Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Mol Sci ; 22(4)2021 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-33671654

RESUMO

Sphingosine-1-phosphate (S1P), is a signaling sphingolipid which acts as a bioactive lipid mediator. We assessed whether S1P had multiplex effects in regulating the large-conductance Ca2+-activated K+ channel (BKCa) in catecholamine-secreting chromaffin cells. Using multiple patch-clamp modes, Ca2+ imaging, and computational modeling, we evaluated the effects of S1P on the Ca2+-activated K+ currents (IK(Ca)) in bovine adrenal chromaffin cells and in a pheochromocytoma cell line (PC12). In outside-out patches, the open probability of BKCa channel was reduced with a mean-closed time increment, but without a conductance change in response to a low-concentration S1P (1 µM). The intracellular Ca2+ concentration (Cai) was elevated in response to a high-dose (10 µM) but not low-dose of S1P. The single-channel activity of BKCa was also enhanced by S1P (10 µM) in the cell-attached recording of chromaffin cells. In the whole-cell voltage-clamp, a low-dose S1P (1 µM) suppressed IK(Ca), whereas a high-dose S1P (10 µM) produced a biphasic response in the amplitude of IK(Ca), i.e., an initial decrease followed by a sustained increase. The S1P-induced IK(Ca) enhancement was abolished by BAPTA. Current-clamp studies showed that S1P (1 µM) increased the action potential (AP) firing. Simulation data revealed that the decreased BKCa conductance leads to increased AP firings in a modeling chromaffin cell. Over a similar dosage range, S1P (1 µM) inhibited IK(Ca) and the permissive role of S1P on the BKCa activity was also effectively observed in the PC12 cell system. The S1P-mediated IK(Ca) stimulation may result from the elevated Cai, whereas the inhibition of BKCa activity by S1P appears to be direct. By the differentiated tailoring BKCa channel function, S1P can modulate stimulus-secretion coupling in chromaffin cells.


Assuntos
Cálcio/metabolismo , Células Cromafins/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Bovinos , Sistema Livre de Células , Células Cromafins/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletrofisiologia/métodos , Lisofosfolipídeos/administração & dosagem , Lisofosfolipídeos/farmacologia , Células PC12 , Ratos , Esfingosina/administração & dosagem , Esfingosina/metabolismo , Esfingosina/farmacologia
2.
J Mol Cell Cardiol ; 123: 185-197, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30261161

RESUMO

AIMS: Phospholamban (PLB) is the key regulator of the cardiac Ca2+ pump (SERCA2a)-mediated sarcoplasmic reticulum Ca2+ stores. We recently reported that PLB is highly concentrated in the nuclear envelope (NE) from where it can modulate perinuclear Ca2+ handling of the cardiomyocytes (CMs). Since inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) mediates nuclear Ca2+ release, we examined whether the nuclear pool of PLB regulates IP3-induced nuclear Ca2+ handling. METHODS AND RESULTS: Fluo-4 based confocal Ca2+ imaging was performed to measure Ca2+ dynamics across both nucleus and cytosol in saponin-permeabilized CMs isolated from wild-type (WT) or PLB-knockout (PLB-KO) mice. At diastolic intracellular Ca2+ ([Ca2+]i = 100 nM), the Fab fragment of the monoclonal PLB antibody (anti-PLB Fab) facilitated the formation and increased the length of spontaneous Ca2+ waves (SCWs) originating from the nuclear region in CMs from WT but not from PLB-KO mice. We next examined nuclear Ca2+ activities at basal condition and after sequential addition of IP3, anti-PLB Fab, and the IP3R inhibitor 2-aminoethoxydiphenyl borate (2-APB) at a series of [Ca2+]i. In WT mice, at 10 nM [Ca2+]i where ryanodine receptor (RyR2) based spontaneous Ca2+ sparks rarely occurred, IP3 increased fluorescence amplitude (F/F0) of overall nuclear region to 1.19 ±â€¯0.02. Subsequent addition of anti-PLB Fab significantly decreased F/F0 to 1.09 ±â€¯0.02. At 50 nM [Ca2+]i, anti-PLB Fab not only decreased the overall nuclear F/F0 previously elevated by IP3, but also increased the amplitude and duration of spark-like nuclear Ca2+ release events. These nuclear Ca2+ releases were blocked by 2-APB. At 100 nM [Ca2+]i, IP3 induced short SCWs originating from nucleus. Anti-PLB Fab transformed those short waves into long SCWs with propagation from the nucleus into the cytosol. In contrast, neither nuclear nor cytosolic Ca2+ dynamics was affected by anti-PLB Fab in CMs from PLB-KO mice in all these conditions. Furthermore, in WT CMs pretreated with RyR2 blocker tetracaine, IP3 and anti-PLB Fab still increased the magnitude of nuclear Ca2+ release but failed to regenerate SCWs. Finally, anti-PLB Fab increased low Ca2+ affinity mag-fluo 4 fluorescence intensity in the lumen of NE of nuclei isolated from WT but not in PLB-KO mice. CONCLUSION: PLB regulates nuclear Ca2+ handling. By increasing Ca2+ uptake into lumen of the NE and perhaps other perinuclear membranes, the acute reversal of PLB inhibition decreases global Ca2+ concentration at rest in the nucleoplasm, and increases Ca2+ release into the nucleus, through mechanisms involving IP3R and RyR2 in the vicinity.


Assuntos
Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Núcleo Celular/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Cães , Camundongos , Camundongos Knockout , Imagem Molecular/métodos , Miócitos Cardíacos/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Tetracaína/farmacologia
3.
J Mol Cell Cardiol ; 100: 1-8, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27642167

RESUMO

AIMS: Phospholamban (PLB) regulates the cardiac Ca2+-ATPase (SERCA2a) in sarcoplasmic reticulum (SR). However, the localization of PLB at subcellular sites outside the SR and possible contributions to Ca2+ cycling remain unknown. We examined the intracellular distribution of PLB and tested whether a pool of PLB exists in the nuclear envelope (NE) that might regulate perinuclear/nuclear Ca2+ (nCa2+) handling in cardiomyocytes (CMs). METHODS AND RESULTS: Using confocal immunofluorescence microscopy and immunoblot analyses of CMs and CM nuclei, we discovered that PLB was highly concentrated in NE. Moreover, the ratio of PLB levels to SERCA levels was greater in NE than in SR. The increased levels of PLB in NE were a consistent finding using a range of antibodies, tissue samples, and species. To address a possible role in affecting Ca2+ handling, we used Fluo-4 based confocal Ca2+ imaging, with scan-lines across cytosol and nuclei, and evaluated the effects of PLB on cytosolic and nCa2+ uptake and release in mouse CMs. In intact CMs, isoproterenol increased amplitude and decreased the decay time of Ca2+ transients not only in cytosol but also in nuclear regions. In saponin-permeabilized mouse CMs ([Ca2+]i=400nM), we measured spontaneous Ca2+ waves after specific reversal of PLB activity by addition of the Fab fragment of an anti-PLB monoclonal antibody (100µg/ml). This highly selective immunological reagent enhanced Ca2+ uptake (faster decay times) and Ca2+ release (greater intensity) in both cytosol and across the nuclear regions. CONCLUSIONS: Besides SR, PLB is concentrated in NE of CMs, and may be involved in modulation of nCa2+ dynamics.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Miócitos Cardíacos/metabolismo , Membrana Nuclear/metabolismo , Animais , Transporte Biológico , Sinalização do Cálcio/efeitos dos fármacos , Nucléolo Celular/metabolismo , Humanos , Espaço Intracelular/metabolismo , Isoproterenol/farmacologia , Camundongos , Microscopia de Fluorescência , Imagem Molecular , Miócitos Cardíacos/efeitos dos fármacos , Coelhos , Especificidade da Espécie
4.
Heart Rhythm ; 18(1): 88-97, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32707174

RESUMO

BACKGROUND: Female sex is a known risk factor for drug-induced long QT syndrome (diLQTS). We recently demonstrated a sex difference in apamin-sensitive small-conductance Ca2+-activated K+ current (IKAS) activation during ß-adrenergic stimulation. OBJECTIVE: The purpose of this study was to test the hypothesis that there is a sex difference in IKAS in the rabbit models of diLQTS. METHODS: We evaluated the sex difference in ventricular repolarization in 15 male and 22 female Langendorff-perfused rabbit hearts with optical mapping techniques during atrial pacing. HMR1556 (slowly activating delayed rectifier K+ current [IKs] blocker), E4031 (rapidly activating delayed rectifier K+ current [IKr] blocker) and sea anemone toxin (ATX-II, late Na+ current [INaL] activator) were used to simulate types 1-3 long QT syndrome, respectively. Apamin, an IKAS blocker, was then added to determine the magnitude of further QT prolongation. RESULTS: HMR1556, E4031, and ATX-II led to the prolongation of action potential duration at 80% repolarization (APD80) in both male and female ventricles at pacing cycle lengths of 300-400 ms. Apamin further prolonged APD80 (pacing cycle length 350 ms) from 187.8±4.3 to 206.9±7.1 (P=.014) in HMR1556-treated, from 209.9±7.8 to 224.9±7.8 (P=.003) in E4031-treated, and from 174.3±3.3 to 188.1±3.0 (P=.0002) in ATX-II-treated female hearts. Apamin did not further prolong the APD80 in male hearts. The Cai transient duration (CaiTD) was significantly longer in diLQTS than baseline but without sex differences. Apamin did not change CaiTD. CONCLUSION: We conclude that IKAS is abundantly increased in female but not in male ventricles with diLQTS. Increased IKAS helps preserve the repolarization reserve in female ventricles treated with IKs and IKr blockers or INaL activators.


Assuntos
Ventrículos do Coração/efeitos dos fármacos , Síndrome do QT Longo/metabolismo , Miocárdio/metabolismo , Animais , Apamina/toxicidade , Diagnóstico por Imagem , Modelos Animais de Doenças , Feminino , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/patologia , Masculino , Miocárdio/patologia , Técnicas de Patch-Clamp , Coelhos , Fatores Sexuais , Canais de Potássio Ativados por Cálcio de Condutância Baixa
5.
Heart Rhythm ; 17(2): 332-340, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31513946

RESUMO

BACKGROUND: Ondansetron, a widely prescribed antiemetic, has been implicated in drug-induced long QT syndrome. Recent patch clamp experiments have shown that ondansetron inhibits the apamin-sensitive small conductance calcium-activated potassium current (IKAS). OBJECTIVE: The purpose of this study was to determine whether ondansetron causes action potential duration (APD) prolongation by IKAS inhibition. METHODS: Optical mapping was performed in rabbit hearts with pacing-induced heart failure (HF) and in normal hearts before and after ondansetron (100 nM) infusion. APD at 80% repolarization (APD80) and arrhythmia inducibility were determined. Additional studies with ondansetron were performed in normal hearts perfused with hypokalemic Tyrode's (2.4 mM) solution before or after apamin administration. RESULTS: The corrected QT interval in HF was 326 ms (95% confidence interval [CI] 306-347 ms) at baseline and 364 ms (95% CI 351-378 ms) after ondansetron infusion (P < .001). Ondansetron significantly prolonged APD80 in the HF group and promoted early afterdepolarizations, steepened the APD restitution curve, and increased ventricular vulnerability. Ventricular fibrillation was not inducible in HF ventricles at baseline, but after ondansetron infusion, ventricular fibrillation was induced in 5 of the 7 ventricles (P = .021). In hypokalemia, apamin prolonged APD80 from 163 ms (95% CI 146-180 ms) to 180 ms (95% CI 156-204 ms) (P = .018). Subsequent administration of ondansetron failed to further prolong APD80 (180 ms [95% CI 156-204 ms] vs 179 ms [95% CI 165-194 ms]; P = .789). The results were similar when ondansetron was administered first, followed by apamin. CONCLUSION: Ondansetron is a specific IKAS blocker at therapeutic concentrations. Ondansetron may prolong the QT interval in HF by inhibiting small conductance calcium-activated potassium channels, which increases the vulnerability to ventricular arrhythmias.


Assuntos
Estimulação Cardíaca Artificial , Insuficiência Cardíaca/terapia , Ventrículos do Coração/fisiopatologia , Ondansetron/farmacologia , Fibrilação Ventricular/complicações , Potenciais de Ação , Animais , Apamina/farmacologia , Modelos Animais de Doenças , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Técnicas de Patch-Clamp , Coelhos , Antagonistas da Serotonina/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Fibrilação Ventricular/fisiopatologia
6.
Heart Rhythm ; 15(5): 761-769, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29325977

RESUMO

BACKGROUND: Apamin-sensitive small conductance calcium-activated K current (IKAS) is up-regulated during ventricular pacing and masks short-term cardiac memory (CM). OBJECTIVE: The purpose of this study was to determine the role of IKAS in long-term CM. METHODS: CM was created with 3-5 weeks of ventricular pacing and defined by a flat or inverted T wave off pacing. Epicardial optical mapping was performed in both paced and normal ventricles. Action potential duration (APD80) was determined during right atrial pacing. Ventricular stability was tested before and after IKAS blockade. Four paced hearts and 4 normal hearts were used for western blotting and histology. RESULTS: There were no significant differences in either echocardiographic parameters or fibrosis levels between groups. Apamin induced more APD80 prolongation in CM than in normal ventricles (mean [95% confidence interval]: 9.6% [8.8%-10.5%] vs 3.1% [1.9%-4.3%]; P <.001). Apamin significantly lengthened APD80 in the CM model at late activation sites, indicating significant IKAS up-regulation at those sites. The CM model also had altered Ca2+ handling, with the 50% Ca2+ transient duration and amplitude increased at distal sites compared to a proximal site (near the pacing site). After apamin, the CM model had increased ventricular fibrillation (VF) inducibility (paced vs control: 33/40 (82.5%) vs 7/20 (35%); P <.001) and longer VF durations (124 vs 26 seconds; P <.001). CONCLUSION: Chronic ventricular pacing increases Ca2+ transients at late activation sites, which activates IKAS to maintain repolarization reserve. IKAS blockade increases VF vulnerability in chronically paced rabbit ventricles.


Assuntos
Apamina/farmacologia , Ventrículos do Coração/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Fibrilação Ventricular/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Animais , Western Blotting , Modelos Animais de Doenças , Ecocardiografia , Feminino , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/efeitos dos fármacos , Imuno-Histoquímica , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , RNA/genética , Coelhos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Fibrilação Ventricular/diagnóstico , Fibrilação Ventricular/genética
7.
JCI Insight ; 3(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30429367

RESUMO

The mechanisms of J wave syndrome (JWS) are incompletely understood. Here, we showed that the concomitant activation of small-conductance calcium-activated potassium (SK) current (IKAS) and inhibition of sodium current by cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA) recapitulate the phenotypes of JWS in Langendorff-perfused rabbit hearts. CyPPA induced significant J wave elevation and frequent spontaneous ventricular fibrillation (SVF), as well as sinus bradycardia, atrioventricular block, and intraventricular conduction delay. IKAS activation by CyPPA resulted in heterogeneous shortening of action potential (AP) duration (APD) and repolarization alternans. CyPPA inhibited cardiac sodium current (INa) and decelerated AP upstroke and intracellular calcium transient. SVFs were typically triggered by short-coupled premature ventricular contractions, initiated with phase 2 reentry and originated more frequently from the right than the left ventricles. Subsequent IKAS blockade by apamin reduced J wave elevation and eliminated SVF. ß-Adrenergic stimulation was antiarrhythmic in CyPPA-induced electrical storm. Like CyPPA, hypothermia (32.0°C) also induced J wave elevation and SVF. It facilitated negative calcium-voltage coupling and phase 2 repolarization alternans with spatial and electromechanical discordance, which were ameliorated by apamin. These findings suggest that IKAS activation contributes to the development of JWS in rabbit ventricles.


Assuntos
Arritmias Cardíacas/etiologia , Doença do Sistema de Condução Cardíaco/etiologia , Sistema de Condução Cardíaco , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Sódio/metabolismo , Animais , Feminino , Masculino , Potássio/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Coelhos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Síndrome , Taquicardia Ventricular/etiologia , Fibrilação Ventricular/etiologia
8.
Life Sci ; 80(4): 378-87, 2007 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17097686

RESUMO

The ATP-sensitive K(+) (K(ATP)) channels are known to provide a functional linkage between the electrical activity of the cell membrane and metabolism. Two types of inwardly rectifying K(+) channel subunits (i.e., Kir6.1 and Kir6.2) with which sulfonylurea receptors are associated were reported to constitute the K(ATP) channels. In this study, we provide evidence to show two types of K(ATP) channels with different biophysical properties functionally expressed in isolated rat ventricular myocytes. Using patch-clamp technique, we found that single-channel conductance for the different two types of K(ATP) channels in these cells was 57 and 21 pS. The kinetic properties, including mean open time and bursting kinetics, did not differ between these two types of K(ATP) channels. Diazoxide only activated the small-conductance K(ATP) channel, while pinacidil and dinitrophenol stimulated both channels. Both of these K(ATP) channels were sensitive to block by glibenclamide. Additionally, western blotting, immunochemistry, and RT-PCR revealed two types of Kir6.X channels, i.e., Kir6.1 and Kir6.2, in rat ventricular myocytes. Single-cell Ca(2+) imaging also revealed that similar to dinitrophenol, diazoxide reduced the concentration of intracellular Ca(2+). The present results suggest that these two types of K(ATP) channels may functionally be related to the activity of heart cells.


Assuntos
Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Diazóxido/farmacologia , Dinitrofenóis/farmacologia , Relação Dose-Resposta a Droga , Técnica Direta de Fluorescência para Anticorpo , Expressão Gênica/efeitos dos fármacos , Glibureto/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Canais KATP , Masculino , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Pinacidil/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Curr Top Med Chem ; 6(10): 1025-30, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16787277

RESUMO

The gating of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channel is primarily controlled by intracellular Ca(2+) and/or membrane depolarization. These channels play a role in the coupling of excitation-contraction and stimulus-secretion. A variety of structurally distinct compounds may influence the activity of these channels. Squamocin, an Annonaceous acetogenin, could interact with the BK(Ca) channel to increase the amplitude of Ca(2+)-activated K(+) current in coronary smooth muscle cells. Its stimulatory effect is related to intracellular Ca(2+) concentrations. In inside-out patches, application of ceramide to the bath suppressed the activity of BK(Ca) channels recorded from pituitary GH(3) cells and from retinal pigment epithelial cells. ICI-182,780, an estrogen receptor antagonist, was found to modulate BK(Ca)-channel activity in cultured endothelial cells and smooth muscle cells in a mechanism unlinked to the inhibition of estrogen receptors. Caffeic acid phenethyl ester (CAPE) and its analogy, cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate, could directly increase the activity of BK(Ca) channels in GH(3) cells. CAPE also reduced the frequency and amplitude of intracellular Ca(2+) oscillations in these cells. The CAPE-stimulated activity in BK(Ca) channels is thought to be unassociated with its inhibition of NF-kappaB activation. Cilostazol, an inhibitor of cyclic nucleotide phosphodiesterase, could stimulate BK(Ca) channel-activity and reduce the firing of action currents simultaneously in GH(3) cells. Therefore, the regulation by these compounds of BK(Ca) channels may in part be responsible for their regulatory actions on cell functions.


Assuntos
Canais de Potássio Cálcio-Ativados/efeitos dos fármacos , Animais , Furanos/farmacologia , Humanos , Lactonas/farmacologia
10.
Chin J Physiol ; 49(1): 1-13, 2006 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-16900700

RESUMO

The effects of changes in membrane cholesterol on ion currents were investigated in pituitary GH3 cells. Depletion of membrane cholesterol by exposing cells to methyl-beta-cyclodextrin (MbetaCD), an oligosaccharide, resulted in an increase in the density of Ca2+-activated K+ current (IK(Ca)). However, no significant change in IK(Ca) density was demonstrated in GH3 cells treated with a mixture of MbetaCD and cholesterol. Cholesterol depletion with MbetaCD (1.5 mg/ml) slightly suppressed the density of voltage-dependent L-type Ca2+ current. In inside-out patches recorded from MbetaCD-treated cells, the activity of large-conductance Ca2+-activated K+ (BK(Ca)) channels was enhanced with no change in single-channel conductance. In MbetaCD-treated cells, voltage-sensitivity of BK(Ca) channels was increased; however, no change in Ca2+-sensitivity could be demonstrated. A negative correlation between adjacent closed and open times in BK(Ca) channels was observed in MbetaCD-treated cells. In inside-out patches from MbetaCD-treated cells, dexamethasone (30 microM) applied to the intracellular surface did not increase BK(Ca)-channel activity, although caffeic acid phenethyl ester and cilostazol still opened its probability effectively. However, no modification in the activity of ATP-sensitive K+ channels could be seen in MbetaCD-treated cells. Current-clamp recordings demonstrated that the cholesterol depletion maneuver with MbetaCD reduced the firing of action potentials. Therefore, the increase in BK(Ca)-channel activity induced by membrane depletion may influence the functional activities of neurons or neuroendocrine cells if similar results occur in vivo.


Assuntos
Colesterol/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Lipídeos de Membrana/fisiologia , Neoplasias Hipofisárias/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Animais , Apamina/farmacologia , Ácidos Cafeicos/farmacologia , Cálcio/farmacologia , Cilostazol , Dexametasona/farmacologia , Diazóxido/farmacologia , Glibureto/farmacologia , Indóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/farmacologia , Ratos , Tetrazóis/farmacologia , Células Tumorais Cultivadas , beta-Ciclodextrinas/farmacologia
11.
Eur J Pharmacol ; 523(1-3): 16-24, 2005 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-16243310

RESUMO

The effects of paeoniflorin, a glycoside isolated from the root of Paeonia lactiflora, on ion currents in a mouse neuroblastoma and rat glioma hybrid cell line, NG108-15 were investigated. Paeoniflorin (1-300 microM) reversibly produced an inhibition of L-type voltage-dependent Ca2+ current (I(Ca,L)) in a concentration-dependent manner. Paeoniflorin caused no change in the overall shape of the current-voltage relationship of I(Ca,L). The IC50 value of paeoniflorin-induced inhibition of I(Ca,L) was 14 microM. However, neither adenosine deaminase (1 U/ml) nor 8-cyclopentyl-1, 3-dipropylxanthine (10 microM) could reverse the inhibition by paeoniflorin of I(Ca,L). Paeoniflorin (30 microM) shifted the steady-state inactivation curve of I(Ca,L) to more negative membrane potentials by approximately -10 mV. It also prolonged the recovery of I(Ca,L). The inhibitory effect of paeoniflorin on I(Ca,L) exhibited tonic and use-dependent characteristics. Paeoniflorin could effectively suppress I(Ca,L) evoked by action potential waveforms. Paeoniflorin at a concentration of 30 microM produce a slight inhibition of voltage-dependent Na+ current and delayed rectifier K+ current. Under current-clamp configuration, unlike adenosine, this compound decreased the firing of action potentials. Taken together, this study indicates that paeoniflorin can block L-type Ca2+ channels in NG108-15 cells in a mechanism unlinked to the binding to adenosine receptors. The effects of paeoniflorin on ion currents may partly, if not entirely, contribute to the underlying mechanisms through which it affects neuronal or neuroendocrine function.


Assuntos
Benzoatos/farmacologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Glucosídeos/farmacologia , Paeonia/química , Potenciais de Ação/efeitos dos fármacos , Animais , Benzoatos/isolamento & purificação , Hidrocarbonetos Aromáticos com Pontes/isolamento & purificação , Bloqueadores dos Canais de Cálcio/isolamento & purificação , Canais de Cálcio Tipo L/metabolismo , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/metabolismo , Relação Dose-Resposta a Droga , Glucosídeos/isolamento & purificação , Camundongos , Monoterpenos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Raízes de Plantas , Potássio/metabolismo , Ratos , Sódio/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Fatores de Tempo , Células Tumorais Cultivadas
12.
J Neurochem ; 102(3): 944-56, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17630986

RESUMO

Bioactive lysophospholipids (LPLs) are released by blood cells and can modulate many cellular activities such as angiogenesis and cell survival. In this study, the effects of sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) on excitability and exocytosis in bovine chromaffin cells were investigated using the whole-cell configuration of the patch-clamp. Voltage-gated Ca(2+) current was inhibited by S1P and LPA pre-treatment in a concentration-dependent manner with IC(50)s of 0.46 and 0.79 mumol/L, respectively. Inhibition was mostly reversible upon washout and prevented by suramin, an inhibitor of G-protein signaling. Na(+) current was inhibited by S1P, but not by LPA. However, recovery of Na(+) channels from inactivation was slowed by both LPLs. The outward K(+) current was also significantly reduced by both LPLs. Chromaffin cells fired repetitive action potentials in response to minimal injections of depolarizing current. Repetitive activity was dramatically reduced by LPLs. Consistent with the reduction in Ca(2+) current, exocytosis elicited by a train of depolarizations and the ensuing endocytosis were both inhibited by LPL pre-treatments. These data demonstrate the interaction between immune and endocrine systems mediated by the inhibitory effects of LPLs on the excitability of adrenal chromaffin cells.


Assuntos
Medula Suprarrenal/metabolismo , Membrana Celular/metabolismo , Células Cromafins/metabolismo , Lisofosfolipídeos/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Medula Suprarrenal/ultraestrutura , Animais , Bovinos , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Células Cromafins/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estimulação Elétrica , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Lisofosfolipídeos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia , Suramina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa