Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 603
Filtrar
1.
Biochim Biophys Acta ; 1840(6): 1640-5, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24412196

RESUMO

BACKGROUND: Rapid trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) to the plasma membrane is considered a fundamental biological process for learning and memory. GluR1 is an AMPAR subunit. We have shown that mice with knockout of excitatory amino acid transporter type 3 (EAAT3), a neuronal glutamate transporter, have impaired learning and memory. The mechanisms for this impairment are not known and may be via regulation of AMPAR trafficking. METHODS: Freshly prepared 300µm coronal hippocampal slices from wild-type or EAAT3 knockout mice were incubated with or without 25mM tetraethylammonium for 10min. The trafficking of GluR1, an AMPAR subunit, to the plasma membrane and its phosphorylation were measured. RESULTS: Tetraethylammonium increased the trafficking of GluR1 and EAAT3 to the plasma membrane in the wild-type mouse hippocampal slices but did not cause GluR1 trafficking in the EAAT3 knockout mice. Tetraethylammonium also increased the phosphorylation of GluR1 at S845, a protein kinase A (PKA) site, in the wild-type mice but not in the EAAT3 knockout mice. The PKA antagonist KT5720 attenuated tetraethylammonium-induced GluR1 phosphorylation and trafficking in the wild-type mice. The PKA agonist 6-BNz-cAMP caused GluR1 trafficking to the plasma membrane in the EAAT3 knockout mice. In addition, EAAT3 was co-immunoprecipitated with PKA. CONCLUSIONS: These results suggest that EAAT3 is upstream of PKA in a pathway to regulate GluR1 trafficking. GENERAL SIGNIFICANCE: Our results provide initial evidence for the involvement of EAAT3 in the biochemical cascade of learning and memory.


Assuntos
Transportador 3 de Aminoácido Excitatório/fisiologia , Hipocampo/metabolismo , Receptores de AMPA/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Masculino , Camundongos , Transporte Proteico , Compostos de Tetraetilamônio/farmacologia
2.
Gen Comp Endocrinol ; 169(3): 231-43, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20850441

RESUMO

Growth hormone release in goldfish is partly dependent on voltage-sensitive Ca(2+) channels but somatotrope electrophysiological events affecting such channel activities have not been elucidated in this system. The electrophysiological properties of goldfish somatotropes in primary culture were studied using the whole-cell and amphotericin B-perforated patch-clamp techniques. Intracellular Ca(2+) concentration ([Ca(2+)]i) of identified somatotropes was measured using Fura-2/AM dye. Goldfish somatotropes had an average resting membrane potential of -78.4 ± 4.6 mV and membrane input resistance of 6.2 ± 0.2 GΩ. Voltage steps from a holding potential of -90 mV elicited a non-inactivating outward current and transient inward currents at potentials more positive than 0 and -30 mV, respectively. Isolated current recordings indicate the presence of 4-aminopyridine- and tetraethylammonium (TEA)-sensitive K(+), tetrodotoxin (TTX)-sensitive Na(+), and nifedipine (L-type)- and ω-conotoxin GVIA (N-type)-sensitive Ca(2+) channels. Goldfish somatotropes rarely fire action potentials (APs) spontaneously, but single APs can be induced at the start of a depolarizing current step; this single AP was abolished by TTX and significantly reduced by nifedipine and ω-conotoxin GVIA. TEA increased AP duration and triggered repetitive AP firing resulting in an increase in [Ca(2+)]i, whereas TTX, nifedipine and ω-conotoxin GVIA inhibited TEA-induced [Ca(2+)]i pulses. These results indicate that in goldfish somatotropes, TEA-sensitive K(+) channels regulate excitability while TTX-sensitive Na(+) channels together with N- and L-type Ca channels mediates the depolarization phase of APs. Opening of voltage-sensitive Ca(2+) channels during AP firing leads to increases in [Ca(2+)]i.


Assuntos
Carpa Dourada/fisiologia , Somatotrofos/fisiologia , 4-Aminopiridina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Bário/fisiologia , Canais de Cálcio/fisiologia , Células Cultivadas , Potenciais da Membrana/fisiologia , Nifedipino , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Canais de Sódio/fisiologia , Compostos de Tetraetilamônio/farmacologia , ômega-Conotoxina GVIA/farmacologia
3.
Biol Pharm Bull ; 30(8): 1477-80, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17666807

RESUMO

The previous data indicated that the testosterone (Tes)-induced relaxation of thoracic aorta is greater in spontaneously hypertensive rats (SHR) than in normotensive rats (Wistar-Kyoto rats; WKY) and that there were differences between SHR and WKY in the functions of K(ATP), K(v), and K(Ca) channels. The present study was carried out to ascertain the mechanisms of the Tes-induced relaxation. Indomethacin (30 muM) pretreatment suppressed the Tes-induced relaxation. Following noradrenalin (NA)-induced vasoconstriction, the relaxation induced by Tes was significantly attenuated by endothelium removal in SHR (not in WKY), but the dilatory effect of Tes following KCl-induced vasoconstriction was not attenuated by endothelium removal. After tetraethylammonium (K(Ca) channel inhibitor) or iberiotoxin (large conductance, Ca(2+) activated BK channel inhibitor) pretreatment, the Tes-induced relaxation was attenuated in SHR, but not in WKY. This attenuation in SHR was not observed after endothelium removal. The above results suggest that the relaxation induced by Tes following NA-induced vasoconstriction in SHR results from hyperpolarization due to BK channel opening.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Testosterona/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Masculino , Relaxamento Muscular/efeitos dos fármacos , Norepinefrina/farmacologia , Peptídeos/farmacologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Compostos de Tetraetilamônio/farmacologia , Vasoconstritores/farmacologia
4.
Vascul Pharmacol ; 47(1): 74-82, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17521967

RESUMO

It is known that myogenic reactivity is a fundamental determinant of the relative constancy of blood flow through the cerebral artery. It is also known that acute alteration of pH significantly affects the cerebral circulation and, therefore, we investigated the effect of mechanism of action of intracellular acidosis on myogenic tone in rabbit basilar artery. Myogenic tone was developed by imposed stretch of basilar artery and intracellular acidosis induced by the bath application of 20 mmol/L sodium acetate. Sodium acetate caused a biphasic increase in myogenic tone. The initial component reached a peak quickly and then fell slowly to a lower steady-state significantly above basal tone. The sodium acetate-induced increase in myogenic tone was completely inhibited by elimination of external Ca2+, or treatment of nifedipine, but not with gadolinium or NPPB. TEA (5 mmol/L) and iberiotoxin (100 nmol/L) inhibited the sodium acetate-induced increase in myogenic tone. In inside-out patch-clamp recordings, decreasing pH of the mock intracellular solution from 7.4 to 6.9 markedly inhibited BKCa currents. Several inhibitors involved in Ca2+ sensitization pathways, 10(-6) mol/L Y-27632, 5 x 10(-7) mol/L calphostin C and 10(-5) mol/L PD98059 had no effect on the sodium acetate-induced increase in myogenic tone. These results suggest that intracellular acidosis increases stretch-induced myogenic tone in rabbit basilar artery. Furthermore, voltage-dependent Ca2+ influx plays a key role in intracellular acidosis-induced increase in myogenic tone and may be mediated, at least in part, by inhibition of BKCa.


Assuntos
Acidose/fisiopatologia , Artéria Basilar/efeitos dos fármacos , Tono Muscular/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Animais , Artéria Basilar/fisiologia , Cálcio/metabolismo , Feminino , Concentração de Íons de Hidrogênio , Masculino , Nifedipino/farmacologia , Peptídeos/farmacologia , Coelhos , Retículo Sarcoplasmático/metabolismo , Compostos de Tetraetilamônio/farmacologia
5.
Mol Pharmacol ; 71(3): 843-51, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17164406

RESUMO

In the present study, we investigated the effect of the antiestrogen compound tamoxifen on BK channels by the use of the patch-clamp technique. The perfusion of 10 nM tamoxifen significantly increased the magnitude of a voltage-dependent K+ current by 22.6 +/- 10.6% (n = 23). The effect of tamoxifen was always obtained in the first minute, peaked at 5.9 +/- 2.2 min (n = 23), and was abolished by the perfusion of tetraethylammonium (0.5 mM), charybdotoxin (50 nM), or iberiotoxin (100 nM). The stimulatory effect of 10 nM tamoxifen was the same at low (50 nM) and high (700 nM) internal calcium concentration and was not additive to that of 17-beta-estradiol (E2) or its membrane-impermeant form, beta-estradiol 6-(O-carboxymethyl)oxime:bovine serum albumin. Furthermore, the effect of tamoxifen was still recorded in the presence of the selective estrogen receptor antagonist faslodex (ICI-182,780; 1 microM). At the single-channel level, tamoxifen significantly increased the open probability of the BK channel by 46.2 +/- 10.1% (n = 4) without changing its unitary conductance. Moreover, we show here that the stimulation of BK channel activity by tamoxifen is involved in MCF-7 cell proliferation. Taken together, these results permitted us to identify the BK channel as the molecular target of tamoxifen that probably acts at the same extracellular molecular level as E2. The site of action of tamoxifen is probably the channel itself or the auxiliary beta subunits.


Assuntos
Neoplasias da Mama/patologia , Antagonistas de Estrogênios/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Tamoxifeno/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Fulvestranto , Humanos , Compostos de Tetraetilamônio/farmacologia
6.
Cell Physiol Biochem ; 18(1-3): 47-56, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16914889

RESUMO

We report here the expression and properties of the intermediate-conductance Ca(2+)-activated K(+) (IK(Ca)) channel in the GL-15 human glioblastoma cell line. Macroscopic IK(Ca) currents on GL-15 cells displayed a mean amplitude of 7.2+/-0.8 pA/pF at 0 mV, at day 1 after plating. The current was inhibited by clotrimazole (CTL, IC(50)=257 nM), TRAM-34 (IC(50)=55 nM), and charybdotoxin (CTX, IC(50)=10.3 nM). RT-PCR analysis demonstrated the expression of mRNA encoding the IK(Ca) channel in GL-15 cells. Unitary currents recorded using the inside-out configuration had a conductance of 25 pS, a K(D) for Ca(2+) of 188 nM at -100 mV, and no voltage dependence. We tested whether the IKCa channel expression in GL-15 cells could be the result of an increased ERK activity. Inhibition of the ERK pathway with the MEK antagonist PD98059 (25 muM, for 5 days) virtually suppressed the IK(Ca) current in GL-15 cells. PD98059 treatment also increased the length of cellular processes and up-regulated the astrocytic differentiative marker GFAP. A significant reduction of the IKCa current amplitude was also observed with time in culture, with mean currents of 7.17+/-0.75 pA/pF at 1-2 days, and 3.11+/-1.35 pA/pF at 5-6 days after plating. This time-dependent downregulation of the IK(Ca) current was not accompanied by changes in the ERK activity, as assessed by immunoblot analysis. Semiquantitative RT-PCR analysis demonstrated a ~35% reduction of the IK(Ca) channel mRNA resulting from ERK inhibition and a approximately 50% reduction with time in culture.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Flavonoides/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/fisiopatologia , Humanos , Immunoblotting , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Bloqueadores dos Canais de Potássio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Compostos de Tetraetilamônio/farmacologia , Fatores de Tempo
7.
Anesth Analg ; 103(2): 443-7, table of contents, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16861431

RESUMO

We examined the effects of equianesthetic concentrations of sevoflurane, desflurane, isoflurane, and halothane on the spontaneous contractility of isolated human pregnant uterine muscles. We also determined if their action was related to potassium channels. Uterine specimens were obtained from normal full-term pregnant women undergoing elective lower-segment cesarean delivery. Longitudinal muscle strips were mounted vertically in tissue chambers. Their isometric tension was recorded while they were exposed to 0.5-3 minimum alveolar concentration (MAC) of volatile anesthetics in the absence and presence of the high conductance calcium-activated potassium channel blocker, tetraethylammonium, or the adenosine triphosphate-sensitive potassium channel (K(ATP))-blocker, glibenclamide. The anesthetics examined produced a dose-dependent depression of contractility. The inhibitory potency of sevoflurane and desflurane was comparable to, whereas that of isoflurane was smaller than, that of halothane: concentrations causing 50% inhibition of the contractile amplitude (ED(50)) were 1.72, 1.44, 2.35, and 1.66 MAC (P < 0.05), respectively. Tetraethylammonium and glibenclamide did not affect the uterine response to the anesthetics, except for glibenclamide, which attenuated the response to isoflurane. These results indicate that the volatile anesthetics have inhibitory effects on the contractility of the human uterus. The inhibitory effect of isoflurane may in part be mediated through activation of K(ATP) channels.


Assuntos
Anestésicos Inalatórios/farmacologia , Contração Uterina/efeitos dos fármacos , Cálcio/metabolismo , Desflurano , Relação Dose-Resposta a Droga , Feminino , Glibureto/farmacologia , Halotano/farmacologia , Humanos , Técnicas In Vitro , Isoflurano/análogos & derivados , Isoflurano/farmacologia , Éteres Metílicos/farmacologia , Gravidez , Sevoflurano , Compostos de Tetraetilamônio/farmacologia
8.
J Pharmacol Exp Ther ; 318(3): 1307-14, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16772540

RESUMO

The epoxyeicosatrienoic acids (EETs) have been identified as endothelium-derived hyperpolarizing factors. Metabolism of the EETs to the dihydroxyeicosatrienoic acids is catalyzed by soluble epoxide hydrolase (sEH). Administration of urea-based sEH inhibitors provides protection from hypertension-induced renal injury at least in part by lowering blood pressure. Here, we investigated the hypothesis that a mechanism by which sEH inhibitors elicit their cardiovascular protective effects is via their action on the vasculature. Mesenteric resistance arteries were isolated from Sprague-Dawley rats, pressurized, and constricted with the thromboxane A2 agonist U46619 (9,11-dideoxy-11,9-epoxymethano-prostaglandin F2alpha). Mesenteric arteries were then incubated with increasing concentrations of the sEH inhibitor 12-(3-adamantan-1-yl-ureido)dodecanoic acid (AUDA). AUDA resulted in a concentration-dependent relaxation of mesenteric arteries, with 10 microM resulting in a 48 +/- 7% relaxation. Chain-shortened analogs of AUDA had an attenuated vasodilatory response. Interestingly, at 10 microM, the sEH inhibitors 1-cyclohexyl-3-dodecylurea, 12-(3-cyclohexylureido)dodecanoic acid, and 950 [adamantan-1-yl-3-{5-[2-(2-ethoxyethoxy)ethoxy]pentyl}urea] were significantly less active, resulting in a 25 +/- 8%, 10 +/- 9%, and -8 +/- 3% relaxation, respectively. Treatment of mesenteric arteries with tetraethylammonium, iberiotoxin, ouabain, or glibenclamide did not alter AUDA-induced relaxation. The AUDA-induced relaxation was completely inhibited when constricted with KCl. In separate experiments, denuding mesenteric resistance vessels did not alter AUDA-induced relaxation. Taken together, these data demonstrate that adamantyl-urea inhibitors have unique dilator actions on vascular smooth muscle compared with other sEH inhibitors and that these dilator actions depend on the adamantyl group and carbon chain length.


Assuntos
Adamantano/análogos & derivados , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Ácidos Láuricos/farmacologia , Artérias Mesentéricas/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Adamantano/farmacologia , Animais , Masculino , Artérias Mesentéricas/fisiologia , Peptídeos/farmacologia , Canais de Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/fisiologia , Compostos de Tetraetilamônio/farmacologia , Resistência Vascular
9.
Cardiovasc Res ; 71(2): 383-92, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16716281

RESUMO

OBJECTIVE: The human radial artery has demonstrated superior long-term results as a graft in coronary bypass surgery, but undesirable post-surgical spasm limits its clinical application. Few have examined its excitatory properties, especially the underlying ion channel mechanisms. In this study, we investigated the kinetic and pharmacological properties of the smooth muscle membrane potassium currents of this important artery. METHODS AND RESULTS: Using whole cell patch-clamp techniques, we found the K(+) current to be voltage-dependent and outwardly rectifying. Voltage-dependent inactivation was observed, being half-maximal at +28.0 mV but incomplete even at +40 mV. The K(+) currents were predominantly sensitive to the K(Ca) blocker tetraethylammonium (TEA; 63.9+/-12.1% inhibition, p<0.05), less sensitive to the Kv blocker 4-aminopyridine (4-AP; 32.8+/-4.4% inhibition, p<0.05), and the K(ATP) blocker glibenclamide (28.7+/-8.5% inhibition), at -20 mV testing potential. Resting membrane potential was -52.0+/-6.8 mV (n=5), and suppression of K(+) currents by TEA and iberiotoxin (IbTx) caused membrane depolarization. Western blot analysis with channel-specific antibodies confirmed the presence of K(Ca) and Kv channel proteins. TEA evoked 20.7+/-9.9% of the contractile response to 60 mM KCl, whereas IbTx caused about 10% of the above response at 10(-7) M. The nitric oxide donor SNAP augmented membrane K(+) currents in a concentration-dependent fashion; the augmentation was completely suppressed by TEA, but was relatively insensitive to the guanylate cyclase inhibitor ODQ. CONCLUSIONS: The radial artery manifests mainly Ca(2+)-dependent K(+) currents at rest; this current is augmented by nitric oxide through a cGMP- and protein kinase G-independent action. The relatively depolarized membrane potential, as well as its muscular structure, predisposes the radial artery to spasm. Agents that activate the Ca(2+)-dependent K(+) current could be of therapeutic value in preventing post-surgical vasospasm.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Doadores de Óxido Nítrico/farmacologia , Penicilamina/análogos & derivados , Canais de Potássio/metabolismo , Artéria Radial , 4-Aminopiridina/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting/métodos , Feminino , Glibureto/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Humanos , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Pessoa de Meia-Idade , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Oxidiazóis/farmacologia , Técnicas de Patch-Clamp , Penicilamina/farmacologia , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Quinoxalinas/farmacologia , Compostos de Tetraetilamônio/farmacologia
10.
Circ Res ; 97(4): 329-36, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-16037573

RESUMO

Ischemic and pharmacological preconditioning can be triggered by an intracellular signaling pathway in which Gi-coupled surface receptors activate a cascade including phosphatidylinositol 3-kinase, endothelial nitric oxide synthase, guanylyl cyclase, and protein kinase G (PKG). Activated PKG opens mitochondrial KATP channels (mitoKATP) which increase production of reactive oxygen species. Steps between PKG and mitoKATP opening are unknown. We describe effects of adding purified PKG and cGMP on K+ transport in isolated mitochondria. Light scattering and respiration measurements indicate PKG induces opening of mitoKATP similar to KATP channel openers like diazoxide and cromakalim in heart, liver, and brain mitochondria. This effect was blocked by mitoKATP inhibitors 5-hydroxydecanoate, tetraphenylphosphonium, and glibenclamide, PKG-selective inhibitor KT5823, and protein kinase C (PKC) inhibitors chelerythrine, Ro318220, and PKC-epsilon peptide antagonist epsilonV(1-2). MitoKATP are opened by the PKC activator 12-phorbol 13-myristate acetate. We conclude PKG is the terminal cytosolic component of the trigger pathway; it transmits the cardioprotective signal from cytosol to inner mitochondrial membrane by a pathway that includes PKC-epsilon.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Citosol/metabolismo , Precondicionamento Isquêmico Miocárdico , Mitocôndrias Cardíacas/metabolismo , Transdução de Sinais/fisiologia , Trifosfato de Adenosina/farmacologia , Animais , Encéfalo/metabolismo , GMP Cíclico/fisiologia , Masculino , Mitocôndrias Hepáticas/metabolismo , Consumo de Oxigênio , Canais de Potássio/fisiologia , Proteína Quinase C/fisiologia , Ratos , Ratos Wistar , Acetato de Tetradecanoilforbol/farmacologia , Compostos de Tetraetilamônio/farmacologia
11.
J Pharm Sci ; 93(2): 262-72, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14705184

RESUMO

Nafamostat mesilate (NM) is a serine-protease inhibitor that is rapidly eliminated from the circulation and accumulated in the kidney. This study was conducted to characterize the mechanism of NM transport in the kidney because a serious side effect of NM-induced hyperkalemia may be related to accumulation of NM in the kidney. Measurements of uptake of NM in vivo by the kidney uptake index (KUI) method and of transport in an in vitro-cultured LLC-PK1 cell system suggested the involvement of an organic cation transporter (OCT). To clarify the involvement of OCTs located in the basolateral membrane of proximal tubules, we evaluated NM transport by OCTs expressed in Xenopus laevis oocytes. The IC(50) values of NM on [(14)C]TEA ([(14)C]tetraethylammonium) uptake by rOCT1, rOCT2, and hOCT2 were 50, 0.5, and 20 microM, respectively, and NM was concluded to be a substrate of OCTs. To investigate the transport of NM across the brush-border membrane, we examined the uptake of NM into brush-border membrane vesicles (BBMVs) isolated from rat renal cortex. NM was taken up into the BBMVs, and the uptake was decreased by unlabeled NM and temperature, implying that a transporter(s) is also involved in NM transport across the apical membrane. NM was not a substrate of hOCTN1, hOCTN2, or P-gp, implying the involvement of some unknown transporter(s). Thus, renal accumulation of NM can be explained by the involvement of the basolateral OCTs, though the influence of the apical membrane transporter remains to be clarified.


Assuntos
Proteínas de Transporte/metabolismo , Guanidinas/metabolismo , Rim/metabolismo , Inibidores de Serina Proteinase/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Benzamidinas , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Técnicas In Vitro , Córtex Renal/metabolismo , Túbulos Renais/metabolismo , Células LLC-PK1 , Masculino , Microvilosidades/metabolismo , Oócitos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Transportador 1 de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico , RNA Complementar/biossíntese , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Suínos , Compostos de Tetraetilamônio/farmacologia , Xenopus laevis
12.
J Pharmacol Exp Ther ; 305(2): 502-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12606650

RESUMO

K(+) channel blockers such as 4-aminopyridine (4-AP) can be toxic to neurons; the cellular mechanism underlying the toxicity, however, is obscure. In cultured mouse cortical neurons, we tested the hypothesis that the toxic effect of 4-AP might result from inhibiting the Na(+),K(+)-ATPase (Na(+),K(+)-pump) and thereafter induction of a hybrid death of concomitant apoptosis and necrosis. The Na(+),K(+)-pump activity, monitored as whole-cell membrane currents, was markedly blocked by 4-AP in concentration- and voltage-dependent manners in low millimolar ranges. At similar concentrations, 4-AP induced a neuronal death sensitive to attenuation by the caspase inhibitor Z-VAD-FMK (Z-Val-Ala-Asp(OMe)-fluoromethyl ketone) or Ca(2+) chelator BAPTA-AM (1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester). Electron microscopy confirmed hybrid ultrastructural features of coexisting apoptotic and necrotic components in same cells. We suggest that 4-AP is a potent antagonist of the Na(+),K(+)-ATPase and an inducer of the hybrid death of central neurons.


Assuntos
4-Aminopiridina/farmacologia , Córtex Cerebral/citologia , Inibidores Enzimáticos/farmacologia , Neurônios/enzimologia , Bloqueadores dos Canais de Potássio/farmacologia , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Eletrofisiologia , Células Híbridas/efeitos dos fármacos , Camundongos , Microscopia Eletrônica , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Estrofantidina/farmacologia , Compostos de Tetraetilamônio/farmacologia
13.
J Biol Chem ; 276(37): 34594-9, 2001 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-11454866

RESUMO

Estrogen (17beta-estradiol; 17betaE) and xenoestrogens, estrogenic compounds that are not steroid hormones, have non-genomic actions at plasma membrane receptors unrelated to the nuclear estrogen receptor. The open probability (P(o)) of large conductance Ca(2+)/voltage-sensitive k(+)(BK) channels is increased by 17betaE through the regulatory beta1 subunit. The pharmacological nature of the putative membrane binding site is unclear. We probed the site by determining whether tamoxifen ((Z)-1-(p-dimethylaminoethoxy-phenyl)-1,2-diphenyl-1-butene; Tx), a chemotherapeutic xenoestrogen, increased P(o) in clinically relevant concentrations (0.1-10 microm). In whole cell patch clamp recordings on canine colonic myocytes, which express the beta1 subunit, Tx activated charybdotoxin-sensitive K(+) current. In single channel experiments, Tx increased the NP(o) (P(o) x number channels; N) and decreased the unitary conductance (gamma) of BK channels. Tx increased NP(o) (EC(50) = 0.65 microm) in excised membrane patches independent of Ca(2+) changes. The Tx mechanism of action requires the beta1 subunit, as Tx increased the NP(o) of Slo alpha expressed in human embryonic kidney cells only in the presence of the beta1 subunit. Tx decreased gamma of the alpha subunit expressed alone, without effect on NP(o). Our data indicate that Tx increases BK channel activity in therapeutic concentrations and reveal novel pharmacological properties attributable to the alpha and beta1 subunits. These data shed light on BK channel structure and function, non-genomic mechanisms of regulation, and physiologically and therapeutically relevant effects of xenoestrogens.


Assuntos
Antagonistas de Estrogênios/farmacologia , Músculo Liso/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Tamoxifeno/farmacologia , Animais , Cálcio/metabolismo , Linhagem Celular , Cães , Humanos , Músculo Liso/metabolismo , Canais de Potássio/química , Subunidades Proteicas , Compostos de Tetraetilamônio/farmacologia
14.
Toxicology ; 150(1-3): 129-36, 2000 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-10996669

RESUMO

The effect of pH on the secretory transport of Cd through the intestinal brush border membrane was investigated using isolated rat intestinal brush border membrane vesicles (BBMV) and the Caco-2 intestinal epithelial cell line. BBMV equilibrated at pH 5.5 or 7.5 (pH(in)) were mixed with an experimental buffer at pH 5.5 or 7.5 (pH(out)) containing CdCl(2). The initial accumulation of Cd in BBMV incubated for 1 or 3 min at pH(in) 5.5 and pH(out) 7.5 (outwardly directed H(+)-gradient) was significantly higher than that at pH(in)=pH(out)=7.5, but the equilibrated Cd accumulation incubated for 30 min was marginally lower. Carbonylcyanide-p-trifluoromethoxyphenylhydrazone (FCCP), a protonophore, diminished the increasing effect of the H(+)-gradient on the initial Cd accumulation. Caco-2 cell monolayers cultured on permeable membranes were incubated with CdCl(2) from the basolateral medium, and the transport of Cd from the basolateral to apical medium and the accumulation of Cd in the monolayers were measured. Cd transport was increased by lowering the pH of the apical medium, and was accompanied by a decrease in the Cd accumulation. Coincubation with CdCl(2) and tetraethylammonium, a typical substrate for H(+)-antiport of the renal organic cation transporter, from the basolateral medium slightly but significantly decreased the basolateral-to-apical transport of Cd, with a concomitant increase in the Cd accumulation. These findings suggest the secretory transport of Cd through the intestinal brush border membrane not only via passive diffusion but also via H(+)-antiport of the putative organic cation transporter.


Assuntos
Cádmio/farmacocinética , Mucosa Intestinal/metabolismo , Transporte Biológico , Células CACO-2 , Humanos , Concentração de Íons de Hidrogênio , Intestinos/ultraestrutura , Microvilosidades/metabolismo , Compostos de Tetraetilamônio/farmacologia
15.
Kaohsiung J Med Sci ; 16(3): 134-40, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10846349

RESUMO

Thallium-201 myocardial perfusion imaging is wildly used to detect and assess the extent of jeopardized myocardial ischemia in the coronary artery disease and the viability of myocardium post infarction. In recent years, there has been a great deal of pharmacological development of blockers and openers of potassium channel. In this study, we will discuss the interference of uptake of thallium-201 ion in cultured neonatal rat myocytes with existence of a variety of pharmacological agents. The cultures of neonatal rat myocardial cells were incubated with different agents such as potassium chloride, sodium-potassium ATPase pump inhibitor (ouabain), cesium compound, variable potassium channel blockers (4 AP, TEA and glibenclamide) and their openers (minoxidil, and cromakalim). The radioactivity of intracellular thallium-201 that could enter rat myocardial cells was detected by gamma counter sixty minutes after thallium-201 was added. In this study we found that thallium and potassium ions behave in an analogous manner in cultured rat myocardial cells. Both 2.5 mM and 5 mM concentration of extracellular potassium ion significantly result in reduction of thallium-201 ion influx in rat myocardial cells. 0.5 mM ouabain, an inhibitor of sodium-potassium ATPase pump, reduced about 40% of influx of thallium-201 ion in cultured rat myocardial cells via active transport. Combination of both potassium ion and ouabain inhibit most of thallium-201 ions influx in myocardial cells, but it is not completely inhibited. Cesium, a potassium antagonist, also interferes with the uptake of thallium-201 in cultured rat myocytes in our study. The most interesting finding in our investigation is that potassium channel blockers such as TEA and glibenclamide, inhibit the influx of thallium-201 in myocytes. However, potassium channel openers have no overt effect on influx of thallium-201 in cultured rat myocytes. We indirectly observe about 60% of influx of thallium-201 ion into cultured rat myocardial cells via active sodium-potassium ATPase pump. Potassium, cesium and potassium channel blockers, such as TEA and glibenclamide, inhibited the different percentage of influx of thallium-201 in cultured rat myocardial cells in this study.


Assuntos
Miocárdio/metabolismo , Canais de Potássio/efeitos dos fármacos , Radioisótopos de Tálio/farmacocinética , Trifosfato de Adenosina/farmacologia , Animais , Células Cultivadas , Cloreto de Potássio/farmacologia , Ratos , Ratos Wistar , Compostos de Tetraetilamônio/farmacologia
16.
Proc Natl Acad Sci U S A ; 97(8): 4374-9, 2000 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-10760304

RESUMO

Pulmonary neuroepithelial bodies (NEBs) are presumed airway chemoreceptors that express the putative O(2) sensor protein NADPH oxidase and O(2)-sensitive K(+) channels K(+)(O(2)). Although there is a consensus that redox modulation of K(+)(O(2)) may be a common O(2)-sensing mechanism, the identity of the O(2) sensor and related coupling pathways are still controversial. To test whether NADPH oxidase is the O(2) sensor in NEB cells, we performed patch-clamp experiments on intact NEBs identified by neutral red staining in fresh lung slices from wild-type (WT) and oxidase-deficient (OD) mice. In OD mice, cytochrome b(558) and oxidase function was disrupted in the gp91(phox) subunit coding region by insertion of a neomycin phosphotransferase (neo) gene. Expression in NEB cells of neo mRNA, a marker for nonfunctional gp91(phox), was confirmed by nonisotopic in situ hybridization. In WT cells, hypoxia (pO(2) = 15-20 mmHg; 1 mmHg = 133 Pa) caused a reversible inhibition ( approximately 46%) of both Ca(2+)-independent and Ca(2+)-dependent K(+) currents. In contrast, hypoxia had no effect on K(+) current in OD cells, even though both K(+) current components were expressed. Diphenylene iodonium (1 microM), an inhibitor of the oxidase, reduced K(+) current by approximately 30% in WT cells but had no effect in OD cells. Hydrogen peroxide (H(2)O(2); 0.25 mM), a reactive oxygen species generated by functional NADPH oxidase, augmented K(+) current by >30% in both WT and OD cells; further, in WT cells, H(2)O(2) restored K(+) current amplitude in the presence of diphenylene iodonium. We conclude that NADPH oxidase acts as the O(2) sensor in pulmonary airway chemoreceptors.


Assuntos
Células Quimiorreceptoras/metabolismo , NADPH Oxidases/metabolismo , Oxigênio/metabolismo , Canais de Potássio/metabolismo , Traqueia/metabolismo , 4-Aminopiridina/farmacologia , Animais , Sequência de Bases , Hipóxia Celular , Primers do DNA , Doença Granulomatosa Crônica/enzimologia , Doença Granulomatosa Crônica/genética , Pulmão/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/genética , Canais de Potássio/efeitos dos fármacos , Compostos de Tetraetilamônio/farmacologia
17.
Eur J Pharmacol ; 392(1-2): 51-9, 2000 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-10748272

RESUMO

We assessed the role of endothelium in the arterial response to thapsigargin, the Ca(2+)-ATPase inhibitor of the endoplasmic reticulum, in rat isolated aortic rings. Thapsigargin induced an endothelium-dependent relaxation of phenylephrine-contracted aortic rings with an EC(50) of 2.6+/-0.4 nM and a 75% maximum relaxation, while it was less effective against 30 mM K(+)-induced contraction. Pretreatment of aortic rings with N(G)-nitro-L-arginine methyl ester (30 microM) or methylene blue (1 microM) reduced thapsigargin-induced relaxation by approximately 85%. Thapsigargin failed to relax the endothelium-denuded rings. L-Arginine (3 mM) partially, but significantly, antagonized the effect of 30 microM N(G)-nitro-L-arginine methyl ester. Pretreatment with indomethacin (3 microM), glibenclamide (1 microM) or iberiotoxin (100 nM) did not alter the thapsigargin-induced relaxation. In contrast, pretreatment with tetrapentylammonium ions (TPA(+), 1-3 microM) or with 300 microM Ba(2+) suppressed the relaxant response to thapsigargin. TPA(+) (3 microM) also attenuated acetylcholine-induced relaxation. Thapsigargin-induced endothelium-dependent relaxation was primarily dependent on the presence of extracellular Ca(2+). Interestingly, when the tissues were exposed to very low concentrations of thapsigargin (1-3 nM) the nitric oxide-dependent relaxation induced by acetylcholine or A23187 was markedly reduced. While thapsigargin (3 nM) did not influence the relaxation induced by endothelium-independent dilators, sodium nitroprusside and verapamil. These results indicate that thapsigargin produced complex vascular effects primarily by acting on the endothelial cells. Thapsigargin causes an endothelial nitric oxide-dependent relaxation; on the other hand, it inhibits nitric oxide-mediated relaxation at the similar concentrations. Activation of TPA(+)- and Ba(2+)-sensitive but not Ca(2+)-activated or ATP-sensitive K(+) channels may be also involved in thapsigargin-induced relaxation of rat isolated aortic rings.


Assuntos
ATPases Transportadoras de Cálcio/antagonistas & inibidores , Endotélio Vascular/fisiologia , Tapsigargina/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Calcimicina/farmacologia , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/fisiologia , Nitroarginina/farmacologia , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Compostos de Tetraetilamônio/farmacologia , Vasodilatação/efeitos dos fármacos
18.
Clin Neurophysiol ; 110(11): 1858-67, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10576480

RESUMO

OBJECTIVES: Roles of a calcium-dependent potassium conductance of slow afterhyperpolarization (AHP) type (gK(AHP)) in generating magnetoencephalographic (MEG) signals were studied in hippocampal longitudinal CA3 slices of the guinea pig. METHODS: The roles of gK(AHP) were experimentally inferred from effects of its blocker, carbamylcholine-chloride (carbachol, CCh), on MEG signals. The MEG signals were compared with extracellular field potentials and intracellular potentials of the pyramidal cells in the slice. RESULTS: CCh profoundly affected MEG waveforms. CCh reduced the initial spike of the evoked MEG signals independently of stimulation of the cell layer and apical dendrites. The slow wave of the evoked MEG signals was reduced by the somatic stimulation, but was enhanced by the apical stimulation. Elevated extracellular calcium and bath-applied tetraethylammonium (TEA) enhanced the CCh effects. CCh also increased spontaneous MEG signals. These effects on MEG and field potentials could be interpreted on the basis of synaptic and intracellular effects of CCh. CONCLUSIONS: Our results indicate that abnormality in this subtle calcium-dependent potassium channel may profoundly influence MEG and EEG signals.


Assuntos
Eletroencefalografia/efeitos dos fármacos , Campos Eletromagnéticos , Hipocampo/fisiologia , Canais de Potássio/fisiologia , Animais , Carbacol/farmacologia , Estimulação Elétrica , Potenciais Evocados/efeitos dos fármacos , Cobaias , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Técnicas In Vitro , Magnetoencefalografia , Agonistas Muscarínicos/farmacologia , Agonistas Nicotínicos/farmacologia , Estimulação Física , Bloqueadores dos Canais de Potássio , Canais de Potássio/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Compostos de Tetraetilamônio/farmacologia
19.
Br J Pharmacol ; 127(5): 1065-74, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10455250

RESUMO

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


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/efeitos dos fármacos , Verapamil/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo L , Cristalização , Canal de Potássio Kv1.3 , Modelos Estruturais , Dados de Sequência Molecular , Mutação , Canais de Potássio/química , Ratos , Canais de Sódio/efeitos dos fármacos , Relação Estrutura-Atividade , Compostos de Tetraetilamônio/farmacologia , Células Tumorais Cultivadas , Verapamil/metabolismo
20.
J Physiol ; 519 Pt 3: 645-55, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10457080

RESUMO

1. K+ channels from the apical membrane of immortalized human kidney epithelial (IHKE-1) cells were investigated in the cell-attached membrane configuration as well as in excised membranes using the patch clamp technique. 2. In cell-attached membrane patches the open probability (Po) of the K+ channel was 0.42 +/- 0.06 (mean +/- s.e.m. , n = 22) and its conductance was 94 +/- 5 pS with 145 mM K+ in the pipette (n = 25). In excised membrane patches the Po of the channel was 0.55 +/- 0.03 (n = 86) and its conductance was 65 +/- 2 pS (n = 68) with 145 mM K+ on one side of the membrane and 3.6 mM K+ on the other. The I-V curve of the K+ channel was not rectifying. 3. The channel was inhibited by several blockers of K+ channels such as 1 mM Ba2+ (cell-attached membrane: 78 +/- 8 %, n = 9; excised: 80 +/- 4 %, n = 26), 10 mM TEA+ (excised inside-out: 48 +/- 5 %, n = 34; excised outside-out: 100 +/- 0 %, n = 26), 0.1 mM verapamil (excised: 73 +/- 9 %, n = 12), and 10 nM charybdotoxin (excised outside-out: 67 +/- 9 %, n = 9). 4. The K+ channel was activated by depolarization and rising cytosolic Ca2+. Half-maximal activity occurred at a cytosolic Ca2+ concentration of 200 nM. In the cell-attached membrane configuration the K+ channel was inhibited in a concentration-dependent manner by atrial natriuretic peptide (ANP). Powas blocked equally well by 10 nM ANP (52 +/- 7 %, n = 10), brain natriuretic peptide (BNP; 37 +/- 11 %, n = 6) and C-type natriuretic peptide (CNP; 44 +/- 13 %, n = 8). 8-Bromoguanosine 3',5' cyclic monophosphate (8-Br-cGMP, 0.1 mM) also inhibited Poof this K+ channel, by 70 +/- 10 % (n = 5). 5. In excised membrane patches cGMP inhibited Po of this K+ channel in a concentration-dependent manner. The first significant effects were measured at a concentration of 1 microM (22 +/- 7 %, n = 6), and greatest effects were obtained at 0.1 mM (34 +/- 5 %, n = 15). cAMP (0.1 mM, n = 5) as well as GTP (0.1 mM, n = 5) had no significant effects on Po of this K+ channel. ATP (0.1 mM) had a weak inhibitory effect (17 +/- 5 %, n = 14). Addition of Mg-ATP to cGMP did not increase the inhibitory effect (30 +/- 4 %, n = 14). KT5823 (1 microM), a specific inhibitor of cGMP-dependent protein kinases, did not significantly alter the cGMP-induced reduction in Po of the K+ channel in three excised membrane patches. 6. The results present the first electrophysiological characterization of a mammalian K+ channel that is directly regulated by cGMP.


Assuntos
GMP Cíclico/fisiologia , Túbulos Renais Proximais/fisiologia , Canais de Potássio/fisiologia , Fator Natriurético Atrial/farmacologia , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Humanos , Canais de Potássio/efeitos dos fármacos , Compostos de Tetraetilamônio/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA