• Title/Summary/Keyword: $Ca^{2+}$ channel blocker

Search Result 150, Processing Time 0.029 seconds

Effect of Lespedezea Cuneata on the Contraction of Rabbit Common Carotid Artery and Corpus Cavernosum (야관문(夜關門)이 토끼의 혈관과 음경해면체 수축에 미치는 영향)

  • Park, Sun Young;Kim, Ho Hyun
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.27 no.6
    • /
    • pp.809-817
    • /
    • 2013
  • The aim of this study was to evaluate the mechanism of vasodilation of Lespedezea cuneata(LC) in rabbit common carotid artery and cavernosal smooth muscle. LC relaxed arterial strips precontracted with norepinephrine and cavernosal strips precontracted with phenylephrine. The arterial relaxation effects of LC was endothelium-dependent. $N{\omega}$-nitro-L-arginine(L-NNA), NOS inhibitor, methylene blue(MB), cGMP inhibitor, indomethacin(IM), cyclo-oxygenase inhibitor and tetraethylammonium chloride(TEA), KCa-channel blocker attenuate the relaxation responses of LC in arterial strips. In $Ca^{2+}$-free krebs-ringer solution, pretreatment of LC extract significantly reduced the contraction induced by addition $Ca^{2+}$. L-NNA reduced LC extract-induced relaxation in cavernosal strips, but IM, TEA and MB didn't affect LC extract-induced relaxation. When LC extract was applicated on human umbilical vein endothelial cell, the nitric oxide concentration was increased. We conclude that in rabbit common carotid artery, LC may suppress influx of extra-cellular $Ca^{2+}$ through the release of endothelium derived relaxing factor including nitric oxide, prostacyclin, endothelium derived hyperpolarizing factor. And LC exerts a relaxing effect on corpus cavernosum through activating the NO.

Effects of Electrolytes and Drugs on the Inhibitory Junction Potentials Recorded from the Antrum of Guinea-pig Stomach (기니피그 유문동에서 기록되는 억제성 접합부 전압에 미치는 전해질과 약물의 효과)

  • Goo, Yong-Sook;Suh, Suk-Hyo;Lee, Suk-Ho;Hwang, Sang-Ik;Kim, Ki-Whan
    • The Korean Journal of Physiology
    • /
    • v.24 no.1
    • /
    • pp.1-13
    • /
    • 1990
  • The effects of electrolytes, adenosine, ATP, 5-hydroxytryptamine (5-HT, serotonin) and ketanserin on the inhibitory junction potentials (IJPs) were investigated to clarify the interactions of these drugs with the neurotransmitters released from non-adrenergic, non-cholinergic nerves in the antrum of guinea-pig stomach. Electrical responses of antral circular muscle cells were recorded intracellularly using glass capillary microelectrode filled with 3 M KCI. All experiments were performed in Tris-buffered Tyrode soluition which was aerated with 100% $O_{2}$ and kept at $35^{\circ}C$. The results obtained were as follows: 1) Inhibitory junction potential (IJP) was recorded in antral strip, while excitatory junction potential (EJP) was recorded in fundic strip. 2) IJP recorded in antral strip was not influenced by atropine $(10^{-6}\;M)$ and guanethidine $(5{\times}10^{-6})$. 3) The amplitude of IJP increased in high $Ca^{2+}$ solution, while that of IJP decreased in high $Mg^{2+}$ solution or by $Ca^{2+}$ antagonist (verapamil). Apamin, $Ca^{2+}$-activated $K^{+}$ channel blocker blocked IJP completely. 4) ATP and adenosine decreased the amplitude of IJP. 5) 5-HT decreased the amplitude of IJP with no change of the amplitude of slow waves, while ketanserin (5-HT type 2 blocker) decreased the amplitude of slow waves markedly with no change in that of IJP. From the above results, the following conclusions could be made. 1) IJP recorded in antral strip is resulted from neurotransmitters released from non-adrenergic, non-cholinergic nerves. 2) An increase in the concentration of external $Ca^{2+}$ enhances the release of neurotransmitters from non-adrenergic, non-cholinergic nerves which activate the $Ca^{2+}$-dependent $K^{+}$ channel.

  • PDF

Calcium Current in the Unfertilized Egg of the Hamster

  • Haan, Jae-Hee;Cho, Soo-Wan;Yang, Young-Sun;Park, Young-Geun;Park, Hong-Gi;Chang, Gyeong-Jae;Kim, Yang-Mi;Park, Choon-Ok;Hong, Seong-Geun
    • The Korean Journal of Physiology
    • /
    • v.28 no.2
    • /
    • pp.215-224
    • /
    • 1994
  • The presence of a calcium current $(i_{Ca^{2+}})$ passed via a specific channel was examined in the unfertilized hamster egg using the whole-cell voltage clamp technique. Pure inward current was isolated using a $Ca^{2+}-rich$ pipette solution containing 10 mM TEA. This current was independent of external $Na^+$ and was highly sensitive to the $Ca^{2+}$ concentration in the bathing solution, indicating that the inward current is carried by $Ca^{2+}$. The maximal amplitude was $-4.12{\pm}0.58nA\;(n=12)$ with 10mM $Ca^{2+}$ at -3OmV from a holding potential of -8OmV. This current reached its maximum within 20ms beyond -3OmV and decayed rapidly with an inactivation time constant $({\tau})$ of 15ms. Activation and inactivation of this $i_{Ca^{2+}}$ was steeply dependent on the membrane potential. The $i_{Ca^{2+}}$ began to activate at the lower voltage of -55 mV and reached its peak at -35 mV, being completely inactivated at potentials more positive than -40 mV. These result suggest that $i_{Ca^{2+}}$ in hamster eggs passes through channels with electrical properties similar to low voltage-activated T-type channels. Other results from the present study support this suggestion; First, the inhibitory effect of $Ni^{2+}\;(IC_{50}=13.7\;{\mu}M)$ was more potent than $Cd^{2+}\;(IC_{50}=123\;{\mu}M)$. Second, $Ba^{2+}$ conductance was equal to or below that of $Ca^{2+}$. Third, $i_{Ca^{2+}}$ in hamster eggs was relatively insensitive to nifedipine $(IC_{50}=96.6\;{\mu}M)$, known to be a specific t-type blocker. The physiological role of $i_{Ca^{2+}}$ in the unfertilized hamster eggs remains unclear. Analysis from steady-state inactivation activation curves reveals that only a small amount of this current will pass in the voltage range $(-70{\sim}-30\;mV)$ which partially overlaps with the resting membrane potential. This current has the property that it can be easily activated by a weak depolarization, thus it may trigger a certain kind of a intracellular event following fertilization which may cause oscillations in the membrane potential.

  • PDF

Carbachol Regulates Pacemaker Activities in Cultured Interstitial Cells of Cajal from the Mouse Small Intestine

  • So, Keum Young;Kim, Sang Hun;Sohn, Hong Moon;Choi, Soo Jin;Parajuli, Shankar Prasad;Choi, Seok;Yeum, Cheol Ho;Yoon, Pyung Jin;Jun, Jae Yeoul
    • Molecules and Cells
    • /
    • v.27 no.5
    • /
    • pp.525-531
    • /
    • 2009
  • 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 $Ca^{2+}$-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 $Ca^{2+}$-ATPase inhibitor in the endoplasmic reticulum. However, carbachol still produced tonic inward pacemaker currents with the removal of external $Ca^{2+}$. In recording of intracellular $Ca^{2+}$ concentrations using fluo 3-AM dye, carbachol increased intracellular $Ca^{2+}$ concentrations with increasing of $Ca^{2+}$ 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 $Ca^{2+}$ release mechanism.

The Mechanism of t-Butylhydroperoxide-Induced Apoptosis in IMR-32 Human Neuroblastoma Cells

  • Kim, Jung-Ae;Lee, Yong-Soo;Huh, Keun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.3 no.1
    • /
    • pp.19-27
    • /
    • 1999
  • Apoptosis has been implicated in the pathophysiological mechanisms of various neurodegenerative diseases. In a variety of cell types, oxidative stress has been demonstrated to play an important role in the apoptotic cell death. However, the exact mechanism of oxidative stress-induced apoptosis in neuronal cells is not known. In this study, we induced oxidative stress in IMR-32 human neuroblastoma cells with tert- butylhydroperoxide (TBHP), which was confirmed by significantly reduced glutathione content and glutathione reductase activity, and increased glutathione peroxidase activity. TBHP induced decrease in cell viability and increase in DNA fragmentation, a hallmark of apoptosis, in a dose-dependent manner. TBHP also induced a sustained increase in intracellular $Ca^{2+}$ concentration, which was completely prevented either by EGTA, an extracellular $Ca^{2+}$ chelator or by flufenamic acid (FA), a non-selective cation channel (NSCC) blocker. These results indicate that the TBHP-induced intracellular $Ca^{2+}$ increase may be due to $Ca^{2+}$ influx through the activation of NSCCs. In addition, treatment with either an intracellular $Ca^{2+}$ chelator (BAPTA/AM) or FA significantly suppressed the TBHP-induced apoptosis. Moreover, TBHP increased the expression of p53 gene but decreased c-myc gene expression. Taken together, these results suggest that the oxidative stress-induced apoptosis in neuronal cells may be mediated through the activation of intracellular $Ca^{2+}$ signals and altered expression of p53 and c-myc.

  • PDF

Magnesium-induced Relaxation in Rat Aorta (Magnesium에 의한 흰쥐 대동맥 이완)

  • Oh, Sung-suck;Lee, Sang-woo;Kang, Hyung-sub;Kim, Jin-shang
    • Korean Journal of Veterinary Research
    • /
    • v.43 no.3
    • /
    • pp.373-382
    • /
    • 2003
  • Magnesium ion ($Mg^{2+}$) is a vasodilator, but little is known about its mechanism of action on vascular system. In vitro, extracellular magnesium sulfate ($MgSO_4$) produced relaxation in phenylephrine (PE) or high KCl-precontracted isolated rat thorocic aorta with (+E) or without (-E) endothelium in a concentration-dependent manner. The $MgSO_4$-induced relaxations were not affected by removal of the endothelium. Pretreatment of +E or -E aortic rings with nitric oxide synthase (NOS) inhibitors ($20{\mu}M$ L-NNA, $100{\mu}M$ L-NAME, $1{\mu}M$ dexamethasone and $400{\mu}M$ aminoguanidine), cyclooxygenase inhibitor ($10{\mu}M$ indomethacin), guanylate cyclase inhibitors ($10{\mu}M$ ODQ and $30{\mu}M$ methylene blue) and $Ca^{2+}$ transport blocker ($10{\mu}M$ ryanodine) did not affect the relaxant effects of $MgSO_4$. $Ca^{2+}$ channel blockers ($0.3{\mu}M$ nifedipine and $0.5{\mu}M$ veropamil) completely decreased the relaxant effects of $MgSO_4$ in +E and -E aortic rings. However, in $Ca^{2+}$-free medium, $MgSO_4$-induced vasorelaxation was potentiated and this response was inhibited by nifedipine. Protein kinase C (PKC) inhibitors ($1.0{\mu}M$ staurosporine, $0.5{\mu}M$ tamoxifen and $0.1{\mu}M$ H7) or PLC inhibitor ($100{\mu}M$ NCDC) markedly decreased the relaxant effects of $MgSO_4$ in +E and -E aortic rings. In vivo, infusion of $MgSO_4$ elicited significant decreases in arterial blood pressure. After intravenous injection of nifedipine ($150{\mu}g/kg$) and NCDC (3 mg/kg), infusion of $MgSO_4$ inhibited the $MgSO_4$-lowered blood pressure markedly. However, after introvenous injection of saponin (15 mg/kg), L-NNA (3 mg/kg), L-NAME (5 mg/kg), indomethacin (2 mg/kg), methylene blue (15 mg/kg) and aminoguanidine (10 mg/kg) failed to inhibit it. These results suggest that endothelial NQ-cGMP or prostaglandin pathway is not involved in vasorelaxant or hypotensive action of $Mg^{2+}$ and that these effects are due to the inhibitory action of $Mg^{2+}$ on the $Ca^{2+}$ channel or PLC-PKC pathway, and are due to the competitive influx of $Mg^{2+}$ and $Ca^{2+}$ through the $Ca^{2+}$ channel.

Influence of Apamin on Catecholamine Secretion from the Rat Adrenal Medulla

  • Lee, Eun-Sook;Park, Hyeon-Gyoon;Lim, Dong-Yoon
    • Biomolecules & Therapeutics
    • /
    • v.10 no.3
    • /
    • pp.142-151
    • /
    • 2002
  • The present study was attempted to investigate the effect of apamin on catecholamine (CA) secretion evoked by ACh, high $K^+$, DMPP, McN-A-343, cyclopiazonic acid and Bay-K-8644 from the isolated perfused rat adrenal gland and to establish the mechanism of its action. The perfusion of apamin (1 nM) into an adrenal vein for 20 min produced greatly potentiation in CA secretion evoked by ACh (5.32 $ imes$ $10^{-3}$ M), high $K^+$, (5.6 $ imes$ $10^{-2}$), DMPP ($10^{-4}$ M for 2 min), McN-A-343 ($10^{-4}$ M for 2 min), cyclopiazonic acid ($10^{-5}$ M for 4 min) and Bay-K-8644 ($10^{-5}$ M for 4 min). However, apamin itself did fail to affect basal catecholamine output. Furthermore, in adrenal glands preloaded with apamin (1 nM) under the presence of glibenclamide ($10^{-6}$ M), an antidiabetic sulfonylurea that has been shown to be a specific blocker of ATP-regulated potassium channels (for 20 min), CA secretion evoked by DMPP and McN-A-343 was not affected. However, the perfusion of high concentration of apamin (100 nM) into an adrenal vein for 20 min rather inhibited significantly CA secretory responses evoked by ACh, high $K^+$, DMPP, McN-A-343, cyclopiazonic acid and Bay-K-8644. Taken together, these results suggest that the low concentration of apamin causes greatly the enhancement of CA secretion evoked by stimulation of cholinergic (both nicotinic and muscarinic) receptors as well as by membrane depolarization. These findings suggests that apamin-sensitive SK ($Ca^{2+}$) channels located in rat adrenal medullary chromaffin cells may play an inhibitory role in the release of catecholamines mediated by stimulation of cholinergic nicotinic and muscarinic receptors as well as membrane depolarization. However, it is thought that high concentration of apamin cause the inhibitory responses in catecholamine secretion evoked by stimulation of cholinergic receptors as well as by membrane depolarization from the rat adrenal gland without relevance with the SK channel blockade.

Different Mechanisms of K-induced Contracture in Isolated Vascular and Intestinal Smooth Muscles (혈관 및 장관 평활근의 K-경축 발생기전)

  • Kim, Ki-Whan;Hwang, Sang-Ik;Nam, Kee-Yong
    • The Korean Journal of Physiology
    • /
    • v.13 no.1_2
    • /
    • pp.41-50
    • /
    • 1979
  • The activation mechanism of K-induced contracture was studied in renal vascular muscle which does not generate an action potential readily and in taenia coli which generates a spike potential spontaneously. Helical strips of arterial muscle from rabbit renal arteries and longitudinal strips of taenia coli from guinea-pig's colons, respectively, were prepared. All experiments were performed in Tris-buffered Tyrode solution which was aerated with 100% $O_2$ and kept $35^{\circ}C$. Renal arterial muscles developed the contracture rapidly, which was composed of a small phasic and a large tonic components, when exposed to a 40 mM K-Tyrode solution. In the absence of external $Ca^{++}$, however, no K-contracture appeared. The contracture induced by K-depolarization was abolished by the treatment with verapamil, which is known to be a selective $Ca^{++}-blocker$ through potential-sensitive $Ca^{++}-channel$. K-contracture of taenia coli showed the contracture composed of a large phasic and a small tonic components. In the $Ca^{++}-free$ Tyrode solution, only the tonic component was abolished and almost no change in the phasic component was observed. The amplitude of tonic component was dependent on the external $Ca^{++}$; The tonic component increased dose-dependently by a stepwise increase of the external $Ca^{++}$, and this component decreased in parallel with the increase of verapamil in the external medium. The results of this experiment suggest that K-contracture of rabbit renal artery is the direct result of the influx of the external $Ca^{++}$, while that of taenia coli is the result of both $Ca^{++}$ influx and the release of sequestered $Ca^{++}$.

  • PDF

Effect of Na, K, Ca and Mg ions on the Action Potential of the Sinoatrial Node in the Rabbit (토끼 동방결절 활동전압에 대한 Na, K, Ca 및 Mg 이온의 영향)

  • Lee, Jeong-Ryeol;Eom, Yung-Ui
    • Journal of Chest Surgery
    • /
    • v.18 no.1
    • /
    • pp.1-6
    • /
    • 1985
  • Isolated sinus node cells of the rabbit were used to assess the effects of extracellular Na, K, Ca and Mg concentrations on cardiac pacemaker activity. With intracellular glass micro-electrodes spontaneous action potentials of SA node were recorded and the effects of various ions and their blockers were analyzed in terms of the cycle length, the amplitude and the duration of action potentials, the results obtained were as follows. 1. Sodium reduction [up to 30%] decreased the amplitude of action potential and lengthened the cycle length. TTX, specific blocker of Na channel slightly lengthened the cycle length. 2. Increasing potassium ion concentration, the duration of action potential decreased and the frequency increased in 6mM, however, spontaneous action potential was stopped in 24 mM. Barium ion known to be decreasing K conductance increased the duration of action potential but no significant change in the cycle length was noticed. 3. Calcium ion has shortening effect on the duration and the cycle length of action potential but not with dose-dependent manner. Cadmium ion .[0.02mM] lengthened cycle length and the duration of action potential. 4. Increasing the concentration of magnesium ion the cycle length was lengthened, significantly.

  • PDF

Inhibitory Effect of Genistein on Agonist-Induced Modulation of Vascular Contractility

  • Je, Hyun Dong;Sohn, Uy Dong
    • Molecules and Cells
    • /
    • v.27 no.2
    • /
    • pp.191-198
    • /
    • 2009
  • The present study was undertaken to determine whether treatment with genistein, the plant-derived estrogen-like compound influences agonist-induced vascular smooth muscle contraction and, if so, to investigate related mechanisms. The measurement of isometric contractions using a computerized data acquisition system was combined with molecular experiments. Genistein completely inhibited KCl-, phorbol ester-, phenylephrine-, fluoride- and thromboxane $A_2$-induced contractions. An inactive analogue, daidzein, completely inhibited only fluoride-induced contraction regardless of endothelial function, suggesting some difference between the mechanisms of RhoA/Rho-kinase activators such as fluoride and thromboxane $A_2$. Furthermore, genistein and daidzein each significantly decreased phosphorylation of MYPT1 at Thr855 had been induced by a thromboxane $A_2$ mimetic. Interestingly, iberiotoxin, a blocker of large-conductance calcium-activated potassium channels, did not inhibit the relaxation response to genistein or daidzein in denuded aortic rings precontracted with fluoride. In conclusion, genistein or daidzein elicit similar relaxing responses in fluoride-induced contractions, regardless of tyrosine kinase inhibition or endothelial function, and the relaxation caused by genistein or daidzein was not antagonized by large conductance $K_{Ca}$-channel inhibitors in the denuded muscle. This suggests that the RhoA/Rho-kinase pathway rather than $K^+$- channels are involved in the genistein-induced vasodilation. In addition, based on molecular and physiological results, only one vasoconstrictor fluoride seems to be a full RhoA/Rho-kinase activator; the others are partial activators.