Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 32(14): 3016-3032.e3, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35688155

RESUMEN

The mechanisms of volatile anesthetic action remain among the most perplexing mysteries of medicine. Across phylogeny, volatile anesthetics selectively inhibit mitochondrial complex I, and they also depress presynaptic excitatory signaling. To explore how these effects are linked, we studied isoflurane effects on presynaptic vesicle cycling and ATP levels in hippocampal cultured neurons from wild-type and complex I mutant (Ndufs4(KO)) mice. To bypass complex I, we measured isoflurane effects on anesthetic sensitivity in mice expressing NADH dehydrogenase (NDi1). Endocytosis in physiologic concentrations of glucose was delayed by effective behavioral concentrations of isoflurane in both wild-type (τ [unexposed] 44.8 ± 24.2 s; τ [exposed] 116.1 ± 28.1 s; p < 0.01) and Ndufs4(KO) cultures (τ [unexposed] 67.6 ± 16.0 s; τ [exposed] 128.4 ± 42.9 s; p = 0.028). Increasing glucose, to enhance glycolysis and increase ATP production, led to maintenance of both ATP levels and endocytosis (τ [unexposed] 28.0 ± 14.4; τ [exposed] 38.2 ± 5.7; reducing glucose worsened ATP levels and depressed endocytosis (τ [unexposed] 85.4 ± 69.3; τ [exposed] > 1,000; p < 0.001). The block in recycling occurred at the level of reuptake of synaptic vesicles into the presynaptic cell. Expression of NDi1 in wild-type mice caused behavioral resistance to isoflurane for tail clamp response (EC50 Ndi1(-) 1.27% ± 0.14%; Ndi1(+) 1.55% ± 0.13%) and halothane (EC50 Ndi1(-) 1.20% ± 0.11%; Ndi1(+) 1.46% ± 0.10%); expression of NDi1 in neurons improved hippocampal function, alleviated inhibition of presynaptic recycling, and increased ATP levels during isoflurane exposure. The clear alignment of cell culture data to in vivo phenotypes of both isoflurane-sensitive and -resistant mice indicates that inhibition of mitochondrial complex I is a primary mechanism of action of volatile anesthetics.


Asunto(s)
Anestésicos por Inhalación , Isoflurano , Adenosina Trifosfato , Anestésicos por Inhalación/farmacología , Animales , Complejo I de Transporte de Electrón/genética , Endocitosis , Glucosa , Isoflurano/farmacología , Ratones
2.
Br J Anaesth ; 128(1): 77-88, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34857359

RESUMEN

BACKGROUND: If anaesthetics cause permanent cognitive deficits in some children, the implications are enormous, but the molecular causes of anaesthetic-induced neurotoxicity, and consequently possible therapies, are still debated. Anaesthetic exposure early in development can be neurotoxic in the invertebrate Caenorhabditis elegans causing endoplasmic reticulum (ER) stress and defects in chemotaxis during adulthood. We screened this model organism for compounds that alleviated neurotoxicity, and then tested these candidates for efficacy in mice. METHODS: We screened compounds for alleviation of ER stress induction by isoflurane in C. elegans assayed by induction of a green fluorescent protein (GFP) reporter. Drugs that inhibited ER stress were screened for reduction of the anaesthetic-induced chemotaxis defect. Compounds that alleviated both aspects of neurotoxicity were then blindly tested for the ability to inhibit induction of caspase-3 by isoflurane in P7 mice. RESULTS: Isoflurane increased ER stress indicated by increased GFP reporter fluorescence (240% increase, P<0.001). Nine compounds reduced induction of ER stress by isoflurane by 90-95% (P<0.001 in all cases). Of these compounds, tetraethylammonium chloride and trehalose also alleviated the isoflurane-induced defect in chemotaxis (trehalose by 44%, P=0.001; tetraethylammonium chloride by 23%, P<0.001). In mouse brain, tetraethylammonium chloride reduced isoflurane-induced caspase staining in the anterior cortical (-54%, P=0.007) and hippocampal regions (-46%, P=0.002). DISCUSSION: Tetraethylammonium chloride alleviated isoflurane-induced neurotoxicity in two widely divergent species, raising the likelihood that it may have therapeutic value. In C. elegans, ER stress predicts isoflurane-induced neurotoxicity, but is not its cause.


Asunto(s)
Isoflurano/toxicidad , Síndromes de Neurotoxicidad/prevención & control , Tetraetilamonio/farmacología , Anestésicos por Inhalación/toxicidad , Animales , Caenorhabditis elegans , Caspasa 3/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Ratones , Síndromes de Neurotoxicidad/etiología , Especificidad de la Especie
3.
Elife ; 102021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34254587

RESUMEN

Volatile anesthetics (VAs) are widely used in medicine, but the mechanisms underlying their effects remain ill-defined. Though routine anesthesia is safe in healthy individuals, instances of sensitivity are well documented, and there has been significant concern regarding the impact of VAs on neonatal brain development. Evidence indicates that VAs have multiple targets, with anesthetic and non-anesthetic effects mediated by neuroreceptors, ion channels, and the mitochondrial electron transport chain. Here, we characterize an unexpected metabolic effect of VAs in neonatal mice. Neonatal blood ß-hydroxybutarate (ß-HB) is rapidly depleted by VAs at concentrations well below those necessary for anesthesia. ß-HB in adults, including animals in dietary ketosis, is unaffected. Depletion of ß-HB is mediated by citrate accumulation, malonyl-CoA production by acetyl-CoA carboxylase, and inhibition of fatty acid oxidation. Adults show similar significant changes to citrate and malonyl-CoA, but are insensitive to malonyl-CoA, displaying reduced metabolic flexibility compared to younger animals.


Asunto(s)
Anestésicos/metabolismo , Anestésicos/farmacología , Ácido 3-Hidroxibutírico , Acetil-CoA Carboxilasa/metabolismo , Animales , Citratos/metabolismo , Ácido Cítrico/metabolismo , Ácidos Grasos/metabolismo , Femenino , Glucosa/metabolismo , Hipoglucemia , Isoflurano/metabolismo , Cetosis , Masculino , Malonil Coenzima A/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias , Oxidación-Reducción
4.
Anesthesiology ; 134(6): 901-914, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33909880

RESUMEN

BACKGROUND: Ndufs4 knockout (KO) mice are defective in mitochondrial complex I function and hypersensitive to inhibition of spinal cord-mediated response to noxious stimuli by volatile anesthetics. It was hypothesized that, compared to wild-type, synaptic or intrinsic neuronal function is hypersensitive to isoflurane in spinal cord slices from knockout mice. METHODS: Neurons from slices of the vestibular nucleus, central medial thalamus, and spinal cord from wild-type and the global Ndufs4 knockout were patch clamped. Unstimulated synaptic and intrinsic neuronal characteristics were measured in response to isoflurane. Norfluoxetine was used to block TREK channel conductance. Cholinergic cells were labeled with tdTomato. RESULTS: All values are reported as means and 95% CIs. Spontaneous synaptic activities were not different between the mutant and control. Isoflurane (0.6%; 0.25 mM; Ndufs4[KO] EC95) increased the holding current in knockout (ΔHolding current, 126 pA [95% CI, 99 to 152 pA]; ΔHolding current P < 0.001; n = 21) but not wild-type (ΔHolding current, 2 7 pA [95% CI, 9 to 47 pA]; ΔHolding current, P = 0.030; n = 25) spinal cord slices. Knockout and wild-type ΔHolding currents were significantly different (P < 0.001). Changes comparable to those in the knockout were seen in the wild type only in 1.8% (0.74 mM) isoflurane (ΔHolding current, 72 pA [95% CI, 43 to 97 pA]; ΔHolding current, P < 0.001; n = 13), the control EC95. Blockade of action potentials indicated that the increased holding current in the knockout was not dependent on synaptic input (ΔHolding current, 154 pA [95% CI, 99 to 232 pA]; ΔHolding current, P = 0.506 compared to knockout without blockade; n = 6). Noncholinergic neurons mediated the increase in holding current sensitivity in Ndufs4 knockout. The increased currents were blocked by norfluoxetine. CONCLUSIONS: Isoflurane increased an outwardly rectifying potassium current in ventral horn neurons of the Ndufs4(KO) mouse at a concentration much lower than in controls. Noncholinergic neurons in the spinal cord ventral horn mediated the effect. Presynaptic functions in Ndufs4(KO) slices were not hypersensitive to isoflurane. These data link anesthetic sensitivity, mitochondrial function, and postsynaptic channel activity.


Asunto(s)
Anestésicos , Isoflurano , Anestésicos/farmacología , Animales , Complejo I de Transporte de Electrón , Isoflurano/farmacología , Ratones , Ratones Noqueados , Mitocondrias , Médula Espinal
5.
Neuroscience ; 349: 35-47, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28237815

RESUMEN

Several phosphorylation signaling pathways have been implicated in the pathogenesis of epilepsy arising from both genetic causes and acquired insults to the brain. Identification of dysfunctional signaling pathways in epilepsy may provide novel targets for antiepileptic therapies. We previously described a deficit in phosphorylation signaling mediated by p38 mitogen-activated protein kinase (p38 MAPK) that occurs in an animal model of temporal lobe epilepsy, and that produces neuronal hyperexcitability measured in vitro. We asked whether in vivo pharmacological manipulation of p38 MAPK activity would influence seizure frequency in chronically epileptic animals. Administration of a p38 MAPK inhibitor, SB203580, markedly worsened spontaneous seizure frequency, consistent with prior in vitro results. However, anisomycin, a non-specific p38 MAPK activator, significantly increased seizure frequency. We hypothesized that this unexpected result was due to activation of a related MAPK, c-Jun N-terminal kinase (JNK). Administration of JNK inhibitor SP600125 significantly decreased seizure frequency in a dose-dependent manner without causing overt behavioral abnormalities. Biochemical analysis showed increased JNK expression and activity in untreated epileptic animals. These results show for the first time that JNK is hyperactivated in an animal model of epilepsy, and that phosphorylation signaling mediated by JNK may represent a novel antiepileptic target.


Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Animales , Anisomicina/farmacología , Antiinflamatorios no Esteroideos/farmacología , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Physiol ; 593(13): 2779-92, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25820761

RESUMEN

KEY POINTS: Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, particularly that of the HCN1 isoform, are enriched in the distal dendrites of hippocampal CA1 pyramidal neurons; these channels have physiological functions with respect to decreasing neuronal excitability. In the present study, we aimed to investigate phosphorylation as a mechanism controlling Ih amplitude and HCN1 surface expression in hippocampal principal neurons under normal physiological conditions. Tyrosine phosphorylation decreased Ih amplitude at maximal activation (maximal Ih ), without altering HCN1 surface expression, in two classes of hippocampal principal neurons. Inhibition of serine/threonine protein phosphatases 1 and 2A decreased maximal Ih and HCN1 surface expression in hippocampal principal neurons. Protein kinase C (PKC) activation irreversibly diminished Ih and HCN1 surface expression, whereas PKC inhibition augmented Ih and HCN1 surface expression. PKC activation increased HCN1 channel phosphorylation. These results demonstrate the novel finding of a phosphorylation mechanism, dependent on PKC activity, which bidirectionally modulates Ih amplitude and HCN1channel surface expression in hippocampal principal neurons under normal physiological conditions. ABSTRACT: Hyperpolarization-activated, cyclic nucleotide-gated (HCN) ion channels attenuate excitability in hippocampal pyramidal neurons. Loss of HCN channel-mediated current (Ih ), particularly that mediated by the HCN1 isoform, occurs with the development of epilepsy. Previously, we showed that, following pilocarpine-induced status epilepticus, there are two independent changes in HCN function in dendrites: decreased Ih amplitude associated with a loss of HCN1 surface expression and a hyperpolarizing shift in voltage-dependence of activation (gating). The hyperpolarizing shift in gating was attributed to decreased phosphorylation as a result of a loss of p38 mitogen-activated protein kinase activity and increased calcineurin activity; however, the mechanisms controlling Ih amplitude and HCN1 surface expression under epileptic or normal physiological conditions are poorly understood. We aimed to investigate phosphorylation as a mechanism regulating Ih amplitude and HCN1 surface expression (i.e. as is the case for HCN gating) in hippocampal principal neurons under normal physiological conditions. We discovered that inhibition of either tyrosine phosphatases or the serine/threonine protein phosphatases 1 and 2A decreased Ih at maximal activation in hippocampal CA1 pyramidal dendrites and pyramidal-like principal neuron somata from naïve rats. Furthermore, we found that inhibition of PP1/PP2A decreased HCN1 surface expression, whereas tyrosine phosphatase inhibition did not. Protein kinase C (PKC) activation reduced Ih amplitude and HCN1 surface expression, whereas PKC inhibition produced the opposite effect. Inhibition of protein phosphatases 1 and 2A and activation of PKC increased the serine phosphorylation state of the HCN1 protein. The effect of PKC activation on Ih was irreversible. These results indicate that PKC bidirectionally modulates Ih amplitude and HCN1 surface expression in hippocampal principal neurons.


Asunto(s)
Potenciales de Acción , Región CA1 Hipocampal/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteína Quinasa C/metabolismo , Células Piramidales/metabolismo , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Membrana Celular/metabolismo , Masculino , Transporte de Proteínas , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley
7.
J Neurosci ; 31(40): 14291-5, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976514

RESUMEN

Epilepsy is associated with loss of expression and function of hyperpolarization-activated, cyclic nucleotide-gated (HCN) ion channels. Previously, we showed that loss of HCN channel-mediated current (I(h)) occurred in the dendrites of CA1 hippocampal pyramidal neurons after pilocarpine-induced status epilepticus (SE), accompanied by loss of HCN1 channel protein expression. However, the precise onset and mechanistic basis of HCN1 channel loss post-SE was unclear, particularly whether it preceded the onset of spontaneous recurrent seizures and could contribute to epileptogenesis or development of the epileptic state. Here, we found that loss of I(h) and HCN1 channel expression began within an hour after SE and involved sequential processes of dendritic HCN1 channel internalization, delayed loss of protein expression, and later downregulation of mRNA expression. We also found that an in vitro SE model reproduced the rapid loss of dendritic I(h), demonstrating that this phenomenon was not specific to in vivo SE. Together, these results show that HCN1 channelopathy begins rapidly and persists after SE, involves both transcriptional and nontranscriptional mechanisms, and may be an early contributor to epileptogenesis.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/antagonistas & inhibidores , Dendritas/metabolismo , Regulación de la Expresión Génica , Hipocampo/patología , Células Piramidales/metabolismo , Células Piramidales/patología , Estado Epiléptico/metabolismo , Estado Epiléptico/patología , Animales , Recuento de Células/métodos , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Dendritas/patología , Hipocampo/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Masculino , Neuronas/metabolismo , Neuronas/patología , Canales de Potasio/genética , Canales de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/genética , Factores de Tiempo
8.
J Neurosci ; 30(19): 6678-88, 2010 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-20463230

RESUMEN

The onset of spontaneous seizures in the pilocarpine model of epilepsy causes a hyperpolarized shift in the voltage-dependent activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel-mediated current (Ih) in CA1 hippocampal pyramidal neuron dendrites, contributing to neuronal hyperexcitability and possibly to epileptogenesis. However, the specific mechanisms by which spontaneous seizures cause downregulation of HCN channel gating are yet unknown. We asked whether the seizure-dependent downregulation of HCN channel gating was due to altered phosphorylation signaling mediated by the phosphatase calcineurin (CaN) or the kinase p38 mitogen-activated protein kinase (p38 MAPK). We first found that CaN inhibition upregulated HCN channel gating and reduced neuronal excitability under normal conditions, showing that CaN is a strong modulator of HCN channels. We then found that an in vitro model of seizures (1 h in 0 Mg2+ and 50 microM bicuculline at 35-37 degrees C) reproduced the HCN channel gating change seen in vivo. Pharmacological inhibition of CaN or activation of p38 MAPK partially reversed the in vitro seizure-induced hyperpolarized shift in HCN channel gating, and the shift was fully reversed by the combination of CaN inhibition and p38 MAPK activation. We then demonstrated enhanced CaN activity as well as reduced p38 MAPK activity in vivo in the CA1 hippocampal area of chronically epileptic animals. Pharmacological reversal of these phosphorylation changes restored HCN channel gating downregulation and neuronal hyperexcitability in epileptic tissue to control levels. Together, these results suggest that alteration of two different phosphorylation pathways in epilepsy contributes to the downregulation of HCN channel gating, which consequently produces neuronal hyperexcitability and thus may be a target for novel antiepileptic therapies.


Asunto(s)
Región CA1 Hipocampal/fisiopatología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Dendritas/fisiología , Epilepsia/fisiopatología , Células Piramidales/fisiopatología , Animales , Bicuculina , Región CA1 Hipocampal/efectos de los fármacos , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Enfermedad Crónica , Dendritas/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Epilepsia/inducido químicamente , Técnicas In Vitro , Compuestos de Magnesio , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Fosforilación/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
J Neurosci ; 27(47): 13012-21, 2007 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18032674

RESUMEN

Ion channelopathy plays an important role in human epilepsy with a genetic cause and has been hypothesized to occur in epilepsy after acquired insults to the CNS as well. Acquired alterations of ion channel function occur after induction of status epilepticus (SE) in animal models of epilepsy, but it is unclear how they correlate with the onset of spontaneous seizures. We examined the properties of hyperpolarization-activated cation (HCN) channels in CA1 hippocampal pyramidal neurons in conjunction with video-EEG (VEEG) recordings to monitor the development of spontaneous seizures in the rat pilocarpine model of epilepsy. Our results showed that dendritic HCN channels were significantly downregulated at an acute time point 1 week postpilocarpine, with loss of channel expression and hyperpolarization of voltage-dependent activation. This downregulation progressively increased when epilepsy was established in the chronic period. Surprisingly, VEEG recordings during the acute period showed that a substantial fraction of animals were already experiencing recurrent seizures. Suppression of these seizures with phenobarbital reversed the change in the voltage dependence of I(h), the current produced by HCN channels, but did not affect the loss of HCN channel expression. These results suggest two mechanisms of HCN channel downregulation after SE, one dependent on and one independent of recurrent seizures. This early and progressive downregulation of dendritic HCN channel function increases neuronal excitability and may be associated with both the process of epileptogenesis and maintenance of the epileptic state.


Asunto(s)
Canalopatías/metabolismo , Canalopatías/fisiopatología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Dendritas/metabolismo , Epilepsia/metabolismo , Epilepsia/fisiopatología , Pilocarpina/toxicidad , Canales de Potasio/fisiología , Animales , Canalopatías/inducido químicamente , Dendritas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epilepsia/inducido químicamente , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Masculino , Ratas , Ratas Sprague-Dawley
10.
J Neurochem ; 96(3): 885-92, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16405501

RESUMEN

Abstract gamma-Aminobutyric acid type A (GABA(A)) receptors are molecular targets for alcohols. Previous work suggests that S270 and A291 residues in the transmembrane (TM) 2 and 3 domains of the GABA(A) receptor alpha subunit are components of an alcohol-binding pocket, and S270I and A291W mutants abolished ethanol potentiation. Our results showed that A295C and F296C residues in the TM3 of the GABA(A) receptor alpha1 subunit are accessible to hexylmethanethiosulfonate (HMTS) in the alcohol-bound state, but not in the resting state. Thus, the A295C and F296C sites become water-accessible as a result of alcohol-induced conformational changes. If S270 or A291 residues are sites of alcohol binding, then S270I or A291W mutations should prevent alcohol-induced conformational movements within the TM3 domain. To investigate this question, the accessibility of HMTS reagent to double mutants (A291W/A295C, A291W/F296C, S270I/A295C or S270I/F296C) in the presence of ethanol or hexanol was tested. The A291W or S270I mutations markedly reduced the accessibility of HMTS to all the double mutants in the ethanol-bound state, and to S270I/F296C, A291W/A295C and A291W/F296C double mutants in the hexanol-bound state, suggesting that the A291 or S270 residues are critical sites for alcohol binding and alcohol-induced conformational changes.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Receptores de GABA-A/química , Animales , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/métodos , Hexanoles/farmacología , Larva , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Potenciales de la Membrana/efectos de la radiación , Mesilatos/farmacología , Mutagénesis Sitio-Dirigida/métodos , Mutación , Oocitos , Técnicas de Placa-Clamp , Conformación Proteica/efectos de los fármacos , Receptores de GABA-A/genética , Relación Estructura-Actividad , Xenopus , Ácido gamma-Aminobutírico/farmacología
11.
J Biol Chem ; 280(1): 308-16, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15522868

RESUMEN

The substituted cysteine accessibility method has proven useful for investigating structural changes of the gamma-aminobutyric acid type A (GABA(A)) receptor during channel gating and allosteric modulation. In the present study, the surface accessibility and reaction rate of propyl- and hexyl-methanethiosulfonate to cysteine residues introduced into the third transmembrane segment of the GABA(A) receptor alpha(1) subunit were examined. GABA-induced currents in Xenopus oocytes expressing wild type and cysteine mutant GABA(A) receptors were recorded before and after application of methanethiosulfonate (MTS) reagents in the resting, GABA- or alcohol-bound (ethanol or hexanol) states. Our results indicate that a water-filled cavity exists around the Ala(291) and Tyr(294) residues of the third transmembrane segment, in agreement with previous results. Furthermore, our data indicate that a conformational change produced by alcohols (200 mM ethanol or 0.5 mM hexanol) exposure induces the water cavity around the A291C and Y294C residues to extend deeper, causing the A295C and F296C residues to become accessible to the MTS reagents. In addition, exposure of the A291C, Y294C, F296C, and V297C mutants to MTS reagents in the presence of GABA had significant effects on their GABA-induced currents, indicating that the water cavity around A291C and Y294C residues expanded to F296C and V297C by a structural movement caused by GABA binding. Our data show that GABA(A) receptor is a dynamic protein during alcohol modulation and channel gating.


Asunto(s)
Activación del Canal Iónico , Receptores de GABA-A , Regulación Alostérica , Animales , Etanol/farmacología , Hexanoles/farmacología , Activación del Canal Iónico/efectos de los fármacos , Mesilatos/farmacología , Mutación , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Ratas , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...