Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 276(16): 12730-5, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11278630

RESUMO

We investigated the mechanism of interaction of individual L-type channel amino acid residues with dihydropyridines within a dihydropyridine-sensitive alpha1A subunit (alpha1A(DHP)). Mutation of individual residues in repeat III and expression in Xenopus oocytes revealed that Thr(1393) is not required for dihydropyridine interaction but that bulky side chains (tyrosine, phenylalanine) in this position sterically inhibit dihydropyridine coordination. In position 1397 a side chain carbonyl group was required for high antagonist sensitivity. Agonist function required the complete amide group of a glutamine residue. Val(1516) and Met(1512) side chains were required for agonist (Val(1516)) and antagonist (Val(1516), Met(1512)) sensitivity. Replacement of Ile(1504) and Ile(1507) by alpha1A phenylalanines was tolerated. Substitution of Thr(1393) by phenylalanine or Val(1516) by alanine introduced voltage dependence of antagonist action into alpha1A(DHP), suggesting that these residues form part of a mechanism mediating voltage dependence of dihydropyridine sensitivity. Our data provide important insight into dihydropyridine binding to alpha1A(DHP) which could facilitate the development of alpha1A-selective modulators. By modulating P/Q-type Ca(2+) channels such drugs could serve as new anti-migraine therapeutics.


Assuntos
Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/fisiologia , Di-Hidropiridinas/farmacologia , Oócitos/fisiologia , Alanina , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Bloqueadores dos Canais de Cálcio/farmacologia , Di-Hidropiridinas/farmacocinética , Feminino , Isoleucina , Isradipino/farmacologia , Cinética , Metionina , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oócitos/efeitos dos fármacos , Fenilalanina , Estrutura Secundária de Proteína , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Valina , Xenopus laevis
2.
Biochem J ; 347 Pt 3: 829-36, 2000 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10769189

RESUMO

Sensitivity to 1,4-dihydropyridines (DHPs) can be transferred from L-type (alpha1C) to non-L-type (alpha1A) Ca(2+) channel alpha1 subunits by the mutation of nine pore-associated non-conserved amino acid residues, yielding mutant alpha1A(DHP). To determine whether the hallmarks of reversible DHP binding to L-type Ca(2+) channels (nanomolar dissociation constants, stereoselectivity and modulation by other chemical classes of Ca(2+) antagonist drugs) were maintained in alpha1A(DHP), we analysed the pharmacological properties of (+)-[(3)H]isradipine-labelled alpha1A(DHP) Ca(2+) channels after heterologous expression. Binding of (+)-isradipine (K(i) 7.4 nM) and the non-benzoxadiazole DHPs nifedipine (K(i) 86 nM), (+/-)-nitrendipine (K(i) 33 nM) and (+/-)-nimodipine (K(i) 67 nM) to alpha1A(DHP) occurred at low nanomolar K(i) values. DHP binding was highly stereoselective [25-fold higher affinity for (+)-isradipine]. As with native channels it was stimulated by (+)-cis-diltiazem, (+)-tetrandrine and mibefradil. This suggested that the three-dimensional architecture of the channel pore was maintained within the non-L-type alpha1A subunit. To predict the three-dimensional arrangement of the DHP-binding residues we exploited the X-ray structure of a recently crystallized bacterial K(+) channel (KcsA) as a template. Our model is based on the assumption that the Ca(2+) channel S5 and S6 segments closely resemble the KcsA transmembrane folding architecture. In the absence of three-dimensional structural data for the alpha1 subunit this is currently the most reasonable approach for modelling this drug-interaction domain. Our model predicts that the previously identified DHP-binding residues form a binding pocket large enough to co-ordinate a single DHP molecule. It also implies that the four homologous Ca(2+) channel repeats are arranged in a clockwise manner.


Assuntos
Substituição de Aminoácidos/genética , Proteínas de Bactérias , Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio/química , Canais de Cálcio/metabolismo , Di-Hidropiridinas/metabolismo , Dobramento de Proteína , Sequência de Aminoácidos , Animais , Sítios de Ligação , Canais de Cálcio/genética , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Canais de Potássio/química , Ligação Proteica , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Estereoisomerismo , Especificidade por Substrato , Termodinâmica
3.
J Biol Chem ; 274(10): 6154-60, 1999 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-10037699

RESUMO

The molecular basis of the Ca2+ channel block by (+)-cis-diltiazem was studied in class A/L-type chimeras and mutant alpha1C-a Ca2+ channels. Chimeras consisted of either rabbit heart (alpha1C-a) or carp skeletal muscle (alpha1S) sequence in transmembrane segments IIIS6, IVS6, and adjacent S5-S6 linkers. Only chimeras containing sequences from alpha1C-a were efficiently blocked by (+)-cis-diltiazem, whereas the phenylalkylamine (-)-gallopamil efficiently blocked both constructs. Carp skeletal muscle and rabbit heart Ca2+ channel alpha1 subunits differ with respect to two nonconserved amino acids in segments IVS6. Transfer of a single leucine (Leu1383, located at the extracellular mouth of the pore) from IVS6 alpha1C-a to IVS6 of alpha1S significantly increased the (+)-cis-diltiazem sensitivity of the corresponding mutant L1383I. An analysis of the role of the two heterologous amino acids in a L-type alpha1 subunit revealed that corresponding amino acids in position 1487 (outer channel mouth) determine recovery of resting Ca2+ channels from block by (+)-cis-diltiazem. The second heterologous amino acid in position 1504 of segment IVS6 (inner channel mouth) was identified as crucial inactivation determinant of L-type Ca2+ channels. This residue simultaneously modulates drug binding during membrane depolarization. Our study provides the first evidence for a guarded and modulated benzothiazepine receptor on L-type channels.


Assuntos
Canais de Cálcio/genética , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Canais de Cálcio/metabolismo , Carpas , Dados de Sequência Molecular , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade
4.
J Bioenerg Biomembr ; 30(4): 319-34, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9758329

RESUMO

Different types of voltage-gated Ca2+ channels exist in the plasma membrane of electrically excitable cells. By controlling depolarization-induced Ca2+ entry into cells they serve important physiological functions, such as excitation-contraction coupling, neurotransmitter and hormone secretion, and neuronal plasticity. Their function is fine-tuned by a variety of modulators, such as enzymes and G-proteins. Block of so-called L-type Ca2+ channels by drugs is exploited as a therapeutic principle to treat cardiovascular disorders, such as hypertension. More recently, block of so-called non-L-type Ca2+ channels was found to exert therapeutic effects in the treatment of severe pain and ischemic stroke. As the subunits of different Ca2+ channel types have been cloned, the modulatory sites for enzymes, G-proteins, and drugs can now be determined using molecular engineering and heterologous expression. Here we summarize recent work that has allowed us to determine the sites of action of L-type Ca2+ channel modulators. Together with previous biochemical, electrophysiological, and drug binding data these results provide exciting insight into the molecular pharmacology of this voltage-gated Ca2+ channel family.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Marcadores de Afinidade , Regulação Alostérica , Sítio Alostérico , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Anti-Hipertensivos/farmacologia , Sítios de Ligação , Cálcio/metabolismo , Canais de Cálcio/química , Canais de Cálcio/genética , Canais de Cálcio/imunologia , Canais de Cálcio Tipo L , Sinalização do Cálcio/efeitos dos fármacos , Desenho de Fármacos , Epitopos/imunologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade
5.
Trends Pharmacol Sci ; 19(3): 108-15, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9584627

RESUMO

At least five different types of voltage-gated Ca2+ channels exist in electrically excitable mammalian cells. Only one type, the family of L-type Ca2+ channels (L channels), contains high-affinity binding domains within their alpha 1-subunits for different chemical classes of drugs (Ca2+ channel antagonists; exemplified by isradipine, verapamil and diltiazem). Their stereoselective, high-affinity binding induces block of channel-mediated Ca2+ inward currents in heart and smooth muscle, resulting in antihypertensive, cardiodepressive and antiarrhythmic effects. Amino acids involved in drug binding have recently been identified using photoaffinity labelling, chimeric alpha 1-subunits and site-directed mutagenesis. Insertion of the drug-binding amino acids enabled the transfer of drug-sensitivity into Ca2+ channels that are insensitive to Ca2+ channel antagonists ('gain-of-function' approach). In this review, Jörg Striessing and colleagues summarize the present knowledge about the molecular architecture of L channel drug-binding domains and the implications for Ca2+ channel pharmacology and drug development.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio/metabolismo , Animais , Canais de Cálcio/química , Humanos , Conformação Proteica
6.
J Biol Chem ; 273(15): 9110-8, 1998 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-9535900

RESUMO

To study the interactions of the alpha1S subunit of the skeletal muscle L-type Ca2+ channel with the skeletal beta1a and the cardiac beta2a, these subunits were expressed alone or in combination in tsA201 cells. Immunofluorescence- and green fluorescent protein-labeling showed that, when expressed alone, beta1a was diffusely distributed throughout the cytoplasm, beta2a was localized in the plasma membrane, and alpha1S was concentrated in a tubular/reticular membrane system, presumably the endoplasmic reticulum (ER). Upon coexpression with alpha1S, beta1a became colocalized with alpha1S in the ER. Upon coexpression with beta2a, alpha1S redistributed to the plasma membrane, where it aggregated in large clusters. Coexpression of alpha1S with beta1a but not with beta2a increased the frequency at which cells expressed L-type currents. A point mutation (alpha1S-Y366S) or deletion (alpha1S-Delta351-380) in the beta interaction domain of alpha1S blocked both translocation of beta1a to the ER and beta2a-induced translocation of the alpha1S mutants to the plasma membrane. However, the point mutation did not interfere with beta1a-induced current stimulation. Thus, beta1a and beta2a are differentially distributed in tsA201 cells and upon coexpression with alpha1S, form alpha1S. beta complexes in different cellular compartments. Complex formation but not current stimulation requires the intact beta interaction domain in the I-II cytoplasmic loop of alpha1S.


Assuntos
Canais de Cálcio/química , Canais de Cálcio/fisiologia , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Animais , Canais de Cálcio/biossíntese , Canais de Cálcio Tipo L , Linhagem Celular , Primers do DNA , Proteínas de Fluorescência Verde , Humanos , Rim , Proteínas Luminescentes/biossíntese , Substâncias Macromoleculares , Potenciais da Membrana , Mutagênese Sítio-Dirigida , Mutação Puntual , Reação em Cadeia da Polimerase , Multimerização Proteica , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Deleção de Sequência , Frações Subcelulares/metabolismo , Transfecção
7.
J Biol Chem ; 273(5): 2639-44, 1998 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-9446567

RESUMO

Five novel peptidyl inhibitors of Shaker-type (Kv1) K+ channels have been purified to homogeneity from venom of the scorpion Centruroides limbatus. The complete primary amino acid sequence of the major component, hongotoxin-1 (HgTX1), has been determined and confirmed after expression of the peptide in Escherichia coli. HgTX1 inhibits 125I-margatoxin binding to rat brain membranes as well as depolarization-induced 86Rb+ flux through homotetrameric Kv1.1, Kv1. 2, and Kv1.3 channels stably transfected in HEK-293 cells, but it displays much lower affinity for Kv1.6 channels. A HgTX1 double mutant (HgTX1-A19Y/Y37F) was constructed to allow high specific activity iodination of the peptide. HgTX1-A19Y/Y37F and monoiodinated HgTX1-A19Y/Y37F are equally potent in inhibiting 125I-margatoxin binding to rat brain membranes as HgTX1 (IC50 values approximately 0.3 pM). 125I-HgTX1-A19Y/Y37F binds with subpicomolar affinities to membranes derived from HEK-293 cells expressing homotetrameric Kv1.1, Kv1.2, and Kv1.3 channels and to rat brain membranes (Kd values 0.1-0.25 pM, respectively) but with lower affinity to Kv1.6 channels (Kd 9.6 pM), and it does not interact with either Kv1.4 or Kv1.5 channels. Several subpopulations of native Kv1 subunit oligomers that contribute to the rat brain HgTX1 receptor have been deduced by immunoprecipitation experiments using antibodies specific for Kv1 subunits. HgTX1 represents a novel and useful tool with which to investigate subclasses of voltage-gated K+ channels and Kv1 subunit assembly in different tissues.


Assuntos
Química Encefálica , Ativação do Canal Iônico , Canais de Potássio/química , Venenos de Escorpião/farmacologia , Vesículas Sinápticas/química , Sequência de Aminoácidos , Animais , Ligantes , Dados de Sequência Molecular , Neurotoxinas/farmacologia , Canais de Potássio/classificação , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Venenos de Escorpião/química , Venenos de Escorpião/genética , Venenos de Escorpião/metabolismo , Análise de Sequência , Homologia de Sequência de Aminoácidos , Superfamília Shaker de Canais de Potássio
8.
J Biol Chem ; 272(44): 27686-93, 1997 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-9346909

RESUMO

Pharmacological modulation by 1,4-dihydropyridines is a central feature of L-type calcium channels. Recently, eight L-type amino acid residues in transmembrane segments IIIS5, IIIS6, and IVS6 of the calcium channel alpha1 subunit were identified to substantially contribute to 1,4-dihydropyridine sensitivity. To determine whether these eight L-type residues (Thr1066, Gln1070, Ile1180, Ile1183, Tyr1490, Met1491, Ile1497, and Ile1498; alpha1C-a numbering) are sufficient to form a high affinity 1,4-dihydropyridine binding site in a non-L-type calcium channel, we transferred them to the 1, 4-dihydropyridine-insensitive alpha1A subunit using site-directed mutagenesis. 1,4-Dihydropyridine agonist and antagonist modulation of barium inward currents mediated by the mutant alpha1A subunits, coexpressed with alpha2delta and beta1a subunits in Xenopus laevis oocytes, was investigated with the two-microelectrode voltage clamp technique. The resulting mutant alpha1A-DHPi displayed low sensitivity for 1,4-dihydropyridines. Analysis of the 1,4-dihydropyridine binding region of an ancestral L-type alpha1 subunit previously cloned from Musca domestica body wall muscle led to the identification of Met1188 (alpha1C-a numbering) as an additional critical constituent of the L-type 1,4-dihydropyridine binding domain. The introduction of this residue into alpha1A-DHPi restored full sensitivity for 1,4-dihydropyridines. It also transferred functional properties considered hallmarks of 1, 4-dihydropyridine agonist and antagonist effects (i.e. stereoselectivity, voltage dependence of drug modulation, and agonist-induced shift in the voltage-dependence of activation). Our gain-of-function mutants provide an excellent model for future studies of the structure-activity relationship of 1, 4-dihydropyridines to obtain critical structural information for the development of drugs for neuronal, non-L-type calcium channels.


Assuntos
Aminoácidos/metabolismo , Canais de Cálcio/efeitos dos fármacos , Di-Hidropiridinas/farmacologia , Neurônios/efeitos dos fármacos , Sequência de Aminoácidos , Aminoácidos/genética , Animais , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Di-Hidropiridinas/metabolismo , Dados de Sequência Molecular , Mutagênese , Neurônios/metabolismo , Ligação Proteica , Coelhos , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Xenopus laevis
9.
Biophys J ; 73(1): 157-67, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9199780

RESUMO

The pore-forming alpha 1 subunit of L-type calcium (Ca2+) channels is the molecular target of Ca2+ channel blockers such as phenylalkylamines (PAAs). Association and dissociation rates of (-)devapamil were compared for a highly PAA-sensitive L-type Ca2+ channel chimera (Lh) and various class A Ca2+ channel mutants. These mutants carry the high-affinity determinants of the PAA receptor site in a class A sequence environment. Apparent drug association and dissociation rate constants were significantly affected by the sequence environment (class A or L-type) of the PAA receptor site. Single point mutations affecting the high-affinity determinants in segments IVS6 of the PAA receptor site, introduced into a class A environment, reduced the apparent drug association rates. Mutation I1811M in transmembrane segment IVS6 (mutant AL25/-I) had the highest impact and decreased the apparent association rate for (-)devapamil by approximately 30-fold, suggesting that this pore-lining isoleucine in transmembrane segment IVS6 plays a key role in the formation of the PAA receptor site. In contrast, apparent drug dissociation rates of Ca2+ channels in the resting state were almost unaffected by point mutations of the PAA receptor site.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Verapamil/análogos & derivados , Animais , Sítios de Ligação , Canais de Cálcio/química , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo L , Substâncias Macromoleculares , Potenciais da Membrana/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Deleção de Sequência , Verapamil/farmacologia , Xenopus
10.
J Biol Chem ; 271(48): 30330-5, 1996 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-8939992

RESUMO

The transmembrane segment IIIS5 of the L-type calcium channel alpha1 subunit participates in the formation of the 1,4-dihydropyridine (DHP) interaction domain (Grabner, M., Wang, Z., Hering, S., Striessnig, J., and Glossmann, H. (1996) Neuron 16, 207-218). We applied mutational analysis to identify amino acid residues within this segment that contribute to DHP sensitivity. DHP agonist and antagonist modulation of Ba2+ inward currents was assessed after coexpression of chimeric and mutant calcium channel alpha1 subunits with alpha2delta and beta1a subunits in Xenopus oocytes. Whereas DHP antagonists required Thr-1066, DHP agonist modulation crucially depended on the additional presence of Gln-1070 (numbering according to alpha1C-a), which also further increased the sensitivity to DHP antagonists. Asp-955, which is found at the corresponding position in the calcium channel alpha1S subunit from carp skeletal muscle, displayed functional similarity to Gln-1070 with respect to DHP interaction. We conclude that these residues (Thr-1066 plus Gln-1070 or Asp-955), which are located in close vicinity on the same side of the putative alpha-helix of transmembrane segment IIIS5, form a crucial DHP binding motif.


Assuntos
Bloqueadores dos Canais de Cálcio/química , Canais de Cálcio/química , Proteínas de Transporte/biossíntese , Di-Hidropiridinas/farmacologia , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Ativação do Canal Iônico , Glicoproteínas de Membrana/química , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Secundária de Proteína , Coelhos , Relação Estrutura-Atividade , Xenopus laevis
11.
J Biol Chem ; 271(40): 24471-5, 1996 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-8798706

RESUMO

To investigate the molecular basis of the calcium channel block by diltiazem, we transferred amino acids of the highly sensitive and stereoselective L-type (alpha1S or alpha1C) to a weakly sensitive, nonstereoselective class A (alpha1A) calcium channel. Transfer of three amino acids of transmembrane segment IVS6 of L-type alpha1 into the alpha1A subunit (I1804Y, S1808A, and M1811I) was sufficient to support a use-dependent block by diltiazem and by the phenylalkylamine (-)-gallopamil after expression in Xenopus oocytes. An additional mutation F1805M increased the sensitivity for (-)-gallopamil but not for diltiazem. Our data suggest that the receptor domains for diltiazem and gallopamil have common but not identical molecular determinants in transmembrane segment IVS6. These mutations also identified single amino acid residues in segment IVS6 that are important for class A channel inactivation.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Diltiazem/farmacologia , Sequência de Aminoácidos , Animais , Bário/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L , Galopamil/farmacologia , Transporte de Íons , Dados de Sequência Molecular , Alinhamento de Sequência , Xenopus
12.
Biochemistry ; 35(29): 9400-6, 1996 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-8755718

RESUMO

Full length L-type calcium channel alpha 1 subunits are rapidly phosphorylated by protein kinase A (PK-A) in vitro and in vivo at sites located in their long carboxyl terminal tails. In skeletal muscle, heart, and brain the majority of biochemically isolated alpha 1 subunits lacks these phosphorylation sites due to posttranslational proteolytic processing. Truncation may therefore modify the regulation of channel activity by PK-A. We combined site-directed mutagenesis and heterologous expression to investigate the extent to which putative cAMP-dependent phosphorylation sites in the C-terminus of alpha 1 subunits from skeletal muscle, heart, and brain are phosphorylated in vitro. The full length size form of wild-type and mutant calcium channel alpha 1 subunits was obtained at high yield after heterologous expression in Saccharomyces cerevisiae. Like in fetal rabbit myotubes [Rotman, E.I., et al. (1995) J. Biol. Chem. 270, 16371-16377], the rabbit skeletal muscle alpha 1 C-terminus was phosphorylated at serine residues 1757 and 1854. In the carboxyl terminus of alpha 1S from carp skeletal muscle and alpha 1C from rabbit heart a single serine residue was phosphorylated by PK-A in vitro. The C-terminus of alpha 1D was phosphorylated at more than one site. Employing deletion mutants, most of the phosphorylation ( > 70%) was found to occur between amino acid residues 1805 and 2072. Serine 1743 was identified as additional phosphorylation site in alpha 1D. We conclude that in class S and C calcium channels the most C-terminal phosphorylation sites are substrate for PK-A in vitro, whereas in class D calcium channels phosphorylation also occurs at a site which is likely to be retained even after posttranslational truncation.


Assuntos
Canais de Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Encéfalo/enzimologia , Canais de Cálcio/química , Carpas/metabolismo , Clonagem Molecular , AMP Cíclico/farmacologia , Primers do DNA , Immunoblotting , Dados de Sequência Molecular , Peso Molecular , Músculo Esquelético/enzimologia , Mutagênese Sítio-Dirigida , Miocárdio/enzimologia , Fosforilação , Coelhos , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Serina/metabolismo
13.
Biochemistry ; 34(29): 9350-5, 1995 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-7626604

RESUMO

The molecular determinants for Ca2+ modulation of dihydropyridine (DHP) binding to cardiac Ca2+ channels were identified by mutational neutralization of the glutamate residues that comprise the Ca2+ channel selectivity filter. The binding activity of the DHP (+)-[3H]isradipine, monitored after expression of wild-type and mutant alpha 1 subunits in COS-7 cells, was markedly reduced in four single mutants and a double mutant. Evidence for decreased Ca2+ affinity was obtained for two single mutants in kinetic and equilibrium binding studies. Mutational destabilization of Ca2+ binding resulted in a concomitant decrease of (+)-[3H]isradipine binding affinity. Recovery of (+)-[3H]isradipine binding activity by the allosteric modulator (+)-tetrandrine in two single mutants was associated with a recovery of Ca2+ and DHP binding kinetics to wild-type values. Our findings demonstrate that high-affinity DHP binding is dependent on Ca2+ coordination by glutamate residues which form the selectivity filter of the channel pore.


Assuntos
Benzilisoquinolinas , Canais de Cálcio/química , Canais de Cálcio/metabolismo , Di-Hidropiridinas/metabolismo , Miocárdio/metabolismo , Estrutura Secundária de Proteína , Alcaloides/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Carpas , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Chlorocebus aethiops , Primers do DNA , Feminino , Rim , Cinética , Substâncias Macromoleculares , Potenciais da Membrana , Modelos Estruturais , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transfecção , Xenopus laevis/metabolismo
14.
Biochem Pharmacol ; 49(5): 603-9, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7887974

RESUMO

It has previously been shown that dexniguldipine-HCl (B8509-035) is a potent chemosensitizer in multidrug resistant cells [Hofmann et al., J Cancer Res Clin Oncol 118: 361-366, 1992]. It is shown here that dexniguldipine-HCl causes a dose-dependent reduction of the labeling of the P-glycoprotein by azidopine, indicating a competition of dexniguldipine-HCl with the photoaffinity label for the multidrug resistance gene 1 (MDR-1) product. Exposure to dexniguldipine-HCl results in a dose-dependent accumulation of rhodamine 123 in MDR-1 overexpressing cells. In the presence of 1 microM dexniguldipine-HCl, rhodamine 123 accumulated in multidrug resistant cells to similar levels as in the sensitive parental cell lines. At this concentration, dexniguldipine-HCl enhances the cytotoxicities of Adriamycin and vincristine. The resistance modulating factors (RMF), i.e. IC50 drug/IC50 drug + modulator, were found to be proportional to the expression of MDR-1, ranging from 8 to 42 for Adriamycin and from 16 to 63 for vincristine. Transfection with the MDR-1 gene was found to be sufficient to sensitize cells to the modulation by dexniguldipine-HCl. The compound does not affect the expression of the MDR-1 gene. Dexniguldipine-HCl has no effect on a multidrug resistant phenotype caused by a mutation of topoisomerase II. It is concluded that dexniguldipine-HCl modulates multidrug resistance by direct interaction with the P-glycoprotein.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Di-Hidropiridinas/farmacologia , Resistência a Múltiplos Medicamentos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Marcadores de Afinidade , Azidas/metabolismo , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA Topoisomerases Tipo II/genética , Di-Hidropiridinas/metabolismo , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Interações Medicamentosas , Resistência a Múltiplos Medicamentos/genética , Humanos , Rodamina 123 , Rodaminas/metabolismo , Transfecção , Vincristina/farmacologia
15.
J Biol Chem ; 269(38): 23668-74, 1994 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-8089136

RESUMO

The primary structure of a calcium channel beta subunit (beta M) from housefly (Musca domestica) has been deduced by cDNA cloning and sequence analysis. The open reading frame encodes a 441-amino acid polypeptide with a calculated molecular mass of 48,755 Da. Whole-mount in situ hybridization indicates that beta M mRNA is predominantly expressed in neuronal tissues. Transcription of beta M mRNA is evident from stage 13/14 of embryogenesis up to adulthood. Different expression patterns of splice variants were found in larvae and in adult fly heads. Amino acid identity between beta M and mammalian beta subunits is lower (66-68%) than within mammalian beta subunits (74-80%). Calculation of a phylogenetic tree indicates that beta M is an ancestral form of the four distinct beta subunit gene products identified in mammalian tissues so far. Despite these sequence differences, beta M is able to enhance endogenous calcium channel activity in Xenopus laevis oocytes as well as dihydropyridine binding to membranes from COS 7 cells transfected with rabbit heart alpha 1 cDNA in the same manner as was previously shown for mammalian beta subunits.


Assuntos
Canais de Cálcio/genética , Dípteros/genética , Proteínas do Tecido Nervoso/genética , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Canais de Cálcio/química , Canais de Cálcio/efeitos dos fármacos , Clonagem Molecular , Primers do DNA/química , Di-Hidropiridinas/farmacologia , Condutividade Elétrica , Larva , Dados de Sequência Molecular , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Solubilidade
16.
FEBS Lett ; 352(2): 141-5, 1994 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-7925963

RESUMO

A Ca2+ channel alpha 1-subunit derived from rabbit heart was transiently expressed in COS-7 cells. The dihydropyridine (+)-isradipine had low affinity (Ki = 34.3 nM) for the alpha 1-subunit in the absence of the beta-subunit due to rapid dissociation (k-1 = 0.11 min-1). Co-expression of the beta-subunit resulted in a > 35-fold increase in (+)-isradipine binding affinity (Ki = 0.9 nM) due to decreased dissociation (k-1 of 0.007 min-1). Higher DHP binding affinity was associated with an increase of the apparent affinity of Ca2+ ions for the channel. Our data suggest that the beta-subunit affects the coordination of Ca2+ ions with sites that are coupled to the dihydropyridine binding domain and by this mechanism increases the affinity for these ligands.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Di-Hidropiridinas/metabolismo , Animais , Sítios de Ligação , Canais de Cálcio/química , Canais de Cálcio/genética , Linhagem Celular , DNA Complementar , Expressão Gênica , Cinética , Miocárdio/metabolismo , Transfecção
17.
J Biol Chem ; 265(1): 363-70, 1990 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-2152924

RESUMO

The purified dihydropyridine-sensitive calcium channel from skeletal muscle transverse tubules consists of several subunits, termed alpha 1, alpha 2, beta, gamma and delta. From its associated drug receptors, those for 1,4-dihydropyridines and phenylalkylamines have been shown previously by photoaffinity labeling to reside on the alpha 1 subunit. In the present study the arylazide photo-affinity ligand, (+)-cis-azidodiltiazem ((+)-cis-(2S,3S)-5-[2-(4- azidobenzoyl)aminoethyl]-2,3,4,5-tetrahydro-3-hydroxy-2-(4-methoxyphenyl )-4- oxo-1,5-benzothiazepine), and the respective tritiated derivative, (+)-cis-[3H]azidodiltiazem (45 Ci/mmol), were developed to identify directly the benzothiazepine binding subunit. (+)-cis-Azidodiltiazem binds competitively to the benzothiazepine receptor in rabbit skeletal muscle transverse tubule membranes. Upon ultraviolet irradiation of the (+)-cis-[3H]azidodiltiazem-purified calcium channel complex, the ligand photoincorporates exclusively into the alpha 1 subunit. Photoincorporation is protected by 100 microM (-)-desmethoxyverapamil and 100 microM (+)-cis-diltiazem. A polyclonal antiserum directed against (+)-cis-azidodiltiazem was employed to detect (+)-cis-azidodiltiazem immunoreactivity photoincorporated into the purified calcium channel complex, confirming the exclusive labeling of the alpha 1 subunit. Our data provide direct evidence that, together with the drug receptors for 1,4-dihydropyridines and phenylalkylamines, the benzothiazepine binding domain of skeletal muscle calcium channels is located on the alpha 1 subunit. We conclude that our anti-ligand antibodies could be used successfully to affinity purify the photolabeled proteolytic fragments of the alpha 1 subunit which are expected to form part of the benzothiazepine binding domain.


Assuntos
Azidas , Canais de Cálcio/análise , Músculos/análise , Receptores de Droga/análise , Tiazepinas , Marcadores de Afinidade , Animais , Azidas/metabolismo , Ligação Competitiva , Canais de Cálcio/metabolismo , Ensaio de Imunoadsorção Enzimática , Substâncias Macromoleculares , Fotoquímica , Coelhos , Receptores de Droga/metabolismo , Receptores Nicotínicos/metabolismo , Tiazepinas/metabolismo , Trítio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...