Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 165(2): 434-448, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26997484

RESUMEN

Mutations in the Kv3.3 potassium channel (KCNC3) cause cerebellar neurodegeneration and impair auditory processing. The cytoplasmic C terminus of Kv3.3 contains a proline-rich domain conserved in proteins that activate actin nucleation through Arp2/3. We found that Kv3.3 recruits Arp2/3 to the plasma membrane, resulting in formation of a relatively stable cortical actin filament network resistant to cytochalasin D that inhibits fast barbed end actin assembly. These Kv3.3-associated actin structures are required to prevent very rapid N-type channel inactivation during short depolarizations of the plasma membrane. The effects of Kv3.3 on the actin cytoskeleton are mediated by the binding of the cytoplasmic C terminus of Kv3.3 to Hax-1, an anti-apoptotic protein that regulates actin nucleation through Arp2/3. A human Kv3.3 mutation within a conserved proline-rich domain produces channels that bind Hax-1 but are impaired in recruiting Arp2/3 to the plasma membrane, resulting in growth cones with deficient actin veils in stem cell-derived neurons.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteína 2 Relacionada con la Actina/metabolismo , Proteína 3 Relacionada con la Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Canales de Potasio Shaw/metabolismo , Ataxias Espinocerebelosas/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Secuencia de Aminoácidos , Membrana Celular/metabolismo , Datos de Secuencia Molecular , Mutación , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Canales de Potasio Shaw/química , Canales de Potasio Shaw/genética , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo
2.
Nature ; 604(7904): 190-194, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355020

RESUMEN

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


Asunto(s)
Benzodiazepinas , Receptores de GABA-A , Transducción de Señal , Benzodiazepinas/farmacología , Sitios de Unión , Microscopía por Crioelectrón , Histamina/metabolismo , Humanos , Ligandos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , RNA-Seq , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Receptores de GABA-A/ultraestructura , Análisis de la Célula Individual , Ácido gamma-Aminobutírico/metabolismo
3.
Nature ; 565(7740): 516-520, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30602789

RESUMEN

Type A γ-aminobutyric acid (GABAA) receptors are pentameric ligand-gated ion channels and the main drivers of fast inhibitory neurotransmission in the vertebrate nervous system1,2. Their dysfunction is implicated in a range of neurological disorders, including depression, epilepsy and schizophrenia3,4. Among the numerous assemblies that are theoretically possible, the most prevalent in the brain are the α1ß2/3γ2 GABAA receptors5. The ß3 subunit has an important role in maintaining inhibitory tone, and the expression of this subunit alone is sufficient to rescue inhibitory synaptic transmission in ß1-ß3 triple knockout neurons6. So far, efforts to generate accurate structural models for heteromeric GABAA receptors have been hampered by the use of engineered receptors and the presence of detergents7-9. Notably, some recent cryo-electron microscopy reconstructions have reported 'collapsed' conformations8,9; however, these disagree with the structure of the prototypical pentameric ligand-gated ion channel the Torpedo nicotinic acetylcholine receptor10,11, the large body of structural work on homologous homopentameric receptor variants12 and the logic of an ion-channel architecture. Here we present a high-resolution cryo-electron microscopy structure of the full-length human α1ß3γ2L-a major synaptic GABAA receptor isoform-that is functionally reconstituted in lipid nanodiscs. The receptor is bound to a positive allosteric modulator 'megabody' and is in a desensitized conformation. Each GABAA receptor pentamer contains two phosphatidylinositol-4,5-bisphosphate molecules, the head groups of which occupy positively charged pockets in the intracellular juxtamembrane regions of α1 subunits. Beyond this level, the intracellular M3-M4 loops are largely disordered, possibly because interacting post-synaptic proteins are not present. This structure illustrates the molecular principles of heteromeric GABAA receptor organization and provides a reference framework for future mechanistic investigations of GABAergic signalling and pharmacology.


Asunto(s)
Microscopía por Crioelectrón , Membrana Dobles de Lípidos/química , Receptores de GABA-A/química , Receptores de GABA-A/ultraestructura , Regulación Alostérica , Secuencia de Aminoácidos , Sitios de Unión , Conductividad Eléctrica , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Nanoestructuras/química , Nanoestructuras/ultraestructura , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/ultraestructura , Estructura Cuaternaria de Proteína , Receptores de GABA-A/metabolismo
4.
Nature ; 565(7740): 454-459, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30602790

RESUMEN

Type-A γ-aminobutyric (GABAA) receptors are ligand-gated chloride channels with a very rich pharmacology. Some of their modulators, including benzodiazepines and general anaesthetics, are among the most successful drugs in clinical use and are common substances of abuse. Without reliable structural data, the mechanistic basis for the pharmacological modulation of GABAA receptors remains largely unknown. Here we report several high-resolution cryo-electron microscopy structures in which the full-length human α1ß3γ2L GABAA receptor in lipid nanodiscs is bound to the channel-blocker picrotoxin, the competitive antagonist bicuculline, the agonist GABA (γ-aminobutyric acid), and the classical benzodiazepines alprazolam and diazepam. We describe the binding modes and mechanistic effects of these ligands, the closed and desensitized states of the GABAA receptor gating cycle, and the basis for allosteric coupling between the extracellular, agonist-binding region and the transmembrane, pore-forming region. This work provides a structural framework in which to integrate previous physiology and pharmacology research and a rational basis for the development of GABAA receptor modulators.


