Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cell Mol Life Sci ; 78(23): 7899-7914, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34727194

RESUMEN

The lipophilic polycyclic aromatic hydrocarbon (PAH) phenanthrene is relatively abundant in polluted air and water and can access and accumulate in human tissue. Phenanthrene has been reported to interact with cardiac ion channels in several fish species. This study was undertaken to investigate the ability of phenanthrene to interact with hERG (human Ether-à-go-go-Related Gene) encoded Kv11.1 K+ channels, which play a central role in human ventricular repolarization. Pharmacological inhibition of hERG can be proarrhythmic. Whole-cell patch clamp recordings of hERG current (IhERG) were made from HEK293 cells expressing wild-type (WT) and mutant hERG channels. WT IhERG1a was inhibited by phenanthrene with an IC50 of 17.6 ± 1.7 µM, whilst IhERG1a/1b exhibited an IC50 of 1.8 ± 0.3 µM. WT IhERG block showed marked voltage and time dependence, indicative of dependence of inhibition on channel gating. The inhibitory effect of phenanthrene was markedly impaired by the attenuated inactivation N588K mutation. Remarkably, mutations of S6 domain aromatic amino acids (Y652, F656) in the canonical drug binding site did not impair the inhibitory action of phenanthrene; the Y652A mutation augmented IhERG block. In contrast, the F557L (S5) and M651A (S6) mutations impaired the ability of phenanthrene to inhibit IhERG, as did the S624A mutation below the selectivity filter region. Computational docking using a cryo-EM derived hERG structure supported the mutagenesis data. Thus, phenanthrene acts as an inhibitor of the hERG K+ channel by directly interacting with the channel, binding to a distinct site in the channel pore domain.


Asunto(s)
Canal de Potasio ERG1/antagonistas & inhibidores , Fenómenos Electrofisiológicos , Simulación del Acoplamiento Molecular , Mutación , Fenantrenos/farmacología , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida
2.
Biochem Biophys Res Commun ; 526(4): 1085-1091, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32321643

RESUMEN

The human Ether-à-go-go Related Gene (hERG) encodes a potassium channel responsible for the cardiac rapid delayed rectifier K+ current, IKr, which regulates ventricular repolarization. Loss-of-function hERG mutations underpin the LQT2 form of congenital long QT syndrome. This study was undertaken to elucidate the functional consequences of a variant of uncertain significance, T634S, located at a highly conserved position at the top of the S6 helix of the hERG channel. Whole-cell patch-clamp recordings were made at 37 °C of hERG current (IhERG) from HEK 293 cells expressing wild-type (WT) hERG, WT+T634S and hERG-T634S alone. When the T634S mutation was expressed alone little or no IhERG could be recorded. Co-expressing WT and hERG-T634S suppressed IhERG tails by ∼57% compared to WT alone, without significant alteration of voltage dependent activation of IhERG. A similar suppression of IhERG was observed under action potential voltage clamp. Comparable reduction of IKr in a ventricular AP model delayed repolarization and led to action potential prolongation. A LI-COR® based On/In-Cell Western assay showed that cell surface expression of hERG channels in HEK 293 cells was markedly reduced by the T634S mutation, whilst total cellular hERG expression was unaffected, demonstrating impaired trafficking of the hERG-T634S mutant. Incubation with E-4031, but not lumacaftor, rescued defective hERG-T634S channel trafficking and IhERG density. In conclusion, these data identify hERG-T634S as a rescuable trafficking defective mutation that reduces IKr sufficiently to delay repolarization and, thereby, potentially produce a LQT2 phenotype.


Asunto(s)
Secuencia Conservada , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Mutación con Pérdida de Función/genética , Serina/genética , Treonina/genética , Potenciales de Acción , Secuencia de Aminoácidos , Canal de Potasio ERG1/química , Humanos , Activación del Canal Iónico , Transporte de Proteínas
3.
J Mol Cell Cardiol ; 135: 22-30, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31362019

RESUMEN

Rett Syndrome (RTT) is an X-linked neurodevelopmental disorder associated with respiratory abnormalities and, in up to ~40% of patients, with prolongation of the cardiac QTc interval. QTc prolongation calls for cautious use of drugs with a propensity to inhibit hERG channels. The STARS trial has been undertaken to investigate the efficacy of sarizotan, a 5-HT1A receptor agonist, at correcting RTT respiratory abnormalities. The present study investigated whether sarizotan inhibits hERG potassium channels and prolongs ventricular repolarization. Whole-cell patch-clamp measurements were made at 37 °C from hERG-expressing HEK293 cells. Docking analysis was conducted using a recent cryo-EM structure of hERG. Sarizotan was a potent inhibitor of hERG current (IhERG; IC50 of 183 nM) and of native ventricular IKr from guinea-pig ventricular myocytes. 100 nM and 1 µM sarizotan prolonged ventricular action potential (AP) duration (APD90) by 14.1 ±â€¯3.3% (n = 6) and 29.8 ±â€¯3.1% (n = 5) respectively and promoted AP triangulation. High affinity IhERG inhibition by sarizotan was contingent upon channel gating and intact inactivation. Mutagenesis experiments and docking analysis implicated F557, S624 and Y652 residues in sarizotan binding, with weaker contribution from F656. In conclusion, sarizotan inhibits IKr/IhERG, accessing key binding residues on channel gating. This action and consequent ventricular AP prolongation occur at concentrations relevant to those proposed to treat breathing dysrhythmia in RTT. Sarizotan should only be used in RTT patients with careful evaluation of risk factors for QTc prolongation.


Asunto(s)
Miocitos Cardíacos/efectos de los fármacos , Síndrome de Rett/tratamiento farmacológico , Potenciales de Acción/efectos de los fármacos , Animales , Canales de Potasio Éter-A-Go-Go , Regulación de la Expresión Génica/efectos de los fármacos , Cobayas , Células HEK293 , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Masculino , Miocitos Cardíacos/metabolismo , Compuestos Orgánicos/farmacología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/patología , Síndrome de Rett/genética , Síndrome de Rett/patología , Factores de Riesgo , Regulador Transcripcional ERG/antagonistas & inhibidores , Regulador Transcripcional ERG/genética
4.
J Biol Chem ; 293(18): 7040-7057, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29545312

RESUMEN

Cardiac potassium channels encoded by human ether-à-go-go-related gene (hERG) are major targets for structurally diverse drugs associated with acquired long QT syndrome. This study characterized hERG channel inhibition by a minimally structured high-affinity hERG inhibitor, Cavalli-2, composed of three phenyl groups linked by polymethylene spacers around a central amino group, chosen to probe the spatial arrangement of side chain groups in the high-affinity drug-binding site of the hERG pore. hERG current (IhERG) recorded at physiological temperature from HEK293 cells was inhibited with an IC50 of 35.6 nm with time and voltage dependence characteristic of blockade contingent upon channel gating. Potency of Cavalli-2 action was markedly reduced for attenuated inactivation mutants located near (S620T; 54-fold) and remote from (N588K; 15-fold) the channel pore. The S6 Y652A and F656A mutations decreased inhibitory potency 17- and 75-fold, respectively, whereas T623A and S624A at the base of the selectivity filter also decreased potency (16- and 7-fold, respectively). The S5 helix F557L mutation decreased potency 10-fold, and both F557L and Y652A mutations eliminated voltage dependence of inhibition. Computational docking using the recent cryo-EM structure of an open channel hERG construct could only partially recapitulate experimental data, and the high dependence of Cavalli-2 block on Phe-656 is not readily explainable in that structure. A small clockwise rotation of the inner (S6) helix of the hERG pore from its configuration in the cryo-EM structure may be required to optimize Phe-656 side chain orientations compatible with high-affinity block.


Asunto(s)
Canal de Potasio ERG1/antagonistas & inhibidores , Canal de Potasio ERG1/química , Bloqueadores de los Canales de Potasio/farmacología , Alanina/química , Simulación por Computador , Microscopía por Crioelectrón , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Células HEK293 , Humanos , Concentración 50 Inhibidora , Activación del Canal Iónico/efectos de los fármacos , Simulación del Acoplamiento Molecular , Mutación , Técnicas de Placa-Clamp , Fenilalanina/química , Bloqueadores de los Canales de Potasio/administración & dosificación , Unión Proteica , Conformación Proteica
5.
J Mol Cell Cardiol ; 86: 42-53, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26159617

RESUMEN

The class Ic antiarrhythmic drug flecainide inhibits KCNH2-encoded "hERG" potassium channels at clinically relevant concentrations. The aim of this study was to elucidate the underlying molecular basis of this action. Patch clamp recordings of hERG current (IhERG) were made from hERG expressing cells at 37°C. Wild-type (WT) IhERG was inhibited with an IC50 of 1.49µM and this was not significantly altered by reversing the direction of K(+) flux or raising external [K(+)]. The use of charged and uncharged flecainide analogues showed that the charged form of the drug accesses the channel from the cell interior to produce block. Promotion of WT IhERG inactivation slowed recovery from inhibition, whilst the N588K and S631A attenuated-inactivation mutants exhibited IC50 values 4-5 fold that of WT IhERG. The use of pore-helix/selectivity filter (T623A, S624A V625A) and S6 helix (G648A, Y652A, F656A) mutations showed <10-fold shifts in IC50 for all but V625A and F656A, which respectively exhibited IC50s 27-fold and 142-fold their WT controls. Docking simulations using a MthK-based homology model suggested an allosteric effect of V625A, since in low energy conformations flecainide lay too low in the pore to interact directly with that residue. On the other hand, the molecule could readily form π-π stacking interactions with aromatic residues and particularly with F656. We conclude that flecainide accesses the hERG channel from the cell interior on channel gating, binding low in the inner cavity, with the S6 F656 residue acting as a principal binding determinant.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Flecainida/administración & dosificación , Torsades de Pointes/tratamiento farmacológico , Transactivadores/genética , Antiarrítmicos/administración & dosificación , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Mutación , Técnicas de Placa-Clamp , Conformación Proteica , Torsades de Pointes/genética , Torsades de Pointes/patología , Transactivadores/biosíntesis , Transactivadores/química , Regulador Transcripcional ERG
6.
Biophys J ; 107(10): L25-8, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25418316

RESUMEN

Relaxation of a hERG K(+) channel model during molecular-dynamics simulation in a hydrated POPC bilayer was accompanied by transitions of an arginine gating charge across a charge transfer center in two voltage sensor domains. Inspection of the passage of arginine side chains across the charge transfer center suggests that the unique hydration properties of the arginine guanidine cation facilitates charge transfer during voltage sensor responses to changes in membrane potential, and underlies the preference of Arg over Lys as a mobile charge carrier in voltage-sensitive ion channels.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Activación del Canal Iónico , Simulación de Dinámica Molecular , Secuencia de Aminoácidos , Transporte de Electrón , Humanos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Datos de Secuencia Molecular , Fosfatidilcolinas/metabolismo , Conformación Proteica
7.
J Mol Cell Cardiol ; 74: 220-30, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24877995

RESUMEN

The antianginal drug ranolazine, which combines inhibitory actions on rapid and sustained sodium currents with inhibition of the hERG/IKr potassium channel, shows promise as an antiarrhythmic agent. This study investigated the structural basis of hERG block by ranolazine, with lidocaine used as a low potency, structurally similar comparator. Recordings of hERG current (IhERG) were made from cell lines expressing wild-type (WT) or mutant hERG channels. Docking simulations were performed using homology models built on MthK and KvAP templates. In conventional voltage clamp, ranolazine inhibited IhERG with an IC50 of 8.03µM; peak IhERG during ventricular action potential clamp was inhibited ~62% at 10µM. The IC50 values for ranolazine inhibition of the S620T inactivation deficient and N588K attenuated inactivation mutants were respectively ~73-fold and ~15-fold that for WT IhERG. Mutations near the bottom of the selectivity filter (V625A, S624A, T623A) exhibited IC50s between ~8 and 19-fold that for WT IhERG, whilst the Y652A and F656A S6 mutations had IC50s ~22-fold and 53-fold WT controls. Low potency lidocaine was comparatively insensitive to both pore helix and S6 mutations, but was sensitive to direction of K(+) flux and particularly to loss of inactivation, with an IC50 for S620T-hERG ~49-fold that for WT IhERG. Docking simulations indicated that the larger size of ranolazine gives it potential for a greater range of interactions with hERG pore side chains compared to lidocaine, in particular enabling interaction of its two aromatic groups with side chains of both Y652 and F656. The N588K mutation is responsible for the SQT1 variant of short QT syndrome and our data suggest that ranolazine is unlikely to be effective against IKr/hERG in SQT1 patients.


Asunto(s)
Acetanilidas/química , Antiarrítmicos/química , Canales de Potasio Éter-A-Go-Go/química , Piperazinas/química , Potasio/metabolismo , Acetanilidas/farmacología , Potenciales de Acción/efectos de los fármacos , Antiarrítmicos/farmacología , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Expresión Génica , Células HEK293 , Humanos , Transporte Iónico , Lidocaína/química , Lidocaína/farmacología , Simulación del Acoplamiento Molecular , Mutación , Técnicas de Placa-Clamp , Piperazinas/farmacología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ranolazina , Relación Estructura-Actividad , Transgenes
8.
J Chem Inf Model ; 54(2): 601-12, 2014 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-24471705

RESUMEN

Many structurally and therapeutically diverse drugs interact with the human heart K+ channel hERG by binding within the K+ permeation pathway of the open channel, leading to drug-induced 'long QT syndrome'. Drug binding to hERG is often stabilized by inactivation gating. In the absence of a crystal structure, hERG pore homology models have been used to characterize drug interactions. Here we assess potentially inactivated states of the bacterial K+ channel, KcsA, as templates for inactivated state hERG pore models in the context of drug binding using computational docking. Although Flexidock and GOLD docking produced low energy score poses in the models tested, each method selected a MthK K+ channel-based model over models based on the putative inactivated state KcsA structures for each of the 9 drugs tested. The variety of docking poses found indicates that an optimal arrangement for drug binding of aromatic side chains in the hERG pore can be achieved in several different configurations. This plasticity of the drug "binding site" is likely to be a feature of the hERG inactivated state. The results demonstrate that experimental data on specific drug interactions can be used as structural constraints to assess and refine hERG homology models.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Simulación del Acoplamiento Molecular , Preparaciones Farmacéuticas/metabolismo , Secuencia de Aminoácidos , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Haloperidol/metabolismo , Haloperidol/farmacología , Humanos , Datos de Secuencia Molecular , Porosidad , Bloqueadores de los Canales de Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Conformación Proteica , Compuestos de Amonio Cuaternario/metabolismo , Homología de Secuencia de Aminoácido
9.
J Chem Inf Model ; 53(6): 1358-70, 2013 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-23672495

RESUMEN

The hERG K(+) channel is important for establishing normal electrical activity in the human heart. The channel's unique gating response to membrane potential changes indicates specific interactions between voltage sensor and pore domains that are poorly understood. In the absence of a crystal structure we constructed a homology model of the full hERG membrane domain and performed 0.5 µs molecular dynamics (MD) simulations in a hydrated membrane. The simulations identify potential interactions involving residues at the extracellular surface of S1 in the voltage sensor and at the N-terminal end of the pore helix in the hERG model. In addition, a diffuse interface involving hydrophobic residues on S4 (voltage sensor) and pore domain S5 of an adjacent subunit was stable during 0.5 µs of simulation. To assess the ability of the model to give insight into the effects of channel mutation we simulated a hERG mutant that contains a Leu to Pro substitution in the voltage sensor S4 helical segment (hERG L532P). Consistent with the retention of gated K(+) conductance, the L532P mutation was accommodated in the S4 helix with little disruption of helical structure. The mutation reduced the extent of interaction across the S4-S5 interface, suggesting a structural basis for the greatly enhanced deactivation rate in hERG L532P. The study indicates that pairwise comparison of wild-type and mutated channel models is a useful approach to interpreting functional data where uncertainty in model structures exist.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Alineación de Secuencia
10.
Pharmaceuticals (Basel) ; 16(9)2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37765012

RESUMEN

hERG (human Ether-à-go-go Related Gene)-encoded potassium channels underlie the cardiac rapid delayed rectifier (IKr) potassium current, which is a major target for antiarrhythmic agents and diverse non-cardiac drugs linked to the drug-induced form of long QT syndrome. E-4031 is a high potency hERG channel inhibitor from the methanesulphonanilide drug family. This study utilized a methanesulphonate-lacking E-4031 analogue, "E-4031-17", to evaluate the role of the methanesulphonamide group in E-4031 inhibition of hERG. Whole-cell patch-clamp measurements of the hERG current (IhERG) were made at physiological temperature from HEK 293 cells expressing wild-type (WT) and mutant hERG constructs. For E-4031, WT IhERG was inhibited by a half-maximal inhibitory concentration (IC50) of 15.8 nM, whilst the comparable value for E-4031-17 was 40.3 nM. Both compounds exhibited voltage- and time-dependent inhibition, but they differed in their response to successive applications of a long (10 s) depolarisation protocol, consistent with greater dissociation of E-4031-17 than the parent compound between applied commands. Voltage-dependent inactivation was left-ward voltage shifted for E-4031 but not for E-4031-17; however, inhibition by both compounds was strongly reduced by attenuated-inactivation mutations. Mutations of S6 and S5 aromatic residues (F656V, Y652A, F557L) greatly attenuated actions of both drugs. The S624A mutation also reduced IhERG inhibition by both molecules. Overall, these results demonstrate that the lack of a methanesulphonate in E-4031-17 is not an impediment to high potency inhibition of IhERG.

11.
J Mol Cell Cardiol ; 52(1): 185-95, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21989164

RESUMEN

The Class Ia antiarrhythmic drug disopyramide (DISO) causes QT interval prolongation that is potentially dangerous in acquired Long QT Syndrome but beneficial in short QT syndrome, through inhibition of the hERG-encoded channels responsible for rapid delayed rectifier K(+) current (I(Kr)). In this study, alanine mutants of hERG S6 and pore helix residues and MthK-based homology modelling and ligand docking were used to investigate molecular determinants of DISO binding to hERG. Whole-cell hERG current (I(hERG)) recordings were made at 37°C from HEK-293 cells expressing WT or mutant hERG channels. WT outward I(hERG) tails were inhibited with an IC(50) of 7.3µM, whilst inward I(hERG) tails in a high [K(+)](e) of 94mM were blocked with an IC(50) of 25.7µM. The IC(50) for the Y652A mutation was ~55-fold that of WT I(hERG); this mutation also abolished a leftward shift in voltage-dependent I(hERG) activation present for WT hERG. The IC(50) for F656A I(hERG) was ~51 fold its corresponding WT control. In contrast to previously studied methanesulphonanilide hERG inhibitors, neither the G648A S6 nor the T623A and S624A pore helical mutations modified DISO IC(50). Computational docking with the hERG model showed that DISO did not exhibit a single unique binding pose; instead several low energy binding poses at the lower end of the pore cavity favoured interactions with Y652 and F656. In the WT hERG model DISO did not interact directly with residues at the base of the pore helix, consistent with the minimal effect of mutation of these residues on drug block.


Asunto(s)
Antiarrítmicos/farmacología , Disopiramida/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Secuencia de Aminoácidos , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/química , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Conformación Proteica
12.
Biochim Biophys Acta ; 1808(10): 2477-87, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21777565

RESUMEN

hERG (human Ether-à-go-go Related Gene) is responsible for ion channels mediating rapid delayed rectifier potassium current, I(Kr), which is key to cardiac action potential repolarization. Gain-of-function hERG mutations give rise to the SQT1 variant of the Short QT Syndrome (SQTS). Reggae mutant zebrafish, with a S4 zERG mutation (Leucine499Proline; L499P), display arrhythmic features analogous to those seen in the SQTS. The affected S4 domain ERG residue is highly conserved. This study was executed to determine how the homologous hERG mutation (L532P) influences channel function at 37°C. Whole-cell measurements of current (I(hERG)) were made from HEK 293 cells expressing WT or L532P hERG. The half maximal activation voltage (V(0.5)) of L532P I(hERG) was positively shifted by ~+36mV compared to WT I(hERG); however at negative voltages a pronounced L532P I(hERG) was observed. Both activation and deactivation time-courses were accelerated for L532P I(hERG). The inactivation V(0.5) for L532P I(hERG) was shifted by ~+32mV. Under action potential (AP) voltage-clamp, L532P I(hERG) exhibited a dome-shaped current peaking at ~+16mV, compared to ~-31mV for WT-I(hERG). The L532P mutation produced an ~5-fold increase in the IC(50) for dronedarone inhibition of I(hERG). Homology modeling indicated that the L532 residue within the S4 helix lies closely apposed to the S5 region of an adjacent hERG subunit. Alterations to the S4 domain structure and, potentially, to interactions between adjacent hERG subunits are likely to account for the functional effects of this mutation.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/química , Calor , Mutación , Línea Celular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Humanos
13.
Front Pharmacol ; 13: 1010119, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36339618

RESUMEN

Loss of function (LOF) mutations of voltage sensitive K+ channel proteins hERG (Kv11.1) and KCNQ1 (Kv7.1) account for the majority of instances of congenital Long QT Syndrome (cLQTS) with the dominant molecular phenotype being a mistrafficking one resulting from protein misfolding. We explored the use of Evolutionary Coupling (EC) analysis, which identifies evolutionarily conserved pairwise amino acid interactions that may contribute to protein structural stability, to identify regions of the channels susceptible to misfolding mutations. Comparison with published experimental trafficking data for hERG and KCNQ1 showed that the method strongly predicts "scaffolding" regions of the channel membrane domains and has useful predictive power for trafficking phenotypes of individual variants. We identified a region in and around the cytoplasmic S2-S3 loop of the hERG Voltage Sensor Domain (VSD) as susceptible to destabilising mutation, and this was confirmed using a quantitative LI-COR ® based trafficking assay that showed severely attenuated trafficking in eight out of 10 natural hERG VSD variants selected using EC analysis. Our analysis highlights an equivalence in the scaffolding structures of the hERG and KCNQ1 membrane domains. Pathogenic variants of ion channels with an underlying mistrafficking phenotype are likely to be located within similar scaffolding structures that are identifiable by EC analysis.

14.
Biochim Biophys Acta ; 1798(6): 1254-62, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20346912

RESUMEN

RTA3 is an alpha-helical, amphipathic peptide with broad-spectrum activity against Gram-negative bacteria and low mammalian cell toxicity. RTA3 contains a cysteine residue, replacement of which with an alanine or serine (RTA3-C15S) virtually abolishes antimicrobial activity. Much of the activity of RTA3 can be recovered in RTA3-C15L, indicating that the C15 residue functions largely as a bulky hydrophobic side chain promoting target cell membrane interactions. The poorly active RTA3-C15S is a useful variant for assessing the mechanistic aspects of RTA3 activity. Binding and membrane perturbation in vesicles containing different proportions of negative surface charge are analyzed in terms of amino acid-specific free energy contributions to interfacial binding, which likely underlie variations in antimicrobial activity amongst RTA3 variants. Comparison with published free energy scales indicates that the reduced electrostatic contribution to binding to membranes having reduced negative surface charge can be compensated in RTA3 (but not RTA3-C15S) by a slightly deeper insertion of the C-terminus of the peptide to maximize hydrophobic contributions to binding. Analysis of inner membrane (IM)- and outer membrane (OM)-selective permeabilization of Escherichiacoli demonstrates a broad similarity between peptide effects on vesicles with low negative surface charge (20% negatively charged lipids), E.coli membrane perturbation, and antimicrobial activity, supporting a role for membrane perturbation in the killing mechanism of RTA3. The results demonstrate that large variations in antimicrobial activity on subtle changes in amino acid sequence in helical amphipathic peptides can be rationalized in terms of the thermodynamics of peptide binding to membranes, allowing a more systematic understanding of antimicrobial activity in these peptides.


Asunto(s)
Antiinfecciosos/química , Membrana Celular/química , Péptidos/química , Antiinfecciosos/farmacología , Membrana Celular/genética , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Escherichia coli/metabolismo , Péptidos/genética , Péptidos/farmacología , Unión Proteica , Pseudomonas aeruginosa/crecimiento & desarrollo , Pseudomonas aeruginosa/metabolismo , Electricidad Estática
15.
Proteins ; 79(7): 2224-32, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21574187

RESUMEN

Molecular dynamics simulations have been conducted of the helical polypeptide melittin, in concentrated aqueous solutions of the alpha and beta anomers of D-glucopyranose. Glucose is an osmolyte, and it is expected to be preferentially excluded from the surfaces of proteins. This was indeed found to be the case in the simulations. The results indicate that the observed exclusion may have a contribution from an under-representation of hydrogen bonding interactions between glucose groups and exposed side chains, compared to water. However, glucose was found to bind quite specifically to melittin by stacking its hydrophobic face, consisting of aliphatic protons, against the flat hydrophobic face of the indole group of the tryptophan-19 side chain. Although the binding site for this interaction is localized, the binding is weak for both anomers, with a binding free energy estimated as only ∼0.5 kcal/mol (i.e. near k(B)T). The face of the sugar stacked against the Trp indole ring is different for the two anomers of glucose, due to the disruption of the H1-H3-H5 hydrophobic triad of the beta anomer by the axial C1 hydroxyl group in the alpha anomer. The measurable affinity of the sugar for the Trp side chain is consistent with the very frequent occurrence of this group in the binding sites of proteins that complex with sugars.


Asunto(s)
Glucosa/química , Meliteno/química , Simulación de Dinámica Molecular , Sitios de Unión , Galactosa/química , Galactosa/metabolismo , Glucosa/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Meliteno/metabolismo , Termodinámica , Triptófano/química , Triptófano/metabolismo
16.
J Am Chem Soc ; 133(19): 7300-3, 2011 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-21520945

RESUMEN

The effects of chloride and sulfate salts of tetrapropylammonium (TPA(+)) and guanidinium (Gdm(+)) on the conformational stabilities of tryptophan zipper (trpzip) and α-helical (alahel) peptides were measured by circular dichroism spectroscopy. Like Gdm(+), TPA(+) interacts with the planar tryptophan indole group, perturbing the conformational stability of trpzip peptides. TPA(+) effects are largely unaffected by sulfate, indicating an absence of the heteroion pairing that is observed in concentrated Gdm(2)SO(4) solutions. TPA(+) stabilizes helical conformations in alahel peptides, indicating exclusion from the peptide bond. The observations are broadly consistent with predictions of molecular dynamics simulations [Mason, P. E.; et al. J. Phys. Chem. B2009, 113, 3227-3234], indicating that the effects of complex ions on proteins are increasingly predictable in terms of ion hydration, complementary interactions with specific protein groups, and ion-pairing contributions.


Asunto(s)
Cloruros/química , Complejos de Coordinación/química , Guanidina/química , Péptidos/química , Compuestos de Amonio Cuaternario/química , Sulfatos/química , Secuencia de Aminoácidos , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Pliegue de Proteína , Estabilidad Proteica
18.
J Electrocardiol ; 43(5): 440-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20493497

RESUMEN

UNLABELLED: The aim of this study was to determine the effects of desethyl-amiodarone (DEA), the major metabolite of the class III antiarrhythmic drug amiodarone, on human ether-à-go-go-related gene (hERG) encoded potassium channel current. MATERIALS AND METHODS: Whole-cell patch clamp recordings were made at 37 degrees C of ionic current (I(hERG)) carried by recombinant hERG channels expressed in HEK-293 cells. RESULTS: Desethyl-amiodarone inhibited I(hERG) with a half-maximal inhibitory concentration of approximately 158 nmol/L, compared with approximately 47 nmol/L for amiodarone. The inhibitory action of DEA on I(hERG) was contingent on channel gating, showing significant time and voltage dependence. Desethyl-amiodarone also produced an approximately -9 mV shift in the voltage dependence of activation of I(hERG); however, there was no significant preference for activated over inactivated channels. CONCLUSIONS: Because hERG underlies native cardiac "I(Kr)" channels, hERG/I(Kr) inhibition by DEA as well as amiodarone may contribute to the overall effects of amiodarone administration on cardiac repolarization.


Asunto(s)
Amiodarona/análogos & derivados , Antiarrítmicos/farmacología , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Amiodarona/farmacología , Análisis de Varianza , Humanos , Técnicas de Placa-Clamp
19.
Physiol Rep ; 8(20): e14568, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33091232

RESUMEN

The voltage-gated hERG (human-Ether-à-go-go Related Gene) K+ channel plays a fundamental role in cardiac action potential repolarization. Loss-of-function mutations or pharmacological inhibition of hERG leads to long QT syndrome, whilst gain-of-function mutations lead to short QT syndrome. A recent open channel cryo-EM structure of hERG represents a significant advance in the ability to interrogate hERG channel structure-function. In order to suppress protein aggregation, a truncated channel construct of hERG (hERGT ) was used to obtain this structure. In hERGT cytoplasmic domain residues 141 to 350 and 871 to 1,005 were removed from the full-length channel protein. There are limited data on the electrophysiological properties of hERGT channels. Therefore, this study was undertaken to determine how hERGT influences channel function at physiological temperature. Whole-cell measurements of hERG current (IhERG ) were made at 37°C from HEK 293 cells expressing wild-type (WT) or hERGT channels. With a standard +20 mV activating command protocol, neither end-pulse nor tail IhERG density significantly differed between WT and hERGT . However, the IhERG deactivation rate was significantly slower for hERGT . Half-maximal activation voltage (V0.5 ) was positively shifted for hERGT by ~+8 mV (p < .05 versus WT), without significant change to the activation relation slope factor. Neither the voltage dependence of inactivation, nor time course of development of inactivation significantly differed between WT and hERGT , but recovery of IhERG from inactivation was accelerated for hERGT (p < .05 versus WT). Steady-state "window" current was positively shifted for hERGT with a modest increase in the window current peak. Under action potential (AP) voltage clamp, hERGT IhERG showed modestly increased current throughout the AP plateau phase with a significant increase in current integral during the AP. The observed consequences for hERGT IhERG of deletion of the two cytoplasmic regions may reflect changes to electrostatic interactions influencing the voltage sensor domain.


Asunto(s)
Potenciales de Acción , Canal de Potasio ERG1/metabolismo , Microscopía por Crioelectrón , Canal de Potasio ERG1/química , Canal de Potasio ERG1/genética , Eliminación de Gen , Células HEK293 , Humanos , Activación del Canal Iónico , Dominios Proteicos
20.
Physiol Rep ; 8(5): e14385, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32147975

RESUMEN

The macrolide antibiotic erythromycin has been associated with QT interval prolongation and inhibition of the hERG-encoded channels responsible for the rapid delayed rectifier K+ current I(Kr ). It has been suggested that low concentrations of erythromycin may have a protective effect against hERG block and associated drug-induced arrhythmia by reducing the affinity of the pore-binding site for high potency hERG inhibitors. This study aimed to explore further the notion of a potentially protective effect of erythromycin. Whole-cell patch-clamp experiments were performed in which hERG-expressing mammalian (Human Embryonic Kidney; HEK) cells were preincubated with low to moderate concentrations of erythromycin (3 or 30 µM) prior to whole-cell patch clamp recordings of hERG current (IhERG ) at 37°C. In contrast to a previous report, exposure to low concentrations of erythromycin did not reduce pharmacological sensitivity of hERG to the antipsychotic thioridazine and antihistamine terfenadine. The IC50 value for IhERG tail inhibition by terfenadine was decreased by ~32-fold in the presence of 3 µM erythromycin (p < .05 vs. no preincubation). Sensitivity to thioridazine remained unchanged (p > .05 vs. no preincubation). The effects of low concentrations of erythromycin were investigated for a series of pore blocking drugs, and the results obtained were consistent with additive and/or synergistic effects. Experiments with the externally acting blocker BeKm-1 on WT hERG and a pore mutant (F656V) were used to explore the location of the binding site for erythromycin. Our data are inconsistent with the use of erythromycin for the management of drug-induced QT prolongation.


Asunto(s)
Antipsicóticos/administración & dosificación , Canal de Potasio ERG1/antagonistas & inhibidores , Eritromicina/administración & dosificación , Antagonistas de los Receptores Histamínicos H1 no Sedantes/administración & dosificación , Terfenadina/administración & dosificación , Tioridazina/administración & dosificación , Sitios de Unión/efectos de los fármacos , Canal de Potasio ERG1/fisiología , Eritromicina/química , Células HEK293 , Humanos , Concentración 50 Inhibidora , Macrólidos/administración & dosificación , Macrólidos/química , Técnicas de Placa-Clamp
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA