Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Kidney Res Clin Pract ; 35(1): 15-21, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27069853

RESUMEN

BACKGROUND: Chronic treatment with the dietary flavonoid quercetin is known to lower blood pressure and restore endothelial dysfunction in animal models of hypertension. This study investigated the direct effects of quercetin on vascular response in chronic 2-kidney, 1-clip (2K1C) renal hypertensive rats. The effects of antioxidant vitamin ascorbic acid on the vasoreactivity were also examined. METHODS: 2K1C renal hypertension was induced by clipping the left renal artery; age-matched rats that received sham treatment served as controls. Thoracic aortae were mounted in tissue baths for the measurement of isometric tension. RESULTS: Relaxant responses to acetylcholine were significantly attenuated in 2K1C rats in comparison with sham rats. Quercetin or ascorbic acid augmented acetylcholine-induced relaxation in 2K1C rats, whereas no significant differences were noted in sham rats. The relaxation response to sodium nitroprusside was comparable between 2K1C and sham rats, and sodium nitroprusside-induced relaxation was not altered by quercetin or ascorbic acid in either group. The contractile response to phenylephrine was significantly enhanced in 2K1C rats compared with sham rats. Phenylephrine-induced contraction was inhibited by pretreatment with quercetin or ascorbic acid in 2K1C rats, whereas neither chemical affected responses in sham rats. N(w)-nitro-L-arginine methyl ester markedly augmented the contractile response to phenylephrine in sham rats, whereas no significant differences were observed in 2K1C rats. Quercetin or ascorbic acid did not affect phenylephrine-induced contraction in the presence of N(w)-nitro-L-arginine methyl ester in either 2K1C or sham rats. CONCLUSION: Acute exposure to quercetin appears to improve endothelium-dependent relaxation and inhibit the contractile response, similar to the effect of ascorbic acid in 2K1C hypertension. These results partially explain the vascular beneficial effects of quercetin in renal hypertension.

2.
Kidney Res Clin Pract ; 33(1): 19-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26877946

RESUMEN

BACKGROUND: Endothelial dysfunction is linked to exaggerated production of superoxide anions. This study was conducted to examine the effects of oxidative stress on endothelial modulation of contractions in chronic two-kidney, one-clip (2K1C) renal hypertensive rats. METHODS: The 2K1C hypertension was induced by clipping the left renal artery; age-matched rats receiving sham treatment served as controls. Thoracic aortae were isolated and mounted in tissue baths for measurement of isometric tension. RESULTS: Norepinephrine-induced contraction was augmented by the removal of the endothelium, which was more pronounced in sham rats than in 2K1C rats. Nω-nitro-L-arginine methyl ester, an inhibitor of nitric oxide production, had a similar augmenting effect. Vitamin C inhibited the contraction in aortic rings with intact endothelium from 2K1C rats but not from sham rats. The contraction was also suppressed by treatment with diphenyleneiodonium or apocynin, inhibitors of nicotinamide adenine dinucleotide/nicotinamide adenine dinucleotide phosphate (NADH/NADPH) oxidase, in the aortae with intact endothelium from 2K1C rats but not in those from sham rats. Superoxide anions generated by xanthine oxidase/hypoxanthine enhanced the contraction in the aortae with intact endothelium from sham rats, but had no effect in 2K1C rats. Enhanced contractile responses to norepinephrine by xanthine oxidase/hypoxanthine in sham rats were reversed by vitamin C. CONCLUSION: These results suggest that the effect on endothelial modulation of endothelium-derived nitric oxide is impaired in 2K1C hypertension. The impairment is, at least in part, related to increased production of superoxide anions by NADH/NADPH oxidase.

3.
Kidney Res Clin Pract ; 33(4): 181-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26885474

RESUMEN

BACKGROUND: The plant-derived estrogen biochanin A is known to cause vasodilation, but its mechanism of action in hypertension remains unclear. This study was undertaken to investigate the effects and mechanisms of biochanin A on the thoracic aorta in two-kidney, one clip (2K1C) renovascular hypertensive rats. METHODS: Hypertension was induced by clipping the left renal artery, and control age-matched rats were sham treated. Thoracic aortae were mounted in tissue baths to measure isometric tension. RESULTS: Biochanin A caused concentration-dependent relaxation in aortic rings from 2K1C hypertensive and sham-treated rats, which was greater in 2K1C rats than in sham rats. Biochanin A-induced relaxation was significantly attenuated by removing the endothelium in aortic rings from 2K1C rats, but not in sham rats. N (ω)-Nitro-l-arginine methyl ester, a nitric oxide synthase inhibitor, or indomethacin, a cyclooxygenase inhibitor, did not affect the biochanin A-induced relaxation in aortic rings from 2K1C and sham rats. By contrast, treatment with glibenclamide, a selective inhibitor of adenosine triphosphate-sensitive K(+) channels, or tetraethylammonium, an inhibitor of Ca(2+)-activated K(+) channels, significantly reduced biochanin A-induced relaxation in aortic rings from both groups. However, 4-aminopyridine, a selective inhibitor of voltage-dependent K(+) channels, inhibited the relaxation induced by biochanin A in 2K1C rats, whereas no significant differences were observed in sham rats. CONCLUSION: These results suggest that the enhanced relaxation caused by biochanin A in aortic rings from hypertensive rats is endothelium dependent. Vascular smooth muscle K(+) channels may be involved in biochanin A-induced relaxation in aortae from hypertensive and normotensive rats. In addition, an endothelium-derived activation of voltage-dependent K(+) channels contributes, at least in part, to the relaxant effect of biochanin A in renovascular hypertension.

4.
Mol Cells ; 33(5): 509-16, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22441675

RESUMEN

Neurotensin, a tridecapeptide localized in the gut to discrete enteroendocrine cells of the small bowel mucosa, is a hormone that plays an important role in gastrointestinal secretion, growth, and motility. Neurotensin has inhibitory and excitatory effects on peristaltic activity and produces contractile and relaxant responses in intestinal smooth muscle. Our objective in this study is to investigate the effects of neurotensin in small intestinal interstitial cells of Cajal (ICC) and elucidate the mechanism. To determine the electrophysiological effects of neurotensin on ICC, whole-cell patch clamp recordings were performed in cultured ICC from the small intestine. Exposure to neurotensin depolarized the membrane of pacemaker cells and produced tonic inward pacemaker currents. Only neurotensin receptor1 was identified when RT-PCR and immunocytochemistry were performed with mRNA isolated from small intestinal ICC and c-Kit positive cells. Neurotensin-induced tonic inward pacemaker currents were blocked by external Na⁺-free solution and in the presence of flufenamic acid, an inhibitor of non-selective cation channels. Furthermore, neurotensin-induced action is blocked either by treatment with U73122, a phospholipase C inhibitor, or thapsigargin, a Ca²âº-ATPase inhibitor in ICC. We found that neurotensin increased spontaneous intracellular Ca²âº oscillations as seen with fluo4/AM recording. These results suggest that neurotensin modulates pacemaker currents via the activation of non-selective cation channels by intracellular Ca²âº-release through neurotensin receptor1.


Asunto(s)
Células Intersticiales de Cajal/metabolismo , Intestino Delgado/metabolismo , Músculo Liso/metabolismo , Neurotensina/metabolismo , Neurotensina/farmacología , Animales , Calcio/metabolismo , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/metabolismo , Células Cultivadas , Retículo Endoplásmico/metabolismo , Femenino , Técnicas In Vitro , Células Intersticiales de Cajal/efectos de los fármacos , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Canales Iónicos/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Receptores de Neurotensina/metabolismo , Sodio/metabolismo , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
5.
Chonnam Med J ; 47(2): 72-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22111064

RESUMEN

We studied whether nitric oxide (NO) and hydrogen sulfide (H(2)S) have an interaction on the pacemaker activities of interstitial cells of Cajal (ICC) from the mouse small intestine. The actions of NO and H(2)S on pacemaker activities were investigated by using the whole-cell patch-clamp technique and intracellular Ca(2+) analysis at 30℃ in cultured mouse ICC. Exogenously applied (±)-S-nitroso-N-acetylpenicillamine (SNAP), an NO donor, or sodium hydrogen sulfide (NaHS), a donor of H(2)S, showed no influence on pacemaker activity (potentials and currents) in ICC at low concentrations (10 µM SNAP and 100 µM NaHS), but SNAP or NaHS completely inhibited pacemaker amplitude and pacemaker frequency with increases in the resting currents in the outward direction at high concentrations (SNAP 100 µM and NaHS 1 mM). Co-treatment with 10 µM SNAP plus 100 µM NaHS also inhibited pacemaker amplitude and pacemaker frequency with increases in the resting currents in the outward direction. ODQ, a guanylate cyclase inhibitor, or glibenclamide, an ATP-sensitive K(+) channel inhibitor, blocked the SNAP+NaHS-induced inhibition of pacemaker currents in ICC. Also, we found that SNAP+NaHS inhibited the spontaneous intracellular Ca(2+) ([Ca(2+)](i)) oscillations in cultured ICC. In conclusion, this study describes the enhanced inhibitory effects of NO plus H(2)S on ICC in the mouse small intestine. NO+H(2)S inhibited the pacemaker activity of ICC by modulating intracellular Ca(2+). These results may be evidence of a physiological interaction of NO and H(2)S in ICC for modulating gastrointestinal motility.

6.
Korean J Physiol Pharmacol ; 15(3): 129-35, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21860590

RESUMEN

In this study we determined whether or not 5-hydroxytryptamine (5-HT) has an effect on the pacemaker activities of interstitial cells of Cajal (ICC) from the mouse small intestine. The actions of 5-HT on pacemaker activities were investigated using a whole-cell patch-clamp technique, intracellular Ca(2+) ([Ca(2+)](i)) analysis, and RT-PCR in ICC. Exogenously-treated 5-HT showed tonic inward currents on pacemaker currents in ICC under the voltage-clamp mode in a dose-dependent manner. Based on RT-PCR results, we found the existence of 5-HT(2B, 3, 4, and 7) receptors in ICC. However, SDZ 205557 (a 5-HT(4) receptor antagonist), SB 269970 (a 5-HT7 receptor antagonist), 3-tropanylindole - 3 - carboxylate methiodide (3-TCM; a 5-HT(3) antagonist) blocked the 5-HT-induced action on pacemaker activity, but not SB 204741 (a 5-HT(2B) receptor antagonist). Based on [Ca(2+)](i) analysis, we found that 5-HT increased the intensity of [Ca(2+)](i). The treatment of PD 98059 or JNK II inhibitor blocked the 5-HT-induced action on pacemaker activity of ICC, but not SB 203580. In summary, these results suggest that 5-HT can modulate pacemaker activity through 5-HT(3, 4, and 7) receptors via [Ca(2+)](i) mobilization and regulation of mitogen-activated protein kinases.

7.
World J Gastroenterol ; 17(9): 1143-51, 2011 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-21448418

RESUMEN

AIM: To explore the role of prostaglandin F(2α) (PGF(2α))) on pacemaker activity in interstitial cells of Cajal (ICC) from mouse small intestine. METHODS: In this study, effects of PGF(2α) in the cultured ICC cells were investigated with patch clamp technology combined with Ca(2+) image analysis. RESULTS: Externally applied PGF(2α) (10 µmol/L) produced membrane depolarization in current-clamp mode and increased tonic inward pacemaker currents in voltage-clamp mode. The application of flufenamic acid (a non-selective cation channel inhibitor) or niflumic acid (a Cl(-) channel inhibitor) abolished the generation of pacemaker currents but only flufenamic acid inhibited the PGF(2α)-induced tonic inward currents. In addition, the tonic inward currents induced by PGF(2α) were not inhibited by intracellular application of 5'-[-thio]diphosphate trilithium salt. Pretreatment with Ca(2+) free solution, U-73122, an active phospholipase C inhibitor, and thapsigargin, a Ca(2+)-ATPase inhibitor in endoplasmic reticulum, abolished the generation of pacemaker currents and suppressed the PGF(2α)-induced tonic inward currents. However, chelerythrine or calphostin C, protein kinase C inhibitors, did not block the PGF(2α)-induced effects on pacemaker currents. When recording intracellular Ca(2+) ([Ca(2+)](i)) concentration using fluo-3/AM, PGF(2α) broadly increased the spontaneous [Ca(2+)](i) oscillations. CONCLUSION: These results suggest that PGF(2α) can modulate pacemaker activity of ICC by acting non-selective action channels through phospholipase C-dependent pathway via [Ca(2+)]i regulation.


Asunto(s)
Dinoprost/farmacología , Células Intersticiales de Cajal/efectos de los fármacos , Intestino Delgado/citología , Animales , Antiinflamatorios/farmacología , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/fisiología , Calcio/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Ácido Flufenámico/farmacología , Células Intersticiales de Cajal/citología , Canales Iónicos/metabolismo , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ácido Niflúmico/farmacología , Técnicas de Placa-Clamp , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
8.
Korean J Physiol Pharmacol ; 14(2): 83-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20473379

RESUMEN

In this study, we studied whether hydrogen sulfide (H(2)S) has an effect on the pacemaker activity of interstitial cells of Cajal (ICC), in the small intestine of mice. The actions of H(2)S on pacemaker activity were investigated using whole-cell patch-clamp technique, intracellular Ca(2+) analysis at 30 and RT-PCR in cultured mouse intestinal ICC. Exogenously applied sodium hydrogen sulfide (NaHS), a donor of hydrogen sulfide, caused a slight tonic inward current on pacemaker activity in ICC at low concentrations (50 and 100 microM), but at high concentration (500 microM and 1 mM) it seemed to cause light tonic inward currents and then inhibited pacemaker amplitude and pacemaker frequency, and also an increase in the resting currents in the outward direction. Glibenclamide or other potassium channel blockers (TEA, BaCl(2), apamin or 4-aminopydirine) did not have an effect on NaHS-induced action in ICC. The exogenous application of carbonilcyanide p-triflouromethoxyphenylhydrazone (FCCP) and thapsigargin also inhibited the pacemaker activity of ICC as NaHS. Also, we found NaHS inhibited the spontaneous intracellular Ca(2+) ([Ca(2+)](i)) oscillations in cultured ICC. In doing an RT-PCR experiment, we found that ICC enriched population lacked mRNA for both CSE and CBS, but was prominently detected in unsorted muscle. In conclusion, H(2)S inhibited the pacemaker activity of ICC by modulating intracellular Ca(2+). These results can serve as evidence of the physiological action of H(2)S as acting on the ICC in gastrointestinal (GI) motility.

9.
J Cell Mol Med ; 14(1-2): 257-66, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20414970

RESUMEN

Hydrogen peroxide (H(2)O(2)) is involved in intestinal motility through changes of smooth muscle activity. However, there is no report as to the modulatory effects of H(2)O(2) on interstitial cells of Cajal (ICC). We investigated the H(2)O(2) effects and signal transductions to determine whether the intestinal motility can be modulated through ICC. We performed whole-cell patch clamp in cultured ICC from murine intestine and molecular analyses. H(2)O(2) hyperpolarized the membrane and inhibited pacemaker currents. These effects were inhibited by glibenclamide, an inhibitor of ATP-sensitive K+ (K(ATP)) channels. The free-radical scavenger catalase inhibited the H(2)O(2)-induced effects. MAFP and AACOCF3 (a cytosolic phospholipase A2 inhibitors) or SC-560 and NS-398 (a selective COX-1 and 2 inhibitor) or AH6809 (an EP2 receptor antagonist) inhibited the H(2)O(2)-induced effects. PD98059 (a mitogen activated/ERK-activating protein kinase inhibitor) inhibited the H(2)O(2)-induced effects, though SB-203580 (a p38 MAPK inhibitor) or a JNK inhibitor did not affect. H(2)O(2)-induced effects could not be inhibited by LY-294002 (an inhibitor of PI3-kinases), calphostin C (a protein kinase C inhibitor) or SQ-22536 (an adenylate cyclase inhibitor). Adenoviral infection analysis revealed H2O2 stimulated tyrosine kinase activity and AG 1478 (an antagonist of epidermal growth factor receptor tyrosine kinase) inhibited the H(2)O(2)-induced effects. These results suggest H(2)O(2) can modulate ICC pacemaker activity and this occur by the activation of K(ATP) channels through PGE(2) production via receptor tyrosine kinase-dependent MAP kinase activation.


Asunto(s)
Peróxido de Hidrógeno/farmacología , Células Intersticiales de Cajal/efectos de los fármacos , Células Intersticiales de Cajal/enzimología , Intestinos/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oxidantes/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/fisiología , Catalasa/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/metabolismo , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Células Intersticiales de Cajal/citología , Intestinos/fisiología , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/genética , Técnicas de Placa-Clamp , Fosfolipasas A2/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/efectos de los fármacos
10.
Arch Pharm Res ; 33(3): 479-89, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20361315

RESUMEN

The aim of this study was to clarify if phentolamine has proven effects on the pacemaker activities of interstitial cells of Cajal (ICC) from the mouse small intestine involving the ATPsensitive K(+) channels and adrenergic receptor. The actions of phentolamine on pacemaker activities were investigated using whole-cell patch-clamp technique and intracellular Ca(2+) analysis at 30 degrees C in cultured mouse intestinal ICC. ICC generated spontaneous pacemaker currents at a holding potential of -70 mV. Treatment with phentolamine reduced the frequency and amplitude of the pacemaker currents and increased the resting outward currents. Moreover, under current clamping (I = 0), phentolamine hyperpolarized the ICC membrane and decreased the amplitude of the pacemaker potentials. We also observed that phentolamine inhibited spontaneous [Ca(2+)](i) oscillations in ICC. The alpha-adrenergic drugs prazosin, yohimbine, methoxamine, and clonidine had no effect on ICC intestinal pacemaker activity and did not block phentolamine-induced effects. Phentolamine-induced effects on the pacemaker currents and the pacemaker potentials were significantly inhibited by ATP sensitive K(+) channel blocker glibenclamide, but not by TEA, apamin, or 4-aminopyridine. In addition, the NO synthase inhibitor, L-NAME and the guanylate cyclase inhibitor, ODQ were incapable of blocking the phentolamine-induced effects. These results demonstrate that phentolamine regulates the pacemaker activity of ICC via ATP-sensitive K(+) channel activation. Phentolamine could act through an adrenergic receptor- and also through NO-independent mechanism that involves intracellular Ca(2+) signaling.


Asunto(s)
Relojes Biológicos/efectos de los fármacos , Células Intersticiales de Cajal/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Canales KATP/agonistas , Fentolamina/farmacología , Potasio/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Femenino , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Células Intersticiales de Cajal/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Canales KATP/antagonistas & inhibidores , Canales KATP/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Factores de Tiempo
11.
Mol Cells ; 27(5): 525-31, 2009 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-19466600

RESUMEN

We studied the effect of carbachol on pacemaker currents in cultured interstitial cells of Cajal (ICC) from the mouse small intestine by muscarinic stimulation using a whole cell patch clamp technique and Ca2+-imaging. ICC generated periodic pacemaker potentials in the current-clamp mode and generated spontaneous inward pacemaker currents at a holding potential of-70 mV. Exposure to carbachol depolarized the membrane and produced tonic inward pacemaker currents with a decrease in the frequency and amplitude of the pacemaker currents. The effects of carbachol were blocked by 1-dimethyl-4-diphenylacetoxypiperidinium, a muscarinic M(3) receptor antagonist, but not by methotramine, a muscarinic M(2) receptor antagonist. Intracellular GDP-beta-S suppressed the carbachol-induced effects. Carbachol-induced effects were blocked by external Na+-free solution and by flufenamic acid, a non-selective cation channel blocker, and in the presence of thapsigargin, a Ca2+-ATPase inhibitor in the endoplasmic reticulum. However, carbachol still produced tonic inward pacemaker currents with the removal of external Ca2+. In recording of intracellular Ca2+ concentrations using fluo 3-AM dye, carbachol increased intracellular Ca2+ concentrations with increasing of Ca2+ oscillations. These results suggest that carbachol modulates the pacemaker activity of ICC through the activation of non-selective cation channels via muscarinic M(3) receptors by a G-protein dependent intracellular Ca2+ release mechanism.


Asunto(s)
Relojes Biológicos/efectos de los fármacos , Carbacol/farmacología , Fármacos Neuromusculares Despolarizantes/farmacología , Piperidinas/farmacología , Receptor Muscarínico M3/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Relojes Biológicos/fisiología , Calcio/metabolismo , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Proteínas de Transporte de Catión/antagonistas & inhibidores , Células Cultivadas , Diaminas/farmacología , Ácido Flufenámico/farmacología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Intestino Delgado/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Técnicas de Placa-Clamp , Raíces de Plantas , Receptor Muscarínico M2/antagonistas & inhibidores , Thapsia , Tapsigargina/farmacología , Tionucleótidos/farmacología
12.
Mol Cells ; 26(2): 181-5, 2008 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-18677097

RESUMEN

The effects of calcitonin gene-related peptide (CGRP) on pacemaker currents in cultured interstitial cells of Cajal (ICC) from the mouse small intestine were investigated using the whole-cell patch clamp technique at 30 degrees . Under voltage clamping at a holding potential of -70 mV, CGRP decreased the amplitude and frequency of pacemaker currents and activated outward resting currents. These effects were blocked by intracellular GDPbetaS, a G-protein inhibitor and glibenclamide, a specific ATP-sensitive K(+) channels blocker. During current clamping, CGRP hyperpolarized the membrane and this effect was antagonized by glibenclamide. Pretreatment with SQ-22536 (an adenylate cyclase inhibitor) or naproxen (a cyclooxygenase inhibitor) did not block the CGRP-induced effects, whereas pretreatment with ODQ (a guanylate cyclase inhibitor) or L-NAME (an inhibitor of nitric oxide synthase) did. In conclusion, CGRP inhibits pacemaker currents in ICC by generating nitric oxide via G-protein activation and so activating ATP-sensitive K(+) channels. Nitric oxide- and guanylate cyclase- dependent pathways are involved in these effects.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/fisiología , GMP Cíclico/farmacología , Intestino Delgado/fisiología , Canales KATP/fisiología , Óxido Nítrico/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Células Cultivadas , Femenino , Guanilato Ciclasa/antagonistas & inhibidores , Intestino Delgado/citología , Canales KATP/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , NG-Nitroarginina Metil Éster/farmacología , Oxadiazoles/farmacología , Técnicas de Placa-Clamp , Quinoxalinas/farmacología
13.
Korean J Physiol Pharmacol ; 12(1): 25-30, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20157390

RESUMEN

Although many studies show that thromboxane A(2) (TXA(2)) has the action of gastrointestinal (GI) motility using GI muscle cells and tissue, there are no reports on the effects of TXA(2) on interstitial cells of Cajal (ICC) that function as pacemaker cells in GI tract. So, we studied the modulation of pacemaker activities by TXA(2) in ICC with whole cell patch-clamp technique. Externally applied TXA(2) (5microM) produced membrane depolarization in current-clamp mode and increased tonic inward pacemaker currents in voltage-clamp mode. The tonic inward currents by TXA(2) were inhibited by intracellular application of GDP-beta-S. The pretreatment of ICC with Ca(2+) free solution and thapsigargin, a Ca(2+)-ATPase inhibitor in endoplasmic reticulum, abolished the generation of pacemaker currents and suppressed the TXA(2)-induced tonic inward currents. However, chelerythrine or calphostin C, protein kinase C inhibitors, did not block the TXA(2)-induced effects on pacemaker currents. These results suggest that TXA(2) can regulate intestinal motility through the modulation of ICC pacemaker activities. This modulation of pacemaker activities by TXA(2) may occur by the activation of G protein and PKC independent pathway via extra and intracellular Ca(2+) modulation.

14.
Korean J Physiol Pharmacol ; 12(3): 111-5, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20157403

RESUMEN

The effects of (-)-epigallocatechin gallate (EGCG) on pacemaker activities of cultured interstitial cells of Cajal (ICC) from murine small intestine were investigated using whole-cell patch-clamp technique at 30 and Ca(2+) image analysis. ICC generated spontaneous pacemaker currents at a holding potential of -70 mV. The treatment of ICC with EGCG resulted in a dose-dependent decrease in the frequency and amplitude of pacemaker currents. SQ-22536, an adenylate cyclase inhibitor, and ODQ, a guanylate cyclase inhibitor, did not inhibit the effects of EGCG. EGCG-induced effects on pacemaker currents were not inhibited by glibenclamide, an ATP-sensitive K(+) channel blocker and TEA, a Ca(2+)-activated K(+) channel blocker. Also, we found that EGCG inhibited the spontaneous [Ca(2+)](i) oscillations in cultured ICC. In conclusion, EGCG inhibited the pacemaker activity of ICC and reduced [Ca(2+)](i) oscillations by cAMP-, cGMP-, ATP-sensitive K+ channel-independent manner.

15.
Biol Pharm Bull ; 30(11): 2037-42, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17978472

RESUMEN

Although ginsenosides have a variety of physiologic or pharmacologic functions in various regions, there are only a few reports on the effects of ginsenosides on gastrointestinal (GI) motility. We studied the modulation of pacemaker activities by ginseng total saponins in the interstitial cells of Cajal (ICC) using the whole cell patch-clamp technique. Externally applied ginseng total saponins (GTS) produced membrane depolarization in the current-clamp mode and increased tonic inward pacemaker currents in the voltage-clamp mode. The application of flufenamic acid or niflumic acid abolished the generation of pacemaker currents, but only treatment with flufenamic acid inhibited the GTS-induced tonic inward currents. The tonic inward currents induced by GTS were not inhibited by the intracellular application of guanosine 5'-[beta-thio]diphosphate trilithium salt. Pretreatment with a Ca(2+)-free solution, with U-73122, an active phospholipase C inhibitor, and with thapsigargin, a Ca(2')-ATPase inhibitor of the endoplasmic reticulum, abolished the generation of pacemaker currents and suppressed the GTS-induced action. However, treatment with chelerythrine and calphostin C, protein kinase C inhibitors, did not block the GTS-induced effects on the pacemaker currents. These results suggest that ginsenosides modulate the pacemaker activities of the ICC, and the ICC can be targets for ginsenosides, and their interaction can affect intestinal motility.


Asunto(s)
Cuerpos Enrollados/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/fisiología , Potenciales de la Membrana/efectos de los fármacos , Panax/química , Saponinas/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Electrofisiología , Femenino , Intestino Delgado/citología , Masculino , Ratones , Ratones Endogámicos BALB C , Técnicas de Placa-Clamp
16.
Naunyn Schmiedebergs Arch Pharmacol ; 376(3): 175-84, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17932655

RESUMEN

We investigated the role of nitric oxide (NO) in pacemaker activity and signal mechanisms in cultured interstitial cells of Cajal (ICC) of the mouse small intestine using whole cell patch-clamp techniques at 30 degrees C. ICC generated pacemaker potential in the current clamp mode and pacemaker currents at a holding potential of -70 mV. (+/-)-S-nitroso-N-acetylpenicillamine (SNAP; a NO donor) produced membrane hyperpolarization and inhibited the amplitude and frequency of the pacemaker currents, and increased resting currents in the outward direction. These effects were blocked by the use of glibenclamide (an ATP-sensitive K+ channel blocker), but not by the use of 5-hydroxydecanoic acid (a mitochondrial ATP-sensitive K+ channel blocker). Pretreatment with ODQ (a guanylate cyclase inhibitor) almost blocked the NO-induced effects. The use of cell-permeable 8-bromo-cyclic GMP also mimicked the action of SNAP. However, the use of KT-5823 (a protein kinase G inhibitor) did not block the NO-induced effects. Spontaneous [Ca2+]i oscillations in ICC were inhibited by the treatment of SNAP, as seen in recordings of intracellular Ca2+ ([Ca2+]i). These results suggest that NO inhibits pacemaker activity by the activation of ATP-sensitive K+ channels via a cyclic GMP dependent mechanism in ICC, and the activation of ATP-sensitive K+ channels mediates the inhibition of spontaneous [Ca2+]i oscillations.


Asunto(s)
Intestino Delgado/citología , Óxido Nítrico/metabolismo , Canales de Potasio/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Carbazoles/farmacología , Células Cultivadas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Gliburida/farmacología , Indoles/farmacología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/antagonistas & inhibidores , Técnicas de Placa-Clamp , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , S-Nitroso-N-Acetilpenicilamina/farmacología
17.
Cell Physiol Biochem ; 18(4-5): 187-98, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17167224

RESUMEN

The interstitial cells of Cajal (ICC) are pacemaker cells in gastrointestinal tract and generate an electrical rhythm in gastrointestinal muscles. We investigated the possibility that PGE(2) might affect the electrical properties of cultured ICC by activating ATP-dependent K(+) channels and, the EP receptor subtypes and the subunits of ATP-dependent K(+) channels involved in these activities were identified. In addition, the regulation of intracellular Ca(2+) ([Ca(2+)](i)) mobilization may be involved the action of PGE(2) on ICC. Treatments of ICC with PGE(2) inhibited electrical pacemaker activities in the same manner as pinacidil, an ATP-dependent K(+) channel opener and PGE(2) had only a dose-dependent effect. Using RT-PCR technique, we found that ATP-dependent K(+) channels exist in ICC and that these are composed of K(ir) 6.2 and SUR 2B subunits. To characterize the specific membrane EP receptor subtypes in ICC, EP receptor agonists and RT-PCR were used: Butaprost (an EP(2) receptor agonist) showed the actions on pacemaker currents in the same manner as PGE(2). However sulprostone (a mixed EP(1) and EP(3) agonist) had no effects. In addition, RT-PCR results indicated the presence of the EP(2) receptor in ICC. To investigate cAMP involvement in the effects of PGE(2) on ICCs, SQ-22536 (an inhibitor of adenylate cyclase) and cAMP assays were used. SQ-22536 did not affect the effect of PGE(2) on pacemaker currents, and PGE(2) did not stimulate cAMP production. Also, we found PGE(2) inhibited the spontaneous [Ca(2+)](i) oscillations in cultured ICC. These observations indicate that PGE(2) alters pacemaker currents by activating the ATP-dependent K(+) channels comprised of K(ir) 6.2-SUR 2B in ICC and this action of PGE(2) are through EP(2) receptor subtype and also the activation of ATP-dependent K(+) channels involves intracellular Ca(2+) mobilization.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/agonistas , Dinoprostona/farmacología , Intestino Delgado/efectos de los fármacos , Canales de Potasio de Rectificación Interna/agonistas , Canales de Potasio/agonistas , Receptores de Droga/agonistas , Receptores de Prostaglandina E/metabolismo , Transportadoras de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/genética , Adenina/análogos & derivados , Adenina/farmacología , Adenosina Trifosfato/metabolismo , Inhibidores de Adenilato Ciclasa , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Intestino Delgado/química , Intestino Delgado/citología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos BALB C , Canales de Potasio/análisis , Canales de Potasio/genética , Canales de Potasio de Rectificación Interna/análisis , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/análisis , Receptores de Droga/genética , Subtipo EP2 de Receptores de Prostaglandina E , Receptores de Sulfonilureas
18.
Arch Pharm Res ; 29(11): 998-1005, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17146969

RESUMEN

The effects of imipramine on A-type delayed rectifier K+ currents and ATP-sensitive K+ (KATP) currents were studied in isolated murine proximal colonic myocytes using the whole-cell patch-clamp technique. Depolarizing test pulses between -80 mV and +30 mV with 10 mV increments from the holding potential of -80 mV activated voltage-dependent outward K+ currents that peaked within 50 ms followed by slow decreasing sustained currents. Early peak currents were inhibited by the application of 4-aminopyridine, whereas sustained currents were inhibited by the application of TEA. The peak amplitude of A-type delayed rectifier K+ currents was reduced by external application of imipramine. The half-inactivation potential and the half-recovery time of A-type delayed rectifier K+ currents were not changed by imipramine. With 0.1 mM ATP and 140 mM K+ in the pipette and 90 mM K+ in the bath solution and a holding potential of -80 mV, pinacidil activated inward currents; this effect was blocked by glibenclamide. Imipramine also inhibited KATP currents. The inhibitory effects of imipramine in A-type delayed rectifier K+ currents and KATP currents were not changed by guanosine 5-O-(2-thiodiphosphate) (GDPbetaS) and chelerythrine, a protein kinase C inhibitor. These results suggest that imipramine inhibits A-type delayed rectifier K+ currents and KATP currents in a manner independent of G-protein and protein kinase C.


Asunto(s)
Colon/metabolismo , Canales de Potasio de Tipo Rectificador Tardío/efectos de los fármacos , Proteínas de Unión al GTP/fisiología , Imipramina/farmacología , Miocitos del Músculo Liso/metabolismo , Bloqueadores de los Canales de Potasio , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Proteína Quinasa C/fisiología , 4-Aminopiridina/farmacología , Animales , Colon/citología , Colon/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Técnicas In Vitro , Canales KATP , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Miocitos del Músculo Liso/efectos de los fármacos , Técnicas de Placa-Clamp , Proteína Quinasa C/antagonistas & inhibidores
19.
Br J Pharmacol ; 148(7): 918-26, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16783409

RESUMEN

We studied the modulation of pacemaker activities by bradykinin in cultured interstitial cells of Cajal (ICC) from murine small intestine with the whole-cell patch-clamp technique. Externally applied bradykinin produced membrane depolarization in the current-clamp mode and increased tonic inward pacemaker currents in the voltage-clamp mode. Pretreatment with bradykinin B1 antagonist did not block the bradykinin-induced effects on pacemaker currents. However, pretreatment with bradykinin B2 antagonist selectively blocked the bradykinin-induced effects. Also, only externally applied selective bradykinin B2 receptor agonist produced tonic inward pacemaker currents and ICC revealed a colocalization of the bradykinin B2 receptor and c-kit immunoreactivities, but bradykinin B1 receptors did not localize in ICC. External Na(+)-free solution abolished the generation of pacemaker currents and inhibited the bradykinin-induced tonic inward current. However, a Cl(-) channel blocker (DIDS) did not block the bradykinin-induced tonic inward current. The pretreatment with Ca(2+)-free solution and thapsigargin, a Ca(2+)-ATPase inhibitor in endoplasmic reticulum, abolished the generation of pacemaker currents and suppressed the bradykinin-induced action. Chelerythrine and calphostin C, protein kinase C inhibitors or naproxen, an inhibitor of cyclooxygenase, did not block the bradykinin-induced effects on pacemaker currents. These results suggest that bradykinin modulates the pacemaker activities through bradykinin B2 receptor activation in ICC by external Ca(2+) influx and internal Ca(2+) release via protein kinase C- or cyclooxygenase-independent mechanism. Therefore, the ICC are targets for bradykinin and their interaction can affect intestinal motility.


Asunto(s)
Relojes Biológicos/efectos de los fármacos , Bradiquinina/farmacología , Intestino Delgado/metabolismo , Receptor de Bradiquinina B2/efectos de los fármacos , Animales , Calcio/fisiología , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Células Cultivadas , Canales de Cloruro/antagonistas & inhibidores , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Inmunohistoquímica , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Técnicas de Placa-Clamp , Proteína Quinasa C/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sodio/fisiología
20.
Life Sci ; 78(20): 2322-8, 2006 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-16266721

RESUMEN

Tricyclic antidepressants have been widely used for the treatment of depression and as a therapeutic agent for the altered gastrointestinal (GI) motility of irritable bowel syndrome (IBS). The aim of this study was to clarify whether antidepressants directly modulate pacemaker currents in cultured interstitial cells of Cajal (ICC). We used the whole-cell patch-clamp techniques at 30 degrees C in cultured ICC from the mouse small intestine. Treatment of pinacidil, an ATP-sensitive K(+) channel opener, in the ICC using the current clamping mode, produced hyperpolarization of the membrane potential and decreased the amplitude of the pacemaker potentials. With the voltage clamp mode, we observed a decrease in the frequency and amplitude of pacemaker currents and increases in the resting outward currents. These effects of pinacidil on pacemaker potentials and currents were completely suppressed by glibenclamide, an ATP-sensitive K(+) channel blocker. Also, with the current clamp mode, imipramine blocked the affect of pinacidil on the pacemaker potentials. Observations of the voltage clamp mode with imipramine, desipramine and amitryptyline suppressed the action of pinacidil in the ICC. Next, we examined whether protein kinase C (PKC) and the G protein are involved in the action of imipramine on pinacidil induced pacemaker current inhibition. We used chelerythrine, a potent PKC inhibitor and GDPbetaS, a nonhydrolyzable guanosine 5-diphosphate (GDP) analogue that permanently inactivates GTP-binding proteins. We found that pretreatment with chelerythrine and intracellular application of GDPbetaS had no influence on the blocking action of imipramine on inhibited pacemaker currents by pinacidil. We conclude that imipramine inhibited the activated ATP-sensitive K(+) channels in ICC. This action does not appear to be mediated through the G protein and protein kinase C. Furthermore, this study may suggest another possible mechanism for tricyclic antidepressants related modulation of GI motility.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/efectos de los fármacos , Antidepresivos Tricíclicos/farmacología , Imipramina/farmacología , Intestino Delgado/metabolismo , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Animales , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Femenino , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteínas de Unión al GTP/metabolismo , Motilidad Gastrointestinal/efectos de los fármacos , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Canales KATP , Masculino , Ratones , Ratones Endogámicos BALB C , Técnicas de Placa-Clamp , Pinacidilo/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...