Asunto(s)
Alprazolam/química , Bicuculina/química , Microscopía por Crioelectrón , Diazepam/química , Picrotoxina/química , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Alprazolam/farmacología , Benzodiazepinas/química , Benzodiazepinas/farmacología , Bicuculina/farmacología , Unión Competitiva/efectos de los fármacos , Diazepam/farmacología , Moduladores del GABA/química , Moduladores del GABA/farmacología , Humanos , Ligandos , Modelos Moleculares , Nanoestructuras/química , Picrotoxina/farmacología
5.
Nature ; 566(7744): E8, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30733619

RESUMEN

In Fig. 5b, d, the arrows showing transmembrane domain rotations were inadvertently pointing clockwise instead of anticlockwise. Similarly, 'anticlockwise' should have been 'clockwise' in the sentence 'This conformational change of the ECD triggers a clockwise rotation of the TMD.' In Extended Data Table 1, the units of the column 'Model resolution' should have been Å instead of Å2. These errors have been corrected online.

6.
J Biol Chem ; 290(38): 23432-46, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26229099

RESUMEN

In the process of developing safer general anesthetics, isomers of anesthetic ethers and barbiturates have been discovered that act as convulsants and inhibitors of γ-aminobutyric acid type A receptors (GABAARs) rather than potentiators. It is unknown whether these convulsants act as negative allosteric modulators by binding to the intersubunit anesthetic-binding sites in the GABAAR transmembrane domain (Chiara, D. C., Jayakar, S. S., Zhou, X., Zhang, X., Savechenkov, P. Y., Bruzik, K. S., Miller, K. W., and Cohen, J. B. (2013) J. Biol. Chem. 288, 19343-19357) or to known convulsant sites in the ion channel or extracellular domains. Here, we show that S-1-methyl-5-propyl-5-(m-trifluoromethyl-diazirynylphenyl) barbituric acid (S-mTFD-MPPB), a photoreactive analog of the convulsant barbiturate S-MPPB, inhibits α1ß3γ2 but potentiates α1ß3 GABAAR responses. In the α1ß3γ2 GABAAR, S-mTFD-MPPB binds in the transmembrane domain with high affinity to the γ(+)-ß(-) subunit interface site with negative energetic coupling to GABA binding in the extracellular domain at the ß(+)-α(-) subunit interfaces. GABA inhibits S-[(3)H]mTFD-MPPB photolabeling of γ2Ser-280 (γM2-15') in this site. In contrast, within the same site GABA enhances photolabeling of ß3Met-227 in ßM1 by an anesthetic barbiturate, R-[(3)H]methyl-5-allyl-5-(m-trifluoromethyl-diazirynylphenyl)barbituric acid (mTFD-MPAB), which differs from S-mTFD-MPPB in structure only by chirality and two hydrogens (propyl versus allyl). S-mTFD-MPPB and R-mTFD-MPAB are predicted to bind in different orientations at the γ(+)-ß(-) site, based upon the distance in GABAAR homology models between γ2Ser-280 and ß3Met-227. These results provide an explanation for S-mTFD-MPPB inhibition of α1ß3γ2 GABAAR function and provide a first demonstration that an intersubunit-binding site in the GABAAR transmembrane domain binds negative and positive allosteric modulators.


Asunto(s)
Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/farmacología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Células HEK293 , Humanos , Estructura Terciaria de Proteína , Receptores de GABA-A/genética
7.
J Physiol ; 593(22): 4943-61, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26378885

RESUMEN

KEY POINTS: Most barbiturates are anaesthetics but unexpectedly a few are convulsants whose mechanism of action is poorly understood. We synthesized and characterized a novel pair of chiral barbiturates that are capable of photolabelling their binding sites on GABAA receptors. In mice the S-enantiomer is a convulsant, but the R-enantiomer is an anticonvulsant. The convulsant S-enantiomer binds solely at an inhibitory site. It is both an open state inhibitor and a resting state inhibitor. Its action is pH independent, suggesting the pyrimidine ring plays little part in binding. The inhibitory site is not enantioselective because the R-enantiomer inhibits with equal affinity. In contrast, only the anticonvulsant R-enantiomer binds to the enhancing site on open channels, causing them to stay open longer. The enhancing site is enantioselective. The in vivo actions of the convulsant S-enantiomer are accounted for by its interactions with GABAA receptors. ABSTRACT: Most barbiturates are anaesthetics but a few unexpectedly are convulsants. We recently located the anaesthetic sites on GABAA receptors (GABAA Rs) by photolabelling with an anaesthetic barbiturate. To apply the same strategy to locate the convulsant sites requires the creation and mechanistic characterization of a suitable agent. We synthesized enantiomers of a novel, photoactivable barbiturate, 1-methyl-5-propyly-5-(m-trifluoromethyldiazirinyl) phenyl barbituric acid (mTFD-MPPB). In mice, S-mTFD-MPPB acted as a convulsant, whereas R-mTFD-MPPB acted as an anticonvulsant. Using patch clamp electrophysiology and fast solution exchange on recombinant human α1 ß3 γ2L GABAA Rs expressed in HEK cells, we found that S-mTFD-MPPB inhibited GABA-induced currents, whereas R-mTFD-MPPB enhanced them. S-mTFD-MPPB caused inhibition by binding to either of two inhibitory sites on open channels with bimolecular kinetics. It also inhibited closed, resting state receptors at similar concentrations, decreasing the channel opening rate and shifting the GABA concentration-response curve to the right. R-mTFD-MPPB, like most anaesthetics, enhanced receptor gating by rapidly binding to allosteric sites on open channels, initiating a rate-limiting conformation change to stabilized open channel states. These states had slower closing rates, thus shifting the GABA concentration-response curve to the left. Under conditions when most GABAA Rs were open, an inhibitory action of R-mTFD-MPPB was revealed that had a similar IC50 to that of S-mTFD-MPPB. Thus, the inhibitory sites are not enantioselective, and the convulsant action of S-mTFD-MPPB results from its negligible affinity for the enhancing, anaesthetic sites. Interactions with these two classes of barbiturate binding sites on GABAA Rs underlie the enantiomers' different pharmacological activities in mice.


Asunto(s)
Anticonvulsivantes/farmacología , Convulsivantes/farmacología , GABAérgicos/farmacología , Fenobarbital/análogos & derivados , Receptores de GABA-A/metabolismo , Potenciales de Acción , Regulación Alostérica , Animales , Anticonvulsivantes/química , Convulsivantes/química , GABAérgicos/química , Células HEK293 , Humanos , Activación del Canal Iónico , Isomerismo , Masculino , Ratones , Fenobarbital/química , Fenobarbital/farmacología , Receptores de GABA-A/química , Xenopus
8.
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
9.
Mol Pharmacol ; 83(6): 1200-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23525330

RESUMEN

A central axiom of ligand-receptor theory is that agonists bind more tightly to active than to inactive receptors. However, measuring agonist affinity in inactive receptors is confounded by concomitant activation. We identified a cysteine substituted mutant γ-aminobutyric acid type A (GABAA) receptor with unique characteristics allowing the determination of allosteric agonist site occupancy in both inactive and active receptors. Etomidate, the allosteric agonist, is an anesthetic that activates or modulates α1ß2γ2L GABAA receptors via transmembrane sites near ß2M286 residues in M3 domains. Voltage-clamp electrophysiology studies of α1ß2M286Cγ2L receptors show that GABA is an efficacious agonist and that etomidate modulates GABA-activated activity, but direct etomidate agonism is absent. Quantitative analysis of mutant activity using an established Monod-Wyman-Changeux (MWC) allosteric model indicates that the intrinsic efficacy of etomidate, defined as its relative affinity for active versus inactive receptors, is lower than in wild-type receptors. Para-chloromercuribenzene sulfonate covalently modifies ß2M286C side-chain sulfhydryls, irreversibly altering GABA-induced currents. Etomidate concentration dependently reduces the apparent rate of ß2M286C-pCMBS bond formation, tracked electrophysiologically. High etomidate concentrations completely protect the ß2M286C suflhydryl from covalent modification, suggesting close steric interactions. The 50% protective etomidate concentration (PC50) is 14 µM in inactive receptors and 1.1 to 2.2 µM during GABA-activation, experimentally demonstrating that activated receptors bind etomidate more avidly than do inactive receptors. The experimental PC50 values are remarkably close to, and therefore validate, MWC model predictions for etomidate dissociation constants in both inactive and active receptors. Our results support MWC models as valid frameworks for understanding the agonism, coagonism, and modulation of ligand-gated ion channels.


Asunto(s)
Anestésicos Generales/farmacología , Cisteína/genética , Etomidato/farmacología , Agonistas de Receptores de GABA-A/farmacología , Receptores de GABA-A/fisiología , Sitio Alostérico , Animales , Sitios de Unión , Femenino , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Oocitos/efectos de los fármacos , Oocitos/fisiología , Técnicas de Placa-Clamp , Receptores de GABA-A/genética , Xenopus laevis
10.
Anesth Analg ; 116(3): 573-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23400978

RESUMEN

BACKGROUND: 5-Hydroxytryptamine type 3 (5-HT(3)) receptors are excitatory ion channels belonging to the cys-loop family of ligand-gated ion channels. They are involved in nausea and vomiting and their antagonists are used clinically as antiemetic drugs. We previously reported the development of a novel pyrrole analog of etomidate, (R)-ethyl 1-(1-phenylethyl)-1H-pyrrole-2-carboxylate (carboetomidate), which retains etomidate's desirable anesthetic and hemodynamic properties, but lacks its potent inhibitory effect on adrenocorticotropic hormone-stimulated steroid synthesis. Also in contrast to etomidate, carboetomidate potently inhibits nicotinic acetylcholine receptors. Because nicotinic acetylcholine and 5-HT(3) receptors are highly homologous, we hypothesized that carboetomidate might also potently inhibit 5-HT(3) receptors with potentially important implications for the drug's emetogenic activity. In the current studies, we investigated and compared modulation of 5-HT(3A) receptors by carboetomidate and etomidate. METHODS: 5-HT(3) receptors were heterologously expressed in human embryonic kidney cells. Drugs were applied with a multichannel superfusion pipette coupled to piezoelectric elements, and currents were recorded from cells in either the whole-cell or excised outside-out patch configuration of patch-clamp recordings. RESULTS: Carboetomidate and etomidate inhibited integrated 5-HT(3A) receptor-mediated currents with respective half-inhibitory concentrations of 1.9 µM (95% confidence interval [CI] = 1.4-2.7 µM) and 25 µM (95% CI = 17-37 µM). These values may be compared with respective hypnotic concentrations of 5.4 and 2.3 µM. This inhibition reflected hypnotic effects on peak current amplitudes and desensitization rates. Half-inhibitory concentrations for reducing peak current amplitudes were 34 µM (95% CI = 24-48 µM) for carboetomidate and 171 µM (95% CI = 128-228 µM) for etomidate. Half-inhibitory concentrations for reducing the desensitization time constant were 3.5 µM (95% CI = 2.4-5.1 µM) for carboetomidate and 36 µM (95% CI = 21-59 µM) for etomidate. CONCLUSIONS: In contrast to etomidate, carboetomidate inhibits 5-HT(3A) receptor-mediated currents at hypnotic concentrations. This inhibition is primarily the result of enhancing the rate of desensitization. Because carboetomidate potently inhibits 5-HT(3A) receptors, it may be less emetogenic than etomidate.


Asunto(s)
Etomidato/farmacología , Hipnóticos y Sedantes/farmacología , Pirroles/farmacología , Receptores de Serotonina 5-HT3/metabolismo , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Células HEK293 , Humanos , Receptores de Serotonina 5-HT3/fisiología
11.
Anesthesiology ; 111(4): 774-84, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19741491

RESUMEN

BACKGROUND: Etomidate acts at gamma-Aminobutyric acid type A (GABAA) receptors containing beta2 or beta3, but not beta1 subunits. Mutations at beta residue 265 (Ser in beta1; Asn in beta2 or beta3) profoundly affect etomidate sensitivity. Whether these mutations alter etomidate binding remains uncertain. METHODS: Heterologously expressed alpha1beta2gamma2L GABAA receptors and receptors with beta2(N265S) or beta2(N265M) mutations were studied electrophysiologically in both Xenopus oocytes and HEK293 cells. Experiments quantified the impact of beta2N265 mutations or substituting beta1 for beta2 on basal channel activation, GABA EC50, maximal GABA efficacy, etomidate-induced leftward shift in GABA responses, etomidate direct activation, and rapid macrocurrent kinetics. Results were analyzed in the context of an established allosteric co-agonist mechanism. RESULTS: Mutations produced only small changes in basal channel activity, GABA EC50, maximal GABA efficacy, and macrocurrent kinetics. Relative to wild-type, beta2(N265S) reduced etomidate enhancement of apparent GABA affinity six-fold, and it reduced etomidate direct activation efficacy 14-fold. beta2(N265M) totally eliminated both etomidate modulation of GABA responses and direct channel activation. Mechanism-based analysis showed that the function of both mutants remains consistent with the allosteric co-agonist model and that beta2(N265S) reduced etomidate allosteric efficacy five-fold, whereas etomidate-binding affinity dropped threefold. Experiments swapping beta2 subunits for beta1 indicated that etomidate efficacy is reduced 34-fold, whereas binding affinity drops less than two-fold. CONCLUSIONS: Mutations at beta2N265 profoundly alter etomidate sensitivity with only small changes in basal and GABA-dependent channel activity. Mutations at the beta2N265 residue or replacement of beta2 with beta1 influence etomidate efficacy much more than binding to inactive receptors.


Asunto(s)
Etomidato/farmacología , Agonistas del GABA/farmacología , Hipnóticos y Sedantes/farmacología , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/genética , Algoritmos , Animales , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , ADN Complementario/genética , Interpretación Estadística de Datos , Electrofisiología , Femenino , Humanos , Mutación/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Xenopus laevis
12.
J Neurosci ; 27(10): 2617-27, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344399

RESUMEN

The Slack (sequence like a calcium-activated K channel) and Slick (sequence like an intermediate conductance K channel) genes, which encode sodium-activated K+ (K(Na)) channels, are expressed at high levels in neurons of the medial nucleus of the trapezoid body (MNTB) in the auditory brainstem. These neurons lock their action potentials to incoming stimuli with a high degree of temporal precision. Channels with unitary properties similar to those of Slack and/or Slick channels, which are gated by [Na+]i and [Cl-]i and by changes in cytoplasmic ATP levels, are present in MNTB neurons. Manipulations of the level of K(Na) current in MNTB neurons, either by increasing levels of internal Na+ or by exposure to a pharmacological activator of Slack channels, significantly enhance the accuracy of timing of action potentials at high frequencies of stimulation. These findings suggest that such fidelity of timing at high frequencies may be attributed in part to high-conductance K(Na) channels.


Asunto(s)
Vías Auditivas/fisiología , Neuronas Aferentes/fisiología , Canales de Potasio/fisiología , Tiempo de Reacción/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Bitionol/farmacología , Tronco Encefálico/metabolismo , Simulación por Computador , Conductividad Eléctrica , Estimulación Eléctrica , Electrofisiología , Técnicas In Vitro , Ratones , Modelos Neurológicos , Proteínas del Tejido Nervioso , Neuronas/metabolismo , Neuronas/fisiología , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Canales de potasio activados por Sodio , Tiempo de Reacción/efectos de los fármacos , Sodio/farmacología
13.
Mol Pharmacol ; 74(6): 1687-95, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18805938

RESUMEN

The potent general anesthetic etomidate produces its effects by enhancing GABA(A) receptor activation. Its photolabel analog [(3)H]azi-etomidate labels residues within transmembrane domains on alpha and beta subunits: alphaMet236 and betaMet286. We hypothesized that these methionines contribute to etomidate sites formed at alpha-beta subunit interfaces and that increasing side-chain bulk and hydrophobicity at either locus would mimic etomidate binding and block etomidate effects. Channel activity was electrophysiologically quantified in alpha(1)beta(2)gamma(2L) receptors with alpha(1)M236W or beta(2)M286W mutations, in both the absence and the presence of etomidate. Measurements included spontaneous activation, GABA EC(50), etomidate agonist potentiation, etomidate direct activation, and rapid macrocurrent kinetics. Both alpha(1)M236W and beta(2)M286W mutations induced spontaneous channel opening, lowered GABA EC(50), increased maximal GABA efficacy, and slowed current deactivation, mimicking effects of etomidate on alpha(1)beta(2)gamma(2L) channels. These changes were larger with alpha(1)M236W than with beta(2)M286W. Etomidate (3.2 muM) reduced GABA EC(50) much less in alpha(1)M236Wbeta(2)gamma(2L) receptors (2-fold) than in wild type (23-fold). However, etomidate was more potent and efficacious in directly activating alpha(1)M236Wbeta(2)gamma(2L) compared with wild type. In alpha(1)beta(2)M286Wgamma(2L) receptors, etomidate induced neither agonist-potentiation nor direct channel activation. These results support the hypothesis that alpha(1)Met236 and beta(2)Met286 are within etomidate sites that allosterically link to channel gating. Although alpha(1)M236W produced the larger impact on channel gating, beta(2)M286W produced more profound changes in etomidate sensitivity, suggesting a dominant role in drug binding. Furthermore, quantitative mechanistic analysis demonstrated that wild-type and mutant results are consistent with the presence of only one class of etomidate sites mediating both agonist potentiation and direct activation.


Asunto(s)
Anestésicos Generales/farmacología , Etomidato/análogos & derivados , Etiquetas de Fotoafinidad/farmacología , Receptores de GABA-A/fisiología , Triptófano/genética , Regulación Alostérica , Animales , Sitios de Unión , Línea Celular , Canales de Cloruro/fisiología , Sinergismo Farmacológico , Etomidato/farmacología , Femenino , Agonistas del GABA/farmacología , Humanos , Técnicas In Vitro , Activación del Canal Iónico , Mutación , Oocitos/fisiología , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Receptores de GABA-A/genética , Xenopus laevis , Ácido gamma-Aminobutírico/farmacología
14.
PLoS One ; 13(1): e0191583, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29352320

RESUMEN

The inhibitory γ-aminobutyric acid type A receptors are implicated in numerous physiological processes, including cognition and inhibition of neurotransmission, rendering them important molecular targets for many classes of drugs. Functionally, the entire GABAAR family of receptors can be subdivided into phasic, fast acting synaptic receptors, composed of α-, ß- and γ-subunits, and tonic extrasynaptic receptors, many of which contain the δ-subunit in addition to α- and ß-subunits. Whereas the subunit arrangement of the former group is agreed upon, that of the αßδ GABAARs remains unresolved by electrophysiological and pharmacological research. To resolve such issues will require biophysical techniques that demand quantities of receptor that have been previously unavailable. Therefore, we have engineered a stable cell line with tetracycline inducible expression of human α4-, ß3- and N-terminally Flag-tagged δ-subunits. This cell line achieved a specific activity between 15 and 20 pmol [3H]muscimol sites/mg of membrane protein, making it possible to obtain 1 nmole of purified α4ß3δ GABAAR from sixty 15-cm culture dishes. When induced, these cells exhibited agonist-induced currents with characteristics comparable to those previously reported for this receptor and a pharmacology that included strong modulation by etomidate and the δ-subunit-specific ligand, DS2. Immunoaffinity purification and reconstitution in CHAPS/asolectin micelles resulted in the retention of equilibrium allosteric interactions between the separate agonist, anesthetic and DS2 sites. Moreover, all three subunits retained glycosylation. The establishment of this well-characterized cell line will allow molecular level studies of tonic receptors to be undertaken.


Asunto(s)
Receptores de GABA-A/biosíntesis , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Cinética , Ingeniería de Proteínas , Subunidades de Proteína , Ensayo de Unión Radioligante , Receptores de GABA-A/genética , Receptores de GABA-A/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Transfección , Ácido gamma-Aminobutírico/metabolismo
15.
Eur J Med Chem ; 136: 334-347, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28505538

RESUMEN

Neuroactive steroids are potent positive allosteric modulators of GABAA receptors (GABAAR), but the locations of their GABAAR binding sites remain poorly defined. To discover these sites, we synthesized two photoreactive analogs of alphaxalone, an anesthetic neurosteroid targeting GABAAR, 11ß-(4-azido-2,3,5,6-tetrafluorobenzoyloxy)allopregnanolone, (F4N3Bzoxy-AP) and 11-aziallopregnanolone (11-AziAP). Both photoprobes acted with equal or higher potency than alphaxalone as general anesthetics and potentiators of GABAAR responses, left-shifting the GABA concentration - response curve for human α1ß3γ2 GABAARs expressed in Xenopus oocytes, and enhancing [3H]muscimol binding to α1ß3γ2 GABAARs expressed in HEK293 cells. With EC50 of 110 nM, 11-AziAP is one the most potent general anesthetics reported. [3H]F4N3Bzoxy-AP and [3H]11-AziAP, at anesthetic concentrations, photoincorporated into α- and ß-subunits of purified α1ß3γ2 GABAARs, but labeling at the subunit level was not inhibited by alphaxalone (30 µM). The enhancement of photolabeling by 3H-azietomidate and 3H-mTFD-MPAB in the presence of either of the two steroid photoprobes indicates the neurosteroid binding site is different from, but allosterically related to, the etomidate and barbiturate sites. Our observations are consistent with two hypotheses. First, F4N3Bzoxy-AP and 11-aziAP bind to a high affinity site in such a pose that the 11-photoactivatable moiety, that is rigidly attached to the steroid backbone, points away from the protein. Second, F4N3Bzoxy-AP, 11-aziAP and other steroid anesthetics, which are present at very high concentration at the lipid-protein interface due to their high lipophilicity, act via low affinity sites, as proposed by Akk et al. (Psychoneuroendocrinology2009, 34S1, S59-S66).


Asunto(s)
Pregnanodionas/farmacología , Receptores de GABA-A/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Estructura Molecular , Pregnanodionas/síntesis química , Pregnanodionas/química , Relación Estructura-Actividad
16.
Hear Res ; 206(1-2): 133-45, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16081004

RESUMEN

The firing patterns of neurons in central auditory pathways encode specific features of sound stimuli, such as frequency, intensity and localization in space. The generation of the appropriate pattern depends, to a major extent, on the properties of the voltage-dependent potassium channels in these neurons. The mammalian auditory pathways that compute the direction of a sound source are located in the brainstem and include the connection from bushy cells in the anteroventral cochlear nucleus (AVCN) to the principal neurons of the medial nucleus of the trapezoid body (MNTB). To preserve the fidelity of timing of action potentials that is required for sound localization, these neurons express several types of potassium channels, including the Kv3 and Kv1 families of voltage-dependent channels and the Slick and Slack sodium-dependent channels. These channels determine the pattern of action potentials and the amount of neurotransmitter released during repeated stimulation. The amplitude of currents carried by one of these channels, the Kv3.1b channel, is regulated in the short term by protein phosphorylation, and in the long term, by changes in gene expression, such that the intrinsic excitability of the neurons is constantly being regulated by the ambient auditory environment.


Asunto(s)
Potenciales de Acción/fisiología , Vías Auditivas/fisiología , Nervio Coclear/fisiología , Neuronas/fisiología , Canales de Potasio/metabolismo , Núcleos Vestibulares/fisiología , Animales , Núcleo Coclear/fisiología , Humanos , Fosforilación , Canales de Potasio/genética , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Tiempo de Reacción , Sinapsis/metabolismo , Factores de Tiempo
17.
Protein Sci ; 23(2): 157-66, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24288268

RESUMEN

Gamma-aminobutyric acid type A receptors (GABA(A)Rs) are the most important inhibitory chloride ion channels in the central nervous system and are major targets for a wide variety of drugs. The subunit compositions of GABA(A)Rs determine their function and pharmacological profile. GABAA Rs are heteropentamers of subunits, and (α1)2 (ß3)2 (γ2L)1 is a common subtype. Biochemical and biophysical studies of GABA(A)Rs require larger quantities of receptors of defined subunit composition than are currently available. We previously reported high-level production of active human α1ß3 GABA(A)R using tetracycline-inducible stable HEK293 cells. Here we extend the strategy to receptors containing three different subunits. We constructed a stable tetracycline-inducible HEK293-TetR cell line expressing human (N)-FLAG-α1ß3γ2L-(C)-(GGS)3 GK-1D4 GABA(A)R. These cells achieved expression levels of 70-90 pmol [(3)H]muscimol binding sites/15-cm plate at a specific activity of 15-30 pmol/mg of membrane protein. Incorporation of the γ2 subunit was confirmed by the ratio of [(3)H]flunitrazepam to [(3)H]muscimol binding sites and sensitivity of GABA-induced currents to benzodiazepines and zinc. The α1ß3γ2L GABA(A)Rs were solubilized in dodecyl-D-maltoside, purified by anti-FLAG affinity chromatography and reconstituted in CHAPS/asolectin at an overall yield of ∼ 30%. Typical purifications yielded 1.0-1.5 nmoles of [(3)H]muscimol binding sites/60 plates. Receptors with similar properties could be purified by 1D4 affinity chromatography with lower overall yield. The composition of the purified, reconstituted receptors was confirmed by ligand binding, Western blot, and proteomics. Allosteric interactions between etomidate and [(3)H]muscimol binding were maintained in the purified state.


Asunto(s)
Subunidades de Proteína/biosíntesis , Receptores de GABA-A/biosíntesis , Regulación Alostérica , Células HEK293 , Humanos , Muscimol/química , Unión Proteica , Subunidades de Proteína/química , Receptores de GABA-A/química , Receptores de GABA-A/aislamiento & purificación
18.
Protein Sci ; 19(9): 1728-38, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20662008

RESUMEN

The human neuronal Cys-loop ligand-gated ion channel superfamily of ion channels are important determinants of human behavior and the target of many drugs. It is essential for their structural characterization to achieve high-level expression in a functional state. The aim of this work was to establish stable mammalian cell lines that enable high-level heterologous production of pure receptors in a state that supports agonist-induced allosteric conformational changes. In a tetracycline-inducible stable human embryonic kidney cells (HEK293S) cell line, GABA(A) receptors containing α1 and ß3 subunits could be expressed with specific activities of 29-34 pmol/mg corresponding to 140-170 pmol/plate, the highest expression level reported so far. Comparable figures for serotonin (5-HT(3A)) receptors were 49-63 pmol/mg and 245-315 pmol/plate. The expression of 10 nmol of either receptor in suspension in a bioreactor required 0.3-3.0 L. Both receptor constructs had a FLAG epitope inserted at the N-terminus and could be purified in one step after solubilization using ANTI-FLAG affinity chromatography with yields of 30-40%. Purified receptors were functional. Binding of the agonist [(3)H]muscimol to the purified GABA(A)R was enhanced allosterically by the general anesthetic etomidate, and purified 5-hydroxytryptamine-3A receptor supported serotonin-stimulated cation flux when reconstituted into lipid vesicles.


Asunto(s)
Antibacterianos/farmacología , Expresión Génica , Células HEK293/metabolismo , Receptores de GABA-A/genética , Receptores de Serotonina 5-HT3/genética , Tetraciclina/farmacología , Animales , Cromatografía de Afinidad , Clonación Molecular/métodos , Electrofisiología , Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Unión Proteica , Receptores de GABA-A/aislamiento & purificación , Receptores de GABA-A/metabolismo , Receptores de Serotonina 5-HT3/aislamiento & purificación , Receptores de Serotonina 5-HT3/metabolismo , Solubilidad , Transfección/métodos
19.
J Med Chem ; 53(17): 6432-44, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20704351

RESUMEN

We synthesized the R- and S-enantiomers of ethyl 1-(1-(4-(3-((trifluoromethyl)-3H-diazirin-3-yl)phenyl)ethyl)-1H-imidazole-5-carboxylate (trifluoromethyldiazirinyl-etomidate), or TFD-etomidate, a novel photoactivable derivative of the stereoselective general anesthetic etomidate (R-(2-ethyl 1-(phenylethyl)-1H-imidazole-5-carboxylate)). Anesthetic potency was similar to etomidate's, but stereoselectivity was reversed and attenuated. Relative to etomidate, TFD-etomidate was a more potent inhibitor of the excitatory receptors, nAChR (nicotinic acetylcholine receptor) ((alpha1)(2)beta1delta1gamma1) and 5-HT(3A)R (serotonin type 3A receptor), causing significant inhibition at anesthetic concentrations. S- but not R-TFD-etomidate enhanced currents elicited from inhibitory alpha1beta2gamma2L GABA(A)Rs by low concentrations of GABA, but with a lower efficacy than R-etomidate, and site-directed mutagenesis suggests they act at different sites. [(3)H]TFD-etomidate photolabeled the alpha-subunit of the nAChR in a manner allosterically regulated by agonists and noncompetitive inhibitors. TFD-etomidate's novel pharmacology is unlike that of etomidate derivatives with photoactivable groups in the ester position, which behave like etomidate, suggesting that it will further enhance our understanding of anesthetic mechanisms.


Asunto(s)
Anestésicos Generales/síntesis química , Diazometano/análogos & derivados , Etomidato/análogos & derivados , Canales Iónicos/fisiología , Luz , Regulación Alostérica , Anestésicos Generales/química , Anestésicos Generales/farmacología , Animales , Bovinos , Corteza Cerebral/metabolismo , Diazometano/síntesis química , Diazometano/química , Diazometano/farmacología , Etomidato/síntesis química , Etomidato/química , Etomidato/farmacología , Femenino , Técnicas In Vitro , Activación del Canal Iónico , Larva , Ligandos , Mutagénesis Sitio-Dirigida , Antagonistas Nicotínicos/síntesis química , Antagonistas Nicotínicos/química , Antagonistas Nicotínicos/farmacología , Oocitos/efectos de los fármacos , Oocitos/fisiología , Etiquetas de Fotoafinidad/síntesis química , Etiquetas de Fotoafinidad/química , Etiquetas de Fotoafinidad/farmacología , Subunidades de Proteína/fisiología , Ensayo de Unión Radioligante , Receptores de GABA-A/genética , Receptores de GABA-A/fisiología , Receptores Nicotínicos/fisiología , Receptores de Serotonina 5-HT3/fisiología , Antagonistas del Receptor de Serotonina 5-HT3 , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Torpedo , Xenopus laevis , Ácido gamma-Aminobutírico/farmacología
20.
J Biol Chem ; 283(32): 22283-94, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18539595

RESUMEN

Modulation of some Kv3 family potassium channels by protein kinase C (PKC) regulates their amplitude and kinetics and adjusts firing patterns of auditory neurons in response to stimulation. Nevertheless, little is known about the modulation of Kv3.3, a channel that is widely expressed throughout the nervous system and is the dominant Kv3 family member in auditory brainstem. We have cloned the cDNA for the Kv3.3 channel from mouse brain and have expressed it in a mammalian cell line and in Xenopus oocytes to characterize its biophysical properties and modulation by PKC. Kv3.3 currents activate at positive voltages and undergo inactivation with time constants of 150-250 ms. Activators of PKC increased current amplitude and removed inactivation of Kv3.3 currents, and a specific PKC pseudosubstrate inhibitor peptide prevented the effects of the activators. Elimination of the first 78 amino acids of the N terminus of Kv3.3 produced noninactivating currents suggesting that PKC modulates N-type inactivation, potentially by phosphorylation of sites in this region. To identify potential phosphorylation sites, we investigated the response of channels in which serines in this N-terminal domain were subjected to mutagenesis. Our results suggest that serines at positions 3 and 9 are potential PKC phosphorylation sites. Computer simulations of model neurons suggest that phosphorylation of Kv3.3 by PKC may allow neurons to maintain action potential height during stimulation at high frequencies, and may therefore contribute to stimulus-induced changes in the intrinsic excitability of neurons such as those of the auditory brainstem.


Asunto(s)
Proteína Quinasa C/metabolismo , Canales de Potasio Shaw/antagonistas & inhibidores , Canales de Potasio Shaw/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/fisiología , Células CHO , Simulación por Computador , Cricetinae , Cricetulus , Ratones , Datos de Secuencia Molecular , Mutación , Neuronas/fisiología , Fosforilación , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Serina/metabolismo , Canales de Potasio Shaw/química , Canales de Potasio Shaw/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA