Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Nature ; 585(7824): 303-308, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32879488

RESUMEN

Most general anaesthetics and classical benzodiazepine drugs act through positive modulation of γ-aminobutyric acid type A (GABAA) receptors to dampen neuronal activity in the brain1-5. However, direct structural information on the mechanisms of general anaesthetics at their physiological receptor sites is lacking. Here we present cryo-electron microscopy structures of GABAA receptors bound to intravenous anaesthetics, benzodiazepines and inhibitory modulators. These structures were solved in a lipidic environment and are complemented by electrophysiology and molecular dynamics simulations. Structures of GABAA receptors in complex with the anaesthetics phenobarbital, etomidate and propofol reveal both distinct and common transmembrane binding sites, which are shared in part by the benzodiazepine drug diazepam. Structures in which GABAA receptors are bound by benzodiazepine-site ligands identify an additional membrane binding site for diazepam and suggest an allosteric mechanism for anaesthetic reversal by flumazenil. This study provides a foundation for understanding how pharmacologically diverse and clinically essential drugs act through overlapping and distinct mechanisms to potentiate inhibitory signalling in the brain.


Asunto(s)
Anestésicos Generales/química , Anestésicos Generales/farmacología , Barbitúricos/química , Barbitúricos/farmacología , Benzodiazepinas/química , Benzodiazepinas/farmacología , Microscopía por Crioelectrón , Receptores de GABA-A/química , Regulación Alostérica/efectos de los fármacos , Anestésicos Generales/metabolismo , Barbitúricos/metabolismo , Benzodiazepinas/metabolismo , Bicuculina/química , Bicuculina/metabolismo , Bicuculina/farmacología , Sitios de Unión , Unión Competitiva/efectos de los fármacos , Diazepam/química , Diazepam/metabolismo , Diazepam/farmacología , Electrofisiología , Etomidato/química , Etomidato/metabolismo , Etomidato/farmacología , Flumazenil/farmacología , Antagonistas de Receptores de GABA-A/química , Antagonistas de Receptores de GABA-A/metabolismo , Antagonistas de Receptores de GABA-A/farmacología , Humanos , Ligandos , Modelos Moleculares , Conformación Molecular , Simulación de Dinámica Molecular , Fenobarbital/química , Fenobarbital/metabolismo , Fenobarbital/farmacología , Picrotoxina/química , Picrotoxina/metabolismo , Picrotoxina/farmacología , Propofol/química , Propofol/metabolismo , Propofol/farmacología , Receptores de GABA-A/metabolismo , Receptores de GABA-A/ultraestructura , Ácido gamma-Aminobutírico/química , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología
2.
Mol Pharmacol ; 104(6): 266-274, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37586749

RESUMEN

Multiple approaches, including cryogenic electron microscopy (cryo-EM), indicate that the anesthetics etomidate and propofol modulate α1ß2/3γ2 GABAA receptors by binding in overlapping transmembrane inter-subunit sites near ßM286 and αL232 sidechains. High-precision approaches in functional receptors are needed for comparisons with cryo-EM. We previously used substituted cysteine modification and protection (SCAMP) with n-alkyl-methanethiosulfonate (MTS) reagents and electrophysiology in α1ß3M286Cγ2L receptors to estimate the distance from etomidate to ß3M286 with precision near 1.3 Å. Here, we address three more aims using this approach: (i) SCAMP with etomidate was tested in α1L232Cß3γ2L receptors; (ii) studies in α1L232Wß3M286Cγ2L receptors assessed whether α1L232W displaces etomidate relative to ß3M286C; and (iii) results with propofol were compared with those with etomidate. Voltage-clamp electrophysiology in Xenopus oocytes was used to assess persistent functional changes after exposing cysteine-substituted receptors to methyl-MTS through n-decyl-MTS. Overlap of modified cysteine sidechains with bound anesthetic was inferred when anesthetic co-application with alkyl-MTS reagent blocked the development of persistent effects. In α1L232Cß3γ2L receptors, only pentyl-MTS and hexyl-MTS induced persistent effects that were unaltered by etomidate co-application, precluding a direct estimate of intermolecular distance. In α1L232Wß3M286Cγ2L receptors, sidechain overlap with bound etomidate was inferred for modifications with ethyl-MTS through n-pentyl-MTS, with unambiguous cut-on and cut-off. Comparison with results in α1ß3M286Cγ2L reveals that α1L232W, which increases maximal sidechain length by 2.1 Å, displaces etomidate closer to ß3M286C by about 1.3 Å. Propofol results largely mirrored those with etomidate. These findings indicate that both etomidate and propofol bind within 1 Å of α1L232, consistent with cryo-EM structures. SIGNIFICANCE STATEMENT: We combined electrophysiology, cysteine substitutions, and n-alkyl-methanethiosulfonate modifiers in functional GABAA receptors to enable precise estimates of the distance between ß3M286C sidechains and anesthetics (etomidate and propofol) bound in transmembrane ß+/α- inter-subunit pockets. Comparing results in α1ß3M286Cγ2L and α1L232Wß3M286Cγ2L receptors reveals that α1L232W mutations displace both anesthetics toward ß3M286C, indicating that these anesthetics bind within 1 Å of the α1L232 sidechain in functional receptors, consistent with cryogenic electron microscopy structures derived under nonphysiologic conditions.


Asunto(s)
Anestésicos , Etomidato , Propofol , Receptores de GABA-A/metabolismo , Etomidato/farmacología , Etomidato/química , Propofol/farmacología , Cisteína/genética , Anestésicos/farmacología , Sitios de Unión , Mutación , Ácido gamma-Aminobutírico/genética
3.
Proc Natl Acad Sci U S A ; 116(31): 15706-15715, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31308218

RESUMEN

Intravenous anesthetic agents are associated with cardiovascular instability and poorly tolerated in patients with cardiovascular disease, trauma, or acute systemic illness. We hypothesized that a new class of intravenous (IV) anesthetic molecules that is highly selective for the slow type of γ-aminobutyric acid type A receptor (GABAAR) could have potent anesthetic efficacy with limited cardiovascular effects. Through in silico screening using our GABAAR model, we identified a class of lead compounds that are N-arylpyrrole derivatives. Electrophysiological analyses using both an in vitro expression system and intact rodent hippocampal brain slice recordings demonstrate a GABAAR-mediated mechanism. In vivo experiments also demonstrate overt anesthetic activity in both tadpoles and rats with a potency slightly greater than that of propofol. Unlike the clinically approved GABAergic anesthetic etomidate, the chemical structure of our N-arylpyrrole derivative is devoid of the chemical moieties producing adrenal suppression. Our class of compounds also shows minimal to no suppression of blood pressure, in marked contrast to the hemodynamic effects of propofol. These compounds are derived from chemical structures not previously associated with anesthesia and demonstrate that selective targeting of GABAAR-slow subtypes may eliminate the hemodynamic side effects associated with conventional IV anesthetics.


Asunto(s)
Anestésicos , Agonistas de Receptores de GABA-A , Pirroles , Receptores de GABA-A/metabolismo , Anestésicos/química , Anestésicos/farmacología , Animales , Evaluación Preclínica de Medicamentos , Etomidato/química , Etomidato/farmacología , Agonistas de Receptores de GABA-A/química , Agonistas de Receptores de GABA-A/farmacología , Humanos , Ratones , Pirroles/química , Pirroles/farmacología , Ratas , Receptores de GABA-A/genética , Xenopus laevis
4.
Pharmacology ; 106(11-12): 644-657, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34644710

RESUMEN

INTRODUCTION: The mixture of etomidate and propofol is widely used in clinical practice to improve efficacy of general anesthesia and to minimize side effects. As a thermodynamically unstable system, emulsion is prone to destabilization through mechanisms including coalescence, flocculation, and creaming. Such unwanted phenomenon can induce fat embolism after intravenous administration. This study was aimed to investigate the physical and chemical stability of the mixture of etomidate and propofol in the dosage form of emulsion. METHODS: This compatibility study focused on the critical quality attributes (CQAs) of drug-containing emulsions, such as appearance, pH, particle size and distribution, zeta potential, the observation under centrifugation, and drug content and impurity. RESULTS: As the results, there were no significant changes in the CQAs of the mixed emulsions up to 24 h after mixing at refrigeration temperature (4°C), room temperature (25°C), and body temperature (37°C). CONCLUSIONS: These results demonstrate that etomidate emulsion is physically and chemically compatible with propofol emulsions up to 24 h at 4°C, 25°C, and 37°C, suggesting that etomidate and propofol can be administrated in mixture without adversely affecting product characteristics, at least in vitro.


Asunto(s)
Anestésicos Intravenosos/química , Emulsiones/química , Etomidato/química , Propofol/química , Contaminación de Medicamentos , Estabilidad de Medicamentos , Concentración de Iones de Hidrógeno , Inyecciones Intravenosas , Tamaño de la Partícula , Propiedades de Superficie
5.
Eur Biophys J ; 49(7): 591-607, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32940715

RESUMEN

GABA and glycine act as inhibitory neurotransmitters in the CNS. Inhibitory neurotransmission is mediated via activation of ionotropic GABAA and glycine receptors. We used a modeling approach to explain the opposite effects of the general anesthetic etomidate (ETM) and fenamate mefenamic acid (MFA) on GABA- and glycine-activated currents recorded in isolated cerebellar Purkinje cells and hippocampal pyramidal neurons, respectively. These drugs potentiated GABAARs but blocked GlyRs. We built a homology model of α1ß GlyR based on the cryo-EM structure of open α1 GlyR, used the α1ß3γ2 GABAAR structure from the PDB, and applied Monte-Carlo energy minimization to optimize models of receptors and ligand-receptor complexes. In silico docking suggests that ETM/MFA bind at the transmembrane ß( +)/α( -) intersubunit interface in GABAAR. Our models predict that the bulky side chain of the highly conserved Arg19' residue at the plus interface side wedges the interface and maintains the conducting receptor state. We hypothesized that MFA/ETM binding at the ß( +)/α( -) interface leads to prolongation of receptor life-time in the open state. Having analyzed different GABAAR and GlyR structures available in the PDB, we found that mutual arrangement of the Arg19' and Gln-26' side chains at the plus and minus interface sides, respectively, plays an important role when the receptor switches from the open to closed state. We show that this process is accompanied by narrowing of the intersubunit interfaces, leading to extrusion of the Arg19' side chain from the interface. Our models allow us to explain the lack of GlyR potentiation in our electrophysiological experiments.


Asunto(s)
Etomidato/química , Ácido Mefenámico/química , Neuronas/metabolismo , Proteínas Nucleares/química , Oxidorreductasas/química , Receptores de GABA-A/química , Anestésicos Generales/farmacología , Animales , Sitios de Unión , Simulación por Computador , Bases de Datos de Proteínas , Electrofisiología , Fenamatos/química , Glicina/química , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Método de Montecarlo , Unión Proteica , Ratas , Ratas Wistar , Receptores de Glicina/química , Transmisión Sináptica
6.
Mol Pharmacol ; 95(6): 615-628, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30952799

RESUMEN

GABAA receptors (GABAARs) are targets for important classes of clinical agents (e.g., anxiolytics, anticonvulsants, and general anesthetics) that act as positive allosteric modulators (PAMs). Previously, using photoreactive analogs of etomidate ([3H]azietomidate) and mephobarbital [[3H]1-methyl-5-allyl-5-(m-trifluoromethyl-diazirynylphenyl)barbituric acid ([3H]R-mTFD-MPAB)], we identified two homologous but pharmacologically distinct classes of general anesthetic binding sites in the α1ß3γ2 GABAAR transmembrane domain at ß +-α - (ß + sites) and α +-ß -/γ +-ß - (ß - sites) subunit interfaces. We now use competition photolabeling with [3H]azietomidate and [3H]R-mTFD-MPAB to identify para-substituted propofol analogs and other drugs that bind selectively to intersubunit anesthetic sites. Propofol and 4-chloro-propofol bind with 5-fold selectivity to ß +, while derivatives with bulkier lipophilic substitutions [4-(tert-butyl)-propofol and 4-(hydroxyl(phenyl)methyl)-propofol] bind with ∼10-fold higher affinity to ß - sites. Similar to R-mTFD-MPAB and propofol, these drugs bind in the presence of GABA with similar affinity to the α +-ß - and γ +-ß - sites. However, we discovered four compounds that bind with different affinities to the two ß - interface sites. Two of these bind with higher affinity to one of the ß - sites than to the ß + sites. We deduce that 4-benzoyl-propofol binds with >100-fold higher affinity to the γ +-ß - site than to the α +-ß - or ß +-α - sites, whereas loreclezole, an anticonvulsant, binds with 5- and 100-fold higher affinity to the α +-ß - site than to the ß + and γ +-ß - sites. These studies provide a first identification of PAMs that bind selectively to a single intersubunit site in the GABAAR transmembrane domain, a property that may facilitate the development of subtype selective GABAAR PAMs.


Asunto(s)
Anestésicos/farmacología , Propofol/análogos & derivados , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Regulación Alostérica , Anestésicos/química , Bicuculina/química , Bicuculina/farmacología , Sitios de Unión , Etomidato/química , Etomidato/farmacología , Células HEK293 , Humanos , Propofol/química , Dominios Proteicos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Triazoles/química , Triazoles/farmacología
7.
J Labelled Comp Radiopharm ; 62(4): 190-197, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30690759

RESUMEN

[11 C]metomidate ([11 C]MTO) is a radiotracer widely used to detect disorders of adrenocortical origin by positron emission tomography (PET) imaging. [11 C]MTO PET/computed tomography (PET/CT) is considered a sensitive and specific noninvasive alternative to adrenal vein sampling (AVS) in the management of primary hyperaldosteronism (PHA). Herein, we report a reliable automated procedure for the routine manufacturing of [11 C]MTO in current good manufacturing practice (cGMP) conditions on the commercial Synthra MeIPlus Loop Vessel synthesizer. The method is based on a combination of the captive-solvent 11 C-methylation of the carboxylate salt 1b of the MTO precursor 1a followed by solid phase extraction (SPE) cartridge purification methodology, which substitutes HPLC purification of the crude reaction mixture. Starting from 45 GBq [11 C]CO2 at the end of bombardment (EOB), 3 GBq of pure [11 C]MTO was produced in 18 minutes with 12% decay corrected radiochemical yield (RCY) at the end of synthesis (EOS) and with the modest molar activity of 13 GBq/µmol at the time of application. Each dose produced met all established quality control (QC) criteria. The method can easily be implemented into other commercial automated radiosynthesizers for manufacturing carbon-11 labeled radiotracers.


Asunto(s)
Etomidato/análogos & derivados , Radioquímica/métodos , Extracción en Fase Sólida/instrumentación , Automatización , Técnicas de Química Sintética , Etomidato/síntesis química , Etomidato/química , Etomidato/aislamiento & purificación , Humanos , Control de Calidad
8.
J Pharmacol Exp Ther ; 364(2): 229-237, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29203576

RESUMEN

Cushing's syndrome is characterized by the overproduction of adrenocortical steroids. Steroidogenesis inhibitors are mainstays of medical therapy for Cushing's syndrome; unfortunately, adverse side effects and treatment failures are common with currently available drugs. The general anesthetic induction agent etomidate is among the most potent inhibitors of adrenocortical steroidogenesis. However, its use as a treatment of Cushing's syndrome is complicated by its sedative-hypnotic activity and ability to produce myoclonus, central nervous system actions thought to be mediated by the GABAA receptor. Here, we describe the pharmacology of the novel etomidate analog (R)-ethyl 1-(1-(3,5-dimethoxyphenyl)ethyl)-1H-imidazole-5-carboxylate (dimethoxy-etomidate). In contrast to etomidate, dimethoxy-etomidate minimally enhanced GABA-evoked GABAA receptor-mediated currents even at a near-saturating aqueous concentration. In Sprague-Dawley rats, dimethoxy-etomidate's potency for producing loss of righting reflexes-an animal model of sedation/hypnosis-was 2 orders of magnitude lower than that of etomidate, and it did not produce myoclonus. However, similar to etomidate, dimethoxy-etomidate potently suppressed adrenocortical steroid synthesis primarily by inhibiting 11ß-hydroxylase. [3H]etomidate binding to rat adrenocortical membranes was inhibited by dimethoxy-etomidate in a biphasic manner with IC50 values of 8.2 and 3970 nM, whereas that by etomidate was monophasic with an IC50 of 22 nM. Our results demonstrate that, similar to etomidate, dimethoxy-etomidate potently and dose-dependently suppresses adrenocortical steroid synthesis by inhibiting 11ß-hydroxylase. However, it is essentially devoid of etomidate's GABAA receptor positive modulatory and sedative-hypnotic activities and produces no myoclonus, providing proof of concept for the design of etomidate analogs without important central nervous system actions for the pharmacologic treatment of Cushing's syndrome.


Asunto(s)
Etomidato/análogos & derivados , Etomidato/farmacología , Esteroides/biosíntesis , Animales , Fenómenos Electrofisiológicos/efectos de los fármacos , Etomidato/química , Masculino , Movimiento/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Ácido gamma-Aminobutírico/farmacología
9.
Pharm Dev Technol ; 21(5): 528-34, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25757642

RESUMEN

Etomidate is a unique drug used for induction of general anesthesia and sedation, and is usually used through intravenous injection clinically. Before targeting to the receptor, etomidate binds proteins in blood when it comes into veins. Thus to study the interaction of etomidate and serum albumin would be of great toxicological and pharmacological importance. In this study, the interaction between etomidate and human serum albumin (HSA) was studied using fluorescence spectroscopy, UV-vis absorption spectroscopy, Fourier transform infrared spectroscopy (FT-IR), circular dichroism (CD) spectroscopy, site maker displacement and molecular modeling methods. Investigations of the binding constant (K = 3.55 × 10(5 )M(-1), 295 K), the number of binding sites (n = 1.16), thermodynamic parameters (ΔG = 3.13 × 10(4 )J·mol(-1), ΔS = 364 J·mol(-1)·K(-1) and ΔH = -6.85 × 10(5 )J·mol(-1)) for the reaction and changes to the binding sites and conformation in HSA in response to etomidate were presented. Results show that etomidate can bind HSA tightly through electrostatic forces, and the protein skeleton conformation and secondary structure changes thereby. This is the first spectroscopic report for etomidate-HSA interactions which illustrates the complex nature of this subject.


Asunto(s)
Etomidato/química , Etomidato/metabolismo , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/metabolismo , Albúmina Sérica/química , Albúmina Sérica/metabolismo , Sitios de Unión/fisiología , Fenómenos Biofísicos/fisiología , Humanos , Simulación del Acoplamiento Molecular/métodos , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Espectroscopía Infrarroja por Transformada de Fourier/métodos
10.
J Biol Chem ; 288(42): 30373-30386, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24009076

RESUMEN

Etomidate is a potent general anesthetic that acts as an allosteric co-agonist at GABAA receptors. Photoreactive etomidate derivatives labeled αMet-236 in transmembrane domain M1, which structural models locate in the ß+/α- subunit interface. Other nearby residues may also contribute to etomidate binding and/or transduction through rearrangement of the site. In human α1ß2γ2L GABAA receptors, we applied the substituted cysteine accessibility method to α1-M1 domain residues extending from α1Gln-229 to α1Gln-242. We used electrophysiology to characterize each mutant's sensitivity to GABA and etomidate. We also measured rates of sulfhydryl modification by p-chloromercuribenzenesulfonate (pCMBS) with and without GABA and tested if etomidate blocks modification of pCMBS-accessible cysteines. Cys substitutions in the outer α1-M1 domain impaired GABA activation and variably affected etomidate sensitivity. In seven of eight residues where pCMBS modification was evident, rates of modification were accelerated by GABA co-application, indicating that channel activation increases water and/or pCMBS access. Etomidate reduced the rate of modification for cysteine substitutions at α1Met-236, α1Leu-232 and α1Thr-237. We infer that these residues, predicted to face ß2-M3 or M2 domains, contribute to etomidate binding. Thus, etomidate interacts with a short segment of the outer α1-M1 helix within a subdomain that undergoes significant structural rearrangement during channel gating. Our results are consistent with in silico docking calculations in a homology model that orient the long axis of etomidate approximately orthogonal to the transmembrane axis.


Asunto(s)
4-Cloromercuribencenosulfonato/química , Anestésicos Intravenosos/química , Inhibidores Enzimáticos/química , Etomidato/química , Activación del Canal Iónico/fisiología , Receptores de GABA-A/química , 4-Cloromercuribencenosulfonato/farmacología , Sustitución de Aminoácidos , Anestésicos Intravenosos/farmacología , Animales , Inhibidores Enzimáticos/farmacología , Etomidato/farmacología , Femenino , Humanos , Activación del Canal Iónico/efectos de los fármacos , Simulación del Acoplamiento Molecular , Mutación Missense , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Xenopus laevis
11.
J Biol Chem ; 288(27): 19343-57, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23677991

RESUMEN

GABA type A receptors (GABAAR), the brain's major inhibitory neurotransmitter receptors, are the targets for many general anesthetics, including volatile anesthetics, etomidate, propofol, and barbiturates. How such structurally diverse agents can act similarly as positive allosteric modulators of GABAARs remains unclear. Previously, photoreactive etomidate analogs identified two equivalent anesthetic-binding sites in the transmembrane domain at the ß(+)-α(-) subunit interfaces, which also contain the GABA-binding sites in the extracellular domain. Here, we used R-[(3)H]5-allyl-1-methyl-5-(m-trifluoromethyl-diazirynylphenyl) barbituric acid (R-mTFD-MPAB), a potent stereospecific barbiturate anesthetic, to photolabel expressed human α1ß3γ2 GABAARs. Protein microsequencing revealed that R-[(3)H]mTFD-MPAB did not photolabel the etomidate sites at the ß(+)-α(-) subunit interfaces. Instead, it photolabeled sites at the α(+)-ß(-) and γ(+)-ß(-) subunit interfaces in the transmembrane domain. On the (+)-side, α1M3 was labeled at Ala-291 and Tyr-294 and γ2M3 at Ser-301, and on the (-)-side, ß3M1 was labeled at Met-227. These residues, like those in the etomidate site, are located at subunit interfaces near the synaptic side of the transmembrane domain. The selectivity of R-etomidate for the ß(+)-α(-) interface relative to the α(+)-ß(-)/γ(+)-ß(-) interfaces was >100-fold, whereas that of R-mTFD-MPAB for its sites was >50-fold. Each ligand could enhance photoincorporation of the other, demonstrating allosteric interactions between the sites. The structural heterogeneity of barbiturate, etomidate, and propofol derivatives is accommodated by varying selectivities for these two classes of sites. We hypothesize that binding at any of these homologous intersubunit sites is sufficient for anesthetic action and that this explains to some degree the puzzling structural heterogeneity of anesthetics.


Asunto(s)
Anestésicos Intravenosos/química , Barbitúricos/química , Etomidato/química , Receptores de GABA-A/química , Anestésicos Intravenosos/metabolismo , Barbitúricos/metabolismo , Sitios de Unión , Etomidato/metabolismo , Células HEK293 , Humanos , Ligandos , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Subunidades de Proteína , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Análisis de Secuencia de Proteína
12.
Anesthesiology ; 121(2): 290-301, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24777068

RESUMEN

BACKGROUND: R-etomidate possesses unique desirable properties but potently suppresses adrenocortical function. Consequently, efforts are being made to define structure-activity relationships with the goal of designing analogues with reduced adrenocortical toxicity. The authors explored the pharmacological impact of modifying etomidate's chiral center using R-etomidate, S-etomidate, and two achiral etomidate analogues (cyclopropyl etomidate and dihydrogen etomidate). METHODS: The γ-aminobutyric acid type A receptor modulatory potencies of drugs were assessed in oocyte-expressed α1(L264T)ß3γ2L and α1(L264T)ß1γ2L γ-aminobutyric acid type A receptors (for each drug, n = 6 oocytes per subtype). In rats, hypnotic potencies and durations of action were measured using a righting reflex assay (n = 26 to 30 doses per drug), and adrenocortical potencies were quantified by using an adrenocorticotropic hormone stimulation test (n = 20 experiments per drug). RESULTS: All four drugs activated both γ-aminobutyric acid type A receptor subtypes in vitro and produced hypnosis and suppressed adrenocortical function in rats. However, drug potencies in each model ranged by 1 to 2 orders of magnitude. R-etomidate had the highest γ-aminobutyric acid type A receptor modulatory, hypnotic, and adrenocortical inhibitory potencies. Respectively, R-etomidate, S-etomidate, and cyclopropyl etomidate were 27.4-, 18.9-, and 23.5-fold more potent activators of receptors containing ß3 subunits than ß1 subunits; however, dihydrogen etomidate's subunit selectivity was only 2.48-fold and similar to that of propofol (2.08-fold). S-etomidate was 1/23rd as potent an adrenocortical inhibitor as R-etomidate. CONCLUSION: The linkage between the structure of etomidate's chiral center and its pharmacology suggests that altering etomidate's chiral center may be used as part of a strategy to design analogues with more desirable adrenocortical activities and/or subunit selectivities.


Asunto(s)
Anestésicos Intravenosos/química , Anestésicos Intravenosos/farmacología , Carbono/química , Etomidato/análogos & derivados , Etomidato/farmacología , Corteza Suprarrenal/efectos de los fármacos , Enfermedades de la Corteza Suprarrenal/inducido químicamente , Enfermedades de la Corteza Suprarrenal/patología , Anestésicos Intravenosos/toxicidad , Animales , Etomidato/química , Femenino , Agonistas del GABA/síntesis química , Agonistas del GABA/química , Agonistas del GABA/farmacología , Hipnóticos y Sedantes/síntesis química , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/farmacología , Indicadores y Reactivos , Dosificación Letal Mediana , Masculino , Conformación Molecular , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Xenopus laevis
13.
Chem Commun (Camb) ; 60(35): 4691-4694, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38592772

RESUMEN

The first fluorescent sensor based on the indicator displacement assay (IDA) for on-site determination of etomidate.


Asunto(s)
Etomidato , Colorantes Fluorescentes , Etomidato/análogos & derivados , Etomidato/química , Colorantes Fluorescentes/química , Espectrometría de Fluorescencia , Animales , Humanos
14.
Mol Pharm ; 10(3): 1119-30, 2013 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-23343186

RESUMEN

Metyrapone, metyrapol, and etomidate are competitive inhibitors of 11-deoxycorticosterone hydroxylation by 11ß-hydroxylase. [(3)H]Metyrapol and 4-[(131)I]iodometomidate bind with high affinity to membranes prepared from bovine and rat adrenals. Here we report inhibitory potencies of several compounds structurally related to one or both of these adrenostatic drugs, against the binding of both radioligands to rat adrenal membranes. While derivatives of etomidate inhibited the binding of both radioligands with similar potencies, derivatives of metyrapone inhibited the binding of 4-[(131)I]iodometomidate about 10 times weaker than the binding of [(3)H]metyrapol. By X-ray structure analysis the absolute configuration of (+)-1-(2-fluorophenyl)-2-methyl-2-(pyridin-3-yl)-1-propanol [(+)-11, a derivative of metyrapol] was established as (R). We introduce 1-(2-fluorophenyl)-2-methyl-2-(pyridin-3-yl)-1-propanone (9; Ki = 6 nM), 2-(1-imidazolyl)-2-methyl-1-phenyl-1-propanone (13; 2 nM), and (R)-(+)-[1-(4-iodophenyl)ethyl]-1H-imidazole (34; 4 nM) as new high affinity ligands for the metyrapol binding site on 11ß-hydroxylase and discuss our results in relation to a proposed active site model of 11ß-hydroxylase.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Membrana Celular/metabolismo , Etomidato/análogos & derivados , Metirapona/química , Animales , Sitios de Unión , Bovinos , Etomidato/química , Etomidato/metabolismo , Metirapona/metabolismo , Ratas
15.
Biochemistry ; 51(4): 836-47, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22243422

RESUMEN

The γ-aminobutyric acid type A receptor (GABA(A)R) is a target for general anesthetics of diverse chemical structures, which act as positive allosteric modulators at clinical doses. Previously, in a heterogeneous mixture of GABA(A)Rs purified from bovine brain, [³H]azietomidate photolabeling of αMet-236 and ßMet-286 in the αM1 and ßM3 transmembrane helices identified an etomidate binding site in the GABA(A)R transmembrane domain at the interface between the ß and α subunits [Li, G. D., et.al. (2006) J. Neurosci. 26, 11599-11605]. To further define GABA(A)R etomidate binding sites, we now use [³H]TDBzl-etomidate, an aryl diazirine with broader amino acid side chain reactivity than azietomidate, to photolabel purified human FLAG-α1ß3 GABA(A)Rs and more extensively identify photolabeled GABA(A)R amino acids. [³H]TDBzl-etomidate photolabeled in an etomidate-inhibitable manner ß3Val-290, in the ß3M3 transmembrane helix, as well as α1Met-236 in α1M1, a residue photolabeled by [³H]azietomidate, while no photolabeling of amino acids in the αM2 and ßM2 helices that also border the etomidate binding site was detected. The location of these photolabeled amino acids in GABA(A)R homology models derived from the recently determined structures of prokaryote (GLIC) or invertebrate (GluCl) homologues and the results of computational docking studies predict the orientation of [³H]TDBzl-etomidate bound in that site and the other amino acids contributing to this GABA(A)R intersubunit etomidate binding site. Etomidate-inhibitable photolabeling of ß3Met-227 in ßM1 by [³H]TDBzl-etomidate and [³H]azietomidate also provides evidence of a homologous etomidate binding site at the ß3-ß3 subunit interface in the α1ß3 GABA(A)R.


Asunto(s)
Anestésicos Generales/metabolismo , Diazometano/análogos & derivados , Etomidato/análogos & derivados , Etiquetas de Fotoafinidad/química , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Anestésicos Generales/química , Sitios de Unión , Unión Competitiva , Bases de Datos de Proteínas , Diazometano/química , Diazometano/metabolismo , Etomidato/química , Etomidato/metabolismo , Humanos , Cinética , Ligandos , Metionina/química , Metionina/metabolismo , Simulación de Dinámica Molecular , Mapeo Peptídico , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/genética , Receptores de GABA-A/química , Receptores de GABA-A/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tritio
16.
Anesthesiology ; 117(5): 1027-36, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22929736

RESUMEN

BACKGROUND: Methoxycarbonyl etomidate is the prototypical very rapidly metabolized etomidate analog. Initial studies suggest that it may be too short acting for many clinical uses. We hypothesized that its duration of action could be lengthened and clinical utility broadened by incorporating specific aliphatic groups into the molecule to sterically protect its ester moiety from esterase-catalyzed hydrolysis. To test this hypothesis, we developed a series of methoxycarbonyl etomidate analogs (spacer-linked etomidate esters) containing various aliphatic-protecting groups and spacer lengths. METHODS: Spacer-linked etomidate esters were synthesized and their hypnotic potencies and durations of action following bolus administration were measured in rats using a loss-of-righting reflexes assay. Octanol:water partition coefficients and metabolic half-lives in pooled rat blood were determined chromatographically. RESULTS: All spacer-linked etomidate esters produced hypnosis rapidly and in a dose-dependent manner. ED50s for loss of righting reflexes ranged from 0.69 ± 0.04 mg/kg for cyclopropyl-methoxycarbonyl metomidate to 11.1 ± 0.8 mg/kg for methoxycarbonyl metomidate. The slope of a plot of the duration of loss of righting reflexes versus the logarithm of the dose ranged 12-fold among spacer-linked etomidate esters, implying widely varying brain clearance rates. The in vitro metabolic half-lives of these compounds in rat blood varied by more than two orders of magnitude and were diastereometrically selective. CONCLUSIONS: We created 13 new analogs of methoxycarbonyl etomidate and identified two that have significantly higher potency and potentially address the too-brief duration of action for methoxycarbonyl etomidate. This work may provide a blueprint for optimizing the pharmacological properties of other soft drugs.


Asunto(s)
Etomidato/análogos & derivados , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Ésteres , Etomidato/química , Etomidato/metabolismo , Etomidato/farmacología , Semivida , Hipnóticos y Sedantes/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Reflejo de Enderezamiento/efectos de los fármacos , Reflejo de Enderezamiento/fisiología
17.
Anesth Analg ; 115(1): 70-2, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22543065

RESUMEN

BACKGROUND: Carboetomidate is an etomidate derivative that produces hypnosis without inhibiting adrenal corticosteroid synthesis. Similar to etomidate, carboetomidate modulates γ-aminobutyric acid type A receptors, but its effects on other ion channel targets of general anesthetics are unknown. METHODS: We compared etomidate and carboetomidate effects on human N-methyl-d-aspartate receptors or neuronal nicotinic acetylcholine receptors (nnAChRs) expressed in Xenopus oocytes, using 2-microelectrode voltage clamp electrophysiology. RESULTS: Etomidate did not affect either type of receptor at clinically relevant concentrations, whereas carboetomidate concentrations near 50% effective concentration for anesthesia significantly inhibited nnAChRs. CONCLUSIONS: Compared with etomidate, carboetomidate's higher hydrophobicity is associated with greater inhibition of nnAChRs.


Asunto(s)
Hipnóticos y Sedantes/farmacología , Antagonistas Nicotínicos/farmacología , Pirroles/farmacología , Receptores Nicotínicos/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Etomidato/química , Etomidato/farmacología , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Potenciales de la Membrana , Aceite de Oliva , Oocitos , Técnicas de Placa-Clamp , Aceites de Plantas/química , Pirroles/química , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Xenopus laevis
18.
Biochem Pharmacol ; 199: 114993, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35304861

RESUMEN

(+)-Catharanthine, a coronaridine congener, potentiates the γ-aminobutyric acid type A receptor (GABAAR) and induces sedation through a non-benzodiazepine mechanism, but the specific site of action and intrinsic mechanism have not beendefined. Here, we describe GABAAR subtype selectivity and location of the putative binding site for (+)-catharanthine using electrophysiological, site-directed mutagenesis, functional competition, and molecular docking experiments. Electrophysiological and in silico experiments showed that (+)-catharanthine potentiates the responses to low, subsaturating GABA at ß2/3-containing GABAARs 2.4-3.5 times more efficaciously than at ß1-containing GABAARs. The activity of (+)-catharanthine is reduced by the ß2(N265S) mutation that decreases GABAAR potentiation by loreclezole, but not by the ß3(M286C) or α1(Q241L) mutations that reduce receptor potentiation by R(+)-etomidate or neurosteroids, respectively. Competitive functional experiments indicated that the binding site for (+)-catharanthine overlaps that for loreclezole, but not those for R(+)-etomidate or potentiating neurosteroids. Molecular docking experiments suggested that (+)-catharanthine binds at the ß(+)/α(-) intersubunit interface near the TM2-TM3 loop, where it forms H-bonds with ß2-D282 (TM3), ß2-K279 (TM2-TM3 loop), and ß2-N265 and ß2-R269 (TM2). Site-directed mutagenesis experiments supported the in silico results, demonstrating that the K279A and D282A substitutions, that lead to a loss of H-bonding ability of the mutated residue, and the N265S mutation, impair the gating efficacy of (+)-catharanthine. We infer that (+)-catharanthine potentiates the GABAAR through several H-bond interactions with a binding site located in the ß(+)/α(-) interface in the transmembrane domain, near the TM2-TM3 loop, where it overlaps with loreclezole binding site.


Asunto(s)
Etomidato , Neuroesteroides , Sitios de Unión , Etomidato/química , Etomidato/farmacología , Simulación del Acoplamiento Molecular , Receptores de GABA-A/metabolismo , Alcaloides de la Vinca , Ácido gamma-Aminobutírico/farmacología
19.
Bioengineered ; 13(1): 985-994, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34968169

RESUMEN

Osteoarthritis (OA) is a common chronic inflammatory disease associated with aging. Etomidate is an intravenous anesthetic with profound antioxidant and anti-inflammatory effects. We speculated that etomidate might exert a beneficial effect on OA. Herein, we explored the effects of etomidate on interleukin-1ß (IL-1ß)- induced chondrocytes. Our results prove that etomidate ameliorated the IL-1ß-induced oxidative stress in C28/12 chondrocytes by decreasing and increasing the reactive oxygen species (ROS) and glutathione peroxidase (GPx) levels, respectively. Etomidate prevented the IL-1ß-induced increase in the expressions of matrix metalloproteinase-3 (MMP-3) and matrix metalloproteinase-13 (MMP-13) in C28/I2 chondrocytes at both mRNA and protein levels. It also caused a significant reduction in the percentage of senescence-associated-ß-galactosidase (SA-ß-Gal)-stained chondrocytes, while inducing elevated telomerase activity in IL-1ß-treated C28/I2 chondrocytes. The expression levels of senescence regulators, plasminogen activator inhibitor-1 (PAI-1) and p16, were also inhibited by etomidate in IL-1ß-treated C28/I2 chondrocytes. In addition, etomidate caused the activation of Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), along with upregulated expression levels of phosphorylated AMPKα and phosphorylated acetyl-Co A carboxylase (ACC). Moreover, blockage of AMPK using compound C abolished the protective effects of etomidate on IL-1ß-challenged C28/I2 chondrocytes. Taken together, these results demonstrate that etomidate protected C28/I2 chondrocytes from IL-1ß-induced oxidative stress, ECM degradation, and cellular senescence via activating AMPK signaling.


Asunto(s)
Condrocitos/citología , Etomidato/farmacología , Interleucina-1beta/efectos adversos , Osteoartritis/metabolismo , Adenilato Quinasa/metabolismo , Línea Celular , Senescencia Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Etomidato/química , Matriz Extracelular/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Modelos Biológicos , Estructura Molecular , Osteoartritis/inducido químicamente , Osteoartritis/tratamiento farmacológico , Osteoartritis/genética , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
20.
J Org Chem ; 76(20): 8477-82, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21913645

RESUMEN

Nitrogen-containing fused heterocycles comprise many compounds that demonstrate interesting biological activities. A new synthetic approach involving Mitsunobu alkylation of imidazoles, pyrroles, and indoles followed by palladium-catalyzed cyclization has been developed providing access to fused heterocycles with a defined stereochemistry α to nitrogen. While ethyl imidazole or indole carboxylates are good substrates for Mitsunobu alkylation with optically pure secondary benzylic alcohols, the corresponding pyrroles are poor substrates presumably due to the increased pK(a) of the NH. The presence of a synthetically versatile trichloroacetyl functional group on the pyrroles significantly reduces the pK(a) and thereby facilitates Mitsunobu alkylation. Subsequent cyclization of the alkylated products mediated by palladium in the presence or absence of a ligand gave fused heterocycles in good to excellent yields.


Asunto(s)
Química Farmacéutica/métodos , Agonistas del GABA/síntesis química , Hipnóticos y Sedantes/síntesis química , Imidazoles/síntesis química , Indoles/síntesis química , Pirroles/síntesis química , Alquilación , Ansiedad/tratamiento farmacológico , Catálisis , Ciclización , Etomidato/análisis , Etomidato/química , Etomidato/farmacología , Agonistas del GABA/análisis , Agonistas del GABA/farmacología , Humanos , Hipnóticos y Sedantes/análisis , Hipnóticos y Sedantes/farmacología , Imidazoles/análisis , Indoles/análisis , Espectroscopía de Resonancia Magnética , Estructura Molecular , Nitrógeno/química , Paladio/química , Pirroles/análisis , Estereoisomerismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda