• 제목/요약/키워드: membrane action

Search Result 548, Processing Time 0.027 seconds

Modulation of $Ca^{2+}-Activated$ Potassium Channels by cGMP-Dependent Signal Transduction Mechanism in Cerebral Arterial Smooth Muscle Cell of the Rabbit

  • Han, Jin;Kim, Na-Ri;Lee, Kwang-Bok;Kim, Eui-Yong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.4 no.6
    • /
    • pp.445-453
    • /
    • 2000
  • The present investigation tested the hypothesis that the activation of protein kinase G (PKG) leads to a phosphorylation of $Ca^{2+}-activated$ potassium channel $(K_{Ca}\;channel)$ and is involved in the activation of $K_{Ca}$ channel activity in cerebral arterial smooth muscle cells of the rabbit. Single-channel currents were recorded in cell-attached and inside-out patch configurations of patch-clamp techniques. Both molsidomine derivative 3-morpholinosydnonimine-N-ethylcarbamide $(SIN-1,\;50\;{\mu}M)$ and 8-(4-Chlorophenylthio)-guanosine-3',5'-cyclic monophosphate $(8-pCPT-cGMP,\;100\;{\mu}M),$ a membrane-permeable analogue of cGMP, increased the $K_{Ca}$ channel activity in the cell-attached patch configuration, and the effect was removed upon washout of the drugs. In inside-out patches, single-channel current amplitude was not changed by SIN-1 and 8-pCPT-cGMP. Application of ATP $(100\;{\mu}M),$ cGMP $(100\;{\mu}M),$ ATP+cGMP $(100\;{\mu}M\;each),$ PKG $(5\;U/{\mu}l),$ ATP $(100\;{\mu}M)+PKG\;(5\;U/{\mu}l),$ or cGMP $(100\;{\mu}M)+PKG\;(5\;U/{\mu}l)$ did not increase the channel activity. ATP $(100\;{\mu}M)+cGMP\;(100\;{\mu}M)+PKG\;(5\;U/{\mu}l)$ added directly to the intracellular phase of inside-out patches increased the channel activity with no changes in the conductance. The heat-inactivated PKG had no effect on the channel activity, and the effect of PKG was inhibited by 8-(4-Chlorophenylthio)-guanosine-3',5'-cyclic monophosphate, Rp-isomer $(Rp-pCPT-cGMP,\;100\;{\mu}M),$ a potent inhibitor of PKG or protein phosphatase 2A (PP2A, 1 U/ml). In the presence of okadaic acid (OA, 5 nM), PP2A had no effect on the channel activity. The $K_{Ca}$ channel activity spontaneously decayed to the control level upon washout of ATP, cGMP and PKG, and this was prevented by OA (5 nM) in the medium. These results suggest that the PKG-mediated phosphorylations of $K_{Ca}$ channels, or some associated proteins in the membrane patch increase the activity of the $K_{Ca}$ channel, and the activation may be associated with the vasodilating action.

  • PDF

Influence of Ketamine on Catecholamine Secretion in the Perfused Rat Adrenal Medulla

  • Ko, Young-Yeob;Jeong, Yong-Hoon;Lim, Dong-Yoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.12 no.3
    • /
    • pp.101-109
    • /
    • 2008
  • The aim of the present study was to examine the effects of ketamine, a dissociative anesthetics, on secretion of catecholamines (CA) secretion evoked by cholinergic stimulation from the perfused model of the isolated rat adrenal gland, and to establish its mechanism of action, and to compare ketamine effect with that of thiopental sodium, which is one of intravenous barbiturate anesthetics. Ketamine ($30{\sim}300{\mu}M$), perfused into an adrenal vein for 60 min, dose- and time-dependently inhibited the CA secretory responses evoked by ACh (5.32 mM), high $K^+$ (a direct membrane-depolarizer, 56 mM), DMPP (a selective neuronal nicotinic NN receptor agonist, $100{\mu}M$) and McN-A-343 (a selective muscarinic M1 receptor agonist, $100{\mu}M$). Also, in the presence of ketamine ($100{\mu}M$), the CA secretory responses evoked by veratridine (a voltage-dependent $Na^+$ channel activator, $100{\mu}M$), Bay-K-8644 (an L-type dihydropyridine $Ca^{2+}$ channel activator, $10{\mu}M$), and cyclopiazonic acid (a cytoplasmic $Ca^{2+}$-ATPase inhibitor, $10{\mu}M$) were significantly reduced, respectively. Interestingly, thiopental sodium ($100{\mu}M$) also caused the inhibitory effects on the CA secretory responses evoked by ACh, high $K^+$, DMPP, McN-A-343, veratridine, Bay-K-8644, and cyclopiazonic acid. Collectively, these experimental results demonstrate that ketamine inhibits the CA secretion evoked by stimulation of cholinergic (both nicotinic and muscarinic) receptors and the membrane depolarization from the isolated perfused rat adrenal gland. It seems likely that the inhibitory effect of ketamine is mediated by blocking the influx of both $Ca^{2+}$ and $Na^+$ through voltage-dependent $Ca^{2+}$ and $Na^+$ channels into the rat adrenal medullary chromaffin cells as well as by inhibiting $Ca^{2+}$ release from the cytoplasmic calcium store, which are relevant to the blockade of cholinergic receptors. It is also thought that, on the basis of concentrations, ketamine causes similar inhibitory effect with thiopental in the CA secretion from the perfused rat adrenal medulla.

Pituitary Adenylate Cyclase-activating Polypeptide Inhibits Pacemaker Activity of Colonic Interstitial Cells of Cajal

  • Wu, Mei Jin;Kee, Keun Hong;Na, Jisun;Kim, Seok Won;Bae, Youin;Shin, Dong Hoon;Choi, Seok;Jun, Jae Yeoul;Jeong, Han-Seong;Park, Jong-Seong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.19 no.5
    • /
    • pp.435-440
    • /
    • 2015
  • This study aimed to investigate the effect of pituitary adenylate cyclase-activating peptide (PACAP) on the pacemaker activity of interstitial cells of Cajal (ICC) in mouse colon and to identify the underlying mechanisms of PACAP action. Spontaneous pacemaker activity of colonic ICC and the effects of PACAP were studied using electrophysiological recordings. Exogenously applied PACAP induced hyperpolarization of the cell membrane and inhibited pacemaker frequency in a dose-dependent manner (from 0.1 nM to 100 nM). To investigate cyclic AMP (cAMP) involvement in the effects of PACAP on ICC, SQ-22536 (an inhibitor of adenylate cyclase) and cell-permeable 8-bromo-cAMP were used. SQ-22536 decreased the frequency of pacemaker potentials, and cell-permeable 8-bromo-cAMP increased the frequency of pacemaker potentials. The effects of SQ-22536 on pacemaker potential frequency and membrane hyperpolarization were rescued by co-treatment with glibenclamide (an ATP-sensitive $K^+$ channel blocker). However, neither $N^G$-nitro-L-arginine methyl ester (L-NAME, a competitive inhibitor of NO synthase) nor 1H-[1,2,4]oxadiazolo[4,3-${\alpha}$]quinoxalin-1-one (ODQ, an inhibitor of guanylate cyclase) had any effect on PACAP-induced activity. In conclusion, this study describes the effects of PACAP on ICC in the mouse colon. PACAP inhibited the pacemaker activity of ICC by acting through ATP-sensitive $K^+$ channels. These results provide evidence of a physiological role for PACAP in regulating gastrointestinal (GI) motility through the modulation of ICC activity.

Inhibitory Effects of Olmesartan on Catecholamine Secretion from the Perfused Rat Adrenal Medulla

  • Lim, Hyo-Jeong;Kim, Sang-Yong;Lim, Dong-Yoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.14 no.4
    • /
    • pp.241-248
    • /
    • 2010
  • The present sutdy aimed to determine whether olmesartan, an angiotensin II (Ang II) type 1 ($AT_1$) receptor blocker, can influence the CA release from the isolated perfused model of the rat adrenal medulla. Olmesartan ($5{\sim}50{\mu}M$) perfused into an adrenal vein for 90 min produced dose- and time-dependent inhibition of the CA secretory responses evoked by ACh (5.32 mM), high $K^+$ (56 mM, a direct membrane-depolarizer), DMPP (100 ${\mu}M$) and McN-A-343 (100 ${\mu}M$). Olmesartan did not affect basal CA secretion. Also, in adrenal glands loaded with olmesartan (15 ${\mu}M$), the CA secretory responses evoked by Bay-K-8644 (10 ${\mu}M$, an activator of voltage-dependent L-type $Ca^{2+}$ channels), cyclopiazonic acid (10 ${\mu}M$, an inhibitor of cytoplasmic $Ca^{2+}$-ATPase), veratridine (100 ${\mu}M$, an activator of voltage-dependent $Na^+$ channels), and Ang II (100 nM) were markedly inhibited. However, at high concentrations ($150{\sim}300{\mu}M$), olmesartan rather enhanced the ACh-evoked CA secretion. Taken together, these results show that olmesartan at low concentrations inhibits the CA secretion evoked by cholinergic stimulation (both nicotininc and muscarinic receptors) as well as by direct membrane depolarization from the rat adrenal medulla, but at high concentrations it rather potentiates the ACh-evoked CA secretion. It seems that olmesartan has a dual action, acting as both agonist and antagonist at nicotinic receptors of the isolated perfused rat adrenal medulla, which might be dependent on the concentration. It is also thought that this inhibitory effect of olmesartan may be mediated by blocking the influx of both $Na^+$ and $Ca^{2+}$ into the rat adrenomedullary chromaffin cells as well as by inhibiting the $Ca^{2+}$ release from the cytoplasmic calcium store, which is thought to be relevant to the $AT_1$ receptor blockade, in addition to its enhancement on the CA secreton.

Changes in the $Ca^{2+}\;and\;Mg^{2+}$ - dependent Adenosine Triphosphatase Activity and Ultrastructure of Marine Fishes by Partial Freezing III. Changes in the Ultrastructure of Muscle Tissues of Yellowtail during Low-temperature Preservation (a해산어의 부분동결에 의한 $Ca^{2+}\;및\;Mg^{2+}$ -dependent Adenosin Triphosphatase 활성 및 근섬유의 미세구조 변화 III. 저온저장 과정중 방어 근육조직의 미세구조의 변화)

  • 최경호;박찬성
    • Journal of the Korean Society of Food Science and Nutrition
    • /
    • v.20 no.6
    • /
    • pp.629-636
    • /
    • 1991
  • Yellowtail fishes(Seriola quinqueeradiata) were submitted to the storages using ice-cooling($0^{\circ}C$), partial freezing($-3^{\circ}C$) and freezing $-20^{\circ}C$) method. Changes in the structures of muscle during storage at different temperatures were investigated. The ice-cooling and partial freezing storage caused early decomposition of glycogen granules and mitochondrial inner membrane, but it was accorded to much slower manner comparing with that of ice-cooling storage. The scars of ice crystals were appeared after three days of storage. The number and size of the crystal increased as progressing of the storage. They were circular and mostly located between fibers. When using the freezing storage, glycogen granules were mostly found from the muscle cell even after fourteen days of storage. Mitochonidral inner membrane maintained their integrity. The scars of ice crystals were also found, however, different from those of partial freezing storage. Their existing sites were random and their shapes were irregular. In many cases, they located in the fiber and had keen edges. Fibers were broken mostly at the Z-lines on fourteen days of storage. From these results, it was concluded that partial freezing storage can repress autolytic enzymic action and can reduce the physical damage from ice crystals which is caused by freezing.

  • PDF

The Antitumor Effects of Selenium Compound $Na_5SeV_5O_{18}{\cdot}3H_2O$ in K562 Cell

  • Yang, Jun-Ying;Wang, Zi-Ren
    • Archives of Pharmacal Research
    • /
    • v.29 no.10
    • /
    • pp.859-865
    • /
    • 2006
  • With an approach to study the anti-tumor effects and mechanism of selenium compound, we investigated the anti-tumor activity and mechanism of $Na_5SeV_5O_{18}{\cdot}3H_2O$ (NaSeVO) in K562 cells. The results showed that $0.625{\sim}20\;mg/L$ NaSeVO could significantly inhibit the proliferation of K562 cells in vitro in a time- and concentration-dependent manner as determined by microculture tetrazolium (MTT) assay, the IC50 values were 14.41 (4.45-46.60) and 3.45 (2.29-5.22) mg/L after 48 hand 72 h treatment with NaSeVO respectively. In vivo experiments demonstrated that i.p. administration of 5, 10 mg/kg NaSeVO exhibited an significant inhibitory effect on the growth of transplantation tumor sarcoma 180 (S180) and hepatoma 22 (H22) in mice, with inhibition rate 26.8% and 58.4% on S180 and 31.3% and 47.4% on H22, respectively. Cell cycle studies indicated that the proportion of G0/G1 phase was increased at 2.5 mg/L while decreased at 10 mg/L after treatment for 24, 48 h. Whereas S phase was decreased at 2.5-5 mg/L and markedly increased at 10 mg/L after treatment for 48 h. After treatment for 24 h, 10 mg/L NaSeVO also markedly increased S and G2/M phases. Take together, the result clearly showed that NaSeVO markedly increased S and G2/M phases at 10 mg/L. The study of immunocytochemistry showed that the expression bcl-2 is significantly inhibited by 10 mg/L NaSeVO, and bax increased. Morphology observation also revealed typical apoptotic features. NaSeVO also significantly caused the accumulation of $Ca^{2+}$ and $Mg^{2+}$, reactive oxygen species (ROS) and the reduction of pH value and mitochondrial membrane potential in K562 cells as compared with control by confocal laser scanning microscope. These results suggest that NaSeVO has anti-tumor effects and its mechanism is attributed partially to apoptosis induced by the elevation of intracellular $Ca^{2+}$, $Mg^{2+}$ and ROS concentration, and a reduction of pH value and mitochondria membrane potential (MMP).

The Effect of 1-Propanol on the Rotational Mobility of n-(9-Anthroyloxy) stearic acid in Outer Monolayers of Neuronal and Model Membranes

  • Ahn, Tae-Young;Jin, Seong-Deok;Yang, Hak-Jin;Yoon, Chang-Dae;Kim, Mi-Kyung;An, Taek-Kyung;Bae, Young-Jun;Seo, Sang-Jin;Kim, Gwon-Su;Bae, Moon-Kyoung;Bae, Soo-Kyoung;Jang, Hye-Ock
    • International Journal of Oral Biology
    • /
    • v.42 no.4
    • /
    • pp.175-181
    • /
    • 2017
  • The aim of this study was to provide a basis for the molecular mechanism underlying the pharmacological action of ethanol. We studied the effects of 1-propanol on the location of n-(9-anthroyloxy)palmitic acid or stearic acid (n-AS) within the phospholipids of synaptosomal plasma membrane vesicles (SPMV). The SPMV were isolated from the bovine cerebral cortex and liposomes of total lipids (SPMVTL) and phospholipids (SPMVPL). 1-Propanol increased the rotational mobility of inner hydrocarbons, while decreasing the mobility of membrane interface, in native and model membranes. The degree of rotational mobility varied with the number of carbon atoms at positions 16, 12, 9, 6 and 2 in the aliphatic chain of phospholipids in the neuronal and model membranes. The sensitivity of increasing or decreasing rotational mobility of hydrocarbon interior or surface by 1-propanol varied with the neuronal and model membranes in the following order: SPMV, SPMVPL and SPMVTL.

Biochemical Studies of Ginseng Saponin on RNA and Protein Biosynthesis in the Rat Liver (간에서의 RNA, 단백질 생합성에 미치는 인삼성분의 생화학적 연구)

  • Oura Hikokichi
    • Proceedings of the Ginseng society Conference
    • /
    • 1988.08a
    • /
    • pp.1-10
    • /
    • 1988
  • Previously. we reported that the intraperitoneal administration of ginseng crude saponin increased: (I) nuclear RNA polymerase activity. (2) nuclear RNA synthesis. (3) cytoplasmic RNA synthesis. (4) cytoplasmic heavy polyrioosome content. (5) amino acid incorporation in vitro of microsome and polysome isolated rat liver. and (6) the incorporation rate of labeled amino acids into serum protein. In addition, a spectacular increase in the rough endoplasmic reticulum of hepatocyte administered crude saponin for four weeks orally was shown through electron microscopy. An increase in polysomal content in membrane-hound ribosome was shown through ultracentrifugation. Recently, successive intraperitoneal. administration .of $ginsenosid-Rb_2$ was given to streptozotocin (STZ) diaoetic rats of hypoproteinemia. The blood urea nitrogen and hepatic urea concentration were decreased significantly. The total protein and alhumin levels in the serum were increased in comparison to control values. In contrast. the $ginsenoside-Rb_2$ treated group of STZ diahetic rats showed a significant increase in liver RNA. total ribosome and membrane-bound ribosomal contents. The administration of $ginsenoside-Rb_2$ increased the incorporation rate of labeled - precursor into total serum protein. Additionally $ginsenoside-Rb_2$ improved the nitrogen balance of diabetic rats. On the bases of these experimental results, ginseng saponin has a metabolic stimulatory or anabolic action on RNA and protein synthesis.

  • PDF

R-(-)-TNPA, a Dopaminergic $D_2$ Receptor Agonist, Inhibits Catecholamine Release from the Rat Adrenal Medulla

  • Hong, Soon-Pyo;Seo, Hong-Joo;Lim, Dong-Yoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.10 no.5
    • /
    • pp.273-282
    • /
    • 2006
  • The aim of the present study was to investigate the effects of R-(-)-2,10,11-trihydroxy-N-propylnoraporphine [R-(-)-TNPA], a selective agonist of dopaminergic $D_2$ receptor and S(-)-raclopride, a selective antagonist of dopaminergic $D_2$ receptor, on the secretion of catecholamines (CA) evoked by cholinergic stimulation and membrane-depolarization in the isolated perfused model of the rat adrenal gland, and also to establish its mechanism of action. R-(-)-TNPA $(10{\sim}100\;{\mu}M)$ perfused into an adrenal vein for 60 min produced dose- and time-dependent inhibition in CA secretory responses evoked by ACh (5.32 mM), high $K^+$ (56 mM), DMPP $(100\;{\mu}M)$ and McN-A-343 $(100\;{\mu}M)$. R-(-)-TNPA itself did also fail to affect basal CA output. Also, in adrenal glands loaded with R-(-)-TNPA $(30\;{\mu}M)$, the CA secretory responses evoked by Bay-K-8644 $(10\;{\mu}M)$, an activator of L-type $Ca^2+$ channels and cyclopiazonic acid $(10\;{\mu}M)$, an inhibitor of cytoplasmic $Ca^{2+}-ATPase$ were also inhibited. However, S(-)-raclopride $(1{\sim}10\;{\mu}M)$, given into an adrenal vein for 60 min, enhanced the CA secretory responses evoked by ACh, high $K^+$, DMPP and McN-A-343 only for the first period (4 min), although it alone has weak effect on CA secretion. Moreover, S(-)-raclopride $(3.0\;{\mu}M)$ in to an adrenal vein for 60 min also augmented the CA release evoked by BAY-K-8644 and cyclopiazonic acid only for the first period (4 min). However, after simultaneous perfusion of R-(-)-TNP A $(30\;{\mu}M)$ and S(-)-raclopride $(3.0\;{\mu}M)$, the inhibitory responses of R(-)-TNPA $(30\;{\mu}M)$ on the CA secretion evoked by ACh, high $K^+$, DMPP, McN-A-343, Bay-K-8644, and cyclopiazonic acid were significantly reduced. Taken together, these experimental results suggest that R-(-)-TNPA greatly inhibits the CA secretion from the perfused rat adrenal medulla evoked by cholinergic stimulation (both nicotininc and muscarinic receptors) and membrane depolarization, but S(-)-raclopride rather enhances the CA release by them. It seems that this inhibitory of R-(-)-TNPA may be mediated by stimulation of inhibitory dopaminergic $D_2$ receptors located on the rat adrenomedullary chromaffin cells, while the facilitatory effect of S(-)-raclopride is due to the blockade of dopaminergic $D_2$ receptors, which are relevant to extra- and intracellular calcium mobilization. Therefore, it is thought that dopaminergic $D_2$ receptors may be involved in regulation of CA release in the rat adrenal medulla.

Roles of Dopaminergic $D_1\;and\;D_2$ Receptors in Catecholamine Release from the Rat Adrenal Medulla

  • Baek, Young-Joo;Seo, Yoo-Seong;Lim, Dong-Yoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.12 no.1
    • /
    • pp.13-23
    • /
    • 2008
  • The aim of the present study was designed to establish comparatively the inhibitory effects of $D_1$-like and $D_2$-like dopaminergic receptor agonists, SKF81297 and R(-)-TNPA on the release of catecholamines (CA) evoked by cholinergic stimulation and membrane depolarization from the isolated perfused model of the rat adrenal medulla. SKF81297 $(30{\mu}M)$ and R-(-)-TNPA $(30{\mu}M)$ perfused into an adrenal vein for 60 min, produced great inhibition in the CA secretory responses evoked by ACh $(5.32{\times}10^{-3}\;M)$, DMPP $(10^{-4}\;M)$, McN-A-343 $(10^{-4}\;M)$, high $K^+$ $(5.6{\times}10^{-2}\;M)$, Bay-K-8644 $(10{\mu}M)$, and cyclopiazonic acid $(10{\mu}M)$, respectively. For the release of CA evoked by ACh, high $K^+$, DMPP, McN-A-343, Bay-K-8644 and cyclopiazonic acid, the following rank order of inhibitory potency was obtained: SKF81297>R-(-)-TNPA. However, R(+)-SCH23390, a selectve $D_1$-like dopaminergic receptor antagonist, and S(-)-raclopride, a selectve $D_2$-like dopaminergic receptor antagonist, enhanced the CA secretory responses evoked by ACh, high $K^+$, DMPP, McN-A-343, Bay-K-8644 and cyclopiazonic acid only for $0{\sim}4$ min. The rank order for the enhancement of CA release evoked by high $K^+$, McN-A-343 and cyclopiazonic acid was R(+)-SCH23390>S(-)-raclopride. Also, the rank order for ACh, DMPP and Bay-K-8644 was S(-)-raclopride > R(+)-SCH23390. Taken together, these results demonstrate that both SKF81297 and R-(-)-TNPA inhibit the CA release evoked by stimulation of cholinergic (both nicotinic and muscarinic) receptors and the membrane depolarization from the isolated perfused rat adrenal gland without affecting the basal release, respectively, but both R(+)-SCH23390 and S(-)-raclopride facilitate the CA release evoked by them. It seems likely that the inhibitory effects of SKF81297 and R-(-)-TNPA are mediated by the activation of $D_1$-like and $D_2$-like dopaminergic receptors located on the rat adrenomedullary chromaffin cells, respectively, whereas the facilitatory effects of R(+)-SCH23390 and S(-)-raclopride are mediated by the blockade of $D_1$-like and $D_2$-like dopaminergic receptors, respectively: this action is possibly associated with extra- and intracellular calcium mobilization. Based on these results, it is thought that the presence of dopaminergic $D_1$ receptors may play an important role in regulation of the rat adrenomedullary CA secretion, in addition to well-known dopaminergic $D_2$ receptors.