Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Physiol ; 15: 1386645, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903913

RESUMEN

Ion channels play a pivotal role in regulating cellular excitability and signal transduction processes. Among the various ion channels, G-protein-coupled inwardly rectifying potassium (GIRK) channels serve as key mediators of neurotransmission and cellular responses to extracellular signals. GIRK channels are members of the larger family of inwardly-rectifying potassium (Kir) channels. Typically, GIRK channels are activated via the direct binding of G-protein ßγ subunits upon the activation of G-protein-coupled receptors (GPCRs). GIRK channel activation requires the presence of the lipid signaling molecule, phosphatidylinositol 4,5-bisphosphate (PIP2). GIRK channels are also modulated by endogenous proteins and other molecules, including RGS proteins, cholesterol, and SNX27 as well as exogenous compounds, such as alcohol. In the last decade or so, several groups have developed novel drugs and small molecules, such as ML297, GAT1508 and GiGA1, that activate GIRK channels in a G-protein independent manner. Here, we aim to provide a comprehensive overview focusing on the direct modulation of GIRK channels by G-proteins, PIP2, cholesterol, and novel modulatory compounds. These studies offer valuable insights into the underlying molecular mechanisms of channel function, and have potential implications for both basic research and therapeutic development.

2.
Cell Rep ; 36(8): 109619, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433062

RESUMEN

G-protein-gated inwardly rectifying potassium (GIRK) channels are important for determining neuronal excitability. In addition to G proteins, GIRK channels are potentiated by membrane cholesterol, which is elevated in the brains of people with neurodegenerative diseases such as Alzheimer's dementia and Parkinson's disease. The structural mechanism of cholesterol modulation of GIRK channels is not well understood. In this study, we present cryo- electron microscopy (cryoEM) structures of GIRK2 in the presence and absence of the cholesterol analog cholesteryl hemisuccinate (CHS) and phosphatidylinositol 4,5-bisphosphate (PIP2). The structures reveal that CHS binds near PIP2 in lipid-facing hydrophobic pockets of the transmembrane domain. Our structural analysis suggests that CHS stabilizes PIP2 interaction with the channel and promotes engagement of the cytoplasmic domain onto the transmembrane region. Mutagenesis of one of the CHS binding pockets eliminates cholesterol-dependent potentiation of GIRK2. Elucidating the structural mechanisms underlying cholesterol modulation of GIRK2 channels could facilitate the development of therapeutics for treating neurological diseases. VIDEO ABSTRACT.


Asunto(s)
Colesterol/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Animales , Microscopía por Crioelectrón/métodos , Proteínas de Unión al GTP/metabolismo , Activación del Canal Iónico/fisiología , Ratones , Unión Proteica , Saccharomycetales
3.
Trends Pharmacol Sci ; 42(3): 203-215, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33468322

RESUMEN

G protein-gated inwardly rectifying potassium (GIRK) channels are essential regulators of cell excitability in the brain. While they are implicated in a variety of neurological diseases in both human and animal model studies, their therapeutic potential has been largely untapped. Here, we review recent advances in the development of small molecule compounds that specifically modulate GIRK channels and compare them with first-generation compounds that exhibit off-target activity. We describe the method of discovery of these small molecule modulators, their chemical features, and their effects in vivo. These studies provide a promising outlook on the future development of subunit-specific GIRK modulators to regulate neuronal excitability in a brain region-specific manner.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G , Neuronas , Animales , Encéfalo/metabolismo , Proteínas de Unión al GTP , Humanos , Neuronas/metabolismo
4.
Cell Rep ; 31(11): 107770, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553165

RESUMEN

G-protein-gated inwardly rectifying K+ (GIRK) channels are essential effectors of inhibitory neurotransmission in the brain. GIRK channels have been implicated in diseases with abnormal neuronal excitability, including epilepsy and addiction. GIRK channels are tetramers composed of either the same subunit (e.g., homotetramers) or different subunits (e.g., heterotetramers). Compounds that specifically target subsets of GIRK channels in vivo are lacking. Previous studies have shown that alcohol directly activates GIRK channels through a hydrophobic pocket located in the cytoplasmic domain of the channel. Here, we report the identification and functional characterization of a GIRK1-selective activator, termed GiGA1, that targets the alcohol pocket. GiGA1 activates GIRK1/GIRK2 both in vitro and in vivo and, in turn, mitigates the effects of a convulsant in an acute epilepsy mouse model. These results shed light on the structure-based development of subunit-specific GIRK modulators that could provide potential treatments for brain disorders.


