1.Chronic intermittent hypoxia decreases acute hypoxic inhibition of voltage-gated potassium channel in rat pulmonary arterial smooth muscle cells.
Bi TANG ; Ming TANG ; Yi-Mei DU ; Chang-Jin LIU ; Zhi-Gang HONG ; Hong-Yan LUO ; Xin-Wu HU ; Yuan-Long SONG ; Jiao-Ya XI ; Jurgen HESCHELER
Acta Physiologica Sinica 2004;56(5):625-631
For determination the ionic mechanisms of the hypoxic acclimatization at the level of channels, male Spradue-Dawley rats were divided into two groups: control normoxic group and chronic intermittent hypoxic group [O2 concentration: (10 +/-0.5)%, hypoxia 8 h a day]. Using whole cell patch-clamp technique, voltage-gated potassium channel currents (IK(V)) were recorded in freshly isolated pulmonary arterial smooth muscle cells (PASMCs) of rat with acute isolated method. The effect of acute hypoxia on IK(V) of PASMCs from chronic intermittent hypoxia group was investigated to offer some basic data for clarifying the ionic mechanisms of the hypoxic acclimatization. The results showed: (1) In control normoxic group, after acute hypoxia free-Ca(2+) solution, the resting membrane potential (Em) of PASMCs was depolarized significantly from -47.2+/-2.6 mV to -26.7+/-1.2 mV, and the IK(V) of PASMCs was decreased significantly from 153.4+/-9.5 pA/pF to 70.1+/-0.6 pA/pF, the peak current percent inhibition was up to (57.6+/-3.3)% at +60 mV, and current-voltage relationship curve shifted to the right. (2) In chronic intermittent hypoxic group, the IK(V) of PASMCs was decreased significantly by exposure to intermittent hypoxia in a time-dependent manner, appeared to start on day 10 and continued to day 30 (the longest time tested) of hypoxia, and current-voltage relationship curve shifted to the right in a time-dependent manner. (3) Compared with the control normoxic group, the percent IK(V) inhibition by acute hypoxia was significantly attenuated in the chronic intermittent hypoxia group and this inhibition effect declined with time exposure to hypoxia. The results suggest that K(V) inhibition was significantly attenuated by chronic intermittent hypoxia, and this effect may be a critical mechanism of the body hypoxic acclimatization.
Animals
;
Cell Separation
;
Hypoxia
;
complications
;
physiopathology
;
Male
;
Muscle, Smooth, Vascular
;
cytology
;
metabolism
;
physiology
;
Potassium Channels, Voltage-Gated
;
antagonists & inhibitors
;
Pulmonary Artery
;
metabolism
;
pathology
;
Rats
;
Rats, Sprague-Dawley
2.Arsenic trioxide-induced hela cell death is partially prevented by K+ channel blockers.
De-Li DONG ; Wei-Hua SONG ; Pei-Lin ; Bao-Feng YANG
Acta Pharmaceutica Sinica 2005;40(7):644-648
AIMTo investigate the effects of K+ channel blockers on arsenic trioxide-induced HeLa cell death.
METHODSViability of HeLa cells was assessed by mitochondrial dehydrogenase activity using colorimetric MTT assay and the voltage-dependent K+ currents were recorded by using patch-clamp technique.
RESULTSExposure of As2O3 (5 micromol x L(-1)) for 24 h caused marked HeLa cell death. The rest living cells after As2O3 24 h-incubation showed significant increase of K+ currents densities. At +80 mV, the densities of K+ currents (61 +/- 18) pA/10 pF (n = 8) in As2O3 24 h-incubation group were significantly more than that in the control group (38 +/- 10) pA/10 pF (n = 8, P < 0.05). The HeLa cells were prevented partially from As2O3-induced cell death by co-application for 24 h with typical voltage-dependent K+ channel blockers, 4-aminopyridine (3 mmol x L(-1)) or tetraethylammonium (5 mmol x L(-1)). 4-Aminopyridine (3 mmol x L(-1)) or tetraethylammonium (5 mmol x L(-1)) did not show any toxic effects on HeLa cells.
CONCLUSIONChronic treatment with As2O3 increased voltage-dependent K+ currents in HeLa cells and the cell death induced by As2O3 was reduced partially by voltage-dependent K+ channel blockers, 4-aminopyridine or tetraethylammonium.
4-Aminopyridine ; pharmacology ; Arsenicals ; antagonists & inhibitors ; pharmacology ; Cell Death ; drug effects ; HeLa Cells ; Humans ; Oxides ; antagonists & inhibitors ; pharmacology ; Potassium Channel Blockers ; pharmacology ; Potassium Channels, Voltage-Gated ; drug effects ; Tetraethylammonium ; pharmacology
3.Influence of 4-aminopyridine on voltage-activated K(+) current and cell proliferation in small cell lung cancer.
Liping WANG ; Guoguang SHAO ; Wenjie ZHANG ; Xiping GUO ; Chunguang WANG ; Jihong AN ; Guogan ZHONG ; Hua ZHAO
Chinese Journal of Oncology 2002;24(3):230-233
OBJECTIVETo study the inhibition of voltage-activated K(+) conductance and cell proliferation by 4-aminopyridine (4-AP) in the human small-cell lung cancer (SCLC).
METHODSInhibition of voltage-activated K(+) current by 4-AP through the whole-cell patch-clamp technique in SCLC cell line was studied. The influence on the cell-cycle by 4-AP was observed by flow cytometry to identify the in vitro inhibition by 4-AP to the cell proliferation of the SCLC cell line.
RESULTSExposure of the tumor cells to 5 mmol/L 4-AP reduced the peak outward K(+) current (evoked by a depolarization to +80 mV) from 1.22 +/- 0.11 nA (n = 30) to 0.59 +/- 0.10 nA (n = 28). Flow cytometry results showed that cell population accumulated in the G(0)/G(1) phase and a significantly reduced proportion in the S phase and G(1)/G(2) phase cells after having been exposed to 4-AP for three days. Incubation of the SCLC cells with 0.1, 5, 10, 15, 20 mmol/L 4-AP resulted in a concentration-and time-dependent reduction in the number of viable cells as compared with the control.
CONCLUSIONThe voltage-activated K(+) channels expressed by SCLC play an important role in SCLC cell proliferation. The proliferation of the SCLC cells is inhibited by K(+) channel antagonists.
4-Aminopyridine ; pharmacology ; Carcinoma, Small Cell ; pathology ; Cell Division ; drug effects ; Humans ; Lung Neoplasms ; pathology ; Potassium Channel Blockers ; pharmacology ; Potassium Channels, Voltage-Gated ; antagonists & inhibitors ; metabolism ; Tumor Cells, Cultured
4.Voltage-activated potassium channel blockers inhibit anisodamine-induced relaxation of rabbit aortic smooth muscles precontracted with noradrenaline.
Shu-Qin LIU ; Wei-Jin ZANG ; Zeng-Li LI ; Qiang SUN ; Xiao-Jiang YU ; Hong-Li LUO ; Shu-Ming ZHU
Acta Physiologica Sinica 2005;57(1):21-26
Anisodamine, which is originally extracted from scopolia tangutica and is currently produced in China, is a tropane alkaloid and a muscarinic cholinoceptor blocker. Our previous study found that anisodamine did not alter high K(+)-evoked contraction of rabbit aortic rings using isometric tension recording methods, but could attenuate noradrenaline (NA)-, histamine- or 5-hydroxytryptamine-induced contraction in an endothelium-independent manner. Since the high K(+)-elicited depolarization non-selectively inhibits potassium channels in vascular smooth muscle cell (VSMC) membrane, the vasodilation effect of some potassium channel activators may be inhibited or abolished in high K(+) solution. We hypothesized that some potassium channels in VSMC membrane might play a role in the anisodamine-induced relaxation of blood vessels. The present experiment was designed to investigate whether potassium channel blockers inhibit anisodamine-induced relaxation of the rabbit isolated aortic rings. In a 8-min period, 1, 3 and 10 micromol/L of anisodamine, significantly relaxed the 0.01 micromol/L NA precontracted aortic ring by (19.1+/-3.1)%, (30.1+/-3.8)% and (38.3+/-4.2)%, respectively, compared with the controls [by (4.8+/-2.4)%, (5.1+/-1.8)% and (5.6+/-2.5)%, respectively] (P<0.01). 10 mmol/L of CsCl (a non-selective potassium channel blocker), 1 mmol/L of 4-aminopyridine [a selective voltage-activated potassium channel (K(V)) blocker], 10 mumol/L BaCl2 (a selective inwardly-rectifying potassium channel blocker), 10 micromol/L of glibenclamide (a selective ATP-sensitive potassium channel blocker), 3 micromol/L of charybdotoxin (a large- and intermediate-conductance Ca(2+)-activated potassium channels blocker) and 3 micromol/L of apamin (a selective small conductance Ca(2+)-activated potassium channel blocker) significantly increased the NA-induced contraction by (14.4+/-3.2)%, (16.3+/-5.8)%, (12.7+/-4.2)%, (13.6+/-2.0)%, (11.1+/-5.5)% and (13.4+/-4.3)%, respectively, compared with the control [by (5.6 +/-1.2)%] (P<0.01). In the presence of 10 and 30 mmol/L CsCl or 1 and 3 mmol/L 4-aminopyridine, anisodamine-induced relaxation of the 0.01 micromol/L NA contracted rabbit aortic rings [(28.8+/-3.0)% and (15.9+/-3.7)% or (29.7+/-3.9)% and (19.0+/-5.0)%] significantly deceased, compared with that in the absence of any potassium channel blocker [(38.3+/-4.2)% (P<0.01)] in a 8-min period. However, in the presence of 10, 30 micromol/L of BaCl2, 10, 30 micromol/L of glibenclamide, 3 micromol/L of charybdotoxin, or 3 micromol/L apamin, 10 micromol/L anisodamine-induced relaxation [(37.1+/-3.8)%, (36.2+/-4.7)%, (36.1+/-2.7)%, (35.6+/-3.3)%, (37.8+/-2.0)% and (39.3 +/-4.7) %, respectively] did not decrease, compared with the control [(38.3+/-4.2)%] (P>0.05). This study suggests that K(V) blockers inhibit anisodamine-induced relaxation of the rabbit aortic smooth muscle precontracted with NA and implies that the K(V) in VSMC membrane plays a role in anisodamine-induced relaxation of blood vessels.
Animals
;
Aorta
;
cytology
;
Female
;
Male
;
Muscle Contraction
;
drug effects
;
Muscle Relaxation
;
drug effects
;
Muscle, Smooth, Vascular
;
physiology
;
Norepinephrine
;
antagonists & inhibitors
;
Potassium Channel Blockers
;
pharmacology
;
Potassium Channels, Voltage-Gated
;
Rabbits
;
Solanaceous Alkaloids
;
pharmacology
5.Synthesis, refolding and identification of pharmacological activities of neurotoxin JZTX-XI and R3A-JZTX-XI.
