Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nature ; 606(7915): 820-826, 2022 06.
Article in English | MEDLINE | ID: mdl-35676483

ABSTRACT

γ-Aminobutyric acid (GABA) transporter 1 (GAT1)1 regulates neuronal excitation of the central nervous system by clearing the synaptic cleft of the inhibitory neurotransmitter GABA upon its release from synaptic vesicles. Elevating the levels of GABA in the synaptic cleft, by inhibiting GABA reuptake transporters, is an established strategy to treat neurological disorders, such as epilepsy2. Here we determined the cryo-electron microscopy structure of full-length, wild-type human GAT1 in complex with its clinically used inhibitor tiagabine3, with an ordered part of only 60 kDa. Our structure reveals that tiagabine locks GAT1 in the inward-open conformation, by blocking the intracellular gate of the GABA release pathway, and thus suppresses neurotransmitter uptake. Our results provide insights into the mixed-type inhibition of GAT1 by tiagabine, which is an important anticonvulsant medication. Its pharmacodynamic profile, confirmed by our experimental data, suggests initial binding of tiagabine to the substrate-binding site in the outward-open conformation, whereas our structure presents the drug stalling the transporter in the inward-open conformation, consistent with a two-step mechanism of inhibition4. The presented structure of GAT1 gives crucial insights into the biology and pharmacology of this important neurotransmitter transporter and provides blueprints for the rational design of neuromodulators, as well as moving the boundaries of what is considered possible in single-particle cryo-electron microscopy of challenging membrane proteins.


Subject(s)
GABA Plasma Membrane Transport Proteins , GABA Uptake Inhibitors , gamma-Aminobutyric Acid , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Cryoelectron Microscopy , GABA Plasma Membrane Transport Proteins/chemistry , GABA Plasma Membrane Transport Proteins/metabolism , GABA Plasma Membrane Transport Proteins/ultrastructure , GABA Uptake Inhibitors/chemistry , GABA Uptake Inhibitors/pharmacology , Humans , Neurotransmitter Agents/metabolism , Protein Conformation/drug effects , Tiagabine/chemistry , Tiagabine/metabolism , Tiagabine/pharmacology , gamma-Aminobutyric Acid/metabolism
2.
Molecules ; 25(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081136

ABSTRACT

The human gamma aminobutyric acid transporter subtype 1 (hGAT1) located in the nerve terminals is known to catalyze the neuronal function by the electrogenic reuptake of γ-aminobutyric acid (GABA) with the co-transport of Na+ and Cl- ions. In the past, there has been a major research drive focused on the dysfunction of hGAT1 in several neurological disorders. Thus, hGAT1 of the GABAergic system has been well established as an attractive target for such diseased conditions. Till date, there are various reports about stereo selectivity of -COOH group of tiagabine, a Food and Drug Administration (FDA)-approved hGAT1-selective antiepileptic drug. However, the effect of the stereochemistry of the protonated -NH group of tiagabine has never been scrutinized. Therefore, in this study, tiagabine has been used to explore the binding hypothesis of different enantiomers of tiagabine. In addition, the impact of axial and equatorial configuration of the-COOH group attached at the meta position of the piperidine ring of tiagabine enantiomers was also investigated. Further, the stability of the finally selected four hGAT1-tiagabine enantiomers namely entries 3, 4, 6, and 9 was evaluated through 100 ns molecular dynamics (MD) simulations for the selection of the best probable tiagabine enantiomer. The results indicate that the protonated -NH group in the R-conformation and the -COOH group of Tiagabine in the equatorial configuration of entry 4 provide maximum strength in terms of interaction within the hGAT1 binding pocket to prevent the change in hGAT1 conformational state, i.e., from open-to-out to open-to-in as compared to other selected tiagabine enantiomers 3, 6, and 9.


Subject(s)
GABA Agents/chemistry , GABA Plasma Membrane Transport Proteins/chemistry , Tiagabine/chemistry , gamma-Aminobutyric Acid/chemistry , GABA Agents/pharmacology , Humans , Molecular Dynamics Simulation , Protein Binding/drug effects , Stereoisomerism , Tiagabine/pharmacology
3.
Anal Chem ; 92(5): 3555-3562, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32008316

ABSTRACT

Tiagabine hydrochloride (TGB) is a clinically frequently used drug for anticonvulsion and reducing epileptic frequency. Over administration of TGB could bring about adverse effects, such as speech disorder, depression, and even suicidal tendencies. Therefore, accessible and sensitive assay for analysis of TGB becomes an urgent need toward guiding clinical medication. Here, we present the first report on fluorescence turn-on detection of TGB in urine testing. In this protocol, a fluorescent dye, perylene tetracarboxylic acid imide derivative (PTAI), is found specifically occupying the Sudlow site II of human serum albumin (HSA) and displays a new phenomenon of binding-induced quenching (BIQ). In presence of TGB, competitive binding of the TGB to the site II of HSA will trigger release of PTAI, thus successfully lighting up the fluorescence of PTAI. This label-free assay enjoys a broader working range (1-350 µM) and lower detection limit (0.218 µM) than the traditional liquid chromatography method and is uninterfered by the miscellaneous in the artificial urine. The BIQ probe highlights the merits of HSA as a quencher and a molecular recognition unit, and it opens up a way for studying drug-HSA interaction mechanism and noninvasive pharmaceutical testing.


Subject(s)
Anticonvulsants/analysis , Anticonvulsants/chemistry , Biosensing Techniques/methods , Serum Albumin, Human/chemistry , Tiagabine/analysis , Tiagabine/chemistry , Anticonvulsants/urine , Buffers , Humans , Models, Molecular , Protein Conformation , Spectrometry, Fluorescence , Tiagabine/urine
4.
ACS Chem Neurosci ; 10(1): 337-347, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30222312

ABSTRACT

In this paper, we describe the latest results involving molecular modeling and pharmacodynamic studies of the selected highly lipophilic compounds acting by human GABA transporter 1 (hGAT1) inhibition. The chemical interaction of 17 GABA analogues with a model of hGAT1 is described using the molecular docking method. The biological role of GAT1 is related to the regulation of GABA level in the central nervous system and GAT1 inhibition plays an important role in the control of seizure threshold. To confirm that GAT1 can be also a molecular target for drugs used to treat other neurological and psychiatric diseases (e.g., pain and anxiety), in the in vivo part of this study, potential antinociceptive and anxiolytic-like properties of tiagabine, a selective GAT1 inhibitor, are described.


Subject(s)
GABA Plasma Membrane Transport Proteins/metabolism , GABA Uptake Inhibitors/administration & dosage , Lipids/administration & dosage , Molecular Docking Simulation/methods , Animals , Anxiety/drug therapy , Anxiety/psychology , GABA Uptake Inhibitors/chemistry , Humans , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Structure-Activity Relationship , Tiagabine/administration & dosage , Tiagabine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...