Asunto(s)
Encéfalo/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Proteínas de Unión al GTP/metabolismo , Neuronas/metabolismo , Animales , Epilepsia/metabolismo , Activación del Canal Iónico/fisiología , Ratones Noqueados
5.
Proc Natl Acad Sci U S A ; 116(17): 8370-8379, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30971491

RESUMEN

Metabotropic GABAB receptors mediate a significant fraction of inhibitory neurotransmission in the brain. Native GABAB receptor complexes contain the principal subunits GABAB1 and GABAB2, which form an obligate heterodimer, and auxiliary subunits, known as potassium channel tetramerization domain-containing proteins (KCTDs). KCTDs interact with GABAB receptors and modify the kinetics of GABAB receptor signaling. Little is known about the molecular mechanism governing the direct association and functional coupling of GABAB receptors with these auxiliary proteins. Here, we describe the high-resolution structure of the KCTD16 oligomerization domain in complex with part of the GABAB2 receptor. A single GABAB2 C-terminal peptide is bound to the interior of an open pentamer formed by the oligomerization domain of five KCTD16 subunits. Mutation of specific amino acids identified in the structure of the GABAB2-KCTD16 interface disrupted both the biochemical association and functional modulation of GABAB receptors and G protein-activated inwardly rectifying K+ channel (GIRK) channels. These interfacial residues are conserved among KCTDs, suggesting a common mode of KCTD interaction with GABAB receptors. Defining the binding interface of GABAB receptor and KCTD reveals a potential regulatory site for modulating GABAB-receptor function in the brain.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteínas del Tejido Nervioso , Receptores de GABA-B , Sitios de Unión/genética , Cristalografía , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/genética , Receptores de GABA-B/química , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Transducción de Señal/genética
6.
Sci Rep ; 7(1): 4592, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28676630

RESUMEN

Activation of G protein-gated inwardly rectifying potassium (GIRK) channels leads to a hyperpolarization of the neuron's membrane potential, providing an important component of inhibition in the brain. In addition to the canonical G protein-activation pathway, GIRK channels are activated by small molecules but less is known about the underlying gating mechanisms. One drawback to previous studies has been the inability to control intrinsic and extrinsic factors. Here we used a reconstitution strategy with highly purified mammalian GIRK2 channels incorporated into liposomes and demonstrate that cholesterol or intoxicating concentrations of ethanol, i.e., >20 mM, each activate GIRK2 channels directly, in the absence of G proteins. Notably, both activators require the membrane phospholipid PIP2 but appear to interact independently with different regions of the channel. Elucidating the mechanisms underlying G protein-independent pathways of activating GIRK channels provides a unique strategy for developing new types of neuronal excitability modulators.


Asunto(s)
Colesterol/farmacología , Etanol/farmacología , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Activación Transcripcional , Animales , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/química , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Liposomas/metabolismo , Ratones , Fosfatidilinositol 4,5-Difosfato/metabolismo , Unión Proteica
7.
J Gen Physiol ; 149(8): 799-811, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28720589

RESUMEN

G protein-gated inwardly rectifying potassium (GIRK) channels control neuronal excitability in the brain and are implicated in several different neurological diseases. The anionic phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) is an essential cofactor for GIRK channel gating, but the precise mechanism by which PIP2 opens GIRK channels remains poorly understood. Previous structural studies have revealed several highly conserved, positively charged residues in the "tether helix" (C-linker) that interact with the negatively charged PIP2 However, these crystal structures of neuronal GIRK channels in complex with PIP2 provide only snapshots of PIP2's interaction with the channel and thus lack details about the gating transitions triggered by PIP2 binding. Here, our functional studies reveal that one of these conserved basic residues in GIRK2, Lys200 (6'K), supports a complex and dynamic interaction with PIP2 When Lys200 is mutated to an uncharged amino acid, it activates the channel by enhancing the interaction with PIP2 Atomistic molecular dynamic simulations of neuronal GIRK2 with the same 6' substitution reveal an open GIRK2 channel with PIP2 molecules adopting novel positions. This dynamic interaction with PIP2 may explain the intrinsic low open probability of GIRK channels and the mechanism underlying activation by G protein Gßγ subunits and ethanol.

8.
Int Rev Neurobiol ; 123: 117-60, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26422984

RESUMEN

G protein-gated inwardly rectifying potassium (GIRK; Kir3) channels, which are members of the large family of inwardly rectifying potassium channels (Kir1-Kir7), regulate excitability in the heart and brain. GIRK channels are activated following stimulation of G protein-coupled receptors that couple to the G(i/o) (pertussis toxin-sensitive) G proteins. GIRK channels, like all other Kir channels, possess an extrinsic mechanism of inward rectification involving intracellular Mg(2+) and polyamines that occlude the conduction pathway at membrane potentials positive to E(K). In the past 17 years, more than 20 high-resolution atomic structures containing GIRK channel cytoplasmic domains and transmembrane domains have been solved. These structures have provided valuable insights into the structural determinants of many of the properties common to all inward rectifiers, such as permeation and rectification, as well as revealing the structural bases for GIRK channel gating. In this chapter, we describe advances in our understanding of GIRK channel function based on recent high-resolution atomic structures of inwardly rectifying K(+) channels discussed in the context of classical structure-function experiments.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Potenciales de la Membrana/fisiología , Animales , Humanos , Potasio/metabolismo
9.
Nat Commun ; 3: 706, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22426227

RESUMEN

The cardiac voltage-gated sodium channel (Na(V)1.5) underlies impulse conduction in the heart, and its depolarization-induced inactivation is essential in control of the duration of the QT interval of the electrocardiogram. Perturbation of Na(V)1.5 inactivation by drugs or inherited mutation can underlie and trigger cardiac arrhythmias. The carboxy terminus has an important role in channel inactivation, but complete structural information on its predicted structural domain is unknown. Here we measure interactions between the functionally critical distal carboxy terminus α-helix (H6) and the proximal structured EF-hand motif using transition-metal ion fluorescence resonance energy transfer. We measure distances at three loci along H6 relative to an intrinsic tryptophan, demonstrating the proximal-distal interaction in a contiguous carboxy terminus polypeptide. Using these data together with the existing Na(V)1.5 carboxy terminus nuclear magnetic resonance structure, we construct a model of the predicted structured region of the carboxy terminus. An arrhythmia-associated H6 mutant that impairs inactivation decreases fluorescence resonance energy transfer, indicating destabilization of the distal-proximal intramolecular interaction. These data provide a structural correlation to the pathological phenotype of the mutant channel.


Asunto(s)
Sistema de Conducción Cardíaco/fisiología , Síndrome de QT Prolongado/genética , Canales de Sodio/química , Canales de Sodio/genética , Secuencias de Aminoácidos , Membrana Celular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Corazón/fisiología , Humanos , Activación del Canal Iónico , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Canales de Sodio/metabolismo
10.
Neurosci Lett ; 433(1): 65-70, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18242854

RESUMEN

Mutations in the sodium channel genes SCN1A and SCN2A have been identified in monogenic childhood epilepsies, but SCN3A has not previously been investigated as a candidate gene for epilepsy. We screened a consecutive cohort of 18 children with cryptogenic partial epilepsy that was classified as pharmacoresistant because of nonresponse to carbamazepine or oxcarbazepine, antiepileptic drugs that bind sodium channels. The novel coding variant SCN3A-K354Q was identified in one patient and was not present in 295 neurological normal controls. Twelve novel SNPs were also detected. K354Q substitutes glutamine for an evolutionarily conserved lysine residue in the pore domain of SCN3A. Functional analysis of this mutation in the backbone of the closely related gene SCN5A demonstrated an increase in persistent current that is similar in magnitude to epileptogenic mutations of SCN1A and SCN2A. This observation of a potentially pathogenic mutation of SCN3A (Nav1.3) indicates that this gene should be further evaluated for its contribution to childhood epilepsy.


Asunto(s)
Química Encefálica/genética , Encéfalo/metabolismo , Epilepsia Parcial Compleja/genética , Epilepsia Parcial Compleja/metabolismo , Mutación/genética , Canales de Sodio/genética , Factores de Edad , Sustitución de Aminoácidos/genética , Anticonvulsivantes/farmacología , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Línea Celular , Preescolar , Estudios de Cohortes , Secuencia Conservada/genética , Análisis Mutacional de ADN , Resistencia a Medicamentos/genética , Epilepsia Parcial Compleja/fisiopatología , Frecuencia de los Genes , Predisposición Genética a la Enfermedad/genética , Pruebas Genéticas , Humanos , Masculino , Canal de Sodio Activado por Voltaje NAV1.3 , Polimorfismo de Nucleótido Simple/genética , Estructura Terciaria de Proteína/genética , Canales de Sodio/química
11.
Channels (Austin) ; 1(4): 273-80, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18708744

RESUMEN

Inherited mutations of SCN5A, the gene that encodes Na(V)1.5, the alpha subunit of the principle voltage-gated Na(+) channel in the heart, cause congenital Long QT Syndrome variant 3 (LQT-3) by perturbation of channel inactivation. LQT-3 mutations induce small, but aberrant, inward current that prolongs the ventricular action potential and subjects mutation carriers to arrhythmia risk dictated in part by the biophysical consequences of the mutations. Most previously investigated LQT-3 mutations are associated with increased arrhythmia risk during rest or sleep. Here we report a novel LQT-3 mutation discovered in a pediatric proband diagnosed with LQTS but who experienced cardiac events during periods of mild exercise as well as rest. The mutation, which changes a single amino acid (S1904L) in the Na(V)1.5 carboxy terminal domain, disrupts the channel inactivation gate complex and promotes late Na(+) channel currents, not by promoting a bursting mode of gating, but by increasing the propensity of the channel to reopen during prolonged depolarization. Incorporating a modified version of the Markov model of the Na(V)1.5 channel into a mathematical model of the human ventricular action potential predicts that the biophysical consequences of the S1904L mutation result in action potential prolongation that is seen for all heart rates but, in contrast to other previously-investigated LQT-3 mutant channels, is most pronounced at fast rates resulting in a drastic reduction in the cells ability to adapt APD to heart rate.


Asunto(s)
Arritmias Cardíacas/genética , Sistema de Conducción Cardíaco/metabolismo , Activación del Canal Iónico/genética , Síndrome de QT Prolongado/genética , Proteínas Musculares/genética , Mutación , Canales de Sodio/genética , Sodio/metabolismo , Potenciales de Acción , Sustitución de Aminoácidos , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Niño , Electrocardiografía , Predisposición Genética a la Enfermedad , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca , Humanos , Cinética , Síndrome de QT Prolongado/complicaciones , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología , Masculino , Cadenas de Markov , Modelos Cardiovasculares , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5 , Linaje , Fenotipo , Conformación Proteica , Canales de Sodio/química , Canales de Sodio/metabolismo
12.
J Biol Chem ; 281(33): 24015-23, 2006 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-16798729

RESUMEN

Perturbation of sodium channel inactivation, a finely tuned process that critically regulates the flow of sodium ions into excitable cells, is a common functional consequence of inherited mutations associated with epilepsy, skeletal muscle disease, autism, and cardiac arrhythmias. Understanding the structural basis of inactivation is key to understanding these disorders. Here we identify a novel role for a structural motif in the COOH terminus of the heart NaV1.5 sodium channel in determining channel inactivation. Structural modeling predicts an interhelical hydrophobic interface between paired EF hands in the proximal region of the NaV1.5 COOH terminus. The predicted interface is conserved among almost all EF hand-containing proteins and is the locus of a number of disease-associated mutations. Using the structural model as a guide, we provide biochemical and biophysical evidence that the structural integrity of this interface is necessary for proper Na+ channel inactivation gating. We thus demonstrate a novel role of the sodium channel COOH terminus structure in the control of channel inactivation and in pathologies caused by inherited mutations that disrupt it.


Asunto(s)
Arritmias Cardíacas/genética , Proteínas Musculares/química , Proteínas Musculares/fisiología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/fisiología , Canales de Sodio/química , Canales de Sodio/fisiología , Acrilamida/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Arritmias Cardíacas/congénito , Arritmias Cardíacas/metabolismo , Línea Celular , Biología Computacional/métodos , Motivos EF Hand , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Activación del Canal Iónico/genética , Datos de Secuencia Molecular , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Canal de Sodio Activado por Voltaje NAV1.5 , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Estructura Secundaria de Proteína , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Canales de Sodio/genética , Espectrometría de Fluorescencia , Termodinámica , Triptófano/química
13.
Pflugers Arch ; 451(2): 349-61, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16133267

RESUMEN

The role of the outermost three charged residues of Domain IV/S4 in controlling gating of Ca(v)3.2 was investigated using single substitutions of each arginine with glutamine, cysteine, histidine, and lysine in a Flp-In-293 cell line, in which expression levels could be compared. Channel density, based on gating charge measurements, was ~125,000 channels/cell (10 fC/pF), except for R2Q and R3C, which expressed at lower levels. Channels substituted at Arg-1715 (R1C, R1Q, R1H) demonstrated such modest changes that a role in voltage sensing could not be determined. Arg-1718 (R2) made a contribution to activation voltage sensing, and the channel was sensitive to the geometry of side-chain substitutions at this position. Arg-1721 (R3) substitutions produced complex kinetic changes that together suggested that geometry made a larger contribution than charge. Current decay at positive potentials (O-->I) exponentially approached a constant value for all mutants except R2K channels, which were biphasically dependent on potential. R2K channels also displayed slowed deactivation with reduced voltage dependence despite near control values for conductance. Voltage-dependent accessibility of R to C mutants, evaluated with intracellularly and extracellularly applied methanthiosulfonate (MTS) reagents, showed that both R2 and R3 were exposed only when cells were depolarized, although it was not necessary for channels to open. Together, the data indicate that Domain IV/S4 is an activation domain and is not involved in inactivation from the open state.


Asunto(s)
Arginina/fisiología , Canales de Calcio Tipo T/fisiología , Activación del Canal Iónico/fisiología , Sustitución de Aminoácidos/genética , Arginina/genética , Sitios de Unión/genética , Canales de Calcio Tipo T/genética , Línea Celular , Metanosulfonato de Etilo/análogos & derivados , Metanosulfonato de Etilo/farmacología , Expresión Génica/genética , Humanos , Concentración de Iones de Hidrógeno , Activación del Canal Iónico/efectos de los fármacos , Mesilatos/farmacología , Técnicas de Placa-Clamp , Reactivos de Sulfhidrilo/farmacología , Transfección
14.
J Physiol ; 561(Pt 2): 403-13, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15579536

RESUMEN

The inner pore of the voltage-gated Na+ channel is predicted by the structure of bacterial potassium channels to be lined with the four S6 alpha-helical segments. Our previously published model of the closed pore based on the KcsA structure, and our new model of the open pore based on the MthK structure predict which residues in the mid-portion of S6 face the pore. We produced cysteine mutants of the mid-portion of domain IV-S6 (Ile-1575-Leu-1591) in NaV 1.4 and tested their accessibility to intracellularly and extracellularly placed positively charged methanethiosulfonate (MTS) reagents. We found that only two mutants, F1579C and V1583C, were accessible to both outside and inside 2-(aminoethyl)-methanethiosulfonate hydrobromide (MTSEA) Further study of those mutants showed that efficient closure of the fast inactivation gate prevented block by inside [2-(trimethylammonium)ethyl]methanethiosulfonate bromide (MTSET) at slow stimulation rates. When fast inactivation was inhibited by exposure to anthropleurin B (ApB), increasing channel open time, both mutants were blocked by inside MTSET at a rate that depended on the amount of time the channel was open. Consistent with the fast inactivation gate limiting access to the pore, in the absence of ApB, inside MTSET produced block when the cells were stimulated at 5 or 20 Hz. We therefore suggest that the middle of IV-S6 is an alpha-helix, and we propose a model of the open channel, based on MthK, in which Phe-1579 and Val-1583 face the pore.


Asunto(s)
Mesilatos/metabolismo , Modelos Moleculares , Proteínas Musculares/metabolismo , Fragmentos de Péptidos/metabolismo , Canales de Sodio/metabolismo , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Humanos , Mesilatos/química , Mesilatos/farmacología , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Estructura Secundaria de Proteína/genética , Estructura Terciaria de Proteína/genética , Ratas , Canales de Sodio/genética , Xenopus
15.
Mol Pharmacol ; 66(3): 648-57, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322257

RESUMEN

After opening, Na(+) channels may enter several kinetically distinct inactivated states. Whereas fast inactivation occurs by occlusion of the inner channel pore by the fast inactivation gate, the mechanistic basis of slower inactivated states is much less clear. We have recently suggested that the inner pore of the voltage-gated Na(+) channel may be involved in the process of ultra-slow inactivation (I(US)). The local anesthetic drug lidocaine is known to bind to the inner vestibule of the channel and to interact with slow inactivated states. We therefore sought to explore the effect of lidocaine binding on I(US). rNa(V) 1.4 channels carrying the mutation K1237E in the selectivity filter were driven into I(US) by long depolarizing pulses (-20 mV, 300 s). After repolarization to -120 mV, 53 +/- 5% of the channels recovered with a very slow time constant (tau(rec) = 171 +/- 19 s), typical for recovery from I(US). After exposure to 300 microM lidocaine, the fraction of channels recovering from I(US) was reduced to 13 +/- 4% (P < 0.01, n = 6). An additional mutation in the binding site of lidocaine (K1237E + F1579A) substantially reduced the effect of lidocaine on I(US), indicating that lidocaine has to bind to the inner vestibule of the channel to modulate I(US). We propose that I(US) involves a closure of the inner vestibule of the channel. Lidocaine may interfere with this pore motion by acting as a "foot in the door" in the inner vestibule.


Asunto(s)
Lidocaína/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Electrofisiología , Ácido Glutámico/genética , Lisina/genética , Mutagénesis Sitio-Dirigida , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Canales de Sodio/química , Canales de Sodio/genética , Canales de Sodio/fisiología , Xenopus laevis
16.
J Gen Physiol ; 123(2): 155-65, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14744988

RESUMEN

Electrical activity in nerve, skeletal muscle, and heart requires finely tuned activity of voltage-gated Na+ channels that open and then enter a nonconducting inactivated state upon depolarization. Inactivation occurs when the gate, the cytoplasmic loop linking domains III and IV of the alpha subunit, occludes the open pore. Subtle destabilization of inactivation by mutation is causally associated with diverse human disease. Here we show for the first time that the inactivation gate is a molecular complex consisting of the III-IV loop and the COOH terminus (C-T), which is necessary to stabilize the closed gate and minimize channel reopening. When this interaction is disrupted by mutation, inactivation is destabilized allowing a small, but important, fraction of channels to reopen, conduct inward current, and delay cellular repolarization. Thus, our results demonstrate for the first time that physiologically crucial stabilization of inactivation of the Na+ channel requires complex interactions of intracellular structures and indicate a novel structural role of the C-T domain in this process.


Asunto(s)
Activación del Canal Iónico/fisiología , Fragmentos de Péptidos/fisiología , Canales de Sodio/fisiología , Secuencia de Aminoácidos , Línea Celular , Humanos , Activación del Canal Iónico/genética , Sustancias Macromoleculares , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/genética , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes de Fusión/síntesis química , Proteínas Recombinantes de Fusión/fisiología , Canales de Sodio/síntesis química , Canales de Sodio/genética
17.
Prog Cardiovasc Dis ; 46(3): 259-70, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14685943

RESUMEN

A precise balance of ionic currents underlies normal cardiac excitation and relaxation. Disruption of this equilibrium by genetic defects, polymorphisms, therapeutic intervention, and structural abnormalities can cause arrhythmogenic phenotypes leading to syncope, seizures, and sudden cardiac death. Congenital defects result in an unpredictable expression of phenotypes with variable penetrance, even within single families. Additionally, phenotypically opposite and overlapping cardiac arrhythmogenic syndromes can even stem from the same mutation. Accordingly, the relationship between genetic mutations and clinical syndromes is becoming increasingly complex.


Asunto(s)
Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/genética , Humanos , Fenotipo , Polimorfismo Genético , Sodio/metabolismo
18.
Neurosci Lett ; 337(1): 41-5, 2003 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-12524167

RESUMEN

Local anesthetics (LAs) block Na(+) channels with a higher affinity for the fast or slow inactivated state of the channel. Their binding to the channel may stabilize fast inactivation or induce slow inactivation. We examined the role of the LA binding sites on domain IV, S6 (IVS6) of Na(+) channels in fast and slow inactivation by studying the gating properties of the mutants on IVS6 affecting LA binding. Mutation of the putative LA binding site, F1579C, inhibited fast and slow inactivation. Mutations of another putative LA binding site, Y1586C, and IVS6 residue involved in LA access and binding, I1575C, both enhanced fast and slow inactivation. None of the mutations affected channel activation. These results suggest that the LA binding site on IVS6 is involved in slow inactivation as well as fast inactivation, and these two gatings are coupled at the binding site.


Asunto(s)
Anestésicos Locales/metabolismo , Canales de Sodio/fisiología , Animales , Sitios de Unión , Línea Celular , Humanos , Activación del Canal Iónico , Mutación , Técnicas de Placa-Clamp , Estructura Terciaria de Proteína , Subunidades de Proteína , Ratas , Canales de Sodio/genética , Canales de Sodio/metabolismo , Transfección
19.
J Biol Chem ; 277(40): 37105-15, 2002 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-12138168

RESUMEN

Recently, we reported that mutation A1529D in the domain (D) IV P-loop of the rat skeletal muscle Na(+) channel mu(1) (DIV-A1529D) enhanced entry to an inactivated state from which the channels recovered with an abnormally slow time constant on the order of approximately 100 s. Transition to this "ultra-slow" inactivated state (USI) was substantially reduced by binding to the outer pore of a mutant mu-conotoxin GIIIA. This indicated that USI reflected a structural rearrangement of the outer channel vestibule and that binding to the pore of a peptide could stabilize the pore structure (Hilber, K., Sandtner, W., Kudlacek, O., Glaaser, I. W., Weisz, E., Kyle, J. W., French, R. J., Fozzard, H. A., Dudley, S. C., and Todt, H. (2001) J. Biol. Chem. 276, 27831-27839). Here, we tested the hypothesis that occlusion of the inner vestibule of the Na(+) channel by the fast inactivation gate inhibits ultra-slow inactivation. Stabilization of the fast inactivated state (FI) by coexpression of the rat brain beta(1) subunit in Xenopus oocytes significantly prolonged the time course of entry to the USI. A reduction in USI was also observed when the FI was stabilized in the absence of the beta(1) subunit, suggesting a causal relation between the occurrence of the FI and inhibition of USI. This finding was further confirmed in experiments where the FI was destabilized by introducing the mutations I1303Q/F1304Q/M1305Q. In DIV-A1529D + I1303Q/F1304Q/M1305Q channels, occurrence of USI was enhanced at strongly depolarized potentials and could not be prevented by coexpression of the beta(1) subunit. These results strongly suggest that FI inhibits USI in DIV-A1529D channels. Binding to the inner pore of the fast inactivation gate may stabilize the channel structure and thereby prevent USI. Some of the data have been published previously in abstract form (Hilber, K., Sandtner, W., Kudlacek, O., Singer, E., and Todt, H. (2002) Soc. Neurosci. Abstr. 27, program number 46.12).


Asunto(s)
Activación del Canal Iónico/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Sustitución de Aminoácidos , Animales , Encéfalo/fisiología , Electrofisiología , Activación del Canal Iónico/efectos de los fármacos , Cinética , Potenciales de la Membrana/fisiología , Modelos Moleculares , Músculo Esquelético/fisiología , Mutagénesis Sitio-Dirigida , Mutación Puntual , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/genética , Conformación Proteica , Subunidades de Proteína , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...