Yupeng CHI ; Meichun DENG ; Yuanyuan WU ; Ji LUO ; Minqiang RONG ; Yiya ZHANG ; Dongyi ZHANG ; Xiongzhi ZENG ; Songping LIANG
Chinese Journal of Biotechnology 2011;27(6):900-908
Kv2.1 channel currents in pancreatic beta-cells are thought to contribute to action potential repolarization and thereby modulate insulin secretion. Because of its central role in this important physiological process, Kv2.1 channel is a promising target for the treatment of type 2 diabetes. Jingzhaotoxin-XI (JZTX-XI) is a novel peptide neurotoxin isolated from the venom of the spider Chilobrachys jingzhao. Two-microelectrode voltage clamp experiments had showed that the toxin inhibited Kv2.1 potassium currents expressed in Xenopus Laevis oocytes. In order to investigate the structure-function relationship of JZTX-XI, the natural toxin and a mutant of JZTX-XI in which Arg3 was replaced by Ala, were synthesized by solid-phase chemistry method with Fmoc-protected amino acids on the PS3 automated peptide synthesizer. Reverse-phase high performance liquid chromatography (RP-HPLC) and matrix assisted laser desorption/ ionization time-of-flight mass spectrometry (MALDI-TOF/TOF MS) were used to monitor the oxidative refolding process of synthetic linear peptides to find the optimal renaturation conditions of these toxins. The experiments also proved that the relative molecular masses of refolded peptides were in accordance with their theoretical molecular masses. RP-HPLC chromatogram of co-injected native and refolded JZTX-XI was a single peak. Under the whole-cell patch-clamp mode, JZTX-XI could completely inhibit hKv2.1 and hNav1.5 channels currents expressed in HEK293T cells with IC50 values of 95.8 nmol/L and 437.1 nmol/L respectively. The mutant R3A-JZTX-XI could also inhibit hKv2.1 and hNav1.5 channel currents expressed in HEK293T cells with IC50 values of 1.22 micromol/L and 1.96 micromol/L respectively. However, the prohibitive levels of R3A-JZTX-XI on hKv2.1 and hNav1.5 channels were reduced by about 12.7 times and 4.5 times respectively, indicating that Arg3 was a key amino acid residue relative to the hKv2.1 channel activity of JZTX-XI, but it is also an amino acid residue correlated with the binding activity of JZTX-XI to hNav1.5 channel. Our findings should be helpful to develop JZTX-XI into a molecular probe and drug candidate targeting to Kv2.1 potassium channel in the pancreas.
Animals
;
HEK293 Cells
;
Humans
;
Insulin-Secreting Cells
;
metabolism
;
Mutant Proteins
;
genetics
;
pharmacology
;
NAV1.5 Voltage-Gated Sodium Channel
;
metabolism
;
Neurotoxins
;
chemical synthesis
;
genetics
;
pharmacology
;
Protein Refolding
;
Shab Potassium Channels
;
antagonists & inhibitors
;
metabolism
;
Sodium Channel Blockers
;
pharmacology
;
Spider Venoms
;
genetics
;
pharmacology
;
Transfection
6.Effects of potassium channel blockers on the proliferation of rat bronchial smooth muscle cells.
Xian-sheng LIU ; Yong-jian XU ; Zhen-xiang ZHANG ; Wang NI
Acta Pharmaceutica Sinica 2003;38(5):333-336
AIMTo investigate the effects of blockers of the three kinds of potassium channels: voltage-dependent K+ channel(KV), calcium-activated K+ channel(KCa) and ATP-sensitive K+ channel(KATP), on the proliferation of rat bronchial smooth muscle cells (BSMCs).
METHODSThe effects of three kinds of potassium channel blockers on the proliferation of BSMCs were detected by MTT method, immunocytochemistry staining and flow-cytometry. Their effects on the dynamic changes of Ca2+ concentration in BSMCs were investigated by the fluorophotometry of Fura-2/AM.
RESULTSThe KV blocker 4-aminopyridine (4-AP) was shown to significantly increase the expression of proliferating cell nucleus antigen in cultured rat BSMCs (P < 0.01), but the KCa blocker tetraethylammonium (TEA) and KATP blocker glibenclamide(Glib) did not show such effect (P > 0.05). 4-AP was found to significantly increase the optical density value of the cultured BSMCs (P < 0.01) by MTT method and the numbers of S + G2M BSMCs(P < 0.05) by flow-cytometry. TEA and Glib did not show such effects. 4-AP significantly increased the Ca2+ concentration in cultured BSMCs(P < 0.01). TEA and Glib did not show such effects.
CONCLUSIONThis result suggests that inhibition of KV activity can increase intracellular Ca2+ and proliferation of rat BSMCs, but inhibition of KCa and KATP showed no effect.
4-Aminopyridine ; pharmacology ; Animals ; Bronchi ; cytology ; Calcium ; metabolism ; Cell Division ; drug effects ; Cells, Cultured ; Glyburide ; pharmacology ; Muscle, Smooth ; drug effects ; metabolism ; Potassium Channel Blockers ; pharmacology ; Potassium Channels, Calcium-Activated ; antagonists & inhibitors ; Potassium Channels, Voltage-Gated ; antagonists & inhibitors ; Proliferating Cell Nuclear Antigen ; metabolism ; Rats ; Rats, Sprague-Dawley ; Tetraethylammonium ; pharmacology
7.Effects of amiloride on potassium and calcium currents in guinea pig ventricular myocytes.
Lan CHENG ; Fang WANG ; Hong-Yi ZHOU ; Wei-Xing YAO ; Guo-Jin XIA ; Ming-Xing JIANG
Acta Pharmaceutica Sinica 2004;39(7):509-513
AIMTo elucidate the possible mechanisms underlying antiarrhythmia of the non-selective Na+/H+ exchanger inhibitor--amiloride.
METHODSSingle ventricular cells were isolated using a double-enzyme method. Effects of amiloride on voltage-dependent potassium and calcium currents in isolated guinea pig ventricular myocyte were recorded by using whole-cell patch clamp techniques.
RESULTSExposure to amiloride (10 -100 micromol x L(-1)), the L-type and T-type calcium currents were depressed. Amiloride resulted in a concentration-dependent inhibition of peak (Ca,L), But amiloride did not change the shape of their I - V curves. It only decreased the amplitudes of the currents of the two types. When myocytes were incubated with 100 micromol x L(-1) amiloride, I(Kr) was slightly depressed and I(Ks) did not change. Amiloride (1 - 100 micromol x L(-10) depressed I(K1) in a concentration-dependent manner.
CONCLUSIONAmiloride depressed potassium and calcium currents, which may give support to its uses in some diseases of the cardiovascular system.
Amiloride ; pharmacology ; Animals ; Anti-Arrhythmia Agents ; pharmacology ; Calcium Channels, L-Type ; drug effects ; Calcium Channels, T-Type ; drug effects ; Cell Separation ; Female ; Guinea Pigs ; Heart Ventricles ; cytology ; Male ; Myocytes, Cardiac ; drug effects ; Patch-Clamp Techniques ; Potassium Channels, Inwardly Rectifying ; drug effects ; Potassium Channels, Voltage-Gated ; drug effects ; Sodium-Hydrogen Exchangers ; antagonists & inhibitors
8.The role of subtypes of voltage-gated K+ channels in pulmonary vasoconstriction induced by 15-hydroeicosatetraenoic acid.
Qian LI ; Rong ZHANG ; Chang-Lian LÜ ; Yan LIU ; Zhen WANG ; Da-Ling ZHU
Acta Pharmaceutica Sinica 2006;41(5):412-417
AIMTo observe the effect of subtypes of Kv channels in rat pulmonary artery smooth muscle cells (PASMCs) on the process of pulmonary vasoconstriction induced by 15-HETE.
METHODSIn the present study, ring of rabbit PA with specific Kv channel blockers were employed to functionally identify certain channel subtypes that took part in the process of 15-HETE induced pulmonary vasoconstriction; RT-PCR and Western blotting analysis were also used to measure the expression of subtypes of Kv in PASMCs exposed to 15-HETE,chronic hypoxia.
RESULTSBlocking of Kv1. 1, Kv1. 2, Kv1. 3 and Kv1. 6 channels did not affect 15-HETE induced vasoconstriction in normoxic rats; 15-HETE did not affect expression of Kv1. 1 and Kv1. 2 channels; 15-HETE significantly downregulated the expression of mRNA and protein of Kv1. 5 and Kv2. 1 in rat PASMCs.
CONCLUSIONThe results suggested that hypoxia may block Kv1. 5 and Kv2. 1 channels via 15-HETE mediated mechanism, leading to decrease numbers of functional Kv1. 5 and Kv2. 1 channels in PASMCs, leading to PA vasoconstriction.
Animals ; Cell Hypoxia ; Cells, Cultured ; Hydroxyeicosatetraenoic Acids ; pharmacology ; Hypoxia ; physiopathology ; Kv1.5 Potassium Channel ; biosynthesis ; genetics ; Male ; Muscle, Smooth, Vascular ; cytology ; Myocytes, Smooth Muscle ; cytology ; metabolism ; Potassium Channels, Voltage-Gated ; antagonists & inhibitors ; Pulmonary Artery ; physiopathology ; RNA, Messenger ; biosynthesis ; genetics ; Rats ; Rats, Wistar ; Shab Potassium Channels ; biosynthesis ; genetics ; Vasoconstriction ; drug effects
9.Serotonin contracts the rat mesenteric artery by inhibiting 4-aminopyridine-sensitive Kv channels via the 5-HT2A receptor and Src tyrosine kinase.
Dong Jun SUNG ; Hyun Ju NOH ; Jae Gon KIM ; Sang Woong PARK ; Bokyung KIM ; Hana CHO ; Young Min BAE
Experimental & Molecular Medicine 2013;45(12):e67-
Serotonin (5-hydroxytryptamine (5-HT)) is a neurotransmitter that regulates a variety of functions in the nervous, gastrointestinal and cardiovascular systems. Despite such importance, 5-HT signaling pathways are not entirely clear. We demonstrated previously that 4-aminopyridine (4-AP)-sensitive voltage-gated K+ (Kv) channels determine the resting membrane potential of arterial smooth muscle cells and that the Kv channels are inhibited by 5-HT, which depolarizes the membranes. Therefore, we hypothesized that 5-HT contracts arteries by inhibiting Kv channels. Here we studied 5-HT signaling and the detailed role of Kv currents in rat mesenteric arteries using patch-clamp and isometric tension measurements. Our data showed that inhibiting 4-AP-sensitive Kv channels contracted arterial rings, whereas inhibiting Ca2+-activated K+, inward rectifier K+ and ATP-sensitive K+ channels had little effect on arterial contraction, indicating a central role of Kv channels in the regulation of resting arterial tone. 5-HT-induced arterial contraction decreased significantly in the presence of high KCl or the voltage-gated Ca2+ channel (VGCC) inhibitor nifedipine, indicating that membrane depolarization and the consequent activation of VGCCs mediate the 5-HT-induced vasoconstriction. The effects of 5-HT on Kv currents and arterial contraction were markedly prevented by the 5-HT2A receptor antagonists ketanserin and spiperone. Consistently, alpha-methyl 5-HT, a 5-HT2 receptor agonist, mimicked the 5-HT action on Kv channels. Pretreatment with a Src tyrosine kinase inhibitor, 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine, prevented both the 5-HT-mediated vasoconstriction and Kv current inhibition. Our data suggest that 4-AP-sensitive Kv channels are the primary regulator of the resting tone in rat mesenteric arteries. 5-HT constricts the arteries by inhibiting Kv channels via the 5-HT2A receptor and Src tyrosine kinase pathway.
4-Aminopyridine/pharmacology
;
Action Potentials
;
Animals
;
Calcium Channel Blockers/pharmacology
;
Calcium Channels/metabolism
;
Cells, Cultured
;
Ketanserin/pharmacology
;
Male
;
Mesenteric Arteries/drug effects/*metabolism/physiology
;
Muscle Contraction
;
Muscle, Smooth, Vascular/cytology/drug effects/metabolism/physiology
;
Myocytes, Smooth Muscle/drug effects/metabolism/physiology
;
Nifedipine/pharmacology
;
Potassium Channel Blockers/pharmacology
;
Potassium Channels, Voltage-Gated/antagonists & inhibitors/*metabolism
;
Protein Kinase Inhibitors/pharmacology
;
Rats
;
Rats, Sprague-Dawley
;
Receptor, Serotonin, 5-HT2A/*metabolism
;
Serotonin/*pharmacology
;
Serotonin 5-HT2 Receptor Antagonists/pharmacology
;
Spiperone/pharmacology
;
*Vasoconstriction
;
src-Family Kinases/antagonists & inhibitors/*metabolism