Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Br J Pharmacol ; 178(24): 4842-4858, 2021 12.
Article in English | MEDLINE | ID: mdl-34386973

ABSTRACT

BACKGROUND AND PURPOSE: In addition to binding to the classical high-affinity extracellular benzodiazepine binding site of the GABAA receptor, some benzodiazepines occupy transmembrane inter-subunit anaesthetic sites that bind etomidate (ß+ /α- sites) or the barbiturate derivative R-mTFD-MPAB (α+ /ß- and γ+ /ß- sites). We aimed to define the functional effects of these interactions on GABAA receptor activity and animal behaviour. EXPERIMENTAL APPROACH: With flumazenil blocking classical high-affinity extracellular benzodiazepine site effects, modulation of GABA-activated currents by diazepam, midazolam and flurazepam was measured electrophysiologically in wildtype and M2-15' mutant α1 ß3 γ2L GABAA receptors. Zebrafish locomotive activity was also assessed in the presence of each benzodiazepine plus flumazenil. KEY RESULTS: In the presence of flumazenil, micromolar concentrations of diazepam and midazolam both potentiated and inhibited wildtype GABAA receptor currents. ß3 N265M (M2-15' in the ß+ /α- sites) and α1 S270I (M2-15' in the α+ /ß- site) mutations reduced or abolished potentiation by these drugs. In contrast, the γ2 S280W mutation (M2-15' in the γ+ /ß- site) abolished inhibition. Flurazepam plus flumazenil only inhibited wildtype receptor currents, an effect unaltered by M2-15' mutations. In the presence of flumazenil, zebrafish locomotion was enhanced by diazepam at concentrations up to 30 µM and suppressed at 100 µM, suppressed by midazolam and enhanced by flurazepam. CONCLUSIONS AND IMPLICATIONS: Benzodiazepine binding to transmembrane anaesthetic binding sites of the GABAA receptor can produce positive or negative modulation manifesting as decreases or increases in locomotion, respectively. Selectivity for these sites may contribute to the distinct GABAA receptor and behavioural actions of different benzodiazepines, particularly at high (i.e. anaesthetic) concentrations.


Subject(s)
Anesthetics , Receptors, GABA-A , Animals , Anesthetics/pharmacology , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Binding Sites , Flumazenil/chemistry , Flumazenil/pharmacology , Receptors, GABA-A/metabolism , Zebrafish/metabolism
2.
Mol Biochem Parasitol ; 242: 111350, 2021 03.
Article in English | MEDLINE | ID: mdl-33422580

ABSTRACT

Parasitic infections are a widespread health problem and research of novel anthelmintic compounds is of the utmost importance. In this study we performed a virtual screening campaign by coupling ligand-based pharmacophore, homology modeling and molecular docking. The virtual screening campaign was conducted using a joined pool of the Drugbank database and a library of purchasable compounds in order to identify drug like compounds with similar pharmacological activity. Our aim was to identify compounds with a potential antihelmintic modulatory effect on nicotinic acetylcholine receptors (nAChR). We derived a 3D pharmacophore model based on the chemical features of known Ascaris suum nAChR modulators. To evaluate the in silico predictions, we tested selected hit compounds in contraction assays using somatic muscle flaps of the Ascaris suum neuromuscular tissue. We tested the modulatory effects of GSK575594A, diazepam and flumazenil hit compounds on nematode contractions induced by acetyl choline (ACh). The compound GSK575594A (3 µM) increased the Emax by 21 % with the EC50 dose ratio of 0.96. Diazepam (100 µM) decreased the Emax by 15 % (1.11 g to 0.95 g) with the EC50 ratio of 1.42 (shifted to the left from 11.25 to 7.93). Flumazenil decreased the EC50 value (from 11.22 µM to 4.88 µM) value showing dose ratio of 2.30, and increased the Emax by 4 % (from 1.54 g to 1.59 g). The observed biological activity was rationalized by molecular docking calculations. Docking scores were calculated against several binding sites within the Ascaris suum homology model. We constructed the homology model using the ACR-16 subunit sequence. The compound GSK575594A showed strong affinity for the intersubunit allosteric binding site within the nAChR transmembrane domain. The binding modes of diazepam and flumazenil suggest that these compounds have a comparable affinity for orthosteric and allosteric nAChR binding sites. The selected hit compounds displayed potential for further optimization as lead compounds. Therefore, such compounds may be useful in neutralizing the growing resistance of parasites to drugs, either alone or in combination with existing conventional anthelmintics.


Subject(s)
Anthelmintics/pharmacology , Diazepam/pharmacology , Flumazenil/pharmacology , Helminth Proteins/chemistry , Muscles/drug effects , Piperazines/pharmacology , Receptors, Nicotinic/chemistry , Acetylcholine/pharmacology , Animals , Anthelmintics/chemistry , Ascaris suum/drug effects , Ascaris suum/metabolism , Binding Sites , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Databases, Chemical , Diazepam/chemistry , Flumazenil/chemistry , Gene Expression , Helminth Proteins/agonists , Helminth Proteins/genetics , Helminth Proteins/metabolism , High-Throughput Screening Assays , Humans , Ligands , Molecular Docking Simulation , Piperazines/chemistry , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Structural Homology, Protein , Tissue Culture Techniques , User-Computer Interface
3.
Curr Mol Pharmacol ; 14(1): 36-51, 2021.
Article in English | MEDLINE | ID: mdl-32386503

ABSTRACT

BACKGROUND: Central nervous system disorders such as anxiety, depression and epilepsy are characterized by sharing several molecular mechanisms in common and the involvement of the L-arginine/NO pathway in neurobehavioral studies with ß-caryophyllene is still little discussed. OBJECTIVES: One of the objectives of the present study was to demonstrate the anxiolytic behavioral effect of ß-caryophyllene (ß-CBP) in female Swiss mice, as well as to investigate the molecular mechanisms underlying the results obtained. METHODS: This study evaluated the neurobehavioral effects of ß-CBP using the open field test, rota- rod test, elevated plus maze test, novelty suppressed feeding test, tail suspension test and forced swim test, as well as pilocarpine, pentylenetetrazole and isoniazid-induced epileptic seizure models. RESULTS: The results demonstrated that the neuropharmacological activities of ß-CBP may involve benzodiazepine/GABAergic receptors, since the pre-treatment of ß-CBP (200 mg/kg) associated with flumazenil (5 mg/kg, benzodiazepine receptor antagonist) and bicuculline (1 mg/kg, selective GABAA receptor antagonist) reestablished the anxiety parameters in the elevated plus-maze test, as well as the results of reduced latency to consume food in the novelty suppressed feeding test. In addition to benzodiazepine/GABAergic receptors, the neuropharmacological properties of ß-CBP may be related to inhibition of nitric oxide synthesis, since pre-treatment with L-arginine (500-750 mg/kg) reversed significantly the anxiolytic, antidepressant and anticonvulsant activities of ß-CBP. CONCLUSION: The results obtained provide additional support in understanding the neuromolecular mechanisms underlying the anxiolytic, antidepressant and anticonvulsive properties of ß-CBP in female Swiss mice.


Subject(s)
Anti-Anxiety Agents/chemistry , Anticonvulsants/chemistry , Antidepressive Agents/chemistry , GABA-A Receptor Antagonists/chemistry , Polycyclic Sesquiterpenes/chemistry , Animals , Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Antidepressive Agents/pharmacology , Arginine , Behavior, Animal , Benzodiazepines/metabolism , Bicuculline/chemistry , Bicuculline/pharmacology , Female , Flumazenil/chemistry , Flumazenil/pharmacology , GABA-A Receptor Antagonists/pharmacology , Humans , Maze Learning , Mice , Nitric Oxide/metabolism , Polycyclic Sesquiterpenes/pharmacology , Receptors, GABA-A/metabolism , Seizures/chemically induced , Signal Transduction
4.
Nucl Med Biol ; 88-89: 44-51, 2020.
Article in English | MEDLINE | ID: mdl-32777548

ABSTRACT

INTRODUCTION: Prenatal ethanol exposure (PEE) has been shown to alter the level and function of receptors in the brain, one of which is GABAa receptors (GABAaR), the major inhibitory ligand gated ion channels that mediate neuronal inhibition. High dose PEE in animals resulted in the upregulation of GABAaR, but the effects of low and moderate dose PEE at early gestation have not been investigated. This study aimed at examining GABAaR density in the adult mouse brain following PEE during a period equivalent to the first 3 to 4 weeks in human gestation. It was hypothesized that early moderate PEE would cause alterations in brain GABAaR levels in the adult offspring. METHODS: C57BL/6J mice were given 10% v/v ethanol during the first 8 gestational days. Male offspring were studied using in-vivo Positron Emission Tomography (PET)/Magnetic Resonance Imaging (MRI), biodistribution, in-vitro autoradiography using [18F]AH114726, a novel flumazenil analogue with a high affinity for the benzodiazepine-binding site, and validated using immunohistochemistry. RESULTS: In vivo PET and biodistribution did not detect alteration in brain tracer uptake. In vitro radiotracer studies detected significantly reduced GABAaR in the olfactory bulbs. Immunohistochemistry detected reduced GABAaR in the cerebral cortex, cerebellum and hippocampus, while Nissl staining showed that cell density was significantly higher in the striatum following PEE. CONCLUSION: Early moderate PEE may induce long-term alterations in the GABAaR system that persisted into adulthood.


Subject(s)
Benzodiazepines/chemistry , Brain/metabolism , Ethanol/toxicity , Flumazenil/metabolism , Fluorine Radioisotopes/metabolism , Prenatal Exposure Delayed Effects/pathology , Receptors, GABA-A/metabolism , Animals , Central Nervous System Depressants/toxicity , Disease Models, Animal , Female , Flumazenil/chemistry , Male , Mice , Mice, Inbred C57BL , Positron-Emission Tomography/methods , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/diagnostic imaging , Prenatal Exposure Delayed Effects/metabolism , Radiopharmaceuticals/metabolism , Tissue Distribution
5.
Bioorg Med Chem Lett ; 28(17): 2925-2929, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30122224

ABSTRACT

We designed and synthesized deuterium-substituted [18F]fluoromethyl-PBR28 ([18F]1-d2) as a novel translocator protein 18 kDa (TSPO)-targeted radioligand with enhanced in vivo stability. The comparison studies between [18F]fluoromethyl-PBR28 ([18F]1) and its deuterate analog ([18F]1-d2) were investigated in terms of in vitro binding affinity, lipophilicity and in vivo stability. In addition, the accuracies of both radioligands were determined by comparing the PET imaging data in the same LPS-induced neuroinflammation rat model. Both aryloxyanilide analogs showed similar lipophilicity and in vitro affinity for TSPO. However, [18F]1-d2 provided significantly lower femur uptake than [18F]1 (1.5 ±â€¯1.2 vs. 4.1 ±â€¯1.7%ID/g at 2 h post-injection) in an ex vivo biodistribution study. [18F]1-d2 was also selectively accumulated in the inflammatory lesion with the binding potential of the specifically bound radioligand relative to the non-displaceable radioligand in tissue (BPND = 3.17 ±â€¯0.48), in a LPS-induced acute neuroinflammation rat model, comparable to that of [18F]1 (BPND = 2.13 ±â€¯0.51). These results indicate that [18F]1-d2 had higher in vivo stability, which resulted in an enhanced target-to-background ratio compared to that induced by [18F]1.


Subject(s)
Acetamides/pharmacokinetics , Aminopyridines/pharmacokinetics , Disease Models, Animal , Inflammation/diagnostic imaging , Neurodegenerative Diseases/diagnostic imaging , Radiopharmaceuticals/pharmacokinetics , Acetamides/chemical synthesis , Acetamides/chemistry , Aminopyridines/chemical synthesis , Aminopyridines/chemistry , Animals , Dose-Response Relationship, Drug , Flumazenil/chemistry , Flumazenil/pharmacokinetics , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred ICR , Molecular Structure , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Rats , Structure-Activity Relationship , Tissue Distribution
6.
ACS Chem Biol ; 13(8): 2040-2047, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29989390

ABSTRACT

The anxiolytic, anticonvulsant, muscle-relaxant, and sedative-hypnotic effects of benzodiazepine site ligands are mainly elicited by allosteric modulation of GABAA receptors via their extracellular αx+/γ2- ( x = 1, 2, 3, 5) interfaces. In addition, a low affinity binding site at the homologous α+/ß- interfaces was reported for some benzodiazepine site ligands. Classical benzodiazepines and pyrazoloquinolinones have been used as molecular probes to develop structure-activity relationship models for benzodiazepine site activity. Considering all possible α+/ß- and α+/γ- interfaces, such ligands potentially interact with as many as 36 interfaces, giving rise to undesired side effects. Understanding the binding modes at their binding sites will enable rational strategies to design ligands with desired selectivity profiles. Here, we compared benzodiazepine site ligand interactions in the high affinity α1+/γ2- site with the homologous α1+/ß3- site using a successive mutational approach. We incorporated key amino acids known to contribute to high affinity benzodiazepine binding of the γ2- subunit into the ß3- subunit, resulting in a quadruple mutant ß3(4mut) with high affinity flumazenil (Ro 15-1788) binding properties. Intriguingly, some benzodiazepine site ligands displayed positive allosteric modulation in the tested recombinant α1ß3(4mut) constructs while diazepam remained inactive. Consequently, we performed in silico molecular docking in the wildtype receptor and the quadruple mutant. The results led to the conclusion that different benzodiazepine site ligands seem to use distinct binding modes, rather than a common binding mode. These findings provide structural hypotheses for the future optimization of both benzodiazepine site ligands, and ligands that interact with the homologous α+/ß- sites.


Subject(s)
Flumazenil/chemistry , Receptors, GABA-A/chemistry , Animals , Binding Sites , Female , HEK293 Cells , Humans , Ligands , Models, Chemical , Molecular Docking Simulation , Mutation , Pyrazoles/chemistry , Pyridones/chemistry , Quinolones/chemistry , Receptors, GABA-A/genetics , Xenopus laevis
7.
Nature ; 559(7712): 67-72, 2018 07.
Article in English | MEDLINE | ID: mdl-29950725

ABSTRACT

Fast inhibitory neurotransmission in the brain is principally mediated by the neurotransmitter GABA (γ-aminobutyric acid) and its synaptic target, the type A GABA receptor (GABAA receptor). Dysfunction of this receptor results in neurological disorders and mental illnesses including epilepsy, anxiety and insomnia. The GABAA receptor is also a prolific target for therapeutic, illicit and recreational drugs, including benzodiazepines, barbiturates, anaesthetics and ethanol. Here we present high-resolution cryo-electron microscopy structures of the human α1ß2γ2 GABAA receptor, the predominant isoform in the adult brain, in complex with GABA and the benzodiazepine site antagonist flumazenil, the first-line clinical treatment for benzodiazepine overdose. The receptor architecture reveals unique heteromeric interactions for this important class of inhibitory neurotransmitter receptor. This work provides a template for understanding receptor modulation by GABA and benzodiazepines, and will assist rational approaches to therapeutic targeting of this receptor for neurological disorders and mental illness.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-A/chemistry , Receptors, GABA-A/ultrastructure , Benzodiazepines/antagonists & inhibitors , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Bicuculline/pharmacology , Binding, Competitive/drug effects , Brain Chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , Flumazenil/chemistry , Flumazenil/metabolism , Flumazenil/pharmacology , GABA Modulators/chemistry , GABA Modulators/metabolism , GABA Modulators/pharmacology , Glycosylation , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Ligands , Models, Molecular , Receptors, GABA-A/immunology , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
8.
Bioorg Med Chem ; 26(9): 2475-2487, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29650463

ABSTRACT

A comparison between compounds with pyrazolo[1,5-a]pyrimidine structure (series 4-6) and pyrazolo[5,1-c][1,2,4]triazine core (series 9) as ligands at GABAA-receptor subtype, was evaluated. Moreover, for pyrazolotriazine derivatives having binding recognition, the interaction on recombinant rat α(1-3,5) GABAA receptor subtypes, was performed. Among these latter, emerge compounds 9c, 9k, 9l, 9m and 9n as α1-selective and 9h as α2-selective ligands.


Subject(s)
Pyrazoles/pharmacology , Receptors, GABA-A/metabolism , Triazines/pharmacology , Animals , Binding Sites , Cattle , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Flumazenil/chemistry , Ligands , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rats , Receptors, GABA-A/chemistry , Triazines/chemical synthesis , Triazines/chemistry , Tritium
9.
Mol Imaging Biol ; 20(1): 114-123, 2018 02.
Article in English | MEDLINE | ID: mdl-28631142

ABSTRACT

PURPOSE: This study evaluates the performance of several parametric methods for assessing [11C]flumazenil binding distribution in the rat brain. PROCEDURES: Dynamic (60 min) positron emission tomography data with metabolite-corrected plasma input function were retrospectively analyzed (male Wistar rats, n = 10). Distribution volume (V T) images were generated from basis function method (BFM), Logan graphical analysis (Logan), and spectral analysis (SA). Using the pons as pseudo-reference tissue, binding potential (BP ND and DVR-1) images were obtained from receptor parametric imaging algorithms (RPM and SRTM2) and reference Logan (RLogan). Standardized uptake value images (SUV and SUVR) were also computed for different intervals post-injection. Next, regional averages were extracted from the parametric images, using pre-defined volumes of interest, which were also applied to the regional time-activity curves from the dynamic data. Parametric data were compared to their regional counterparts and to two-tissue compartment model (2TCM)-based values (previously defined as the model of choice for rats). Parameter agreement was assessed by linear regression analysis and Bland-Altman plots. RESULTS: All parametric methods strongly correlated to their regional counterparts (R 2 > 0.97) and to the 2TCM values (R 2 ≥ 0.95). SA and RLogan underestimated V T and BP ND (slope of 0.93 and 0.86, respectively), while SUVR-1 overestimated BP ND (slope higher than 1.07 for all intervals). While BFM and SRTM2 had the smallest bias to 2TCM values (0.05 for both), ratio Bland-Altman plots showed Logan and RLogan displayed relative errors which were comparable between different regions, in contrast with the other methods. Although SUV consistently underestimated V T, the bias in this method was also constant across regions. CONCLUSIONS: All parametric methods performed well for the analysis of [11C]flumazenil distribution and binding in the rat brain. However, Logan and RLogan slightly outperformed the other methods in terms of precision, providing robust parameter estimation and constant bias. Yet, other methods can be of interest, because they can provide tissue perfusion (i.e., K 1 with BFM and SA), relative flow (i.e., R 1 with RPM and SRTM2), and model order (SA) images.


Subject(s)
Brain/metabolism , Carbon Radioisotopes/chemistry , Flumazenil/chemistry , Positron-Emission Tomography , Animals , Flumazenil/pharmacokinetics , Linear Models , Male , Rats, Wistar
10.
Mol Pharmacol ; 90(5): 530-539, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27573669

ABSTRACT

α-Pinene is a major monoterpene of the pine tree essential oils. It has been reported that α-pinene shows anxiolytic and hypnotic effects upon inhaled administration. However, hypnotic effect by oral supplementation and the molecular mechanism of α-pinene have not been determined yet. By combining in vivo sleep behavior, ex vivo electrophysiological recording from brain slices, and in silico molecular modeling, we demonstrate that (-)-α-pinene shows sleep enhancing property through a direct binding to GABAA-benzodiazepine (BZD) receptors by acting as a partial modulator at the BZD binding site. The effect of (-)-α-pinene on sleep-wake profiles was evaluated by recording electroencephalogram and electromyogram. The molecular mechanism of (-)-α-pinene was investigated by electrophysiology and molecular docking study. (-)-α-pinene significantly increased the duration of non-rapid eye movement sleep (NREMS) and reduced the sleep latency by oral administration without affecting duration of rapid eye movement sleep and delta activity. (-)-α-pinene potentiated the GABAA receptor-mediated synaptic response by increasing the decay time constant of sIPSCs in hippocampal CA1 pyramidal neurons. These effects of (-)-α-pinene on sleep and inhibitory synaptic response were mimicked by zolpidem, acting as a modulator for GABAA-BZD receptors, and fully antagonized by flumazenil, an antagonist for GABAA-BZD receptor. (-)-α-pinene was found to bind to aromatic residues of α1- and -γ2 subunits of GABAA-BZD receptors in the molecular model. We conclude that (-)-α-pinene enhances the quantity of NREMS without affecting the intensity of NREMS by prolonging GABAergic synaptic transmission, acting as a partial modulator of GABAA-BZD receptors and directly binding to the BZD binding site of GABAA receptor.


Subject(s)
Benzodiazepines/metabolism , Eye Movements/drug effects , Monoterpenes/pharmacology , Pinus/chemistry , Plant Oils/pharmacology , Receptors, GABA-A/metabolism , Sleep/drug effects , Animals , Bicyclic Monoterpenes , Binding Sites , Flumazenil/chemistry , Flumazenil/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Models, Molecular , Monoterpenes/chemistry , Pentobarbital , Pyridines/chemistry , Pyridines/pharmacology , Sleep, REM/drug effects , Time Factors , Wakefulness/drug effects , Zolpidem
11.
Sci Rep ; 6: 25943, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27198062

ABSTRACT

GABAA receptors are the main inhibitory neurotransmitter receptors in the brain and are targets for numerous clinically important drugs such as benzodiazepines, anxiolytics and anesthetics. We previously identified novel ligands of the classical benzodiazepine binding pocket in α1ß2γ2 GABAA receptors using an experiment-guided virtual screening (EGVS) method. This screen also identified novel ligands for intramembrane low affinity diazepam site(s). In the current study we have further characterized compounds 31 and 132 identified with EGVS as well as 4-O-methylhonokiol. We investigated the site of action of these compounds in α1ß2γ2 GABAA receptors expressed in Xenopus laevis oocytes using voltage-clamp electrophysiology combined with a benzodiazepine site antagonist and transmembrane domain mutations. All three compounds act mainly through the two ß+/α- subunit transmembrane interfaces of the GABAA receptors. We then used concatenated receptors to dissect the involvement of individual ß+/α- interfaces. We further demonstrated that these compounds have anesthetic activity in a small aquatic animal model, Xenopus laevis tadpoles. The newly identified compounds may serve as scaffolds for the development of novel anesthetics.


Subject(s)
Anesthetics/pharmacology , Benzodiazepines/chemistry , Receptors, GABA-A/metabolism , Xenopus laevis/metabolism , Allosteric Regulation/drug effects , Anesthetics/chemistry , Animals , Benzodiazepines/pharmacology , Computer Simulation , Drug Evaluation, Preclinical , Flumazenil/chemistry , Flumazenil/pharmacology , Ligands , Molecular Structure , Patch-Clamp Techniques , Xenopus Proteins/metabolism
12.
Epilepsy Res ; 122: 30-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26921854

ABSTRACT

Benzodiazepines (BZDs) are highly effective to suppress various types of seizures; however, their clinical use is limited due to adverse effects and tolerance and dependence liability. Drugs that act only as partial agonists at the BZD recognition site (initially termed "BZD receptor") of the GABAA receptor chloride ionophore complex or exhibit a GABAA receptor subtype-selectivity are thought to have advantages vs. full agonists such as diazepam and most other clinically used BZDs in that such compounds have less adverse effects and reduced or absent tolerance and dependence liability. One of such compounds, abecarnil, has been clinically evaluated as a novel anxiolytic drug, but, despite its potent preclinical anti-seizure activity, it has not yet been evaluated in patients with epilepsy. In the present proof-of-concept study, we performed a within-subject placebo-controlled, single oral dose study of abecarnil in patients with photosensitive epilepsy. Flumazenil, which is generally considered a BZD receptor antagonist, but has slight partial agonistic properties, was used for comparison. In total, 12 patients were enrolled in this study. Abecarnil, 5 or 10mg, completely abolished the photo-paroxysmal EEG response, while flumazenil, 30, 60 or 100mg, was less effective. The anti-epileptic effect of abecarnil was significantly different from both placebo and flumazenil. Sedative adverse effects were observed after abecarnil but not flumazenil. The study substantiates previous pre-clinical experiments that abecarnil exerts pronounced anti-seizure activity. Epilepsy is often associated with anxiety, so that the anxiolytic activity of abecarnil would be an added advantage when using this compound in epilepsy patients.


Subject(s)
Anticonvulsants/therapeutic use , Carbolines/therapeutic use , Epilepsy, Reflex/drug therapy , Flumazenil/therapeutic use , GABA-A Receptor Agonists/therapeutic use , Administration, Oral , Adolescent , Adult , Anticonvulsants/adverse effects , Anticonvulsants/chemistry , Brain/drug effects , Brain/physiopathology , Carbolines/adverse effects , Carbolines/chemistry , Dose-Response Relationship, Drug , Electroencephalography , Epilepsy, Reflex/physiopathology , Female , Flumazenil/adverse effects , Flumazenil/chemistry , GABA-A Receptor Agonists/adverse effects , GABA-A Receptor Agonists/chemistry , Humans , Male , Middle Aged , Molecular Structure , Photic Stimulation/adverse effects , Pilot Projects , Receptors, GABA-A/metabolism , Single-Blind Method , Young Adult
13.
Sci Rep ; 5: 16347, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26572733

ABSTRACT

High-frequency oscillations in the gamma-band reflect rhythmic synchronization of spike timing in active neural networks. The modulation of gamma oscillations is a widely established mechanism in a variety of neurobiological processes, yet its neurochemical basis is not fully understood. Modeling, in-vitro and in-vivo animal studies suggest that gamma oscillation properties depend on GABAergic inhibition. In humans, search for evidence linking total GABA concentration to gamma oscillations has led to promising -but also to partly diverging- observations. Here, we provide the first evidence of a direct relationship between the density of GABA(A) receptors and gamma oscillatory gamma responses in human primary visual cortex (V1). By combining Flumazenil-PET (to measure resting-levels of GABA(A) receptor density) and MEG (to measure visually-induced gamma oscillations), we found that GABA(A) receptor densities correlated positively with the frequency and negatively with amplitude of visually-induced gamma oscillations in V1. Our findings demonstrate that gamma-band response profiles of primary visual cortex across healthy individuals are shaped by GABA(A)-receptor-mediated inhibitory neurotransmission. These results bridge the gap with in-vitro and animal studies and may have future clinical implications given that altered GABAergic function, including dysregulation of GABA(A) receptors, has been related to psychiatric disorders including schizophrenia and depression.


Subject(s)
Flumazenil/chemistry , Receptors, GABA-A/metabolism , Visual Cortex/metabolism , gamma-Aminobutyric Acid/metabolism , Adult , Female , Humans , Magnetic Resonance Imaging , Magnetoencephalography , Male , Positron-Emission Tomography
14.
Neuropharmacology ; 95: 459-67, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25963418

ABSTRACT

GABAA receptors are the major inhibitory neurotransmitter receptors in the brain and are the target for many clinically important drugs such as the benzodiazepines. Benzodiazepines act at the high-affinity binding site at the α+/γ- subunit interface. Previously, an additional low affinity binding site for diazepam located in the transmembrane (TM) domain has been described. The compound SJM-3 was recently identified in a prospective screening of ligands for the benzodiazepine binding site and investigated for its site of action. We determined the binding properties of SJM-3 at GABAA receptors recombinantly expressed in HEK-cells using radioactive ligand binding assays. Impact on function was assessed in Xenopus laevis oocytes with electrophysiological experiments using the two-electrode voltage clamp method. SJM-3 was shown to act as an antagonist at the α+/γ- site. At the same time it strongly potentiated GABA currents via the binding site for diazepam in the transmembrane domain. Mutation of a residue in M2 of the α subunit strongly reduced receptor modulation by SJM-3 and a homologous mutation in the ß subunit abolished potentiation. SJM-3 acts as a more efficient modulator than diazepam at the site in the trans-membrane domain. In contrast to low concentrations of benzodiazepines, SJM-3 modulates both synaptic and extrasynaptic receptors. A detailed exploration of the membrane site may provide the basis for the design and identification of subtype-selective modulatory drugs.


Subject(s)
GABA Agents/pharmacology , Neurons/drug effects , Quinolines/pharmacology , Receptors, GABA-A/metabolism , Thiazoles/pharmacology , Allosteric Regulation , Animals , Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Binding Sites , Diazepam/chemistry , Diazepam/pharmacology , Flumazenil/chemistry , Flumazenil/pharmacology , GABA Agents/chemistry , HEK293 Cells , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutation , Neurons/metabolism , Oocytes , Patch-Clamp Techniques , Protein Isoforms , Quinolines/metabolism , Radioligand Assay , Receptors, GABA-A/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thiazoles/metabolism , Transfection , Xenopus laevis , gamma-Aminobutyric Acid/metabolism
15.
Mol Imaging Biol ; 16(5): 619-25, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24788440

ABSTRACT

PURPOSE: [(18)F]Flumazenil, which has the advantage of a longer half-life than [(11)C]flumazenil, is well known for determining of the central benzodiazepine receptor concentrations. However, [(18)F]flumazenil has not been widely used because fluctuating and relatively low yields render automatic production insufficient for routine and multicenter clinical trials. Here, we describe the results of a 2.5-year production study of [(18)F]flumazenil using an iodonium tosylate precursor, which allowed us to overcome the limitations of low and fluctuating radiochemical yields. PROCEDURES: We developed a clinically applicable production system by modifying a commercial synthesizer for the reliable and reproducible production of [(18)F]flumazenil for routine clinical studies. [(18)F]Flumazenil was prepared at 150 °C for 5 min in the presence of 4-methylphenyl-mazenil iodonium tosylate (4 mg), a radical scavenger (TEMPO, 1 mg), and [(18)F]KF/kryptofix 2.2.2 complex in N,N-dimethylformamide (1 ml). In the purification step, the final mixture was pretreated using different cartridges before performing high-performance liquid chromatography (HPLC) separation. Finally, we measured the radiochemical yield and performed quality-control assays on 94 batches. RESULTS: After carrying out additional purification before HPLC separation using a C18 plus Sep-Pak cartridge, the radiochemical yield of [(18)F]flumazenil increased from 34.4 ± 9.7 % (without the pretreatment, n = 24) to 53.4 ± 9.0 % (n = 94), and the lifetime of the semi-preparative column was five times that of the column without the C18 plus Sep-Pak cartridge. The mean-specific activity of [(18)F]flumazenil was 572 ± 116 GBq/µmol at the end of synthesis, and the radiochemical purity was more than 99 %, as determined by analytical HPLC and radio-TLC. [(18)F]Flumazenil prepared using this method satisfied all quality-control test standards and was highly stable for up to 6 h after preparation. CONCLUSIONS: The results of the 2.5-year production study using an iodonium tosylate precursor indicate that [(18)F]flumazenil has commercial and routine clinical applicability.


Subject(s)
Flumazenil/chemical synthesis , Fluorine Radioisotopes/chemistry , Onium Compounds/chemistry , Radiochemistry/methods , Tosyl Compounds/chemistry , Chromatography, High Pressure Liquid , Flumazenil/chemistry , Indicators and Reagents , Quality Control
16.
Nucl Med Biol ; 40(7): 901-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23890694

ABSTRACT

INTRODUCTION: Two 7-fluoroimidazobenzodiazepines (AH114726 and GEH120348), analogs of flumazenil, were labeled with fluorine-18 and evaluated as alternative radioligands for in vivo imaging of the GABAA/benzodiazepine receptor by comparing them to [(11)C]flumazenil in rhesus monkey. METHODS: Radiotracers were prepared from the corresponding nitro-precursors in an automated synthesis module, and primate imaging studies were conducted on a Concorde MicroPET P4 scanner. The brain was imaged for 60 (12 × 5 min frames) or 90 min (18 × 5 min frames), and data was reconstructed using the 3D MAP algorithm. Specificity of [(18)F]AH114726 and [(18)F]GEH120348 was confirmed by displacement studies using unlabeled flumazenil. RESULTS: [(18)F]GEH120348 and [(18)F]AH114726 were obtained in 13-24% yields (end of synthesis) with high chemical (>95%) and radiochemical (>99%) purities, and high specific activities (2061 ± 985 Ci/mmol). The in vivo pharmacokinetics of [(18)F]AH114726 and [(18)F]GEH120348 were determined in a non-human primate and directly compared with [(11)C]flumazenil. Both fluorine-18 radioligands showed time-dependent regional brain distributions that correlated with the distribution of [(11)C]flumazenil and the known concentrations of GABAA/benzodiazepine receptors in the monkey brain. [(18)F]AH114726 exhibited maximal brain uptake and tissue time-radioactivity curves that were most similar to [(11)C]flumazenil. In contrast, [(18)F]GEH120348 showed higher initial brain uptake but very different pharmacokinetics with continued accumulation of radioactivity into the cortical regions of high GABA/benzodiazepine receptor concentrations and very little clearance from the regions of low receptor densities. Rapid washout of both radiotracers occurred upon treatment with unlabeled flumazenil. CONCLUSION: The ease of the radiochemical synthesis, together with in vivo brain pharmacokinetics most similar to [(11)C]flumazenil, support that [(18)F]AH114726 is a suitable option for imaging the GABAA receptor.


Subject(s)
Brain/diagnostic imaging , Brain/metabolism , Flumazenil , Fluorine Radioisotopes , Positron-Emission Tomography/methods , Receptors, GABA-A/metabolism , Animals , Female , Flumazenil/chemistry , Flumazenil/pharmacokinetics , Macaca mulatta , Radiochemistry
17.
Bioorg Med Chem Lett ; 23(3): 821-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23265897

ABSTRACT

Positron emission tomography (PET) using the tracer [(11)C]Flumazenil has shown changes in the distribution and expression of the GABA(A) receptor in a range of neurological conditions and injury states. We aim to develop a fluorine-18 labelled PET agent with comparable properties to [(11)C]Flumazenil. In this study we make a direct comparison between the currently known fluorine-18 labelled GABA(A) radiotracers and novel imidazobenzodiazepine ligands. A focussed library of novel compound was designed and synthesised where the fluorine containing moiety and the position of attachment is varied. The in vitro affinity of twenty-two compounds for the GABA(A) receptor was measured. Compounds containing a fluoroalkyl amide or a longer chain ester group were eliminated due to low potency. The fluorine-18 radiochemistry of one compound from each structural type was assessed to confirm that an automated radiosynthesis in good yield was feasible. Eleven of the novel compounds assessed appeared suitable for in vivo assessment as PET tracers.


Subject(s)
Fluorine Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Receptors, GABA-A/chemistry , Flumazenil/chemistry , Humans , Positron-Emission Tomography , Receptors, GABA-A/metabolism , Small Molecule Libraries
18.
Anal Bioanal Chem ; 405(4): 1303-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23180082

ABSTRACT

A general method is presented for the identification of radiometabolites in plasma of human and monkey subjects after administration of positron emission tomography (PET) radioligands. The radiometabolites are first produced in vitro, using liver microsomes, subsequently separated using fast radio-liquid chromatography (radio-LC), and individually collected and identified by ultra high-performance liquid chromatography/quadrupole-time of flight-mass spectrometry in MS and MS(E) mode. Fast radio-LC provided superior resolution compared to conventional radio-LC, resulting in separation of a greater number of metabolites. The radiometabolites produced in vivo are then compared to and identified based on the in vitro results. This approach was applied to three PET radioligands, [(11)C]flumazenil, [(18)F]FE-PE2I, and [(11)C]PBR28, resulting in the identification of five, two, and one radiometabolites, respectively. This procedure can easily be adopted to identify the radiometabolites produced in vivo from a variety of PET radioligands.


Subject(s)
Chromatography, High Pressure Liquid/methods , Cytochrome P-450 Enzyme System/metabolism , Mass Spectrometry/methods , Microsomes, Liver/metabolism , Radiopharmaceuticals/metabolism , Acetamides/chemistry , Acetamides/metabolism , Animals , Cytochrome P-450 Enzyme System/chemistry , Female , Flumazenil/chemistry , Flumazenil/metabolism , Haplorhini , Humans , Kinetics , Male , Microsomes, Liver/chemistry , Positron-Emission Tomography , Pyridines/chemistry , Pyridines/metabolism , Radiopharmaceuticals/chemistry
19.
Mol Imaging Biol ; 14(1): 115-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21365327

ABSTRACT

PURPOSE: We measure the whole-body distribution of IV injected [¹¹C]Flumazenil (FMZ) as a function of time in adult subjects and determine the absorbed radiation doses. PROCEDURES: After injection with 770 MBq of [¹¹C]FMZ (nominal), each of six subjects underwent nine consecutive whole body PET scans. Twelve source organs were identified using PET attenuation and emission images. Activity within each organ as a function of time was determined from the sequence of the nine PET scans. Source organ time activity curves were integrated and normalized by the injected dose to yield source organ residence times for the no voiding situation. Separate bladder residence-time calculations were performed for the cases of a 1- and a 2-h voiding interval. Using the source organ residence times as input, the program OLINDA/EXM (Stabin et al. in J Nucl Med. 46:1023-1027, 2005) was used to perform dosimetry calculations for the various body organs and for the whole body. RESULTS: For the no voiding situation, the average whole-body radiation equivalent dose was 3.02 × 10⁻³ mSv/MBq of injected [¹¹C]FMZ. The average effective dose and effective dose equivalent was 7.57 × 10⁻³ and 1.12 × 10⁻² mSv MBq⁻¹, respectively. The organ receiving the highest equivalent dose was the urinary bladder wall with an average of 6.32 × 10⁻² mSv MBq⁻¹. CONCLUSION: On average, the administration of less than 790 MBq (21 mCi) of [¹¹C]FMZ yields (no voiding model) an organ equivalent dose of under 50 mSv [the single dose limit for research studies under US regulations (21CFR361.1) to body organs other than blood forming organs, gonads or the lens of the eye] to all organs. Equivalent dose to the blood forming organs and gonads from a 790 MBq administered FMZ dose is well under the 30 mSv limit provided under 21CFR361.1. Additionally, administration of less than 1320 MBq (35.7 mCi) yields an effective dose [International Commission on Radiation Protection (ICRP) 60 tissue weighting scheme] of under 10 mSv, which is the ICRP IIb (minor to intermediate) risk category limit.


Subject(s)
Carbon Radioisotopes , Flumazenil/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Adult , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacokinetics , Female , Flumazenil/chemistry , Humans , Male , Positron-Emission Tomography/methods , Radiation Dosage , Radiopharmaceuticals/chemistry , Tissue Distribution , Whole Body Imaging/methods
20.
Org Biomol Chem ; 9(24): 8346-55, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22057475

ABSTRACT

Aromatic radiofluorination of the diaryliodonium tosylate precursor with [(18)F]fluoride ions has been applied successfully to access [(18)F]flumazenil in high radiochemical yields of 67.2 ± 2.7% (decay corrected). The stability and reactivity of the diaryliodonium tosylate precursor plays a key role in increasing the production of (18)F-labelled molecules under the fluorine-18 labelling condition. Various conditions were explored for the preparation of [(18)F]flumazenil from different diaryliodonium tosylate precursors. Optimum incorporation of [(18)F]fluoride ions in the 4-methylphenyl-mazenil iodonium tosylate precursor (5f) was achieved at 150 °C for 5 min by utilizing 4 mg of the precursor, K(2.2.2)/K(2)CO(3) complex, and the radical scavenger in N,N-dimethylformamide. This approach was extended to a viable method for use in automated synthesis with a radiochemical yield of 63.5 ± 3.2% (decay corrected, n = 26) within 60.0 ± 1.1 min. [(18)F]Flumazenil was isolated by preparative HPLC after the reaction was conducted under improved conditions and exhibited sufficient specific activity of 370-450 GBq µmol(-1), with a radiochemical purity of >99%, which will be suitable for human PET studies.


Subject(s)
Flumazenil/chemistry , Hydrocarbons, Iodinated/chemistry , Flumazenil/chemical synthesis , Fluorine Radioisotopes/chemistry , Molecular Structure , Salts/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...