomotor pattern changes in zebrafish larvae. An additional level of analysis for our compound collection is provided by pharmacovigilance alerts using FAERS data. Inspired by the Adverse Outcome Pathway framework, we postulate several candidate pathways leading from specific binding sites to acute seizure induction. The whole workflow can be utilized for any compound collection and should inform about GABAA receptor-mediated seizure risks more comprehensively compared to standard displacement screens, as it rests chiefly on functional data.


Subject(s)
Receptors, GABA-A , Zebrafish , Animals , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Seizures/chemically induced , Binding Sites , gamma-Aminobutyric Acid
12.
J Biochem Mol Toxicol ; 37(1): e23234, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36184906

ABSTRACT

A new series of spiro[indene-1,2'-quinazolin]-4'(3'H)-one derivatives 4a-m were synthesized via a one-pot method and evaluated for anticonvulsant activities using pentylenetetrazole (PTZ) and maximal electroshock (MES)-induced seizures. Obtained results demonstrated that these compounds have not anticonvulsant activity in PTZ test while are active in the MES test. Among the synthesized compounds, the best anticonvulsant activity was obtained with compound 4h. This compound also was not neurotoxic. Given that the title new compounds have the pharmacophore requirement for benzodiazepine (BZD) receptor agonist, the most potent compound was assayed in vivo and in silico as BZD receptor agonist. After treatment with flumazenil as a standard BZD receptor antagonist, anticonvulsant activity of compound 4h decreased. Therefore, the involvement of BZD receptors in anticonvulsant activity of this compound confirmed. Furthermore, docking study of compound 4h in the BZD-binding site of GABAA receptor confirmed that this compound interacted with the important residues.


Subject(s)
Anticonvulsants , Seizures , Humans , Anticonvulsants/pharmacology , Anticonvulsants/chemistry , Binding Sites , Molecular Docking Simulation , Pentylenetetrazole , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/therapeutic use , Seizures/drug therapy , Structure-Activity Relationship
13.
Epilepsy Res ; 189: 107070, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36584483

ABSTRACT

Epilepsy is a complex neurological disease that can be caused by both genetic and environmental factors. Many studies have been conducted to investigate the genetic risk variants and molecular mechanisms of epilepsy. Disruption of excitation-inhibition balance (E/I balance) is one of the widely accepted disease mechanisms of epilepsy. The maintenance of E/I balance is an intricate process that is governed by multiple proteins. Using whole exome sequencing (WES), we identified a novel GABRA1 c.448G>A (p.E150K) variant and ERBB4 c.1972A>T (p.I658F, rs190654033) variant in a Malaysian Chinese family with genetic generalized epilepsy (GGE). The GGE may be triggered by dysregulation of E/I balance mechanism. Segregation of the variants in the family was verified by Sanger sequencing. All family members with GGE inherited both variants. However, family members who carried only one of the variants did not show any symptoms of GGE. Both the GABRA1 and ERBB4 variants were predicted damaging by MutationTaster and CADD, and protein structure analysis showed that the variants had resulted in the formation of additional hydrogen bonds in the mutant proteins. GABRA1 variant could reduce the efficiency of GABAA receptors, and constitutively active ERBB4 receptors caused by the ERBB4 variant promote internalization of GABAA receptors. The interaction between the two variants may cause a greater disruption in E/I balance, which is more likely to induce a seizure. Nevertheless, this disease model was derived from a single small family, further studies are still needed to confirm the verifiability of the purported disease model.


Subject(s)
Epilepsy, Generalized , Epilepsy , Humans , Epilepsy, Generalized/genetics , Epilepsy/genetics , Seizures , Family , Receptors, GABA-A/genetics , Receptors, GABA-A/chemistry , gamma-Aminobutyric Acid , Receptor, ErbB-4/genetics
14.
Eur J Med Chem ; 244: 114780, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36279694

ABSTRACT

The family of GABA-A receptors contains nineteen mammalian subunits from which pentameric, GABA gated anion channels are assembled. The subunit encoded by the GABRA6 gene is highly expressed in the cerebellum and the receptors to which it contributes have recently been demonstrated to be a promising candidate as a novel drug target. Here we examined a series of loreclezole derivatives for potentially selective action at α6ß3γ2 receptors with the help of computational methods and functional testing with the two-electrode voltage clamp technique. The synthetic routes to some previously published ligands were improved, and a new derivative was synthesized based on computational docking results. This new loreclezole derivative, [3-(2-chloro-4-methylphenyl)-3-methylbutanenitrile (40)], was shown to display stronger modulatory action in concatenated α6ß3γ2 receptors compared to their α1ß3γ2 counterpart. The hypothetical bound state structure provides valuable guidance for future design of selective therapeutics.


Subject(s)
Receptors, GABA-A , Triazoles , Ligands , Patch-Clamp Techniques , Receptors, GABA-A/chemistry , Triazoles/chemistry , Triazoles/pharmacology , Allosteric Regulation , Protein Conformation , Humans
15.
Basic Clin Pharmacol Toxicol ; 131(6): 514-524, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36180380

ABSTRACT

Several pyrazoloquinolinone (PQ) ligands were recently discovered as functionally selective positive modulators at the PQ site of α6-containing GABAA receptors. PQs are also neutral modulators at the benzodiazepine site. We assessed the influence of PQ compounds from three structural groups (PZ-II-029 and related deuterated analogues DK-I-56-1, RV-I-029, DK-I-60-3 and DK-I-86-1; LAU 463 and related analogues DK-I-58-1 and DK-II-58-1; and DK-I-87-1), alone and in combination with diazepam, on the behaviour of male Sprague-Dawley rats. An excellent behavioural safety profile of all tested PQs was demonstrated in the spontaneous locomotor activity, rotarod, loss of righting reflex and pentylenetetrazol tests. In interaction studies, only PZ-II-029 and its analogues prevented the ataxic effects of the benzodiazepine, as assessed in the rotarod test and during monitoring of rat locomotor activity after awakening from the loss of righting reflex. Published electrophysiological profiles of PQ ligands imply that positive modulation elicited at α6-GABAA receptors that contain the γ2 and δ subunit, rather than their neutral modulatory action at the benzodiazepine site, may prevent the ataxic action of diazepam. Thus, PZ-II-029 and its deuterated analogues are not prone to untoward interactions with benzodiazepines and may indeed completely abolish their ataxic action, seen at therapeutic, and especially toxic concentrations.


Subject(s)
Diazepam , Receptors, GABA-A , Animals , Rats , Male , Diazepam/pharmacology , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Benzodiazepines/pharmacology , Ligands , gamma-Aminobutyric Acid , Ataxia , GABA Modulators
16.
J Steroid Biochem Mol Biol ; 224: 106158, 2022 11.
Article in English | MEDLINE | ID: mdl-35931327

ABSTRACT

γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter. GABA receptor type A (GABAAR) possesses binding sites for a large group of pharmacological agents which are supposed to interact allosterically with each other. The aim of this work was to study the interaction between the positive allosteric modulators (PAMs) and the competitive antagonists of GABAARs. The GABA-induced chloride current (IGABA) was measured in isolated Purkinje cells of rat cerebellum using the patch-clamp technique. PAMs, neurosteroid allopregnanolone (Allo) and zolpidem (Zolp), a drug that positively modulates the GABAAR through interaction with the benzodiazepine (BDZ) site, doubled the IGABA amplitude in the control solution. Competitive antagonist of GABAARs, bicuculline (Bic, 5 µM) blocked the IGABA by 90%. The addition of 1 µM Allo or 0.5 µM Zolp to the Bic solution caused an unblocking effect, so that the IGABA amplitude increased 10 and 4 times from control value, correspondingly. This unblocking effect developed slowly, as evidenced by a threefold increase in the current rise time. Competitive antagonist of GABAARs, gabazine (GBZ, 0.5 µM) blocked the IGABA by 87%. The addition of 1 µM Allo to the GBZ solution caused an unblocking effect, so that the IGABA amplitude increased 7-fold. However, the addition of 0.5 µM Zolp to the GBZ solution did not cause an unblocking effect. So, Allo appeared to have a stronger unblocking potential than Zolp, and Bic binding site showed a higher sensitivity to the action of unblocking PAMs than GBZ binding site. The results indicate for the first time the existence of an allosteric relationship between the sites binding PAMs and the competitive antagonists of GABAAR.


Subject(s)
Chlorides , Receptors, GABA-A , Rats , Animals , Receptors, GABA-A/chemistry , Chlorides/metabolism , Ligands , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology , Pregnanolone/pharmacology , GABA-A Receptor Antagonists/pharmacology
17.
Biomolecules ; 12(7)2022 06 21.
Article in English | MEDLINE | ID: mdl-35883422

ABSTRACT

GABAA receptors are a major contributor to fast inhibitory neurotransmission in the brain. The receptors are activated upon binding the transmitter GABA or allosteric agonists including a number of GABAergic anesthetics and neurosteroids. Functional receptors can be formed by various combinations of the nineteen GABAA subunits cloned to date. GABAA receptors containing the ε subunit exhibit a significant degree of constitutive activity and have been suggested to be unresponsive to allosteric agents. In this study, we have characterized the functional properties of the rat α1ß2ε GABAA receptor. We confirm that the α1ß2ε receptor exhibits a higher level of constitutive activity than typical of GABAA receptors and show that it is inefficaciously activated by the transmitter and the allosteric agonists propofol, pentobarbital, and allopregnanolone. Manipulations intended to alter ε subunit expression and receptor stoichiometry were largely without effect on receptor properties including sensitivity to GABA and allosteric agonists. Surprisingly, amino acid substitutions at the conserved 9' and 6' positions in the second transmembrane (TM2) domain in the ε subunit did not elicit the expected functional effects of increased constitutive activity and resistance to the channel blocker picrotoxin, respectively. We tested the accessibility of TM2 residues mutated to cysteine using the cysteine-modifying reagent 4-(hydroxymercuri)benzoic acid and found a unique pattern of water-accessible residues in the ε subunit.


Subject(s)
Propofol , Receptors, GABA-A , Animals , Cysteine , Pentobarbital/metabolism , Pentobarbital/pharmacology , Propofol/pharmacology , Rats , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , gamma-Aminobutyric Acid/metabolism
18.
Angew Chem Int Ed Engl ; 61(30): e202205198, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35482315

ABSTRACT

GABAA (γ-aminobutyric acid type A) receptors are ligand-gated ion channels mediating fast inhibitory transmission in the mammalian brain. Here we report the molecular and electronic mechanism governing the turn-on emission of a fluorescein-based imaging probe able to target the human GABAA receptor. Multiscale calculations evidence a drastic conformational change of the probe from folded in solution to extended upon binding to the receptor. Intramolecular ππ-stacking interactions present in the folded probe are responsible for quenching fluorescence in solution. In contrast, unfolding within the GABAA receptor changes the nature of the bright excited state triggering emission. Remarkably, this turn-on effect only manifests for the dianionic prototropic form of the imaging probe, which is found to be the strongest binder to the GABAA receptor. This study is expected to assist the design of new photoactivatable screening tools for allosteric modulators of the GABAA receptor.


Subject(s)
Receptors, GABA-A , gamma-Aminobutyric Acid , Animals , Fluorescein , Fluorescence , Humans , Mammals/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/pharmacology
19.
Int J Mol Sci ; 23(5)2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35269865

ABSTRACT

Mutations of GABAAR have reportedly led to epileptic encephalopathy and neurodevelopmental disorders. We have identified a novel de novo T292S missense variant of GABRA1 from a pediatric patient with grievous global developmental delay but without obvious epileptic activity. This mutation coincidentally occurs at the same residue as that of a previously reported GABRA1 variant T292I identified from a pediatric patient with severe epilepsy. The distinct phenotypes of these two patients prompted us to compare the impacts of the two mutants on the receptor function and to search for suitable therapeutics. In this study, we used biochemical techniques and patch-clamp recordings in HEK293 cells overexpressing either wild-type or mutated rat recombinant GABAARs. We found that the α1T292S variant significantly increased GABA-evoked whole-cell currents, shifting the dose-response curve to the left without altering the maximal response. In contrast, the α1T292I variant significantly reduced GABA-evoked currents, shifting the dose-response curve to the right with a severely diminished maximum response. Single-channel recordings further revealed that the α1T292S variant increased, while the α1T292I variant decreased the GABAAR single-channel open time and open probability. Importantly, we found that the T292S mutation-induced increase in GABAAR function could be fully normalized by the negative GABAAR modulator thiocolchicoside, whereas the T292I mutation-induced impairment of GABAAR function was largely rescued with a combination of the GABAAR positive modulators diazepam and verapamil. Our study demonstrated that α1T292 is a critical residue for controlling GABAAR channel gating, and mutations at this residue may produce opposite impacts on the function of the receptors. Thus, the present work highlights the importance of functionally characterizing each individual GABAAR mutation for ensuring precision medicine.


Subject(s)
Epilepsy, Generalized , Epilepsy , Neurodevelopmental Disorders , Animals , Child , Epilepsy/genetics , HEK293 Cells , Humans , Mutation , Rats , Receptors, GABA-A/chemistry , gamma-Aminobutyric Acid/genetics
20.
Nature ; 604(7904): 190-194, 2022 04.
Article in English | MEDLINE | ID: mdl-35355020

ABSTRACT

Type A γ-aminobutyric acid receptors (GABAARs) are pentameric ligand-gated chloride channels that mediate fast inhibitory signalling in neural circuits1,2 and can be modulated by essential medicines including general anaesthetics and benzodiazepines3. Human GABAAR subunits are encoded by 19 paralogous genes that can, in theory, give rise to 495,235 receptor types. However, the principles that govern the formation of pentamers, the permutational landscape of receptors that may emerge from a subunit set and the effect that this has on GABAergic signalling remain largely unknown. Here we use cryogenic electron microscopy to determine the structures of extrasynaptic GABAARs assembled from α4, ß3 and δ subunits, and their counterparts incorporating γ2 instead of δ subunits. In each case, we identified two receptor subtypes with distinct stoichiometries and arrangements, all four differing from those previously observed for synaptic, α1-containing receptors4-7. This, in turn, affects receptor responses to physiological and synthetic modulators by creating or eliminating ligand-binding sites at subunit interfaces. We provide structural and functional evidence that selected GABAAR arrangements can act as coincidence detectors, simultaneously responding to two neurotransmitters: GABA and histamine. Using assembly simulations and single-cell RNA sequencing data8,9, we calculated the upper bounds for receptor diversity in recombinant systems and in vivo. We propose that differential assembly is a pervasive mechanism for regulating the physiology and pharmacology of GABAARs.


Subject(s)
Benzodiazepines , Receptors, GABA-A , Signal Transduction , Benzodiazepines/pharmacology , Binding Sites , Cryoelectron Microscopy , Histamine/metabolism , Humans , Ligands , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA-Seq , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/ultrastructure , Single-Cell Analysis , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL