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










Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 162(5): 1074-82, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20973779

RESUMEN

BACKGROUND AND PURPOSE: Diltiazem inhibits Ca(V)1.2 channels and is widely used in clinical practice to treat cardiovascular diseases. Binding determinants for diltiazem are located on segments IIIS6, IVS6 and the selectivity filter of the pore forming α1 subunit of Ca(V)1.2. The aim of the present study was to clarify the location of the diltiazem binding site making use of its membrane-impermeable quaternary derivative d-cis-diltiazem (qDil) and mutant α1 subunits. EXPERIMENTAL APPROACH: Ca(V)1.2 composed of α1, α2-δ and ß2a subunits were expressed in tsA-201 cells and barium currents through Ca(V)1.2 channels were recorded using the patch clamp method in the whole cell configuration. qDil was synthesized and applied to the intracellular side (via the patch pipette) or to the extracellular side of the membrane (by bath perfusion). KEY RESULTS: Quaternary derivative d-cis-diltiazem inhibited Ca(V)1.2 when applied to the intracellular side of the membrane in a use-dependent manner (59 ± 4% at 300 µM) and induced only a low level of tonic (non-use-dependent) block (16 ± 2% at 300 µM) when applied to the extracellular side of the membrane. Mutations in IIIS6 and IVS6 that have previously been shown to reduce the sensitivity of Ca(V)1.2 to tertiary diltiazem also had reduced sensitivity to intracellularly applied qDil. CONCLUSION AND IMPLICATIONS: The data show that use-dependent block of in Ca(V)1.2 by diltiazem occurs by interaction with a binding site accessible via a hydrophilic route from the intracellular side of the membrane.


Asunto(s)
Bloqueadores de los Canales de Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Diltiazem/análogos & derivados , Secuencia de Aminoácidos , Sitios de Unión , Canales de Calcio Tipo L/genética , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Diltiazem/metabolismo , Diltiazem/farmacología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Activación del Canal Iónico/efectos de los fármacos , Cinética , Datos de Secuencia Molecular , Proteínas Mutantes/efectos de los fármacos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Técnicas de Placa-Clamp , Subunidades de Proteína , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
2.
Mol Pharmacol ; 58(6): 1264-70, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11093762

RESUMEN

The benzothiazepine diltiazem blocks ionic current through L-type Ca(2+) channels, as do the dihydropyridines (DHPs) and phenylalkylamines (PAs), but it has unique properties that distinguish it from these other drug classes. Wild-type L-type channels containing alpha(1CII) subunits, wild-type P/Q-type channels containing alpha(1A) subunits, and mutants of both channel types were transiently expressed in tsA-201 cells with beta(1B) and alpha(2)delta subunits. Whole-cell, voltage-clamp recordings showed that diltiazem blocks L-type Ca(2+) channels approximately 5-fold more potently than it does P/Q-type channels. Diltiazem blocked a mutant P/Q-type channel containing nine amino acid changes that made it highly sensitive to DHPs, with the same potency as L-type channels. Thus, amino acids specific to the L-type channel that confer DHP sensitivity in an alpha(1A) background also increase sensitivity to diltiazem. Analysis of single amino acid mutations in domains IIIS6 and IVS6 of alpha(1CII) subunits confirmed the role of these L-type-specific amino acid residues in diltiazem block, and also indicated that Y1152 of alpha(1CII), an amino acid critical to both DHP and PA block, does not play a role in diltiazem block. Furthermore, T1039 and Y1043 in domain IIIS5, which are both critical for DHP block, are not involved in block by diltiazem. Conversely, three amino acid residues (I1150, M1160, and I1460) contribute to diltiazem block but have not been shown to affect DHP or PA block. Thus, binding of diltiazem to L-type Ca(2+) channels requires residues that overlap those that are critical for DHP and PA block as well as residues unique to diltiazem.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/fisiología , Diltiazem/farmacología , Alanina/genética , Alanina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/genética , Células Cultivadas , Secuencia Conservada , Electrofisiología , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Datos de Secuencia Molecular , Pruebas de Mutagenicidad , Conformación Proteica , Homología de Secuencia de Aminoácido
3.
J Biol Chem ; 272(30): 18752-8, 1997 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-9228048

RESUMEN

The dihydropyridine Ca2+ antagonist drugs used in the therapy of cardiovacular disorders inhibit L-type Ca2+ channels by binding to a single high affinity site. Photoaffinity labeling and analysis of mutant Ca2+ channels implicate the IIIS6 and IVS6 segments in high affinity binding. The amino acid residues that are required for high affinity binding of dihydropyridine Ca2+ channel antagonists were probed by alanine-scanning mutagenesis of the alpha1C subunit, transient expression in mammalian cells, and analysis by measurements of ligand binding and block of Ba2+ currents through expressed Ca2+ channels. Eleven amino acid residues in transmembrane segments IIIS6 and IVS6 were identified whose mutation reduced the affinity for the Ca2+ antagonist PN200-110 by 2-25-fold. Both amino acid residues conserved among Ca2+ channels and those specific to L-type Ca2+ channels were found to be required for high affinity dihydropyridine binding. In addition, mutation F1462A increased the affinity for the dihydropyridine Ca2+ antagonist PN200-110 by 416-fold with no effect on the affinity for the Ca2+ agonist Bay K8644. The residues in transmembrane segments IIIS6 and IVS6 that are required for high affinity binding are primarily aligned on single faces of these two alpha helices, supporting a "domain interface model" of dihydropyridine binding and action in which the IIIS6 and IVS6 interact to form a high affinity dihydropyridine receptor site on L-type Ca2+ channels.


Asunto(s)
Alanina/química , Canales de Calcio/química , Alanina/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/genética , Canales de Calcio Tipo L , Línea Celular , Humanos , Isradipino/farmacología , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mapeo Peptídico , Estructura Secundaria de Proteína , Conejos
4.
J Biol Chem ; 272(30): 18759-65, 1997 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-9228049

RESUMEN

Recent studies of the phenylalkylamine binding site in the alpha1C subunit of L-type Ca2+ channels have revealed three amino acid residues in transmembrane segment IVS6 that are critical for high affinity block and are unique to L-type channels. We have extended this analysis of the phenylalkylamine binding site to amino acid residues in transmembrane segment IIIS6 and the pore region. Twenty-two consecutive amino acid residues in segment IIIS6 were mutated to alanine and the conserved Glu residues in the pore region of each homologous domain were mutated to Gln. Mutant channels were expressed in tsA-201 cells along with the beta1b and alpha2delta auxiliary subunits. Assay for block of Ba2+ current by (-)-D888 at -60 mV revealed that mutation of five amino acid residues in segment IIIS6 and the pore region that are conserved between L-type and non-L-type channels (Tyr1152, Phe1164, Val1165, Glu1118, and Glu1419) and one L-type-specific amino acid (Ile1153) decreased affinity for (-)-D888 from 10-20-fold. Combination of the four mutations in segment IIIS6 increased the IC50 for block by (-)-D888 to approximately 9 microM, similar to the affinity of non-L-type Ca2+ channels for this drug. These results indicate that there are important determinants of phenylalkylamine binding in both the S6 segments and the pore regions of domains III and IV, some of which are conserved across the different classes of voltage-gated Ca2+ channels. A model of the phenylalkylamine receptor site at the interface between domains III and IV of the alpha1 subunit is presented.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Verapamilo/análogos & derivados , Potenciales de Acción , Secuencia de Aminoácidos , Sitios de Unión/genética , Bloqueadores de los Canales de Calcio/metabolismo , Canales de Calcio Tipo L , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Glutamina/genética , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Relación Estructura-Actividad , Verapamilo/metabolismo , Verapamilo/farmacología
5.
J Neurosci ; 17(4): 1243-55, 1997 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9006969

RESUMEN

Ca2+ influx through skeletal muscle Ca2+ channels and the force of contraction are increased in response to beta-adrenergic stimulation and high-frequency electrical stimulation. These effects are thought to be mediated by cAMP-dependent phosphorylation of the skeletal muscle Ca2+ channel. Modulation of the cloned skeletal muscle Ca2+ channel by cAMP-dependent phosphorylation and by depolarizing prepulses was reconstituted by transient expression in tsA-201 cells and compared to modulation of the native skeletal muscle Ca2+ channel as expressed in mouse 129CB3 skeletal muscle cells. The heterologously expressed Ca2+ channel consisting of alpha1, alpha2delta, and beta subunits gave currents that were similar in time course, current density, and dihydropyridine sensitivity to the native Ca2+ channel. cAMP-dependent protein kinase (PKA) stimulation by Sp-5,6-DCl-cBIMPS (cBIMPS) increased currents through both native and expressed channels two- to fourfold. Tail currents after depolarizations to potentials between -20 and +80 mV increased in amplitude and decayed more slowly as either the duration or potential of the depolarization was increased. The time- and voltage-dependent slowing of channel deactivation required the activity of PKA, because it was enhanced by cBIMPS and reduced or eliminated by the peptide PKA inhibitor PKI (5-24) amide. This voltage-dependent modulation of the cloned skeletal muscle Ca2+ channel by PKA also required anchoring of PKA by A-Kinase Anchoring Proteins because it was blocked by peptide Ht 31, which disrupts such anchoring. The results show that the skeletal muscle Ca2+ channel expressed in heterologous cells is modulated by PKA at rest and during depolarization and that this modulation requires anchored protein kinase, as it does in native skeletal muscle cells.


Asunto(s)
Canales de Calcio/genética , Canales de Calcio/metabolismo , Clonación Molecular , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas de la Membrana/fisiología , Músculo Esquelético/metabolismo , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/farmacología , Animales , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio/fisiología , AMP Cíclico/metabolismo , Diclororribofuranosil Benzoimidazol/análogos & derivados , Diclororribofuranosil Benzoimidazol/farmacología , Dihidropiridinas/agonistas , Conductividad Eléctrica , Ratones , Fosforilación , Tionucleótidos/farmacología
6.
Proc Natl Acad Sci U S A ; 94(4): 1512-6, 1997 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-9037084

RESUMEN

Synaptic transmission is regulated by G protein-coupled receptors whose activation releases G protein betagamma subunits that modulate presynaptic Ca2+ channels. The sequence motif QXXER has been proposed to be involved in the interaction between G protein betagamma subunits and target proteins including adenylyl cyclase 2. This motif is present in the intracellular loop connecting domains I and II (L I-II) of Ca2+ channel alpha1A subunits, which are modulated by G proteins, but not in alpha1C subunits, which are not modulated. Peptides containing the QXXER motif from adenylate cyclase 2 or from alpha1A block G protein modulation but a mutant peptide containing the sequence AXXAA does not, suggesting that the QXXER-containing peptide from alpha1A can competitively inhibit Gbetagamma modulation. Conversion of the R in the QQIER sequence of alpha1A to E as in alpha1C slows channel inactivation and shifts the voltage dependence of steady-state inactivation to more positive membrane potentials. Conversion of the final E in the QQLEE sequence of alpha1C to R has opposite effects on voltage-dependent inactivation, although the changes are not as large as those for alpha1A. Mutation of the QQIER sequence in alpha1A to QQIEE enhanced G protein modulation, and mutation to QQLEE as in alpha1C greatly reduced G protein modulation and increased the rate of reversal of G protein effects. These results indicate that the QXXER motif in L I-II is an important determinant of both voltage-dependent inactivation and G protein modulation, and that the amino acid in the third position of this motif has an unexpectedly large influence on modulation by Gbetagamma. Overlap of this motif with the consensus sequence for binding of Ca2+ channel beta subunits suggests that this region of L I-II is important for three different modulatory influences on Ca2+ channel activity.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Unión al GTP/metabolismo , Activación del Canal Iónico , Fragmentos de Péptidos/metabolismo , Adenilil Ciclasas/metabolismo , Secuencia de Aminoácidos , Canales de Calcio/efectos de los fármacos , Canales de Calcio/genética , Polaridad Celular , Secuencia de Consenso , Análisis Mutacional de ADN , Datos de Secuencia Molecular , Mutagénesis , Unión Proteica , Conformación Proteica , Proteínas Recombinantes/metabolismo
7.
Annu Rev Pharmacol Toxicol ; 37: 361-96, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9131258

RESUMEN

The crucial role of L-type Ca2+ channels in the initiation of cardiac and smooth muscle contraction has made them major therapeutic targets for the treatment of cardiovascular disease. L-type channels share a common pharmacological profile, including high-affinity voltage- and frequency-dependent block by the phenylalkylamines, the benz(othi)azepines, and the dihydropyridines. These drugs are thought to bind to three separate receptor sites on L-type Ca2+ channels that are allosterically linked. Results from different experimental approaches implicate the IIIS5, IIIS6, and IVS6 transmembrane segments of the alpha 1 subunits of L-type Ca2+ channels in binding of all three classes of drugs. Site-directed mutagenesis has identified single amino acid residues within the IIIS5, IIIS6, and IVS6 transmembrane segments that are required for high-affinity binding of phenylalkylamines and/or dihydropyridines, providing further support for identification of these transmembrane segments as critical elements of the receptor sites for these two classes of drugs. The close proximity of the receptor sites for phenylalkylamines, benz(othi)azepines, and dihydropyridines raises the possibility that individual amino acid residues may be required for high-affinity binding of more than one of these ligands. Therefore, we suggest that phenylalkylamines and dihydropyridines bind to different faces of the IIIS6 and IVS6 transmembrane segments and, in some cases, bind to opposite sides of the side chains of the same amino acid residues. The results support the domain interface model for binding and channel modulation by these three classes of drugs.


Asunto(s)
Canales de Calcio/metabolismo , Secuencia de Aminoácidos , Benzazepinas/metabolismo , Benzazepinas/farmacología , Sitios de Unión , Bloqueadores de los Canales de Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/química , Canales de Calcio/efectos de los fármacos , Dihidropiridinas/metabolismo , Dihidropiridinas/farmacología , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Fenetilaminas/metabolismo , Fenetilaminas/farmacología , Unión Proteica/efectos de los fármacos
8.
Proc Natl Acad Sci U S A ; 94(26): 14906-11, 1997 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-9405712

RESUMEN

The activity of L-type Ca2+ channels is increased by dihydropyridine (DHP) agonists and inhibited by DHP antagonists, which are widely used in the therapy of cardiovascular disease. These drugs bind to the pore-forming alpha1 subunits of L-type Ca2+ channels. To define the minimal requirements for DHP binding and action, we constructed a high-affinity DHP receptor site by substituting a total of nine amino acid residues from DHP-sensitive L-type alpha1 subunits into the S5 and S6 transmembrane segments of domain III and the S6 transmembrane segment of domain IV of the DHP-insensitive P/Q-type alpha1A subunit. The resulting chimeric alpha1A/DHPS subunit bound DHP antagonists with high affinity in radioligand binding assays and was inhibited by DHP antagonists with high affinity in voltage clamp experiments. Substitution of these nine amino acid residues yielded 86% of the binding energy of the L-type alpha1C subunit and 92% of the binding energy of the L-type alpha1S subunit for the high-affinity DHP antagonist PN200-110. The activity of chimeric Ca2+ channels containing alpha1A/DHPS was increased 3.5 +/- 0.7-fold by the DHP agonist (-)Bay K8644. The effect of this agonist was stereoselective as in L-type Ca2+ channels since (+) Bay K8644 inhibited the activity of alpha1A/DHPS. The results show conclusively that DHP agonists and antagonists bind to a single receptor site at which they have opposite effects on Ca2+ channel activity. This site contains essential components from both domains III and IV, consistent with a domain interface model for binding and allosteric modulation of Ca2+ channel activity by DHPs.


Asunto(s)
Canales de Calcio/genética , Canales de Calcio/metabolismo , Dihidropiridinas/metabolismo , Sustitución de Aminoácidos , Aminoácidos/genética , Aminoácidos/metabolismo , Sitios de Unión , Canales de Calcio Tipo L , Línea Celular , Dihidropiridinas/agonistas , Dihidropiridinas/antagonistas & inhibidores , Humanos
9.
Mol Pharmacol ; 50(5): 1388-400, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8913371

RESUMEN

The phenylalkylamines (-)-D888, verapamil, and D600, cause voltage- and use-dependent block of L-type Ca2+ channels and differ from each other only in the number of methoxy groups on each of their two terminal phenyl rings. To study the effects of mutations in the phenylalkylamine receptor site on block by these drugs, wild-type and mutant Ca2+ channels were transiently expressed in the tsA-201 clone of human embryonic kidney 293 cells. The combined mutations Y1463A, A1467S, and I1470A (mutant YAI) in transmembrane segment S6 of domain IV of the alpha 1c subunit disrupted block by all three phenylalkylamines. Surprisingly, although this mutation reduced both resting block at -60 mV and depolarized block at +10 mV by (-)-D888, resting and depolarized block by verapamil and D600 were relatively unaffected. In contrast, for all three drugs, use-dependent block during repetitive stimulations was sharply reduced, and the rate of recovery from depolarized block was accelerated for YAI channels. Thus, the effects of the YAI mutation on apparent affinity were specific to (-)-D888, whereas effects on the kinetics of block were observed for all three drugs. Additional experiments with substitution of phenylalanine for Y1463 suggested that (-)-D888 affinity is specifically sensitive to removal of the hydroxyl group of Y1463, whereas effects on the kinetics of block by all three phenylalkylamines require larger molecular changes, perhaps related to residue size and hydrophobicity. Analysis of the data using a state-dependent model of drug block suggests that these kinetic differences are caused by both changes in drug access to the receptor site and affinity for binding to the inactivated state of the channel. The different effects of the YAI mutations on the actions of (-)-D888, verapamil, and D600 indicate that these residues interact differently with these closely related drugs.


Asunto(s)
Compuestos de Anilina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/genética , Mutación , Secuencia de Aminoácidos , Canales de Calcio/fisiología , Canales de Calcio Tipo L , Electrofisiología , Galopamilo/farmacología , Cinética , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Relación Estructura-Actividad , Verapamilo/análogos & derivados , Verapamilo/farmacología
10.
Mol Pharmacol ; 49(6): 1021-32, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8649340

RESUMEN

The interactions between beta-adrenergic receptor (beta AR) antagonists and the beta(2)AR were studied with the use of photoaffinity labels. A proteolytic map of the receptor was made and confirmed through amino-terminal amino acid sequencing by locating sites of derivatization. [125I]Iodoazidothiophenylalprenolol (IAPTA) is a photoaffinity derivative of the beta AR antagonist alprenolol with a photoactivatable group on the aryloxy end of the molecule. IAPTA exclusively derivatizes a peptide consisting of transmembrane domains (TMs) 6 and 7 of the hamster lung beta(2)AR, supporting the contention that TMs 6 and 7 interact with the aryloxy portion of the beta AR antagonist pharmacophore. The beta AR antagonist photoaffinity labels [125I]iodoazidobenzylpindolol (IABP), [125I]iodoazidophenyl CGP-12177A (IAPCGP), and [125I]iodocyanopindololdiazarene (ICYPdz) are similar in that their photoactive moieties are attached to the amino end of the antagonist pharmacophore. IABP derivatized TMs 5-7 and a peptide containing TM 1 to approximately equal extents. IAPCGP derivatized Tms 6 and 7 >> TM 5 = TM 4 = TMs 2 and 3 = TM 1. ICYPdz derivatized TM 1 >> TMs 6 and 7 > Tm 4. We conclude that the aryloxy end of the beta AR antagonist pharmacophore is highly constrained within TMs 6 and 7, whereas the amino terminus is much less constrained and able to assume multiple conformations. Molecular dynamics simulations predict that IABP, IAPCGP, and ICYPdz favor a folded conformation, with both ends close together. Derivatization of TMs 6 and 7 by IABP, IAPCGP, and ICYPdz suggests the folded conformation of these compounds in the ligand binding pocket.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 2 , Antagonistas Adrenérgicos beta/química , Marcadores de Afinidad , Secuencia de Aminoácidos , Animales , Cricetinae , Conformación Molecular , Datos de Secuencia Molecular , Peso Molecular , Mapeo Peptídico , Receptores Adrenérgicos beta 2/química , Serina Endopeptidasas/farmacología , Spodoptera , Tripsina/farmacología
11.
J Biol Chem ; 270(38): 22119-22, 1995 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-7673189

RESUMEN

The high affinity phenylalkylamine (-)D888 blocks ion currents through L-type Ca2+ channels containing the alpha 1C subunit with an apparent Kd of 50 nM, but N-type Ca2+ channels in the pheochromocytoma cell line PC12 are blocked with a 100-fold higher Kd value of 5 microM. L-type Ca2+ channels containing alpha 1C subunits with the site-directed mutations Y1463A, A1467S, or I1470A in the putative transmembrane segment S6 in domain IV (IVS6) were 6-12 times less sensitive to block by (-)D888 than control alpha 1C. Ca2+ channels containing paired combinations of these mutations were even less sensitive to block by (-)D888 than the single mutants, and channels containing all three mutations were > 100 times less sensitive to (-)D888 block, similar to N-type Ca2+ channels. In addition, the Y1463A mutant and all combination mutants including the Y1463A mutation had altered ion selectivity, suggesting that Tyr-1463 faces the pore and is involved in ion permeation. Since these three critical amino acid residues are aligned on the same face of the putative IVS6 alpha-helix, we propose that they contribute to a receptor site in the pore that confers a high affinity block of L-type channels by (-)D888.


Asunto(s)
Canales de Calcio/química , Verapamilo/análogos & derivados , Secuencia de Aminoácidos , Animales , Canales de Calcio/fisiología , Activación del Canal Iónico/efectos de los fármacos , Glicoproteínas de Membrana/química , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Ratas , Relación Estructura-Actividad , Verapamilo/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA