Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 168(2): 432-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22891655

RESUMO

BACKGROUND AND PURPOSE: The K(Ca) 3.1 channel is a potential target for therapy of immune disease. We identified a compound from a new chemical class of K(Ca) 3.1 inhibitors and assessed in vitro and in vivo inhibition of immune responses. EXPERIMENTAL APPROACH: We characterized the benzothiazinone NS6180 (4-[[3-(trifluoromethyl)phenyl]methyl]-2H-1,4-benzothiazin-3(4H)-one) with respect to potency and molecular site of action on K(Ca) 3.1 channels, selectivity towards other targets, effects on T-cell activation as well as pharmacokinetics and inflammation control in colitis induced by 2,4-dinitrobenzene sulfonic acid, a rat model of inflammatory bowel disease (IBD). KEY RESULTS: NS6180 inhibited cloned human K(Ca) 3.1 channels (IC(50) = 9 nM) via T250 and V275, the same amino acid residues conferring sensitivity to triarylmethanes such as like TRAM-34. NS6180 inhibited endogenously expressed K(Ca) 3.1 channels in human, mouse and rat erythrocytes, with similar potencies (15-20 nM). NS6180 suppressed rat and mouse splenocyte proliferation at submicrolar concentrations and potently inhibited IL-2 and IFN-γ production, while exerting smaller effects on IL-4 and TNF-α and no effect on IL-17 production. Antibody staining showed K(Ca) 3.1 channels in healthy colon and strong up-regulation in association with infiltrating immune cells after induction of colitis. Despite poor plasma exposure, NS6180 (3 and 10 mg·kg(-1) b.i.d.) dampened colon inflammation and improved body weight gain as effectively as the standard IBD drug sulfasalazine (300 mg·kg(-1) q.d.). CONCLUSIONS AND IMPLICATIONS: NS6180 represents a novel class of K(Ca) 3.1 channel inhibitors which inhibited experimental colitis, suggesting K(Ca) 3.1 channels as targets for pharmacological control of intestinal inflammation.


Assuntos
Doenças Inflamatórias Intestinais/tratamento farmacológico , Bloqueadores dos Canais de Potássio/uso terapêutico , Tiazinas/uso terapêutico , Animais , Dinitrofluorbenzeno/análogos & derivados , Modelos Animais de Doenças , Eritrócitos/efeitos dos fármacos , Eritrócitos/fisiologia , Humanos , Inflamação/tratamento farmacológico , Doenças Inflamatórias Intestinais/imunologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Bloqueadores dos Canais de Potássio/sangue , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Wistar , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tiazinas/sangue , Tiazinas/farmacologia
2.
J Pharmacol Exp Ther ; 328(1): 28-39, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18820135

RESUMO

Calcium-activated potassium channels are attractive targets for the development of therapeutics for overactive bladder. In the current study, we addressed the role of calcium-activated potassium channels of small (SK; K(Ca)2) and intermediate (IK; K(Ca)3) conductance in bladder function pharmacologically. We identified and characterized a novel positive modulator of SK/IK channels, 4,5-dichloro-1,3-diethyl-1,3-dihydro-benzoimidazol-2-one (NS4591). In whole-cell patch-clamp experiments, NS4591 doubled IK-mediated currents at a concentration of 45 +/- 6 nM(n = 16), whereas 530 +/- 100 nM (n = 7) was required for doubling of SK3-mediated currents. In acutely dissociated bladder primary afferent neurons, the presence of SK channels was verified using apamin and 1-ethyl-2-benzimidazolinone. In these neurons, NS4591 (10 microM) inhibited the number of action potentials generated by suprathreshold depolarizing pulses. NS4591 also reduced carbachol-induced twitches in rat bladder detrusor rings in an apamin-sensitive manner. In vivo, NS4591 (30 mg/kg) inhibited bladder overactivity in rats and cats induced by capsaicin and acetic acid, respectively. In conclusion, the present study supports the involvement of calcium-activated potassium channels in bladder function and identifies NS4591 as a potent modulator of IK and SK channels that is effective in animal models of bladder overactivity.


Assuntos
Vias Aferentes/efeitos dos fármacos , Benzimidazóis/farmacologia , Canais de Cloreto/fisiologia , Neurônios/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Bexiga Urinária/inervação , Bexiga Urinária/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Cloreto de Cálcio/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Canais de Cloreto/efeitos dos fármacos , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Humanos , Rim , Cloreto de Magnésio/farmacologia , Neurônios/efeitos dos fármacos , Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Bexiga Urinária/efeitos dos fármacos , Micção/efeitos dos fármacos , Micção/fisiologia
3.
Br J Pharmacol ; 151(5): 655-65, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17486140

RESUMO

BACKGROUND AND PURPOSE: Positive modulators of small conductance Ca(2+)-activated K(+) channels (SK1, SK2, and SK3) exert hyperpolarizing effects that influence the activity of excitable and non-excitable cells. The prototype compound 1-EBIO or the more potent compound NS309, do not distinguish between the SK subtypes and they also activate the related intermediate conductance Ca(2+)-activated K(+) channel (IK). This paper demonstrates, for the first time, subtype-selective positive modulation of SK channels. EXPERIMENTAL APPROACH: Using patch clamp and fluorescence techniques we studied the effect of the compound cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA) on recombinant hSK1-3 and hIK channels expressed in HEK293 cells. CyPPA was also tested on SK3 and IK channels endogenously expressed in TE671 and HeLa cells. KEY RESULTS: CyPPA was found to be a positive modulator of hSK3 (EC(50) = 5.6 +/- 1.6 microM, efficacy 90 +/- 1.8 %) and hSK2 (EC(50) = 14 +/- 4 microM, efficacy 71 +/- 1.8 %) when measured in inside-out patch clamp experiments. CyPPA was inactive on both hSK1 and hIK channels. At hSK3 channels, CyPPA induced a concentration-dependent increase in the apparent Ca(2+)-sensitivity of channel activation, changing the EC(50)(Ca(2+)) from 429 nM to 59 nM. CONCLUSIONS AND IMPLICATIONS: As a pharmacological tool, CyPPA may be used in parallel with the IK/SK openers 1-EBIO and NS309 to distinguish SK3/SK2- from SK1/IK-mediated pharmacological responses. This is important for the SK2 and SK1 subtypes, since they have overlapping expression patterns in the neocortical and hippocampal regions, and for SK3 and IK channels, since they co-express in certain peripheral tissues.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Algoritmos , Benzimidazóis/farmacologia , Linhagem Celular , Eletrofisiologia , Corantes Fluorescentes , Humanos , Indóis/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Oximas/farmacologia , Técnicas de Patch-Clamp , Tálio
4.
J Pharmacol Exp Ther ; 314(1): 282-92, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15814569

RESUMO

Neuronal Kv7 channels are recognized as potential drug targets for treating hyperexcitability disorders such as pain, epilepsy, and mania. Hyperactivity of the amygdala has been described in clinical and preclinical studies of anxiety, and therefore, neuronal Kv7 channels may be a relevant target for this indication. In patch-clamp electrophysiology on cell lines expressing Kv7 channel subtypes, Maxipost (BMS-204352) exerted positive modulation of all neuronal Kv7 channels, whereas its R-enantiomer was a negative modulator. By contrast, at the Kv7.1 and the large conductance Ca2+-activated potassium channels, the two enantiomers showed the same effect, namely, negative and positive modulation at the two channels, respectively. At GABA(A) receptors (alpha1beta2gamma2s and alpha2beta2gamma2s) expressed in Xenopus oocytes, BMS-204352 was a negative modulator, and the R-enantiomer was a positive modulator. The observation that the S- and R-forms exhibited opposing effects on neuronal Kv7 channel subtypes allowed us to assess the potential role of Kv7 channels in anxiety. In vivo, BMS-204352 (3-30 mg/kg) was anxiolytic in the mouse zero maze and marble burying models of anxiety, with the effect in the burying model antagonized by the R-enantiomer (3 mg/kg). Likewise, the positive Kv7 channel modulator retigabine was anxiolytic in both models, and its effect in the burying model was blocked by the Kv7 channel inhibitor 10,10-bis-pyridin-4-ylmethyl-10H-anthracen-9-one (XE-991) (1 mg/kg). Doses at which BMS-204352 and retigabine induce anxiolysis could be dissociated from effects on sedation or memory impairment. In conclusion, these in vitro and in vivo studies provide compelling evidence that neuronal Kv7 channels are a target for developing novel anxiolytics.


Assuntos
Ansiolíticos/farmacologia , Carbamatos/farmacologia , Indóis/farmacologia , Neurônios/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/agonistas , Algoritmos , Animais , Ansiolíticos/química , Ansiedade/fisiopatologia , Ansiedade/psicologia , Ataxia/induzido quimicamente , Comportamento Animal/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Emoções/efeitos dos fármacos , Comportamento Exploratório/efeitos dos fármacos , Feminino , Força da Mão , Indóis/química , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Masculino , Potenciais da Membrana , Camundongos , Oócitos/efeitos dos fármacos , Técnicas de Patch-Clamp , RNA Complementar/biossíntese , Ratos , Ratos Wistar , Receptores de GABA-A/efeitos dos fármacos , Receptores de GABA-A/genética , Estereoisomerismo , Xenopus laevis
5.
J Membr Biol ; 196(2): 83-94, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14724745

RESUMO

The endogenous volume-regulated anion channel (VRAC) from HEK293 cells was pharmacologically characterized using the whole-cell patch-clamp technique. Under isotonic conditions a small (1.3 nS), Ca(2+)-independent Cl conductance was measured. However, swelling at 75% tonicity activated a VRAC identified as an outward-rectifying anion current ( P(l) > P(Cl) > P(gluconate)), which was ATP-dependent and showed inactivation at positive potentials. Activation of this current followed a sigmoid time course, reaching a plateau conductance of 42.6 nS after 12-15 min ( t(1/2) = 7 min). The pharmacology of this VRAC was investigated using standard Cl(-)-channel blockers (NPPB, DIDS, and tamoxifen) as well as a new group (acidic di-aryl ureas) of Cl(-)-channel blockers (NS1652, NS3623, NS3749, and NS3728). The acidic di-aryl ureas were originally synthezised for inhibition of the human erythrocyte Cl(-) conductance in vivo. NS3728 was the most potent VRAC blocker in this series ( IC(50) = 0.40 micro M) and even more potent than tamoxifen (2.2 micro M). NS3728 accelerated channel inactivation at positive potentials. These results show that acidic di-aryl ureas constitute a promising starting point for the synthesis of potent inhibitors of VRAC.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/fisiologia , Ativação do Canal Iônico/fisiologia , Rim/fisiologia , Potenciais da Membrana/fisiologia , Compostos de Fenilureia/farmacologia , Benzoatos/farmacologia , Linhagem Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/fisiologia , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/embriologia , Potenciais da Membrana/efeitos dos fármacos , Tetrazóis/farmacologia , Ureia/farmacologia
6.
Curr Drug Targets ; 2(4): 401-22, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11732639

RESUMO

This review discusses the Ca2+-activated K+ channels of intermediate conductance (IK channels), and their historical discovery in erythrocytes, their classical biophysical characteristics, physiological function, molecular biology as well as their role as possible molecular targets for pharmacological intervention in various diseases. The first described Ca2+-activated K+ channel ever - the so-called Gard6s channel from human erythrocytes--is an IK channel. The "I" denominates the intermediate conductance that distinguishes the IK channels from the related Ca2+-activated K+ channels of small (SK) or large (BK) conductance. The recent cloning of the human IK channel gene (KCNN4) enabled a detailed mapping of the expression in various tissues. IK channel expression is found predominantly in cells of the blood, in epithelia and endothelia. An important physiological role of IK channels is to set the membrane potential at fairly negative values and thereby to build up large electrical gradients for the passive transport of ions such as Cl- efflux driving water and Na+ secretion from epithelia, and Ca2+ influx controlling T-lymphocyte proliferation. The molecular cloning of IK and SK channels has revealed that both channels gain their Ca2+-sensitivity from tightly bound calmodulin (CaM). The IK channel is potently blocked by the scorpion toxin charybdotoxin (ChTx) and the antimycotic clotrimazole (CLT). CLT has been in clinical trials for the treatment of sickle cell disease, diarrhea and ameliorates the symptoms of rheumatoid arthritis. However, inhibition of cytochrome P450 enzymes by CLT limits its therapeutic value, but new drug candidates are entering the stage. It is discussed whether pharmacological modulation of IK channels may be beneficial in sickle cell anemia, cystic fibrosis, secretory diarrhea, craft-versus-host disease and autoimmune diseases.


Assuntos
Cálcio/metabolismo , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Sequência de Aminoácidos , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Clonagem Molecular , Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Diarreia/tratamento farmacológico , Diarreia/metabolismo , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Eritrócitos/metabolismo , Humanos , Imunossupressores/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Ativação do Canal Iônico/efeitos dos fármacos , Dados de Sequência Molecular , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos
7.
Neuropharmacology ; 40(7): 888-98, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11378159

RESUMO

Activation of potassium channels generally reduces cellular excitability, making potassium channel openers potential drug candidates for the treatment of diseases related to hyperexcitabilty such as epilepsy, neuropathic pain, and neurodegeneration. Two compounds, BMS-204352 and retigabine, presently in clinical trials for the treatment of stroke and epilepsy, respectively, have been proposed to exert their protective action via an activation of potassium channels. Here we show that KCNQ4 channels, stably expressed in HEK293 cells, were activated by retigabine and BMS-204352 in a reversible and concentration-dependent manner in the concentration range 0.1-10 microM. Both compounds shifted the KCNQ4 channel activation curves towards more negative potentials by about 10 mV. Further, the maximal current obtainable at large positive voltages was also increased concentration-dependently by both compounds. Finally, a pronounced slowing of the deactivation kinetics was induced in particular by BMS-204352. The M-current blocker linopirdine inhibited the baseline current, as well as the BMS-204352-induced activation of the KCNQ4 channels. KCNQ2, KCNQ2/Q3, and KCNQ3/Q4 channels were activated to a similar degree as KCNQ4 channels by 10 microM of BMS-204352 and retigabine, respectively. The compounds are, thus, likely to be general activators of M-like currents.


Assuntos
Anticonvulsivantes/farmacologia , Carbamatos/farmacologia , Indóis/farmacologia , Fenilenodiaminas/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/efeitos dos fármacos , Anticonvulsivantes/química , Carbamatos/química , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Indóis/química , Canais de Potássio KCNQ , Fenilenodiaminas/química , Canais de Potássio/fisiologia
8.
J Biol Chem ; 276(13): 9868-76, 2001 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-11136720

RESUMO

The isolation of the peptide inhibitor of M-type K(+) current, BeKm-1, from the venom of the Central Asian scorpion Buthus eupeus has been described previously (Fillipov A. K., Kozlov, S. A., Pluzhnikov, K. A., Grishin, E. V., and Brown, D. A. (1996) FEBS Lett. 384, 277-280). Here we report the cloning, expression, and selectivity of BeKm-1. A full-length cDNA of 365 nucleotides encoding the precursor of BeKm-1 was isolated using the rapid amplification of cDNA ends polymerase chain reaction technique from mRNA obtained from scorpion telsons. Sequence analysis of the cDNA revealed that the precursor contains a signal peptide of 21 amino acid residues. The mature toxin consists of 36 amino acid residues. BeKm-1 belongs to the family of scorpion venom potassium channel blockers and represents a new subgroup of these toxins. The recombinant BeKm-1 was produced as a Protein A fusion product in the periplasm of Escherichia coli. After cleavage and high performance liquid chromatography purification, recombinant BeKm-1 displayed the same properties as the native toxin. Three BeKm-1 mutants (R27K, F32K, and R27K/F32K) were generated, purified, and characterized. Recombinant wild-type BeKm-1 and the three mutants partly inhibited the native M-like current in NG108-15 at 100 nm. The effect of the recombinant BeKm-1 on different K(+) channels was also studied. BeKm-1 inhibited hERG1 channels with an IC(50) of 3.3 nm, but had no effect at 100 nm on hEAG, hSK1, rSK2, hIK, hBK, KCNQ1/KCNE1, KCNQ2/KCNQ3, KCNQ4 channels, and minimal effect on rELK1. Thus, BeKm-1 was shown to be a novel specific blocker of hERG1 potassium channels.


Assuntos
Proteínas de Transporte de Cátions , Proteínas de Ligação a DNA , Bloqueadores dos Canais de Potássio , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Venenos de Escorpião/biossíntese , Venenos de Escorpião/química , Venenos de Escorpião/genética , Transativadores , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Clonagem Molecular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Eletrofisiologia , Escherichia coli/metabolismo , Canais de Potássio Éter-A-Go-Go , Humanos , Concentração Inibidora 50 , Canais de Potássio KCNQ , Canal de Potássio KCNQ1 , Cinética , Espectrometria de Massas , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Fases de Leitura Aberta , Reação em Cadeia da Polimerase , Canais de Potássio/metabolismo , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Escorpiões , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Fatores de Tempo , Regulador Transcricional ERG , Células Tumorais Cultivadas
9.
Pflugers Arch ; 440(6): 809-18, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11041545

RESUMO

This study demonstrated that the methylxanthines, theophylline, IBMX and caffeine, activate the human, intermediate-conductance, Ca2+-activated K+ channel (hIK) stably expressed in HEK-293 cells. Whole-cell voltage-clamp experiments showed that the hIK current increased reversibly and voltage independently after the addition of methylxanthines. In current-clamp experiments, theophylline dose-dependently hyperpolarised the cell membrane from a resting potential of -18 mV to -56 mV. The methylxanthines did not affect large-conductance (BK) or small-conductance (SK2), Ca2+-activated K+ channels, demonstrating that the effects were not secondary to a rise in intracellular Ca2+. However, the activation of hIK by theophylline required an intracellular [Ca2+] above 30 nM. The hIK current was insensitive to 8-bromoadenosine cyclic 3',5'-monophosphate (8-bromo-cAMP), forskolin, 8-bromoguanosine cyclic 3',5'-monophosphate (8-bromo-cGMP) and sodium nitroprusside. Moreover, in the presence of inhibitors of protein kinase A (PKA) or protein kinase G (PKG) theophylline still activated the current. Finally, mutation of the putative PKA/PKG consensus phosphorylation site (Ser334) had no effect on the theophylline-induced activation of hIK. Since the observed activation is independent of changes in PKA/PKG-phosphorylation and of fluctuations in intracellular Ca2+, we suggest that the methylxanthines interact directly with the hIK protein.


Assuntos
Cálcio/farmacologia , GMP Cíclico/análogos & derivados , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Xantinas/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Cafeína/farmacologia , Cálcio/metabolismo , Linhagem Celular , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , GMP Cíclico/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Condutividade Elétrica , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Humanos , Magnésio/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Nitroprussiato/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/genética , Proteínas Recombinantes/metabolismo , Teofilina/administração & dosagem , Teofilina/farmacologia , Transfecção
10.
Br J Pharmacol ; 129(5): 991-9, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10696100

RESUMO

Three genes encode the small-conductance Ca(2+)-activated K(+) channels (SK channels). We have stably expressed hSK1 and rSK2 in HEK 293 cells and addressed the pharmacology of these subtypes using whole-cell patch clamp recordings. The bee venom peptide apamin blocked hSK1 as well as rSK2 with IC(50) values of 3.3 nM and 83 pM, respectively. The pharmacological separation between the subtypes was even more prominent when applying the scorpion peptide blocker scyllatoxin, which blocked hSK1 with an IC(50) value of 80 nM and rSK2 at 287 pM. The potent small molecule blockers showed little differentiation between the channel subtypes. The bis-quinolinium cyclophane UCL 1684 blocked hSK1 with an IC(50) value of 762 pM and rSK2 at 364 pM. The antiseptic compound dequalinium chloride blocked hSK1 and rSK2 with IC(50) values of 444 nM and 162 nM, respectively. The nicotinic acetylcholine receptor antagonist d-tubocurarine was found to block hSK1 and rSK2 with IC(50) values of 27 microM and 17 microM when measured at +80 mV. The inhibition by d-tubocurarine was voltage-dependent with increasing affinities at more hyperpolarized potentials. The GABA(A) receptor antagonist bicuculline methiodide also blocked hSK1 and rSK2 in a voltage-dependent manner with IC(50) values of 15 and 25 microM when measured at +80 mV. In conclusion, the pharmacological separation between SK channel subtypes expressed in mammalian cells is too small to support the notion that the apamin-insensitive afterhyperpolarization of neurones is mediated by hSK1.


Assuntos
Canais de Potássio Cálcio-Ativados , Canais de Potássio/efeitos dos fármacos , Sequência de Aminoácidos , Apamina/farmacologia , Bicuculina/farmacologia , Linhagem Celular , Clonagem Molecular , Eletrofisiologia , Antagonistas GABAérgicos/farmacologia , Humanos , Cinética , Dados de Sequência Molecular , Fármacos Neuromusculares não Despolarizantes/farmacologia , Técnicas de Patch-Clamp , Plasmídeos , Canais de Potássio/biossíntese , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Toxinas Biológicas/farmacologia , Tubocurarina/farmacologia
11.
Biochim Biophys Acta ; 1420(1-2): 231-40, 1999 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-10446306

RESUMO

Modulation of the cloned human intermediate-conductance Ca(2+)-activated K(+) channel (hIK) by the compound 1-ethyl-2-benzimidazolinone (EBIO) was studied by patch-clamp technique using human embryonic kidney cells (HEK 293) stably expressing the hIK channels. In whole-cell studies, intracellular concentrations of free Ca(2+) were systematically varied, by buffering the pipette solutions. In voltage-clamp, the hIK specific currents increased gradually from 0 to approximately 300 pA/pF without reaching saturation even at the highest Ca(2+) concentration tested (300 nM). In the presence of EBIO (100 microM), the Ca(2+)-activation curve was shifted leftwards, and maximal currents were attained at 100 nM Ca(2+). In current-clamp, steeply Ca(2+)-dependent membrane potentials were recorded and the cells gradually hyperpolarised from -20 to -85 mV when Ca(2+) was augmented from 0 to 300 nM. EBIO strongly hyperpolarised cells buffered at intermediate Ca(2+) concentrations. In contrast, no effects were detected either below 10 nM (no basic channel activation) or at 300 nM Ca(2+) (V(m) close to E(K)). Without Ca(2+), EBIO-induced hyperpolarisations were not obtainable, indicating an obligatory Ca(2+)-dependent mechanism of action. When applied to inside-out patches, EBIO exerted a Ca(2+)-dependent increase in the single-channel open-state probability, showing that the compound modulates hIK channels by a direct action on the alpha-subunit or on a closely associated protein. In conclusion, EBIO activates hIK channels in whole-cell and inside-out patches by a direct mechanism, which requires the presence of internal Ca(2+).


Assuntos
Benzimidazóis/farmacologia , Cálcio/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Linhagem Celular , Humanos , Potenciais da Membrana , Técnicas de Patch-Clamp , Canais de Potássio/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
13.
Am J Physiol ; 275(3): C848-56, 1998 09.
Artigo em Inglês | MEDLINE | ID: mdl-9730970

RESUMO

The human intermediate-conductance, Ca2+-activated K+ channel (hIK) was identified by searching the expressed sequence tag database. hIK was found to be identical to two recently cloned K+ channels, hSK4 and hIK1. RNA dot blot analysis showed a widespread tissue expression, with the highest levels in salivary gland, placenta, trachea, and lung. With use of fluorescent in situ hybridization and radiation hybrid mapping, hIK mapped to chromosome 19q13.2 in the same region as the disease Diamond-Blackfan anemia. Stable expression of hIK in HEK-293 cells revealed single Ca2+-activated K+ channels exhibiting weak inward rectification (30 and 11 pS at -100 and +100 mV, respectively). Whole cell recordings showed a noninactivating, inwardly rectifying K+ conductance. Ionic selectivity estimated from bi-ionic reversal potentials gave the permeability (PK/PX) sequence K+ = Rb+ (1.0) > Cs+ (10.4) >> Na+, Li+, N-methyl-D-glucamine (>51). NH+4 blocked the channel completely. hIK was blocked by the classical inhibitors of the Gardos channel charybdotoxin (IC50 28 nM) and clotrimazole (IC50 153 nM) as well as by nitrendipine (IC50 27 nM), Stichodactyla toxin (IC50 291 nM), margatoxin (IC50 459 nM), miconazole (IC50 785 nM), econazole (IC50 2.4 microM), and cetiedil (IC50 79 microM). Finally, 1-ethyl-2-benzimidazolinone, an opener of the T84 cell IK channel, activated hIK with an EC50 of 74 microM.


Assuntos
Cromossomos Humanos Par 19 , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Azepinas/farmacologia , Benzimidazóis/farmacologia , Cálcio/metabolismo , Linhagem Celular , Charibdotoxina/farmacologia , Mapeamento Cromossômico , Clonagem Molecular , Clotrimazol/farmacologia , Venenos de Cnidários/farmacologia , Humanos , Hibridização in Situ Fluorescente , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Cetoconazol/farmacologia , Rim , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miconazol/farmacologia , Neurotoxinas/farmacologia , Nitrendipino/farmacologia , Especificidade de Órgãos , Canais de Potássio/biossíntese , Canais de Potássio/genética , Proteínas Recombinantes/biossíntese , Venenos de Escorpião , Transfecção
14.
FEBS Lett ; 415(1): 67-70, 1997 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-9326371

RESUMO

We have generated HEK293 cell lines stably expressing high levels of either the human BK channel alpha-subunit alone or the BK channel alpha-subunit and beta-subunit together. For co-expression a plasmid with three expression cassettes was constructed. Patch-clamp recordings on inside-out patches from the transfected cells resulted in macroscopic currents reflecting the expression of 200-800 BK channels per patch. No decrease in channel expression could be detected in cells grown for more than 50 passages. The alpha-subunit when expressed alone conducted currents which were sensitive to intracellular Ca2+ in the physiological range. In the presence of the beta-subunit the steady-state activation curves were shifted by -20 to -30 mV and channel deactivation kinetics were slowed. The BK channel opener NS1608 (10 microM) shifted the steady-state activation curves for the alpha-subunit as well as for the alphabeta-subunits by -40 to -50 mV.


Assuntos
Canais de Potássio Cálcio-Ativados , Canais de Potássio/genética , Cálcio/metabolismo , Cálcio/farmacologia , Linhagem Celular , Eletrofisiologia , Regulação da Expressão Gênica , Humanos , Indóis/farmacologia , Cinética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta , Técnicas de Patch-Clamp , Compostos de Fenilureia/farmacologia , Plasmídeos , Reação em Cadeia da Polimerase , Canais de Potássio/metabolismo , Conformação Proteica , Transfecção
15.
Am J Physiol ; 271(5 Pt 1): C1463-71, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8944628

RESUMO

Coronary artery smooth muscle cells express G protein-coupled purinoceptors, and we report here for the first time how receptor activation by extracellular ATP influences cell membrane currents and membrane potential in human cells. ATP (100 microM) stimulated a triphasic change in membrane potential lasting several seconds, which was caused by sequential opening of transient inward and outward conductances. The inward current was carried by Cl- and the outward current by K+, as shown by ion substitution and changes in holding potential. Both currents were independent of the presence of external Ca2+ but were blocked by strong buffering of Ca2+ in the internal solution. The P2u- and P2y-purinoceptor agonists UTP and 2-methylthioadenosine 5'-triphosphate activated similar currents, whereas the P2x-receptor agonist alpha, beta-methyleneadenosine 5'-triphosphate and the P1-receptor agonist adenosine failed to stimulate any whole cell currents. The ATP-activated K+ current was inhibited by iberiotoxin (200 nM), and it was potentiated by the BK channel activator NS-1619 (30 microM). In cell-attached recordings, ATP activated a 230-pS BK channel. In conclusion, ATP acting via P2 purinoceptors stimulated release of Ca2+ from internal stores and transiently activated depolarizing Cl- and hyperpolarizing BK channels in human coronary artery smooth muscle cells.


Assuntos
Trifosfato de Adenosina/farmacologia , Cálcio/metabolismo , Canais de Cloreto/fisiologia , Vasos Coronários/fisiologia , Músculo Liso Vascular/fisiologia , Canais de Potássio/fisiologia , Receptores Purinérgicos P2/fisiologia , Trifosfato de Adenosina/análogos & derivados , Células Cultivadas , Canais de Cloreto/efeitos dos fármacos , Ácido Egtázico/farmacologia , Humanos , Indóis/farmacologia , Cinética , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Receptores Purinérgicos P2/efeitos dos fármacos , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia , Tionucleotídeos/farmacologia , Fatores de Tempo , Uridina Trifosfato/farmacologia
16.
Br J Pharmacol ; 118(7): 1645-52, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8842427

RESUMO

1. The effects of extracellular adenosine 5'-triphosphate (ATP) on smooth muscles are mediated by a variety of purinoceptors. In this study we addressed the identity of the purinoceptors on smooth muscle cells (SMC) cultured from human large coronary arteries. Purinoceptor-mediated increases in [Ca2+]i were measured in single fura-2 loaded cells by applying a digital imaging technique, and the formation of inositol phosphate compounds was quantified after separation on an anion exchange column. 2. Stimulation of the human coronary artery SMC (HCASMC) with extracellular ATP at concentrations of 0.1-100 microM induced a transient increase in [Ca2+]i from a resting level of 49 +/- 21 nM to a maximum of 436 +/- 19 nM. The effect was dose-dependent with an EC50 value for ATP of 2.2 microM. 3. The rise in [Ca2+]i was independent of the presence of external Ca2+, but was abolished after depletion of intracellular stores by incubation with 100 nM thapsigargin. 4. [Ca2+]i was measured upon stimulation of the cells with 0.1-100 microM of the more specific P2-purinoceptor agonists alpha, beta-methyleneadenosine 5'-triphosphate (alpha,beta-MeATP), 2-methylthioadenosine 5'-triphosphate (2MeSATP) and uridine 5'-triphosphate (UTP). alpha, beta-MeATP was without effect, whereas 2MeSATP and UTP induced release of Ca2+ from internal stores with 2MeSATP being the most potent agonist (EC50 = 0.17 microM), and UTP having a potency similar to ATP. The P1 purinoceptor agonist adenosine (100 microM) did not induce any changes in [Ca2+]i. 5. Stimulation with a submaximal concentration of UTP (10 microM) abolished a subsequent ATP-induced increase in [Ca2+]i, whereas an increase was induced by ATP after stimulation with 10 microM 2MeSATP. 6. The phospholipase C (PLC) inhibitor U73122 (5 microM) abolished the purinoceptor-activated rise in [Ca2+]i, whereas pretreatment with the Gi protein inhibitor pertussis toxin (PTX, 500 ng ml-1) was without effect on ATP-evoked [Ca2+]i increases. 7. Receptor activation with UTP and ATP resulted in formation of inositol phosphates with peak levels of inositol 1, 4, 5-trisphosphate (Ins(1, 4, 5)P3) observed 5-20 s after stimulation. 8. These findings show, that cultured HCASMC express G protein-coupled purinoceptors, which upon stimulation activate PLC to induce enhanced Ins(1, 4, 5)P3 production causing release of Ca2+ from internal stores. Since a release of Ca2+ was induced by 2MeSATP as well as by UTP, the data indicate that P2y- as well as P2U-purinoceptors are expressed by the HCASMC.


Assuntos
Cálcio/metabolismo , Vasos Coronários/fisiologia , Fosfatos de Inositol/biossíntese , Músculo Liso Vascular/fisiologia , Receptores Purinérgicos P2/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Células Cultivadas , Vasos Coronários/citologia , Vasos Coronários/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Toxina Pertussis , Agonistas do Receptor Purinérgico P2 , Antagonistas do Receptor Purinérgico P2 , Tapsigargina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo , Fatores de Virulência de Bordetella/farmacologia
17.
Proc Natl Acad Sci U S A ; 93(17): 9200-5, 1996 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-8799178

RESUMO

We have cloned and expressed a Ca(2+)-activated K+ channel beta-subunit from human brain. The open reading frame encodes a 191-amino acid protein possessing significant homology to a previously described subunit cloned from bovine muscle. The gene for this subunit is located on chromosome 5 at band q34 (hslo-beta). There is no evidence for alternative RNA splicing of this gene product. hslo-beta mRNA is abundantly expressed in smooth muscle, but expression levels are low in most other tissues, including brain. Brain subregions in which beta-subunit mRNA expression is relatively high are the hippocampus and corpus callosum. The coexpression of hslo-beta mRNA together with hslo-alpha subunits in either Xenopus oocytes or stably transfected HEK 293 cells give rise to Ca(2+)-activated potassium currents with a much increased calcium and/or voltage sensitivity. These data indicate that the beta-subunit shows a tissue distribution different to that of the alpha-subunit, and in many tissues there may be no association of alpha-subunits with beta-subunits. These beta-subunits can play a functional role in the regulation of neuronal excitability by tuning the Ca2+ and/or the voltage dependence of alpha-subunits.


Assuntos
Química Encefálica/genética , Cálcio/farmacologia , Ativação do Canal Iônico , Canais de Potássio Cálcio-Ativados , Canais de Potássio/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Mapeamento Cromossômico , Cromossomos Humanos Par 5/genética , Clonagem Molecular , Corpo Caloso/química , Hipocampo/química , Humanos , Hibridização in Situ Fluorescente , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Canais de Potássio/biossíntese , Canais de Potássio/efeitos dos fármacos , RNA Mensageiro/isolamento & purificação , Proteínas Recombinantes/biossíntese , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Distribuição Tecidual , Xenopus
18.
Neuropharmacology ; 35(7): 903-14, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8938721

RESUMO

The high-conductance Ca(2+)-activated K channel (BK channel) is not only regulated by a number of physiological stimuli, but it is also sensitive to pharmacological modulation. We have stably expressed the alpha-subunit of the human BK channel, hslo, in HEK 293 cells and studied by patch-clamp technique how its gating is modulated by the channel activator NS 1608, by the selective channel blocker paxilline, as well as by changes in [Ca2+]i and Vm. The cells expressed 200-800 hslo channels per patch. The channel activity was determined by tail current analysis, and the activation curves were fitted to single Boltzmann functions, from which a gating charge for the hslo channel of 1.2 elementary charges was deduced. The hslo channel was very sensitive to changes in [Ca2+]i within the physiological range, whereas Ca(2+)-independent openings were seen at Ca2+ concentrations of 15 nM or below. NS 1608 shifted the hslo channel activation curve towards negative membrane potentials with an EC50 of 2.1 microM and a maximal shift of -74 mV. The channels activated by NS 1608 were sensitive to block by paxilline, but the two molecules apparently did not interact within the same site, since paxilline reduced the size of the tail current at all voltages, whereas NS 1608 shifted the activation curve along the voltage axis. Further, the effect of paxilline was Ca(2+)-sensitive, whereas NS 1608 elicited identical effects in the presence of either < 0.5 nM or 500 nM [Ca2+]i. NS 1608 hyperpolarized the cells by -50 to -70 mV, and paxilline depolarized them towards 0 mV. In addition to the effects on the steady state current NS 1608 also significantly influenced the non-stationary channel kinetics. In the presence of NS 1608 the time constants for deactivation of tail currents were more than tripled at all potentials. We have shown, that NS 1608 modulates steady-state BK currents and channel gating kinetics through a Ca(2+)-independent interaction with the alpha-subunit of the channel.


Assuntos
Cálcio/metabolismo , Indóis/farmacologia , Compostos de Fenilureia/farmacologia , Bloqueadores dos Canais de Potássio , Canais de Potássio Cálcio-Ativados , Linhagem Celular Transformada , Células Cultivadas , Embrião de Mamíferos , Vetores Genéticos , Humanos , Rim , Cinética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta , Potenciais da Membrana , Técnicas de Patch-Clamp , Canais de Potássio/biossíntese , Canais de Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...