Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biophys J ; 2023 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-37752702

RESUMEN

The properties of a potentiator are typically evaluated by measuring its ability to enhance the magnitude of the control response. Analysis of the ability of drugs to potentiate responses from receptor channels takes place in the context of particular models to extract parameters for functional effects. In the often-used coagonist model, the agonist generating control activity and the potentiator enhancing the control activity make additive energetic contributions to stabilize the active state of the receptor. The energetic contributions are fixed and, once known, enable calculation of predicted receptor behavior at any concentration combination of agonist and potentiator. Here, we have examined the applicability of the coagonist model by measuring the relationship between the magnitude of receptor potentiation and the level of background activity. Ternary αßγ GABAA receptors were activated by GABA or the allosteric agonist propofol, or by a gain-of-function mutation, and etiocholanolone- or propofol-mediated potentiation of peak responses was measured. We show that the free energy change contributed by the modulators etiocholanolone or propofol is reduced at higher levels of control activity, thereby being in disagreement with basic principles of the coagonist model. Possible mechanisms underlying this discrepancy are discussed.

2.
Mol Pharmacol ; 104(3): 115-131, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37316350

RESUMEN

Acrylamide-derived compounds have been previously shown to act as modulators of members of the Cys-loop transmitter-gated ion channel family, including the mammalian GABAA receptor. Here we have synthesized and functionally characterized the GABAergic effects of a series of novel compounds (termed "DM compounds") derived from the previously characterized GABAA and the nicotinic α7 receptor modulator (E)-3-furan-2-yl-N-p-tolyl-acrylamide (PAM-2). Fluorescence imaging studies indicated that the DM compounds increase apparent affinity to the transmitter by up to 80-fold in the ternary αßγ GABAA receptor. Using electrophysiology, we show that the DM compounds, and the structurally related (E)-3-furan-2-yl-N-phenylacrylamide (PAM-4), have concurrent potentiating and inhibitory effects that can be isolated and observed under appropriate recording conditions. The potentiating efficacies of the DM compounds are similar to those of neurosteroids and benzodiazepines (ΔG ∼ -1.5 kcal/mol). Molecular docking, functionally confirmed by site-directed mutagenesis experiments, indicate that receptor potentiation is mediated by interactions with the classic anesthetic binding sites located in the transmembrane domain of the intersubunit interfaces. Inhibition by the DM compounds and PAM-4 was abolished in the receptor containing the α1(V256S) mutation, suggestive of similarities in the mechanism of action with that of inhibitory neurosteroids. Functional competition and mutagenesis experiments, however, indicate that the sites mediating inhibition by the DM compounds and PAM-4 differ from those mediating the action of the inhibitory steroid pregnenolone sulfate. SIGNIFICANCE STATEMENT: We have synthesized and characterized the actions of novel acrylamide-derived compounds on the mammalian GABAA receptor. We show that the compounds have concurrent potentiating effects mediated by the classic anesthetic binding sites, and inhibitory actions that bear mechanistic resemblance to but do not share binding sites with, the inhibitory steroid pregnenolone sulfate.


Asunto(s)
Anestésicos , Neuroesteroides , Animales , Receptores de GABA-A/metabolismo , Acrilamida/farmacología , Simulación del Acoplamiento Molecular , Sitios de Unión , Esteroides , Furanos/farmacología , Mamíferos/metabolismo
3.
Mol Pharmacol ; 101(2): 68-77, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34853153

RESUMEN

The GABAA receptor is inhibited by the endogenous sulfated steroids pregnenolone sulfate (PS) and dehydroepiandrosterone sulfate (DHEAS). It has been proposed in previous work that these steroids act by enhancing desensitization of the receptor. Here, we have investigated the modulatory effects of the steroids on the human α1ß3γ2L GABAA receptor. Using electrophysiology and quantitative model-based data analysis, we show that exposure to the steroid promotes occupancy of a nonconducting state that retains high affinity to the transmitter but whose properties differ from those of the classic, transmitter-induced desensitized state. From the analysis of the inhibitory actions of two combined steroids, we infer that PS and DHEAS act through shared or overlapping binding sites. SIGNIFICANCE STATEMENT: Previous work has proposed that sulfated neurosteroids inhibit the GABAA receptor by enhancing the rate of entry into the desensitized state. This study shows that the inhibitory steroids pregnenolone sulfate and dehydroepiandrosterone sulfate act through a common interaction site by stabilizing a distinct nonconducting state.


Asunto(s)
Sulfato de Deshidroepiandrosterona/farmacología , Antagonistas del GABA/farmacología , Pregnenolona/farmacología , Receptores de GABA-A/metabolismo , Animales , Sulfato de Deshidroepiandrosterona/química , Relación Dosis-Respuesta a Droga , Femenino , Antagonistas del GABA/química , Humanos , Neuroesteroides/química , Neuroesteroides/farmacología , Pregnenolona/química , Estabilidad Proteica , Receptores de GABA-A/química , Xenopus laevis
4.
PLoS Biol ; 17(3): e3000157, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30845142

RESUMEN

Neurosteroids are endogenous modulators of neuronal excitability and nervous system development and are being developed as anesthetic agents and treatments for psychiatric diseases. While gamma amino-butyric acid Type A (GABAA) receptors are the primary molecular targets of neurosteroid action, the structural details of neurosteroid binding to these proteins remain ill defined. We synthesized neurosteroid analogue photolabeling reagents in which the photolabeling groups were placed at three positions around the neurosteroid ring structure, enabling identification of binding sites and mapping of neurosteroid orientation within these sites. Using middle-down mass spectrometry (MS), we identified three clusters of photolabeled residues representing three distinct neurosteroid binding sites in the human α1ß3 GABAA receptor. Novel intrasubunit binding sites were identified within the transmembrane helical bundles of both the α1 (labeled residues α1-N408, Y415) and ß3 (labeled residue ß3-Y442) subunits, adjacent to the extracellular domains (ECDs). An intersubunit site (labeled residues ß3-L294 and G308) in the interface between the ß3(+) and α1(-) subunits of the GABAA receptor pentamer was also identified. Computational docking studies of neurosteroid to the three sites predicted critical residues contributing to neurosteroid interaction with the GABAA receptors. Electrophysiological studies of receptors with mutations based on these predictions (α1-V227W, N408A/Y411F, and Q242L) indicate that both the α1 intrasubunit and ß3-α1 intersubunit sites are critical for neurosteroid action.


Asunto(s)
Proteínas de la Membrana/metabolismo , Receptores de GABA/metabolismo , Animales , Sitios de Unión , Línea Celular , Electrofisiología , Femenino , Citometría de Flujo , Humanos , Espectrometría de Masas , Simulación del Acoplamiento Molecular , Muscimol/metabolismo , Neurotransmisores/metabolismo , Oocitos/metabolismo , Xenopus laevis
5.
Mol Pharmacol ; 100(1): 19-31, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33958479

RESUMEN

Prior work employing functional analysis, photolabeling, and X-ray crystallography have identified three distinct binding sites for potentiating steroids in the heteromeric GABAA receptor. The sites are located in the membrane-spanning domains of the receptor at the ß-α subunit interface (site I) and within the α (site II) and ß subunits (site III). Here, we have investigated the effects of mutations to these sites on potentiation of the rat α1ß2γ2L GABAA receptor by the endogenous neurosteroid allopregnanolone (3α5αP). The mutations were introduced alone or in combination to probe the additivity of effects. We show that the effects of amino acid substitutions in sites I and II are energetically additive, indicating independence of the actions of the two steroid binding sites. In site III, none of the mutations tested reduced potentiation by 3α5αP, nor did a mutation in site III modify the effects of mutations in sites I or II. We infer that the binding sites for 3α5αP act independently. The independence of steroid action at each site is supported by photolabeling data showing that mutations in either site I or site II selectively change steroid orientation in the mutated site without affecting labeling at the unmutated site. The findings are discussed in the context of linking energetic additivity to empirical changes in receptor function and ligand binding. SIGNIFICANCE STATEMENT: Prior work has identified three distinct binding sites for potentiating steroids in the heteromeric γ-aminobutyric acid type A receptor. This study shows that the sites act independently and additively in the presence of the steroid allopregnanolone and provide estimates of energetic contributions made by steroid binding to each site.


Asunto(s)
Sustitución de Aminoácidos , Pregnanolona/farmacología , Receptores de GABA-A/química , Animales , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Conformación Molecular , Simulación del Acoplamiento Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Pregnanolona/química , Ratas , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
6.
Mol Pharmacol ; 98(4): 303-313, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32873746

RESUMEN

Muscimol is a psychoactive isoxazole derived from the mushroom Amanita muscaria and a potent orthosteric agonist of the GABAA receptor. The binding of [3H]muscimol has been used to evaluate the distribution of GABAA receptors in the brain, and studies of modulation of [3H]muscimol binding by allosteric GABAergic modulators such as barbiturates and steroid anesthetics have provided insight into the modes of action of these drugs on the GABAA receptor. It has, however, not been feasible to directly apply interaction parameters derived from functional studies to describe the binding of muscimol to the receptor. Here, we employed the Monod-Wyman-Changeux concerted transition model to analyze muscimol binding isotherms. We show that the binding isotherms from recombinant α1ß3 GABAA receptors can be qualitatively predicted using electrophysiological data pertaining to properties of receptor activation and desensitization in the presence of muscimol. The model predicts enhancement of [3H]muscimol binding in the presence of the steroids allopregnanolone and pregnenolone sulfate, although the steroids interact with distinct sites and either enhance (allopregnanolone) or reduce (pregnenolone sulfate) receptor function. We infer that the concerted transition model can be used to link radioligand binding and electrophysiological data. SIGNIFICANCE STATEMENT: The study employs a three-state resting-active-desensitized model to link radioligand binding and electrophysiological data. We show that the binding isotherms can be qualitatively predicted using parameters estimated in electrophysiological experiments and that the model accurately predicts the enhancement of [3H]muscimol binding in the presence of the potentiating steroid allopregnanolone and the inhibitory steroid pregnenolone sulfate.


Asunto(s)
Agonistas de Receptores de GABA-A/farmacología , Muscimol/farmacología , Receptores de GABA-A/metabolismo , Esteroides/farmacología , Regulación Alostérica/efectos de los fármacos , Sitios de Unión , Células HEK293 , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Muscimol/química , Pregnanolona/farmacología , Pregnenolona/farmacología , Receptores de GABA-A/química , Receptores de GABA-A/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tritio/química
7.
Mol Pharmacol ; 98(6): 762-769, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32978327

RESUMEN

Synaptic GABAA receptors are alternately exposed to short pulses of a high, millimolar concentration of GABA and prolonged periods of low, micromolar concentration of the transmitter. Prior work has indicated that exposure to micromolar concentrations of GABA can both activate the postsynaptic receptors generating sustained low-amplitude current and desensitize the receptors, thereby reducing the peak amplitude of subsequent synaptic response. However, the precise relationship between tonic activation and reduction of peak response is not known. Here, we have measured the effect of prolonged exposure to GABA or the combination of GABA and the neurosteroid allopregnanolone, which was intended to desensitize a fraction of receptors, on a subsequent response to a high concentration of agonist in human α1ß3γ2L receptors expressed in Xenopus oocytes. We show that the reduction in the peak amplitude of the post-exposure test response correlates with the open probability of the preceding desensitizing response. Curve fitting of the inhibitory relationship yielded an IC50 of 12.5 µM and a Hill coefficient of -1.61. The activation and desensitization data were mechanistically analyzed in the framework of a three-state Resting-Active-Desensitized model. Using the estimated affinity, efficacy, and desensitization parameters, we calculated the amount of desensitization that would accumulate during a long (2-minute) application of GABA or GABA plus allopregnanolone. The results indicate that accumulation of desensitization depends on the level of activity rather than agonist or potentiator concentration per se. We estimate that in the presence of 1 µM GABA, approximately 5% of α1ß3γ2L receptors are functionally eliminated because of desensitization. SIGNIFICANCE STATEMENT: We present an analytical approach to quantify and predict the loss of activatable GABAA receptors due to desensitization in the presence of transmitter and the steroid allopregnanolone. The findings indicate that the peak amplitude of the synaptic response is influenced by ambient GABA and that changes in ambient concentrations of the transmitter and other GABAergic agents can modify tonically and phasically activated synaptic receptors in opposite directions.


Asunto(s)
Agonistas de Receptores de GABA-A/farmacología , Receptores de GABA-A/metabolismo , Potenciales Sinápticos/efectos de los fármacos , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Oocitos , Técnicas de Placa-Clamp , Pregnanolona/farmacología , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Xenopus laevis , Ácido gamma-Aminobutírico/farmacología
8.
Mol Pharmacol ; 98(4): 280-291, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32675382

RESUMEN

The ρ1 GABAA receptor is prominently expressed in the retina and is present at lower levels in several brain regions and other tissues. Although the ρ1 receptor is insensitive to many anesthetic drugs that modulate the heteromeric GABAA receptor, it maintains a rich and multifaceted steroid pharmacology. The receptor is negatively modulated by 5ß-reduced steroids, sulfated or carboxylated steroids, and ß-estradiol, whereas many 5α-reduced steroids potentiate the receptor. In this study, we analyzed modulation of the human ρ1 GABAA receptor by several neurosteroids, individually and in combination, in the framework of the coagonist concerted transition model. Experiments involving coapplication of two or more steroids revealed that the receptor contains at least three classes of distinct, nonoverlapping sites for steroids, one each for the inhibitory steroids pregnanolone (3α5ßP), 3α5ßP sulfate, and ß-estradiol. The site for 3α5ßP can accommodate the potentiating steroid 5αTHDOC. The findings are discussed with respect to receptor modulation by combinations of endogenous neurosteroids. SIGNIFICANCE STATEMENT: The study describes modulation of the ρ1 GABAA receptor by neurosteroids. The coagonist concerted transition model was used to determine overlap of binding sites for several inhibitory and potentiating steroids.


Asunto(s)
Desoxicorticosterona/análogos & derivados , Neuroesteroides/farmacología , Pregnanolona/farmacología , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Xenopus laevis/genética , Animales , Animales Modificados Genéticamente , Sitios de Unión , Desoxicorticosterona/química , Desoxicorticosterona/farmacología , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Modelos Moleculares , Estructura Molecular , Neuroesteroides/química , Pregnanolona/química , Receptores de GABA-A/genética
9.
Mol Pharmacol ; 95(1): 70-81, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30337372

RESUMEN

Under both physiologic and clinical conditions GABAA receptors are exposed to multiple agonists, including the transmitter GABA, endogenous or exogenous neuroactive steroids, and various GABAergic anesthetic and sedative drugs. The functional output of the receptor reflects the interplay among all active agents. We have investigated the activation of the concatemeric α1ß2γ2L GABAA receptor by combinations of agonists. Simulations of receptor activity using the coagonist concerted transition model demonstrate that the response amplitude in the presence of agonist combinations is highly dependent on whether the paired agonists interact with the same or distinct sites. The experimental data for receptor activation by agonist combinations were in agreement with the established views of the overlap of binding sites for several pairs of orthosteric (GABA, ß-alanine, and piperidine-4-sulfonic acid) and/or allosteric agents (propofol, pentobarbital, and several neuroactive steroids). Conversely, the degree of potentiation when two GABAergic agents are coapplied can be used to determine whether the compounds act by binding to the same or distinct sites. We show that common interaction sites mediate the actions of 5α- and 5ß-reduced neuroactive steroids, and natural and enantiomeric steroids. Furthermore, the results indicate that the anesthetics propofol and pentobarbital interact with partially shared binding sites. We propose that the findings may be used to predict the efficacy of drug mixtures in combination therapy and thus have potential clinical relevance.


Asunto(s)
Sitios de Unión/efectos de los fármacos , Agonistas del GABA/farmacología , Receptores de GABA-A/metabolismo , Regulación Alostérica/efectos de los fármacos , Anestésicos/farmacología , Animales , Neurotransmisores/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Pentobarbital/farmacología , Propofol/farmacología , Xenopus laevis , Ácido gamma-Aminobutírico/metabolismo
10.
Mol Pharmacol ; 96(3): 320-329, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31263018

RESUMEN

The two-state coagonist model has been successfully used to analyze and predict peak current responses of the γ-aminobutyric acid type A (GABAA) receptor. The goal of the present study was to provide a model-based description of GABAA receptor activity under steady-state conditions after desensitization has occurred. We describe the derivation and properties of the cyclic three-state resting-active-desensitized (RAD) model. The relationship of the model to receptor behavior was tested using concatemeric α1ß2γ2 GABAA receptors expressed in Xenopus oocytes. The receptors were activated by the orthosteric agonists GABA or ß-alanine, the allosteric agonist propofol, or combinations of GABA, propofol, pentobarbital, and the steroid allopregnanolone, and the observed steady-state responses were compared with those predicted by the model. A modified RAD model was employed to analyze and describe the actions on steady-state current of the inhibitory steroid pregnenolone sulfate. The findings indicate that the steady-state activity in the presence of multiple active agents that interact with distinct binding sites follows standard energetic additivity. The derived equations enable prediction of peak and steady-state activity in the presence of orthosteric and allosteric agonists, and the inhibitory steroid pregnenolone sulfate. SIGNIFICANCE STATEMENT: The study describes derivation and properties of a three-state resting-active-desensitized model. The model and associated equations can be used to analyze and predict peak and steady-state activity in the presence of one or more active agents.


Asunto(s)
Agonistas de Receptores de GABA-A/farmacología , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Xenopus laevis/genética , Regulación Alostérica , Animales , Complejos Multiproteicos/metabolismo , Pentobarbital/farmacología , Pregnanolona/farmacología , Propofol/farmacología , Receptores de GABA-A/genética , Xenopus laevis/metabolismo , beta-Alanina/farmacología
11.
Mol Pharmacol ; 93(2): 90-100, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29150461

RESUMEN

The concerted transition model for multimeric proteins is a simple formulation for analyzing the behavior of transmitter-gated ion channels. We used the model to examine the relationship between the EC50 for activation of the GABA type A (GABAA) receptor by the transmitter GABA and basal activity employing concatemeric ternary GABAA receptors expressed in Xenopus oocytes. Basal activity, reflecting the receptor function in the absence of the transmitter, can be changed either by mutation to increase constitutive activity or by the addition of a second agonist (acting at a different site) to increase background activity. The model predicts that either mechanism for producing a change in basal activity will result in identical effects on the EC50 We examined receptor activation by GABA while changing the level of basal activity with the allosterically acting anesthetics propofol, pentobarbital, or alfaxalone. We found that the relationship between EC50 and basal activity was well described by the concerted transition model. Changes in the basal activity by gain-of-function mutations also resulted in predictable changes in the EC50 Finally, we altered the number of GABA-binding sites by a mutation and again found that the relationship could be well described by the model. Overall, the results support the idea that interactions between the transmitter GABA and the allosteric agonists propofol, pentobarbital, or alfaxalone can be understood as reflecting additive and independent free energy changes, without assuming any specific interactions.


Asunto(s)
Agonistas del GABA/farmacología , Modelos Teóricos , Receptores de GABA-A/metabolismo , Regulación Alostérica , Anestésicos/farmacología , Animales , Sitios de Unión , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Agonistas del GABA/administración & dosificación , Modelos Biológicos , Mutación , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Pentobarbital/administración & dosificación , Pentobarbital/farmacología , Pregnanodionas/administración & dosificación , Pregnanodionas/farmacología , Propofol/administración & dosificación , Propofol/farmacología , Xenopus laevis , Ácido gamma-Aminobutírico/genética , Ácido gamma-Aminobutírico/metabolismo
12.
Mol Pharmacol ; 93(5): 468-476, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29439087

RESUMEN

GABAA receptors activated by the transmitter GABA are potentiated by several allosterically acting drugs, including the intravenous anesthetic propofol. Propofol can also directly activate the receptor, albeit at higher concentrations. Previous functional studies have identified amino acid residues whose substitution reduces potentiation of GABA-activated receptors by propofol while enhancing the ability of propofol to directly activate the receptor. One interpretation of such observations is that the mutation has specific effects on the sites or processes involved in potentiation or activation. We show here that divergent effects on potentiation and direct activation can be mediated by increased constitutive open probability in the mutant receptor without any specific effect on the interactions between the allosteric drug and the receptor. By simulating GABAA receptor activity using the concerted transition model, we demonstrate that the predicted degree of potentiation is reduced as the level of constitutive activity increases. The model further predicts that a potentiating effect of an allosteric modulator is a computable value that depends on the level of constitutive activity, the amplitude of the response to the agonist, and the amplitude of the direct activating response to the modulator. Specific predictions were confirmed by electrophysiological data from the binary α1ß3 and concatemeric ternary ß2α1γ2L+ß2α1 GABAA receptors. The corollaries of reduced potentiation due to increased constitutive activity are isobolograms that conform to simple additivity and a loss of separation between the concentration-response relationships for direct activation and potentiation.


Asunto(s)
Anestésicos Intravenosos/farmacología , Mutación , Propofol/farmacología , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/genética , Regulación Alostérica , Anestésicos Intravenosos/metabolismo , Animales , Células Cultivadas , Sinergismo Farmacológico , Agonistas del GABA/farmacología , Humanos , Propofol/metabolismo , Receptores de GABA-A/metabolismo , Xenopus , Ácido gamma-Aminobutírico/metabolismo
13.
Mol Pharmacol ; 93(2): 178-189, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29192122

RESUMEN

GABAA receptors can be directly activated and potentiated by the intravenous anesthetic propofol. Previous photolabeling, modeling, and functional data have identified two binding domains through which propofol acts on the GABAA receptor. These domains are defined by the ß(M286) residue at the ß"+"-α"-" interface in the transmembrane region and the ß(Y143) residue near the ß"-" surface in the junction between the extracellular and transmembrane domains. In the ternary receptor, there are predicted to be two copies of each class of sites, for a total of four sites per receptor. We used ß2α1γ2L and ß2α1 concatemeric constructs to determine the functional effects of the ß(Y143W) and ß(M286W) mutations to gain insight into the number of functional binding sites for propofol and the energetic contributions stemming from propofol binding to the individual sites. A mutation of each of the four sites affected the response to propofol, indicating that each of the four sites is functional in the wild-type receptor. The mutations mainly impaired stabilization of the open state by propofol, i.e., reduced gating efficacy. The effects were similar for mutations at either site and were largely additive and independent of the presence of other Y143W or M286W mutations in the receptor. The two classes of sites appeared to differ in affinity for propofol, with the site affected by M286W having about a 2-fold higher affinity. Our analysis indicates there may be one or two additional functionally equivalent binding sites for propofol, other than those modified by substitutions at ß(Y143) and ß(M286).


Asunto(s)
Anestésicos Intravenosos/farmacología , Propofol/farmacología , Receptores de GABA-A/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/metabolismo , Animales , Sitios de Unión , Relación Dosis-Respuesta a Droga , Activación del Canal Iónico/efectos de los fármacos , Mutación , Propofol/administración & dosificación , Propofol/metabolismo , Receptores de GABA-A/química , Receptores de GABA-A/genética , Xenopus laevis
14.
Mol Pharmacol ; 92(5): 556-563, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28790148

RESUMEN

Drug interactions are often analyzed in terms of isobolograms. In the isobologram, the line connecting the axial points corresponding to the concentrations of two different drugs that produce an effect of the same magnitude is termed an isobole of additivity. Although the isobole of additivity can be a straight line in some special cases, previous work has proposed that it is curvilinear when the two drugs differ in their maximal effects or Hill slopes. Modulators of transmitter-gated ion channels have a wide range of maximal effects as well as Hill slopes, suggesting that the isoboles for drug actions on ion channel function are not linear. In this study, we have conducted an analysis of direct activation and potentiation of the human α1ß2γ2L GABAA receptor to demonstrate that: 1) curvilinear isoboles of additivity are predicted by a concerted transition model where the binding of each GABAergic drug additively and independently reduces the free energy of the open receptor compared with the closed receptor; and 2) experimental data for receptor activation using the agonist pair of GABA and propofol or potentiation of responses to a low concentration of GABA by the drug pair of alfaxalone and propofol agree very well with predictions. The approach assuming independent energetic contributions from GABAergic drugs enables, at least for the drug combinations tested, a straightforward method to accurately predict functional responses to any combination of concentrations.


Asunto(s)
Agonistas del GABA/metabolismo , Propofol/metabolismo , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Sitios de Unión/fisiología , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Femenino , Agonistas del GABA/administración & dosificación , Humanos , Propofol/administración & dosificación , Xenopus laevis , Ácido gamma-Aminobutírico/administración & dosificación
15.
Mol Pharmacol ; 91(2): 100-109, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27895161

RESUMEN

Physostigmine is a well known inhibitor of acetylcholinesterase, which can also activate, potentiate, and inhibit acetylcholine receptors, including neuronal nicotinic receptors comprising α4 and ß2 subunits. We have found that the two stoichiometric forms of this receptor differ in the effects of physostigmine. The form containing three copies of α4 and two of ß2 was potentiated at low concentrations of acetylcholine chloride (ACh) and physostigmine, whereas the form containing two copies of α4 and three of ß2 was inhibited. Chimeric constructs of subunits indicated that the presence of inhibition or potentiation depended on the source of the extracellular ligand binding domain of the subunit. Further sets of chimeric constructs demonstrated that a portion of the ACh binding domain, the E loop, is a key determinant. Transferring the E loop from the ß2 subunit to the α4 subunit resulted in strong inhibition, whereas the reciprocal transfer reduced inhibition. To control the number and position of the incorporated chimeric subunits, we expressed chimeric constructs with subunit dimers. Surprisingly, incorporation of a subunit with an altered E loop had similar effects whether it contributed either to an intersubunit interface containing a canonical ACh binding site or to an alternative interface. The observation that the α4 E loop is involved suggests that physostigmine interacts with regions of subunits that contribute to the ACh binding site, whereas the lack of interface specificity indicates that interaction with a particular ACh binding site is not the critical factor.


Asunto(s)
Fisostigmina/farmacología , Receptores Nicotínicos/química , Receptores Nicotínicos/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Humanos , Dominios Proteicos , Estructura Secundaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad , Xenopus
16.
Mol Pharmacol ; 92(3): 318-326, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28630263

RESUMEN

Physostigmine can potentiate and inhibit neuronal nicotinic receptors, in addition to inhibiting the activity of acetylcholinesterase. We found that receptors containing three copies of the α2 subunit are inhibited by low concentrations of physostigmine in contrast to receptors containing three copies of the α4 subunit that are potentiated. We exploited this observation to determine the regions required for the actions of physostigmine. Chimeric constructs of the α2 and α4 subunits located two regions in the extracellular amino-terminal domain of the subunit: the E loop (a loop of the transmitter-binding domain) and a region closer to the amino-terminus that collectively could completely determine the different effects of physostigmine. Point mutations then identified a single residue, α2(I92) versus α4(R92), that, when combined with transfer of the E loop, could convert the inhibition seen with α2 subunits to potentiation and the potentiation seen with α4 subunits to inhibition. In addition, other point mutations could affect the extent of potentiation or inhibition, indicating that a more extensive set of interactions in the amino-terminal domain plays some role in the actions of physostigmine.


Asunto(s)
Antagonistas Nicotínicos/farmacología , Fisostigmina/farmacología , Receptores Nicotínicos/química , Animales , Humanos , Ratones , Dominios Proteicos , Subunidades de Proteína , Receptores Nicotínicos/efectos de los fármacos , Relación Estructura-Actividad , Xenopus laevis
17.
ACS Chem Neurosci ; 15(5): 909-915, 2024 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-38386612

RESUMEN

Activation of the GABAA receptor is associated with numerous behavioral end points ranging from anxiolysis to deep anesthesia. The specific behavioral effect of a GABAergic compound is considered to correlate with the degree of its functional effect on the receptor. Here, we tested the hypothesis that a low-efficacy allosteric potentiator of the GABAA receptor may act, due to a ceiling effect, as a sedative with reduced and limited action. We synthesized a derivative, named (3α,5ß)-20-methyl-pregnane-3,20-diol (KK-235), of the GABAergic neurosteroid 5ß-pregnane-3α,20α-diol. Using electrophysiology, we showed that KK-235 is a low-efficacy potentiator of the synaptic-type α1ß2γ2L GABAA receptor. In the zebrafish larvae behavioral assay, KK-235 was found to only partially block the inverted photomotor response (PMR) and to weakly reduce swimming behavior, whereas the high-efficacy GABAergic steroid (3α,5α,17ß)-3-hydroxyandrostane-17-carbonitrile (ACN) fully blocked PMR and spontaneous swimming. Coapplication of KK-235 reduced the potentiating effect of ACN in an electrophysiological assay and dampened its sedative effect in behavioral experiments. We propose that low-efficacy GABAergic potentiators may be useful as sedatives with limited action.


Asunto(s)
Neuroesteroides , Receptores de GABA-A , Animales , Pez Cebra , Esteroides/farmacología , Pregnanos
18.
Br J Pharmacol ; 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38978389

RESUMEN

BACKGROUND AND PURPOSE: Neurosteroids are allosteric modulators of GABAA currents, acting through several functional binding sites although their affinity and specificity for each site are unknown. The goal of this study was to measure steady-state binding affinities of various neurosteroids for specific sites on the GABAA receptor. EXPERIMENTAL APPROACH: Two methods were developed to measure neurosteroid binding affinity: (1) quenching of specific tryptophan residues in neurosteroid binding sites by the neurosteroid 17-methylketone group, and (2) FRET between MQ290 (an intrinsically fluorescent neurosteroid) and tryptophan residues in the binding sites. The assays were developed using ELIC-α1GABAAR, a chimeric receptor containing transmembrane domains of the α1-GABAA receptor. Tryptophan mutagenesis was used to identify specific interactions. KEY RESULTS: Allopregnanolone (3α-OH neurosteroid) was shown to bind at intersubunit and intrasubunit sites with equal affinity, whereas epi-allopregnanolone (3ß-OH neurosteroid) binds at the intrasubunit site. MQ290 formed a strong FRET pair with W246, acting as a site-specific probe for the intersubunit site. The affinity and site-specificity of several neurosteroid agonists and inverse agonists was measured using the MQ290 binding assay. The FRET assay distinguishes between competitive and allosteric inhibition of MQ290 binding and demonstrated an allosteric interaction between the two neurosteroid binding sites. CONCLUSIONS AND IMPLICATIONS: The affinity and specificity of neurosteroid binding to two sites in the ELIC-α1GABAAR were directly measured and an allosteric interaction between the sites was revealed. Adaptation of the MQ290 FRET assay to a plate-reader format will enable screening for high affinity agonists and antagonists for neurosteroid binding sites.

19.
Biomolecules ; 13(4)2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37189445

RESUMEN

The positive allosteric modulators (PAMs) of the α7 nicotinic receptor N-(5-Cl-2-hydroxyphenyl)-N'-[2-Cl-5-(trifluoromethyl)phenyl]-urea (NS-1738) and (E)-3-(furan-2-yl)-N-(p-tolyl)-acrylamide (PAM-2) potentiate the α1ß2γ2L GABAA receptor through interactions with the classic anesthetic binding sites located at intersubunit interfaces in the transmembrane domain of the receptor. In the present study, we employed mutational analysis to investigate in detail the involvement and contributions made by the individual intersubunit interfaces to receptor modulation by NS-1738 and PAM-2. We show that mutations to each of the anesthetic-binding intersubunit interfaces (ß+/α-, α+/ß-, and γ+/ß-), as well as the orphan α+/γ- interface, modify receptor potentiation by NS-1738 and PAM-2. Furthermore, mutations to any single interface can fully abolish potentiation by the α7-PAMs. The findings are discussed in the context of energetic additivity and interactions between the individual binding sites.


Asunto(s)
Anestésicos , Receptores de GABA-A , Regulación Alostérica , Receptor Nicotínico de Acetilcolina alfa 7/genética , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Sitios de Unión , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Humanos , Animales
20.
Biomolecules ; 13(2)2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36830708

RESUMEN

The neurosteroid allopregnanolone (ALLO) and pregnanolone (PREG), are equally effective positive allosteric modulators (PAMs) of GABAA receptors. Interestingly, the PAM effects of ALLO are strongly enantioselective, whereas those of PREG are not. This study was aimed at determining the basis for this difference in enantioselectivity. The oocyte electrophysiology studies showed that ent-ALLO potentiates GABA-elicited currents in α1ß3 GABAA receptors with lower potency and efficacy than ALLO, PREG or ent-PREG. The small PAM effect of ent-ALLO was prevented by the α1(Q242L) mutation in the intersubunit neurosteroid binding site between the ß3 and α1 subunits. Consistent with this result, neurosteroid analogue photolabeling with mass spectrometric readout, showed that ent-ALLO binds weakly to the ß3-α1 intersubunit binding site in comparison to ALLO, PREG and ent-PREG. Rigid body docking predicted that ent-ALLO binds in the intersubunit site with a preferred orientation 180° different than ALLO, PREG or ent-PREG, potentially explaining its weak binding and effect. Photolabeling studies did not identify differences between ALLO and ent-ALLO binding to the α1 or ß3 intrasubunit binding sites that also mediate neurosteroid modulation of GABAA receptors. The results demonstrate that differential binding of ent-ALLO and ent-PREG to the ß3-α1 intersubunit site accounts for the difference in enantioselectivity between ALLO and PREG.


Asunto(s)
Neuroesteroides , Receptores de GABA-A , Receptores de GABA-A/metabolismo , Estereoisomerismo , Pregnanolona/farmacología , Ácido gamma-Aminobutírico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA