Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 163(3): 662-74, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21306584

RESUMEN

BACKGROUND AND PURPOSE: Voltage-gated K(v)1.3 channels appear on T-lymphocytes and are characterized by their typical C-type inactivation. In order to develop drugs stabilizing the C-type inactivated state and thus potentially useful in treatment of autoimmune diseases, it is important to know more about the three-dimensional structure of this inactivated state of the channel. EXPERIMENTAL APPROACH: The patch-clamp technique was used to study effects of methanethiosulphonate (MTS) compounds on currents through wild-type human K(v)1.3 (hK(v)1.3) and two mutant channels, hK(v)1.3 V417C and hK(v) 1.3 H399T-V417C, in the closed, open and inactivated states. KEY RESULTS: Extracellular application of 2-aminoethyl methanethiosulphonate (MTSEA) irreversibly reduced currents through hK(v) 1.3 V417C channels in the open and inactivated, but not in the closed state, indicating that a modification was possible. Co-application of verapamil prevented this reduction. Intracellular application of MTSEA and [2-(trimethylammonium)ethyl] methanethiosulphonate (MTSET) also modified the mutant channels, whereas extra- and intracellular application of sodium (2-sulfonatoethyl)methanethiosulphonate (MTSES) and intracellular application of MTSET did not. CONCLUSIONS AND IMPLICATIONS: Our experiments showed that the binding site for MTS compounds was intracellular in the mutant channels and that the V417C mutant channels were modified in the open and the inactivated states, and this modification was prevented by verapamil. Therefore, the activation gate on the intracellular side of the selectivity filter must be open during inactivation. Furthermore, although the S6 segment is moving further apart during inactivation, this change does not include a movement of the side chain of the amino acid at position 417, away from lining the channel pore.


Asunto(s)
Metanosulfonato de Etilo/análogos & derivados , Canal de Potasio Kv1.3/fisiología , Mesilatos/farmacología , Verapamilo/farmacología , Animales , Células COS , Chlorocebus aethiops , Metanosulfonato de Etilo/farmacología , Humanos , Activación del Canal Iónico , Canal de Potasio Kv1.3/genética , Mutación , Técnicas de Placa-Clamp
2.
Br J Pharmacol ; 157(5): 757-68, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19371328

RESUMEN

BACKGROUND AND PURPOSE: Verapamil blocks current through the voltage-gated K(+) channel K(v)1.3 in the open and inactivated state of the channel but not the closed state. The binding site for verapamil was proposed to be close to the selectivity filter and the occupancy of the selectivity filter might therefore influence verapamil affinity. EXPERIMENTAL APPROACH: We investigated the influence of intra- and extracellular K(+) and Rb(+) on the effect of verapamil by patch-clamp studies, in COS-7 cells transfected with hK(v)1.3 channels. KEY RESULTS: Verapamil affinity was highest in high intracellular K(+) concentrations ([K(+)](i)) and lowest in low [Rb(+)](i), indicating an influence of intracellular cations on verapamil affinity. Experiments with a mutant channel (H399T), exhibiting a strongly reduced C-type inactivated state, demonstrated that part of this changed verapamil affinity in wild-type channels could be caused by altered C-type inactivation. External K(+) and Rb(+) could influence verapamil affinity by a voltage-dependent entry into the channel thereby modifying the verapamil off-rate and in addition causing a voltage-dependent verapamil off-rate. CONCLUSIONS AND IMPLICATIONS: Recovery from verapamil block was mainly due to the voltage-dependent closing of channels (state-dependent block), implying a second open state of the channel. This hypothesis was confirmed by the dependency of the tail current time course on duration of the prepulse. We conclude that the wild-type hK(v)1.3 channel undergoes at least two different conformational changes before finally closing with a low verapamil affinity in one open state and a high verapamil affinity in the other open state.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio Kv1.3/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Potasio/metabolismo , Rubidio/metabolismo , Verapamilo/farmacología , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Humanos , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Potenciales de la Membrana , Modelos Biológicos , Mutación , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/metabolismo , Conformación Proteica , Factores de Tiempo , Transfección , Verapamilo/metabolismo
3.
FEBS Lett ; 488(1-2): 45-50, 2001 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-11163793

RESUMEN

Using the whole-cell recording mode of the patch-clamp technique we studied the effects of removal of extracellular potassium, [K(+)](o), on a mammalian Shaker-related K(+) channel, hKv1.5. In the absence of [K(+)](o), current through hKv1.5 was similar to currents obtained in the presence of 4.5 mM [K(+)](o). This observation was not expected as earlier results had suggested that either positively charged residues or the presence of a nitrogen-containing residue at the external TEA(+) binding site (R487 in hKv1.5) caused current loss upon removal of [K(+)](o). However, the current loss in hKv1.5 was observed when the extracellular pH, pH(o), was reduced from 7.4 to 6.0, a behavior similar to that observed previously for current through mKv1.3 with a histidine at the equivalent position (H404). These observations suggested that the charge at R487 in hKv1.5 channels was influenced by other amino acids in the vicinity. Replacement of a histidine at position 463 in hKv1.5 by glycine confirmed this hypothesis making this H463G mutant channel sensitive to removal of [K(+)](o) even at pH(o) 7.4. We conclude that the protonation of H463 at pH 7.4 might induce a pK(a) shift of R487 that influences the effective charge at this position leading to a not fully protonated arginine. Furthermore, we assume that the charge at position 487 in hKv1.5 can directly or indirectly disturb the occupation of a K(+) binding site within the channel pore possibly by electrostatic interaction. This in turn might interfere with the concerted transition of K(+) ions resulting in a loss of K(+) conduction.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Potasio/metabolismo , Potasio/farmacología , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Sitios de Unión , Humanos , Concentración de Iones de Hidrógeno , Canal de Potasio Kv1.5 , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Técnicas de Placa-Clamp , Canales de Potasio/química , Canales de Potasio/genética , Unión Proteica , Conformación Proteica , Ratas , Alineación de Secuencia , Electricidad Estática , Células Tumorales Cultivadas
4.
J Membr Biol ; 178(1): 11-20, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11058683

RESUMEN

The calcium indicator fura-2 was used to study the effect of hypotonic solutions on the intracellular calcium concentration, [Ca(2+)](i), in a human osteoblast-like cell line. Decreasing the tonicity of the extracellular solution to 50% leads to an increase in [Ca(2+)](i) from approximately 150 nm up to 1.3 microm. This increase in [Ca(2+)](i) was mainly due to an influx of extracellular Ca(2+) since removing of extracellular Ca(2+) reduced this increase to approximately 250 nm. After cell swelling most of the cells were able to regulate their volume to the initial level within 800 sec. The whole-cell recording mode of the patch-clamp technique was also used to study the effect of an increase in [Ca(2+)](i) on membrane currents in these cells. An increase in [Ca(2+)](i) revealed two types of Ca(2+)-activated K(+) channels, K(Ca) channels. Current through both channel types could not be observed below voltage of +80 mV with [Ca(2+)](i) buffered to 100 nm or less. With patch-electrodes filled with solutions buffering [Ca(2+)](i) to 10 microm both channels types could be readily observed. The activation of the first type was apparently voltage-independent since current could be observed over the entire voltage range used from -160 to +100 mV. In addition, the current was also blocked by charybdotoxin (CTX). The second type of K(Ca) channels in these cells could be activated with depolarizations more positive than -40 mV from a holding potential of -80 mV. This type was blocked by CTX and paxilline. Adding paxilline to the extracellular solution inhibited regulatory volume decrease (RVD), but could not abolish RVD. We conclude that two K(Ca) channel types exist in human osteoblasts, an intermediate conductance K(Ca) channel and a MaxiK-like K(Ca) channel. MaxiK channels might get activated either directly or by an increase in [Ca(2+)](i) elicited through hypotonic solutions. In combination with the volume-regulated Cl(-) conductance in the same cells this K(+) channel seems to play a vital role in volume regulation in human osteoblasts.


Asunto(s)
Osteoblastos/metabolismo , Canales de Potasio Calcio-Activados , Canales de Potasio/fisiología , Calcio/metabolismo , Línea Celular , Caribdotoxina/farmacología , Electrofisiología , Humanos , Soluciones Hipotónicas , Indoles/farmacología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio , Canales de Potasio de Gran Conductancia Activados por el Calcio , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Bloqueadores de los Canales de Potasio
5.
Br J Pharmacol ; 131(7): 1275-84, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11090098

RESUMEN

1. Phenylalkylamines (PAA) usually known for their action on L-type Ca(2+) channels potently block the C-type inactivating lymphocyte Kv1.3 channel resulting in inhibition of activation of T lymphocytes. In order to design PAAs blocking Kv1.3 specifically over L-type Ca(2+) channels, we investigated the state-dependent manner of mKv1. 3 block by the PAA verapamil. 2. Verapamil seems to have access to the open state (OB) and, once bound to the channel, the channel-verapamil complex is absorbed into a slowly recovering state. This state was proposed to be the inactivated blocked state (IB). Here we present a quantitative description of the transition into this state and provide evidence for the IB state through experiments with an inactivation lacking mutant channel. Since the inactivated state cannot be reached in this case the IB state cannot be reached either. 3. We show that the transition OB-->IB is accelerated by verapamil most likely through a mechanism involving the reduction of [K(+)] at an inactivation modulating low affinity binding site for K(+) at the outer vestibule. 4. Measurements of the voltage-dependence of the off-rate constants for verapamil suggest that verapamil can reach the channel in its neutral form and might get partially protonated while bound. Thus only those verapamil molecules that are protonated can more easily dissociate at hyperpolarizing voltages. 5. Since open block kinetics were shown to be similar for wild type mKv1.3 and the H404T mutant mKv1.3 channel, and since the block of the H404T mutant channels by verapamil could be described exactly by a simple three-state open block model, the mutant channel could serve as a screening channel to determine open block affinities of new PAA derivatives in high through-put experiments.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Potasio , Canales de Potasio con Entrada de Voltaje , Verapamilo/farmacología , Sustitución de Aminoácidos , Animales , Relación Dosis-Respuesta a Droga , Cinética , Canal de Potasio Kv1.3 , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Mutación Puntual , Canales de Potasio/genética , Canales de Potasio/fisiología , Células Tumorales Cultivadas
6.
J Biol Chem ; 275(50): 39345-53, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10962004

RESUMEN

Using a peptide toxin, kaliotoxin (KTX), we gained new insight into the topology of the pore region of a voltage-gated potassium channel, mKv1.1. In order to find new interactions between mKv1.1 and KTX, we investigated the pH dependence of KTX block which was stronger at pH(o) 6.2 compared with pH(o) 7.4. Using site-directed mutagenesis on the channel and the toxin, we found that protonation of His(34) in KTX caused the pH(o) dependence of KTX block. Glu(350) and Glu(353) in mKv1.1, which interact with His(34) in KTX, were calculated to be 4 and 7 A away from His(34)/KTX, respectively. Docking of KTX into a homology model of mKv1.1 based on the KcsA crystal structure using this and other known interactions as constraints showed structural differences between mKv1.1 and KcsA within the turret (amino acids 348-357). To satisfy our data, we would have to modify the KcsA crystal structure for the mKv1.1 channel orienting Glu(350) 7 A and Glu(353) 4 A more toward the center of the pore compared with KcsA. This would place Glu(350) 15 A and Glu(353) 11 A away from the center of the pore instead of the distances for the equivalent KcsA residues with 22 A for Gly(53) and 15 A for Gly(56), respectively. Bacterial and mammalian potassium channels may have structural differences regarding the turret of the outer pore vestibule. This topological difference between both channel types may have substantial influence on structure-guided development of new drugs for mammalian potassium channels by rational drug design.


Asunto(s)
Bacterias/química , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/química , Secuencia de Aminoácidos , Animales , Relación Dosis-Respuesta a Droga , Electrofisiología , Ácido Glutámico/química , Concentración de Iones de Hidrógeno , Cinética , Canal de Potasio Kv.1.1 , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Plásmidos/metabolismo , Canales de Potasio/metabolismo , Unión Proteica , Ratas , Proteínas Recombinantes/química , Venenos de Escorpión/química , Venenos de Escorpión/metabolismo , Homología de Secuencia de Aminoácido , Termodinámica , Células Tumorales Cultivadas
7.
Proc Natl Acad Sci U S A ; 97(14): 8151-6, 2000 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-10884437

RESUMEN

The antimycotic clotrimazole, a potent inhibitor of the intermediate-conductance calcium-activated K(+) channel, IKCa1, is in clinical trials for the treatment of sickle cell disease and diarrhea and is effective in ameliorating the symptoms of rheumatoid arthritis. However, inhibition of cytochrome P450 enzymes by clotrimazole limits its therapeutic value. We have used a rational design strategy to develop a clotrimazole analog that selectively inhibits IKCa1 without blocking cytochrome P450 enzymes. A screen of 83 triarylmethanes revealed the pharmacophore for channel block to be different from that required for cytochrome P450 inhibition. The "IKCa1-pharmacophore" consists of a (2-halogenophenyl)diphenylmethane moiety substituted by an unsubstituted polar pi-electron-rich heterocycle (pyrazole or tetrazole) or a -C≡N group, whereas cytochrome P450 inhibition absolutely requires the imidazole ring. A series of pyrazoles, acetonitriles, and tetrazoles were synthesized and found to selectively block IKCa1. TRAM-34 (1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole) inhibits the cloned and the native IKCa1 channel in human T lymphocytes with a K(d) of 20-25 nM and is 200- to 1,500-fold selective over other ion channels. Using TRAM-34, we show that blocking IKCa1 in human lymphocytes, in the absence of P450-inhibition, results in suppression of mitogen-stimulated [(3)H]thymidine incorporation of preactivated lymphocytes with EC(50)-values of 100 nM-1 microM depending on the donor. Combinations of TRAM-34 and cyclosporin A are more effective in suppressing lymphocyte mitogenesis than either compound alone. Our studies suggest that TRAM-34 and related compounds may hold therapeutic promise as immunosuppressants.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Inmunosupresores/farmacología , Canales de Potasio , Pirazoles/farmacología , Clotrimazol/química , Ciclosporina/farmacología , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Diseño de Fármacos , Interacciones Farmacológicas , Conductividad Eléctrica , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio , Activación del Canal Iónico , Ionomicina/farmacología , Activación de Linfocitos/efectos de los fármacos , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología
8.
FEBS Lett ; 469(2-3): 196-202, 2000 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-10713270

RESUMEN

T cells express two different types of voltage-independent Ca(2+)-activated K(+) channels with small (SK) and intermediate (IK) conductance that serve important roles in the activation of T lymphocytes. In contrast to the IK channels from T lymphocytes which are upregulated upon mitogen stimulation, SK channels of Jurkat T cells, a human leukemic T cell line, are constitutively expressed even in the absence of mitogenic stimulation. We have used patch-clamp recordings from transfected or injected mammalian cells to show that the cloned SK2 channel demonstrates the biophysical and pharmacological properties of the majority of K(Ca) channels in Jurkat T cells. The cloned and native channels are voltage-independent, Ca(2+)-activated, apamin-sensitive, show an equivalent voltage-dependent Ba(2+) block and possess a similar ion selectivity. In addition, we used the polymerase chain reaction to demonstrate the presence of SK2 mRNA in Jurkat T cells, whereas SK3 transcripts encoding the other cloned apamin-sensitive SK channel were not detected. These data suggest that the voltage-independent apamin-sensitive K(Ca) channel in Jurkat T cells represents the recently cloned SK2 channel.


Asunto(s)
Apamina/farmacología , Calcio/metabolismo , Canales de Potasio Calcio-Activados , Canales de Potasio/metabolismo , Línea Celular , Humanos , Células Jurkat , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio , Canales de Potasio/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio de Pequeña Conductancia Activados por el Calcio , Transfección
9.
J Membr Biol ; 173(1): 57-66, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10612692

RESUMEN

Using the patch-clamp technique, we have characterized membrane currents in single detrusor smooth muscle cells from rat and human urinary bladder. From the voltage- and Ca(2+)-dependence of the current as well as the single channel conductance we conclude that rat and human urinary bladder smooth muscle cells express MaxiK channels. In smooth muscle cells from rat urinary bladder we tested the action of NS1608 on current through these MaxiK channels. Application of 10 microm NS1608 increased the amplitude of the current and this increase could be explained by a shift in the activation voltage of the MaxiK channels approximately 100 mV towards more negative potentials. Charybdotoxin as well as paxilline, well known blockers of MaxiK channels, were able to reduce current through MaxiK channels in our cell preparation. In addition, application of 10 microm NS1608 hyperpolarized the membrane potential of the investigated cells. This hyperpolarization could be antagonized by the application of paxilline. We conclude that application of NS1608 results in the opening of MaxiK channels under physiological conditions that leads to a hyperpolarization of the cells. This hyperpolarization in turn could relax urinary bladder smooth muscle cells. MaxiK channels in these cells could therefore play a role in directly controlling muscle tone by regulating the membrane potential. This opens up the possibility of MaxiK channels being targets for the treatment of urge incontinence.


Asunto(s)
Músculo Liso/fisiología , Compuestos de Fenilurea/farmacología , Canales de Potasio Calcio-Activados , Canales de Potasio/fisiología , Vejiga Urinaria/fisiología , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Células Cultivadas , Caribdotoxina/farmacología , Proteínas del Citoesqueleto/farmacología , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio , Canales de Potasio de Gran Conductancia Activados por el Calcio , Potenciales de la Membrana/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Técnicas de Placa-Clamp , Paxillin , Fosfoproteínas/farmacología , Canales de Potasio/efectos de los fármacos , Ratas
10.
Br J Pharmacol ; 127(5): 1065-74, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10455250

RESUMEN

We investigated the action of the phenylalkylamines verapamil and N-methyl-verapamil on the Kv1.3 potassium channel using the whole-cell configuration of the patch-clamp technique. Our goal was to identify their binding as a prerequisite for using the phenylalkylamines as small, well-defined molecular probes, not only to expand the structural findings made with peptide toxins or by crystallization, but also to use them as lead compounds for the generation of more potent and therefore more specific K+ channel modulators. Competition experiments with charybdotoxin, known to interact with external residues of Kv1.3, showed no interaction with verapamil. The internal application of quarternary N-methyl-verapamil in combination with verapamil suggested competition for the same internal binding site. Verapamil affinity was decreased 6 fold by a mutation (M395V) in a region of the internal pore which forms part of the internal tetraethylammonium (TEA+) binding site, although mutations at neighbouring residues (T396 and T397) were without effect. Modification of C-type inactivation by mutations in the internal pore suggest that this region participates in the inactivation process. The action of phenylalkylamines and local anaesthetics on L-type Ca2+ channels and Na channels, respectively, and verapamil on Kv1.3 indicate very similar blocking mechanisms. This might allow the use of these compounds as molecular probes to map the internal vestibule of all three channel types.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio/efectos de los fármacos , Verapamilo/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Canales de Calcio/efectos de los fármacos , Canales de Calcio Tipo L , Cristalización , Canal de Potasio Kv1.3 , Modelos Estructurales , Datos de Secuencia Molecular , Mutación , Canales de Potasio/química , Ratas , Canales de Sodio/efectos de los fármacos , Relación Estructura-Actividad , Compuestos de Tetraetilamonio/farmacología , Células Tumorales Cultivadas , Verapamilo/metabolismo
11.
Eur J Neurosci ; 11(7): 2403-12, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10383630

RESUMEN

Episodic ataxia type 1 (EA-1) is a rare neurological disorder and was the first ionic channel disease to be associated with defects in a potassium channel. Until now 10 different point mutations in the KCNA1-gene have been reported to cause this disorder. We have investigated the functional consequences of two mutations leading to amino acid substitutions in the first and sixth transmembrane segments of a Kv1.1 channel subunit, by means of the patch-clamp technique; we injected cRNA coding for, respectively, F184C and V408A mutant Kv1.1 channels into mammalian cells and compared the resulting currents with those in the wild-type. The expression levels of F184C and V408A mutant channels relative to that of the wild-type was 38 and 68%, respectively. Since the single-channel conductance of the F184C mutant was similar to that of the wild-type (12 pS) without an apparent change in the maximum open probability, we conclude that the lower expression level in the F184C mutant channels is due to a reduced number of functional channels on the cell surface. F184C activated slower, and at more depolarized potentials, and deactivated faster compared with the wild-type. V408A channels deactivated and inactivated faster compared with the wild-type. Studies with different extracellular cations and tetraethylammonium gave no indication that the pore structure was changed in the mutant channels. Acetazolamide, that is helpful in some patients suffering from EA-1, was without effect on Kv1.1 wild-type or mutant channels. This study confirms and extends earlier studies on the functional consequences of Kv1.1 mutations associated with EA-1, in an attempt to understand the pathophysiology of the disease.


Asunto(s)
Ataxia/genética , Mutación/fisiología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Canales de Potasio/fisiología , Acetazolamida/farmacología , Animales , Conductividad Eléctrica , Electrofisiología , Canal de Potasio Kv.1.1 , Bloqueadores de los Canales de Potasio , Canales de Potasio/metabolismo , Ratas , Tetraetilamonio/farmacología , Células Tumorales Cultivadas
12.
Br J Pharmacol ; 126(8): 1707-16, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10372812

RESUMEN

1. UK-78,282, a novel piperidine blocker of the T lymphocyte voltage-gated K+ channel, Kv1.3, was discovered by screening a large compound file using a high-throughput 86Rb efflux assay. This compound blocks Kv1.3 with a IC50 of approximately 200 nM and 1:1 stoichiometry. A closely related compound, CP-190,325, containing a benzyl moiety in place of the benzhydryl in UK-78,282, is significantly less potent. 2 Three lines of evidence indicate that UK-78,282 inhibits Kv1.3 in a use-dependent manner by preferentially blocking and binding to the C-type inactivated state of the channel. Increasing the fraction of inactivated channels by holding the membrane potential at - 50 mV enhances the channel's sensitivity to UK-78,282. Decreasing the number of inactivated channels by exposure to approximately 160 mM external K+ decreases the sensitivity to UK-78,282. Mutations that alter the rate of C-type inactivation also change the channel's sensitivity to UK-78,282 and there is a direct correlation between tau(h) and IC50 values. 3. Competition experiments suggest that UK-78,282 binds to residues at the inner surface of the channel overlapping the site of action of verapamil. Internal tetraethylammonium and external charybdotoxin do not compete UK-78,282's action on the channel. 4. UK-78,282 displays marked selectivity for Kv1.3 over several other closely related K+ channels, the only exception being the rapidly inactivating voltage-gated K+ channel, Kv1.4. 5. UK-78,282 effectively suppresses human T-lymphocyte activation.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Piperidinas/farmacología , Bloqueadores de los Canales de Potasio , Linfocitos T/efectos de los fármacos , Animales , Unión Competitiva , Células COS , Bovinos , Caribdotoxina/metabolismo , Caribdotoxina/farmacología , Células HeLa , Humanos , Radioisótopos de Yodo , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Canales de Potasio/metabolismo , Canales de Potasio/fisiología , Ratas , Ratas Endogámicas Lew , Radioisótopos de Rubidio , Linfocitos T/inmunología , Tetraetilamonio/metabolismo , Tetraetilamonio/farmacología
13.
Biophys J ; 76(5): 2351-60, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10233054

RESUMEN

External tetraethylammonium (TEA+) blocked currents through Kv1.1 channels in a voltage-independent manner between 0 and 100 mV. Lowering extracellular pH (pHo) increased the Kd for TEA+ block. A histidine at position 355 in the Kv1.1 channel protein (homologous to Shaker 425) was responsible for this pH-dependent reduction of TEA+ sensitivity, since the TEA+ effect became independent of pHo after chemical modification of the Kv1.1 channel at H355 and in the H355G and H355K mutant Kv1.1 channels. The Kd values for TEA+ block of the two mutant channels (0.34 +/- 0.06 mM, n = 7 and 0.84 +/- 0. 09 mM, n = 13, respectively) were as expected for a vestibule containing either no or a total of four positive charges at position 355. In addition, the pH-dependent TEA+ effect in the wt Kv1.1 channel was sensitive to the ionic strength of the solution. All our observations are consistent with the idea that lowering pHo increased protonation of H355. This increase in positive charge at H355 will repel TEA+ electrostatically, resulting in a reduction of the effective [TEA+]o at the receptor site. From this reduction we can estimate the distance between TEA+ and each of the four histidines at position 355 to be approximately 10 A, assuming fourfold symmetry of the channel and assuming that TEA+ binds in the central axis of the pore. This determination of the dimensions of the outer vestibule of Kv1.1 channels confirms and extends earlier reports on K+ channels using crystal structure data as well as peptide toxin/channel interactions and points out a striking similarity between vestibules of Kv1.1 and KcsA channels.


Asunto(s)
Proteínas Bacterianas , Bloqueadores de los Canales de Potasio , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/química , Tetraetilamonio/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Fenómenos Biofísicos , Biofisica , Línea Celular , Concentración de Iones de Hidrógeno , Canal de Potasio Kv.1.1 , Canal de Potasio Kv1.3 , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Canales de Potasio/genética , Conformación Proteica , Ratas , Homología de Secuencia de Aminoácido
14.
J Biol Chem ; 273(49): 32697-707, 1998 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-9830012

RESUMEN

The voltage-gated potassium channel in T lymphocytes, Kv1.3, is an important molecular target for immunosuppressive agents. A structurally defined polypeptide, ShK, from the sea anemone Stichodactyla helianthus inhibited Kv1.3 potently and also blocked Kv1.1, Kv1.4, and Kv1.6 at subnanomolar concentrations. Using mutant cycle analysis in conjunction with complementary mutagenesis of ShK and Kv1.3, and utilizing the structure of ShK, we determined a likely docking configuration for this peptide in the channel. Based upon this topological information, we replaced the critical Lys22 in ShK with the positively charged, non-natural amino acid diaminopropionic acid (ShK-Dap22) and generated a highly selective and potent blocker of the T-lymphocyte channel. ShK-Dap22, at subnanomolar concentrations, suppressed anti-CD3 induced human T-lymphocyte [3H]thymidine incorporation in vitro. Toxicity with this mutant peptide was low in a rodent model, with a median paralytic dose of approximately 200 mg/kg body weight following intravenous administration. The overall structure of ShK-Dap22 in solution, as determined from NMR data, is similar to that of native ShK toxin, but there are some differences in the residues involved in potassium channel binding. Based on these results, we propose that ShK-Dap22 or a structural analogue may have use as an immunosuppressant for the prevention of graft rejection and for the treatment of autoimmune diseases.


Asunto(s)
Inmunosupresores/metabolismo , Péptidos/metabolismo , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Inmunosupresores/química , Inmunosupresores/farmacología , Canal de Potasio Kv1.3 , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Péptidos/farmacología , Estructura Secundaria de Proteína , Proteínas Recombinantes/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
15.
J Med Chem ; 41(23): 4542-9, 1998 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-9804693

RESUMEN

A series of psoralens and structurally related 5,7-disubstituted coumarins was synthesized and investigated for their K+ channel blocking activity as well as for their phototoxicity to Artemia salina and their ability to generate singlet oxygen and to photomodify DNA. After screening the compounds on Ranvier nodes of the toad Xenopus laevis, the affinities of the most promising compounds, which proved to be psoralens bearing alkoxy substituents in the 5-position or alkoxymethyl substituents in the neighboring 4- or 4'-position, to a number of homomeric K+ channels were characterized. All compounds exhibited the highest affinity to Kv1.2. 5,8-Diethoxypsoralen (10d) was found to be an equally potent inhibitor of Kv1.2 and Kv1.3, while lacking the phototoxicity normally inherent in psoralens. The reported compounds represent a novel series of nonpeptide blockers of Shaker-type K+ channels that could be further developed into selective inhibitors of Kv1.2 or Kv1. 3.


Asunto(s)
Furocumarinas/síntesis química , Bloqueadores de los Canales de Potasio , Canales de Potasio , Rayos Ultravioleta , Animales , Artemia/efectos de los fármacos , Artemia/efectos de la radiación , Axones/efectos de los fármacos , Cumarinas/síntesis química , Cumarinas/farmacología , Cumarinas/toxicidad , ADN/efectos de los fármacos , ADN/metabolismo , ADN/efectos de la radiación , Evaluación Preclínica de Medicamentos , Furocumarinas/farmacología , Furocumarinas/toxicidad , Técnicas In Vitro , Oxígeno/metabolismo , Oxígeno/efectos de la radiación , Nódulos de Ranvier/efectos de los fármacos , Nódulos de Ranvier/ultraestructura , Canales de Potasio de la Superfamilia Shaker , Xenopus laevis
16.
J Physiol ; 506 ( Pt 2): 291-301, 1998 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9490854

RESUMEN

1. Using the whole-cell recording mode we have characterized two non-conducting states in mammalian Shaker-related voltage-gated K+ channels induced by the removal of extracellular potassium, K+o. 2. In the absence of K+o, current through Kv1.4 was almost completely abolished due to the presence of a charged lysine residue at position 533 at the entrance to the pore. Removal of K+o had a similar effect on current through Kv1.3 when the histidine at the homologous position (H404) was protonated (pH 6.0). Channels containing uncharged residues at the corresponding position (Kv1.1: Y; Kv1.2: V) did not exhibit this behaviour. 3. To characterize the nature of the interaction between Kv1.3 and K+o concentration ([K+]o), we replaced H404 with amino acids of different character, size and charge. Substitution of hydrophobic residues (A, V and L) either in all four subunits or in only two subunits in the tetramer made the channel insensitive to the removal of K+o, possibly by stabilizing the channel complex. Replacement of H404 with the charged residue arginine, or the polar residue asparagine, enhanced the sensitivity of the channel to 0 mM K+o, possibly by making the channel unstable in the absence of K+o. Mutation at a neighbouring position (400) had a similar effect. 4. The effect of removing K+o on current amplitude does not seem to be correlated with the rate of C-type inactivation since the slowly inactivating G380F mutant channel exhibited a similar [K+]o dependence as the wild-type Kv1.3 channel. 5. CP-339,818, a drug that recognizes only the inactivated conformation of Kv1.3, could not block current in the absence of K+o unless the channels were inactivated through depolarizing pulses. 6. We conclude that removal of K+o induces the Kv1.3 channel to transition to a non-conducting 'closed' state which can switch into a non-conducting 'inactivated' state upon depolarization.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio/efectos de los fármacos , Células 3T3 , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Electrofisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Histidina/química , Histidina/efectos de los fármacos , Histidina/genética , Activación del Canal Iónico/fisiología , Canal de Potasio Kv.1.1 , Canal de Potasio Kv.1.2 , Canal de Potasio Kv1.3 , Canal de Potasio Kv1.4 , Células L , Ratones , Datos de Secuencia Molecular , Mutación/genética , Mutación/fisiología , Potasio/metabolismo , Potasio/farmacología , Canales de Potasio/genética , Canales de Potasio/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
18.
J Physiol ; 500 ( Pt 3): 653-60, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9161983

RESUMEN

1. The whole-cell recording mode of the patch-clamp technique was used to study the effect of extracellular K+ and Rb+ on membrane currents in human osteoblasts, in a human osteoblast-like cell line, and in the Jurkat human leukaemic T cell line. 2. Increasing the extracellular concentration of K+ increased the membrane conductance of the cells in a concentration-dependent manner. This increase in membrane conductance was due to the activation of a Cl- conductance. Rb+ also induced this conductance, but conductance was less than half that seen in K+. 3. The Cl- channel blockers 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid (DIDS) and 4-acetamido-4'-isothiocyanatostilbene 2,2'-disulphonic acid (SITS) blocked the K(+)-induced Cl- current in a voltage-dependent manner. The degree of blockade increased with membrane depolarization to a maximum level at 40 mV. At potentials above this value the block appeared to decrease. 4. Both tonicity and K+ were required for maximal activation of the Cl- conductance since the K(+)-induced Cl- conductance could be inhibited by hypertonic solutions and the activation of a volume-sensitive Cl- conductance by hypotonic solutions could be enhanced by extracellular K+. 5. We conclude that an outwardly rectifying Cl- conductance can be activated either upon osmotic swelling or by an increase in extracellular K+. Both activation pathways may be involved in cell volume regulation and seem to apply to volume-sensitive Cl- channels in general since we observe this phenomenon in two different cell types, in human osteoblasts as well as in human leukaemic T lymphocytes.


Asunto(s)
Canales de Cloruro/metabolismo , Leucemia-Linfoma de Células T del Adulto/metabolismo , Osteoblastos/metabolismo , Potasio/farmacología , Linfocitos T/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Ácido 4-Acetamido-4'-isotiocianatostilbeno-2,2'-disulfónico/farmacología , Línea Celular , Canales de Cloruro/efectos de los fármacos , Electrofisiología , Humanos , Potenciales de la Membrana/fisiología , Concentración Osmolar , Osteoblastos/efectos de los fármacos , Técnicas de Placa-Clamp , Rubidio/farmacología , Linfocitos T/efectos de los fármacos
19.
Mol Pharmacol ; 50(6): 1625-34, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8967986

RESUMEN

We characterized the action of verapamil and N-methyl-verapamil on current through the delayed-rectifier potassium channel Kv1.3 mouse (mKv1.3). The whole-cell and inside-out configuration of the patch-clamp technique was used to examine the channel properties after injection of in vitro transcribed cRNA into rat basophilic leukemia cells. The action of verapamil on current through mKv1.3 channels could be separated into an acceleration of the rate of current decay during depolarizing pulses and a reduction of steady state peak current when applied either extracellularly or intracellularly. Both effects were greatly reduced when the membrane-impermeable N-methyl-verapamil was applied extracellularly, but it affected current through mKv1.3 channels similar to verapamil if applied to the intracellular side of the membrane. Mutations in the outer vestibule of the mKv1.3 channel did not change the ability of verapamil to accelerate the mKv1.3 current decay during depolarizing pulses, whereas the reduction of the steady state peak current by verapamil applied either extracellularly and intracellularly and by N-methyl-verapamil applied intracellularly was decreased approximately 25-fold in all three cases. Substances known to interact with an extracellular site of the channel (e.g., extracellularly applied tetraethylammonium or kaliotoxin) did not compete with extracellularly applied verapamil on blocking steady state peak current, whereas intracellularly applied tetraethylammonium, which is known to interact with an intracellular site of the channel, was able to reduce the effect of extracellularly applied verapamil on blocking steady state peak current, suggesting competition for a common binding site between verapamil and intracellularly applied tetraethylammonium. The results from the competition experiments as well as from the mutations in the outer vestibule of mKv1.3 are compatible with the idea that verapamil applied extracellularly moves through the membrane to reach its internal binding site on the mKv1.3 channel.


Asunto(s)
Canales de Potasio/efectos de los fármacos , Verapamilo/farmacología , Secuencia de Aminoácidos , Animales , Activación del Canal Iónico , Ratones , Datos de Secuencia Molecular , Canales de Potasio/química , Ratas , Verapamilo/análogos & derivados
20.
Mol Pharmacol ; 50(6): 1672-9, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8967992

RESUMEN

The nonpeptide agent CP-339,818 (1-benzyl-4-pentylimino-1,4-dihydroquinoline) and two analogs (CP-393,223 and CP-394,322) that differ only with respect to the type of substituent at the N1 position, potently blocked the Kv1.3 channel in T lymphocytes. A fourth compound (CP-393,224), which has a smaller and less-lipophilic group at N1, was 100-200-fold less potent, suggesting that a large lipophilic group at this position is necessary for drug activity. CP-339,818 blocked Kv1.3 from the outside with a IC50 value of approximately 200 nM and 1:1 stoichiometry and competitively inhibited 125I-charybdotoxin from binding to the external vestibule of Kv1.3. This drug inhibited Kv1.3 in a use-dependent manner by preferentially blocking the C-type inactivated state of the channel. CP-339,818 was a significantly less potent blocker of Kv1.1, Kv1.2, Kv1.5, Kv1.6, Kv3.1-4, and Kv4.2; the only exception was Kv1.4, a cardiac and neuronal A-type K+ channel. CP-339,818 had no effect on two other T cell channels (I(CRAC) and intermediate-conductance K(Ca)) implicated in T cell mitogenesis. This drug suppresses human T cell activation, suggesting that blockade of Kv1.3 alone is sufficient to inhibit this process.


Asunto(s)
Activación de Linfocitos/efectos de los fármacos , Bloqueadores de los Canales de Potasio , Linfocitos T/efectos de los fármacos , Células HeLa , Humanos , Mutación , Canales de Potasio/química , Conformación Proteica , Canales de Potasio Shal , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA