27 results on '"Potassium Channels, Calcium-Activated agonists"'
Search Results
2. Nitric oxide regulates neuronal activity via calcium-activated potassium channels.
- Author
-
Zhong LR, Estes S, Artinian L, and Rehder V
- Subjects
- 4-Aminopyridine pharmacology, Action Potentials, Animals, Apamin pharmacology, Calcium Channels metabolism, Cells, Cultured, Ganglia, Autonomic cytology, Growth Cones physiology, Helix, Snails, Nitric Oxide Donors pharmacology, Patch-Clamp Techniques, Peptides pharmacology, Potassium Channel Blockers pharmacology, Potassium Channels, Calcium-Activated agonists, Tetraethylammonium pharmacology, Motor Neurons physiology, Nitric Oxide physiology, Potassium Channels, Calcium-Activated metabolism
- Abstract
Nitric oxide (NO) is an unconventional membrane-permeable messenger molecule that has been shown to play various roles in the nervous system. How NO modulates ion channels to affect neuronal functions is not well understood. In gastropods, NO has been implicated in regulating the feeding motor program. The buccal motoneuron, B19, of the freshwater pond snail Helisoma trivolvis is active during the hyper-retraction phase of the feeding motor program and is located in the vicinity of NO-producing neurons in the buccal ganglion. Here, we asked whether B19 neurons might serve as direct targets of NO signaling. Previous work established NO as a key regulator of growth cone motility and neuronal excitability in another buccal neuron involved in feeding, the B5 neuron. This raised the question whether NO might modulate the electrical activity and neuronal excitability of B19 neurons as well, and if so whether NO acted on the same or a different set of ion channels in both neurons. To study specific responses of NO on B19 neurons and to eliminate indirect effects contributed by other cells, the majority of experiments were performed on single cultured B19 neurons. Addition of NO donors caused a prolonged depolarization of the membrane potential and an increase in neuronal excitability. The effects of NO could mainly be attributed to the inhibition of two types of calcium-activated potassium channels, apamin-sensitive and iberiotoxin-sensitive potassium channels. NO was found to also cause a depolarization in B19 neurons in situ, but only after NO synthase activity in buccal ganglia had been blocked. The results suggest that NO acts as a critical modulator of neuronal excitability in B19 neurons, and that calcium-activated potassium channels may serve as a common target of NO in neurons.
- Published
- 2013
- Full Text
- View/download PDF
3. Exchange protein activated by cAMP (Epac) induces vascular relaxation by activating Ca2+-sensitive K+ channels in rat mesenteric artery.
- Author
-
Roberts OL, Kamishima T, Barrett-Jolley R, Quayle JM, and Dart C
- Subjects
- Action Potentials, Animals, Apamin pharmacology, Calcium metabolism, Cells, Cultured, Cyclic AMP analogs & derivatives, Cyclic AMP pharmacology, Guanine Nucleotide Exchange Factors agonists, Male, Mesenteric Arteries cytology, Mesenteric Arteries physiology, Muscle Cells drug effects, Muscle Cells physiology, Muscle Contraction, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular physiology, NG-Nitroarginine Methyl Ester pharmacology, Peptides pharmacology, Potassium pharmacology, Potassium Channel Blockers pharmacology, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated antagonists & inhibitors, Pyrazoles pharmacology, Rats, Rats, Wistar, Guanine Nucleotide Exchange Factors metabolism, Mesenteric Arteries metabolism, Muscle Cells metabolism, Potassium Channels, Calcium-Activated metabolism, Vasodilation
- Abstract
Vasodilator-induced elevation of intracellular cyclic AMP (cAMP) is a central mechanism governing arterial relaxation but is incompletely understood due to the diversity of cAMP effectors. Here we investigate the role of the novel cAMP effector exchange protein directly activated by cAMP (Epac) in mediating vasorelaxation in rat mesenteric arteries. In myography experiments, the Epac-selective cAMP analogue 8-pCPT-2-O-Me-cAMP-AM (5 μM, subsequently referred to as 8-pCPT-AM) elicited a 77.6 ± 7.1% relaxation of phenylephrine-contracted arteries over a 5 min period (mean ± SEM; n = 6). 8-pCPT-AM induced only a 16.7 ± 2.4% relaxation in arteries pre-contracted with high extracellular K(+) over the same time period (n = 10), suggesting that some of Epac's relaxant effect relies upon vascular cell hyperpolarization. This involves Ca(2+)-sensitive, large-conductance K(+) (BK(Ca)) channel opening as iberiotoxin (100 nM) significantly reduced the ability of 8-pCPT-AM to reverse phenylephrine-induced contraction (arteries relaxed by only 35.0 ± 8.5% over a 5 min exposure to 8-pCPT-AM, n = 5; P < 0.05). 8-pCPT-AM increased Ca(2+) spark frequency in Fluo-4-AM-loaded mesenteric myocytes from 0.045 ± 0.008 to 0.103 ± 0.022 sparks s(-1) μm(-1) (P < 0.05) and reversibly increased both the frequency (0.94 ± 0.25 to 2.30 ± 0.72 s(-1)) and amplitude (23.9 ± 3.3 to 35.8 ± 7.7 pA) of spontaneous transient outward currents (STOCs) recorded in isolated mesenteric myocytes (n = 7; P < 0.05). 8-pCPT-AM-activated STOCs were sensitive to iberiotoxin (100 nM) and to ryanodine (30 μM). Current clamp recordings of isolated myocytes showed a 7.9 ± 1.0 mV (n = 10) hyperpolarization in response to 8-pCPT-AM that was sensitive to iberiotoxin (n = 5). Endothelial disruption suppressed 8-pCPT-AM-mediated relaxation in phenylephrine-contracted arteries (24.8 ± 4.9% relaxation after 5 min of exposure, n = 5; P < 0.05), as did apamin and TRAM-34, blockers of Ca(2+)-sensitive, small- and intermediate-conductance K(+) (SK(Ca) and IK(Ca)) channels, respectively, and N(G)-nitro-L-arginine methyl ester, an inhibitor of nitric oxide synthase (NOS). In Fluo-4-AM-loaded mesenteric endothelial cells, 8-pCPT-AM induced a sustained increase in global Ca(2+). Our data suggest that Epac hyperpolarizes smooth muscle by (1) increasing localized Ca(2+) release from ryanodine receptors (Ca(2+) sparks) to activate BK(Ca) channels, and (2) endothelial-dependent mechanisms involving the activation of SK(Ca)/IK(Ca) channels and NOS. Epac-mediated smooth muscle hyperpolarization will limit Ca(2+) entry via voltage-sensitive Ca(2+) channels and represents a novel mechanism of arterial relaxation.
- Published
- 2013
- Full Text
- View/download PDF
4. Emerging role of calcium-activated potassium channel in the regulation of cell viability following potassium ions challenge in HEK293 cells and pharmacological modulation.
- Author
-
Tricarico D, Mele A, Calzolaro S, Cannone G, Camerino GM, Dinardo MM, Latorre R, and Conte Camerino D
- Subjects
- Bendroflumethiazide pharmacology, Cell Line, Charybdotoxin pharmacology, Dichlorphenamide pharmacology, Ethoxzolamide pharmacology, Humans, Methazolamide pharmacology, Peptides pharmacology, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated antagonists & inhibitors, Tetraethylammonium pharmacology, Cell Survival drug effects, Potassium metabolism, Potassium Channels, Calcium-Activated metabolism
- Abstract
Emerging evidences suggest that Ca(2+)activated-K(+)-(BK) channel is involved in the regulation of cell viability. The changes of the cell viability observed under hyperkalemia (15 mEq/L) or hypokalemia (0.55 mEq/L) conditions were investigated in HEK293 cells expressing the hslo subunit (hslo-HEK293) in the presence or absence of BK channel modulators. The BK channel openers(10(-11)-10(-3)M) were: acetazolamide(ACTZ), Dichlorphenamide(DCP), methazolamide(MTZ), bendroflumethiazide(BFT), ethoxzolamide(ETX), hydrochlorthiazide(HCT), quercetin(QUERC), resveratrol(RESV) and NS1619; and the BK channel blockers(2 x 10(-7)M-5 x 10(-3)M) were: tetraethylammonium(TEA), iberiotoxin(IbTx) and charybdotoxin(ChTX). Experiments on cell viability and channel currents were performed using cell counting kit-8 and patch-clamp techniques, respectively. Hslo whole-cell current was potentiated by BK channel openers with different potency and efficacy in hslo-HEK293. The efficacy ranking of the openers at -60 mV(Vm) was BFT> ACTZ >DCP ≥RESV≥ ETX> NS1619> MTZ≥ QUERC; HCT was not effective. Cell viability after 24 h of incubation under hyperkalemia was enhanced by 82+6% and 33+7% in hslo-HEK293 cells and HEK293 cells, respectively. IbTx, ChTX and TEA enhanced cell viability in hslo-HEK293. BK openers prevented the enhancement of the cell viability induced by hyperkalemia or IbTx in hslo-HEK293 showing an efficacy which was comparable with that observed as BK openers. BK channel modulators failed to affect cell currents and viability under hyperkalemia conditions in the absence of hslo subunit. In contrast, under hypokalemia cell viability was reduced by -22+4% and -23+6% in hslo-HEK293 and HEK293 cells, respectively; the BK channel modulators failed to affect this parameter in these cells. In conclusion, BK channel regulates cell viability under hyperkalemia but not hypokalemia conditions. BFT and ACTZ were the most potent drugs either in activating the BK current and in preventing the cell proliferation induced by hyperkalemia. These findings may have relevance in disorders associated with abnormal K(+) ion homeostasis including periodic paralysis and myotonia.
- Published
- 2013
- Full Text
- View/download PDF
5. Cerebellar ataxia by enhanced Ca(V)2.1 currents is alleviated by Ca2+-dependent K+-channel activators in Cacna1a(S218L) mutant mice.
- Author
-
Gao Z, Todorov B, Barrett CF, van Dorp S, Ferrari MD, van den Maagdenberg AM, De Zeeuw CI, and Hoebeek FE
- Subjects
- Action Potentials drug effects, Action Potentials physiology, Animals, Behavior, Animal physiology, Benzimidazoles pharmacology, Calcium physiology, Calcium Channels, N-Type drug effects, Calcium Channels, N-Type genetics, Calcium Signaling drug effects, Cerebellar Ataxia psychology, Chlorzoxazone therapeutic use, Extracellular Space physiology, Female, Homeostasis physiology, Male, Mice, Muscle Relaxants, Central pharmacology, Mutation genetics, Mutation physiology, Patch-Clamp Techniques, Psychomotor Performance physiology, Purkinje Cells physiology, Calcium Channels, N-Type physiology, Calcium Channels, P-Type genetics, Calcium Channels, Q-Type genetics, Cerebellar Ataxia drug therapy, Cerebellar Ataxia genetics, Potassium Channels, Calcium-Activated agonists
- Abstract
Mutations in the CACNA1A gene are associated with neurological disorders, such as ataxia, hemiplegic migraine, and epilepsy. These mutations affect the pore-forming α(1A)-subunit of Ca(V)2.1 channels and thereby either decrease or increase neuronal Ca(2+) influx. A decreased Ca(V)2.1-mediated Ca(2+) influx has been shown to reduce the regularity of cerebellar Purkinje cell activity and to induce episodic cerebellar ataxia. However, little is known about how ataxia can be caused by CACNA1A mutations that increase the Ca(2+) influx, such as the S218L missense mutation. Here, we demonstrate that the S218L mutation causes a negative shift of voltage dependence of Ca(V)2.1 channels of mouse Purkinje cells and results in lowered thresholds for somatic action potentials and dendritic Ca(2+) spikes and in disrupted firing patterns. The hyperexcitability of Cacna1a(S218L) Purkinje cells was counteracted by application of the activators of Ca(2+)-dependent K(+) channels, 1-EBIO and chlorzoxazone (CHZ). Moreover, 1-EBIO also alleviated the irregularity of Purkinje cell firing both in vitro and in vivo, while CHZ improved the irregularity of Purkinje cell firing in vitro as well as the motor performance of Cacna1a(S218L) mutant mice. The current data suggest that abnormalities in Purkinje cell firing contributes to cerebellar ataxia induced by the S218L mutation and they advocate a general therapeutic approach in that targeting Ca(2+)-dependent K(+) channels may be beneficial for treating ataxia not only in patients suffering from a decreased Ca(2+) influx, but also in those suffering from an increased Ca(2+) influx in their Purkinje cells.
- Published
- 2012
- Full Text
- View/download PDF
6. Tuning electrical conduction along endothelial tubes of resistance arteries through Ca(2+)-activated K(+) channels.
- Author
-
Behringer EJ and Segal SS
- Subjects
- Acetylcholine pharmacology, Animals, Benzothiazoles pharmacology, Electric Conductivity, Epigastric Arteries drug effects, Indicators and Reagents pharmacokinetics, Male, Mice, Mice, Inbred C57BL, Microelectrodes, Nitric Oxide metabolism, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated antagonists & inhibitors, Propidium pharmacokinetics, Regional Blood Flow physiology, Signal Transduction physiology, Vascular Resistance drug effects, Vasodilator Agents pharmacology, Endothelium, Vascular physiology, Epigastric Arteries physiology, Gap Junctions physiology, Potassium Channels, Calcium-Activated physiology, Vascular Resistance physiology
- Abstract
Rationale: Electrical conduction through gap junction channels between endothelial cells of resistance vessels is integral to blood flow control. Small and intermediate-conductance Ca(2+)-activated K(+) channels (SK(Ca)/IK(Ca)) initiate electrical signals in endothelial cells, but it is unknown whether SK(Ca)/IK(Ca) activation alters signal transmission along the endothelium., Objective: We tested the hypothesis that SK(Ca)/IK(Ca) activity regulates electrical conduction along the endothelium of resistance vessels., Methods and Results: Freshly isolated endothelial cell tubes (60 μm wide; 1-3 mm long; cell length, ≈35 μm) from mouse skeletal muscle feed (superior epigastric) arteries were studied using dual intracellular microelectrodes. Current was injected (±0.1-3 nA) at site 1 while recording membrane potential (V(m)) at site 2 (separation distance=50-2000 μm). SK(Ca)/IK(Ca) activation (NS309, 1 μmol/L) reduced the change in V(m) along endothelial cell tubes by ≥50% and shortened the electrical length constant (λ) from 1380 to 850 μm (P<0.05) while intercellular dye transfer (propidium iodide) was maintained. Activating SK(Ca)/IK(Ca) with acetylcholine or SKA-31 also reduced electrical conduction. These effects of SK(Ca)/IK(Ca) activation persisted when hyperpolarization (>30 mV) was prevented with 60 mmol/L [K(+)](o). Conversely, blocking SK(Ca)/IK(Ca) (apamin+charybdotoxin) depolarized cells by ≈10 mV and enhanced electrical conduction (ie, changes in V(m)) by ≈30% (P<0.05)., Conclusions: These findings illustrate a novel role for SK(Ca)/IK(Ca) activity in tuning electrical conduction along the endothelium of resistance vessels by governing signal dissipation through changes in membrane resistance. Voltage-insensitive ion channels can thereby tune intercellular electrical signaling independent from gap junction channels.
- Published
- 2012
- Full Text
- View/download PDF
7. Hydrogen sulfide preconditioning or neutrophil depletion attenuates ischemia-reperfusion-induced mitochondrial dysfunction in rat small intestine.
- Author
-
Liu Y, Kalogeris T, Wang M, Zuidema MY, Wang Q, Dai H, Davis MJ, Hill MA, and Korthuis RJ
- Subjects
- Animals, Benzimidazoles administration & dosage, Cytochromes c metabolism, Hydrogen Sulfide metabolism, Intestinal Diseases etiology, Male, Membrane Potential, Mitochondrial drug effects, Mitochondria drug effects, Mitochondria enzymology, Mitochondrial Diseases etiology, Peroxidase analysis, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated antagonists & inhibitors, Rats, Rats, Sprague-Dawley, Tumor Necrosis Factor-alpha analysis, Intestinal Diseases prevention & control, Intestine, Small blood supply, Ischemic Preconditioning methods, Leukocyte Reduction Procedures, Mitochondrial Diseases prevention & control, Neutrophils, Reperfusion Injury complications, Sulfides administration & dosage
- Abstract
The objectives of this study were to determine whether neutrophil depletion with anti-neutrophil serum (ANS) or preconditioning with the hydrogen sulfide (H(2)S) donor NaHS (NaHS-PC) 24 h prior to ischemia-reperfusion (I/R) would prevent postischemic mitochondrial dysfunction in rat intestinal mucosa and, if so, whether calcium-activated, large conductance potassium (BK(Ca)) channels were involved in this protective effect. I/R was induced by 45-min occlusion of the superior mesenteric artery followed by 60-min reperfusion in rats preconditioned with NaHS (NaHS-PC) or a BK(Ca) channel activator (NS-1619-PC) 24 h earlier or treated with ANS. Mitochondrial function was assessed by measuring mitochondrial membrane potential, mitochondrial dehydrogenase function, and cytochrome c release. Mucosal myeloperoxidase (MPO) and TNF-α levels were also determined, as measures of postischemic inflammation. BK(Ca) expression in intestinal mucosa was detected by immunohistochemistry and Western blotting. I/R induced mitochondrial dysfunction and increased tissue MPO and TNF-α levels. Although mitochondrial dysfunction was attenuated by NaHS-PC or NS-1619-PC, the postischemic increases in mucosal MPO and TNF-α levels were not. The protective effect of NaHS-PC or NS-1619-PC on postischemic mitochondrial function was abolished by coincident treatment with BK(Ca) channel inhibitors. ANS prevented the I/R-induced increase in tissue MPO levels and reversed mitochondrial dysfunction. These data indicate that neutrophils play an essential role in I/R-induced mucosal mitochondrial dysfunction. In addition, NaHS-PC prevents postischemic mitochondrial dysfunction (but not inflammation) by a BK(Ca) channel-dependent mechanism.
- Published
- 2012
- Full Text
- View/download PDF
8. CyPPA, a positive modulator of small-conductance Ca(2+)-activated K(+) channels, inhibits phasic uterine contractions and delays preterm birth in mice.
- Author
-
Skarra DV, Cornwell T, Solodushko V, Brown A, and Taylor MS
- Subjects
- Abortifacient Agents pharmacology, Animals, Apamin pharmacology, Calcium metabolism, Calcium physiology, Dinoprost pharmacology, Female, Mice, Mice, Inbred C57BL, Mifepristone pharmacology, Myometrium drug effects, Oxytocin pharmacology, Pregnancy, Obstetric Labor, Premature drug therapy, Potassium Channels, Calcium-Activated agonists, Premature Birth prevention & control, Pyrazoles pharmacology, Pyrimidines pharmacology, Uterine Contraction drug effects
- Abstract
Organized uterine contractions, including those necessary for parturition, are dependent on calcium entry through voltage-gated calcium channels in myometrial smooth muscle cells. Recent evidence suggests that small-conductance Ca(2+)-activated potassium channels (K(Ca)2), specifically isoforms K(Ca)2.2 and 2.3, may control these contractions through negative feedback regulation of Ca(2+) entry. We tested whether selective pharmacologic activation of K(Ca)2.2/2.3 channels might depress uterine contractions, providing a new strategy for preterm labor intervention. Western blot analysis and immunofluorescence microscopy revealed expression of both K(Ca)2.2 and K(Ca)2.3 in the myometrium of nonpregnant (NP) and pregnant (gestation day 10 and 16; D10 and D16, respectively) mice. Spontaneous phasic contractions of isolated NP, D10, and D16 uterine strips were all suppressed by the K(Ca)2.2/2.3-selective activator CyPPA in a concentration-dependent manner. This effect was antagonized by the selective K(Ca)2 inhibitor apamin. Whereas CyPPA sensitivity was reduced in D10 and D16 versus NP strips (pIC(50) 5.33 ± 0.09, 4.64 ± 0.03, 4.72 ± 0.10, respectively), all contractions were abolished between 30 and 60 μM. Blunted contractions were associated with CyPPA depression of spontaneous Ca(2+) events in myometrial smooth muscle bundles. Augmentation of uterine contractions with oxytocin or prostaglandin F(2α) did not reduce CyPPA sensitivity or efficacy. Finally, in an RU486-induced preterm labor model, CyPPA significantly delayed time to delivery by 3.4 h and caused a 2.5-fold increase in pup retention. These data indicate that pharmacologic stimulation of myometrial K(Ca)2.2/2.3 channels effectively suppresses Ca(2+)-mediated uterine contractions and delays preterm birth in mice, supporting the potential utility of this approach in tocolytic therapies.
- Published
- 2011
- Full Text
- View/download PDF
9. Baicalin, a flavonoid from Scutellaria baicalensis Georgi, activates large-conductance Ca2+-activated K+ channels via cyclic nucleotide-dependent protein kinases in mesenteric artery.
- Author
-
Lin YL, Dai ZK, Lin RJ, Chu KS, Chen IJ, Wu JR, and Wu BN
- Subjects
- Animals, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular physiology, Female, Flavonoids isolation & purification, Mesenteric Arteries enzymology, Mesenteric Arteries metabolism, Patch-Clamp Techniques, Rats, Rats, Sprague-Dawley, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Cyclic GMP metabolism, Cyclic GMP-Dependent Protein Kinases metabolism, Flavonoids pharmacology, Mesenteric Arteries drug effects, Potassium Channels, Calcium-Activated agonists, Scutellaria baicalensis chemistry
- Abstract
Baicalin isolated from Scutellaria baicalensis is a traditional Chinese herbal medicine used for cardiovascular dysfunction. The ionic mechanism of the vasorelaxant effects of baicalin remains unclear. We investigated whether baicalin relaxes mesenteric arteries (MAs) via large-conductance Ca2+-activated K+ (BK(Ca)) channel activation and voltage-dependent Ca2+ channel (VDCC) inhibition. The contractility of MA was determined by dual wire myograph. BK(Ca) channels and VDCCs were measured using whole-cell recordings in single myocytes, enzymatically dispersed from rat MAs. Baicalin (10-100 microM) attenuated 80 mM KCl-contracted MA in a concentration-related manner. L-NAME (30 microM) and indomethacin (10 microM) little affected baicalin (100 microM)-induced vasorelaxations. Contractions induced by iberiotoxin (IbTX, 0.1 microM), Bay K8644 (0.1 microM) or PMA (10 microM) were abolished by baicalin 100 microM. In MA myocytes, baicalin (0.3-30 microM) enhanced BK(Ca) channel activity in a concentration-dependent manner. Increased BK(Ca) currents were abolished by IbTX (0.1 microM). Baicalin-mediated (30 microM) BK(Ca) current activation was significantly attenuated by an adenylate cyclase inhibitor (SQ 22536, 10 microM), a soluble guanylate cyclase inhibitor (ODQ, 10 microM), competitive antagonists of cAMP and cGMP (Rp-cAMP, 100 microM and Rp-cGMP, 100 microM), and cAMP- and cGMP-dependent protein kinase inhibitors (KT5720, 0.3 microM and KT5823, 0.3 microM). Perfusate with PMA (0.1 microM) abolished baicalin-enhanced BK(Ca) currents. Additionally, baicalin (0.3-30 microM) reduced the amplitude of VDCC currents in a concentration-dependent manner and abolished VDCC activator Bay K8644-enhanced (0.1 microM) currents. Baicalin produced MA relaxation by activating BK(Ca) and inhibiting VDCC channels by endothelium-independent mechanisms and by stimulating the cGMP/PKG and cAMP/PKA pathways., (2010 Elsevier GmbH. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
10. Endothelial Ca+-activated K+ channels in normal and impaired EDHF-dilator responses--relevance to cardiovascular pathologies and drug discovery.
- Author
-
Grgic I, Kaistha BP, Hoyer J, and Köhler R
- Subjects
- Animals, Blood Pressure drug effects, Blood Pressure physiology, Endothelium, Vascular drug effects, Humans, Models, Biological, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated antagonists & inhibitors, Potassium Channels, Calcium-Activated physiology, Signal Transduction drug effects, Signal Transduction physiology, Vasodilator Agents pharmacology, Biological Factors physiology, Cardiovascular Diseases drug therapy, Cardiovascular Diseases physiopathology, Drug Discovery, Endothelium, Vascular physiopathology, Potassium Channels, Calcium-Activated biosynthesis, Vasodilation drug effects, Vasodilation physiology, Vasodilator Agents therapeutic use
- Abstract
The arterial endothelium critically contributes to blood pressure control by releasing vasodilating autacoids such as nitric oxide, prostacyclin and a third factor or pathway termed 'endothelium-derived hyperpolarizing factor' (EDHF). The nature of EDHF and EDHF-signalling pathways is not fully understood yet. However, endothelial hyperpolarization mediated by the Ca(2+)-activated K(+) channels (K(Ca)) has been suggested to play a critical role in initializing EDHF-dilator responses in conduit and resistance-sized arteries of many species including humans. Endothelial K(Ca) currents are mediated by the two K(Ca) subtypes, intermediate-conductance K(Ca) (KCa3.1) (also known as, a.k.a. IK(Ca)) and small-conductance K(Ca) type 3 (KCa2.3) (a.k.a. SK(Ca)). In this review, we summarize current knowledge about endothelial KCa3.1 and KCa2.3 channels, their molecular and pharmacological properties and their specific roles in endothelial function and, particularly, in the EDHF-dilator response. In addition we focus on recent experimental evidences derived from KCa3.1- and/or KCa2.3-deficient mice that exhibit severe defects in EDHF signalling and elevated blood pressures, thus highlighting the importance of the KCa3.1/KCa2.3-EDHF-dilator system for blood pressure control. Moreover, we outline differential and overlapping roles of KCa3.1 and KCa2.3 for EDHF signalling as well as for nitric oxide synthesis and discuss recent evidence for a heterogeneous (sub) cellular distribution of KCa3.1 (at endothelial projections towards the smooth muscle) and KCa2.3 (at inter-endothelial borders and caveolae), which may explain their distinct roles for endothelial function. Finally, we summarize the interrelations of altered KCa3.1/KCa2.3 and EDHF system impairments with cardiovascular disease states such as hypertension, diabetes, dyslipidemia and atherosclerosis and discuss the therapeutic potential of KCa3.1/KCa2.3 openers as novel types of blood pressure-lowering drugs.
- Published
- 2009
- Full Text
- View/download PDF
11. Loss of Ca-mediated ion transport during colitis correlates with reduced ion transport responses to a Ca-activated K channel opener.
- Author
-
Hirota CL and McKay DM
- Subjects
- Animals, Bethanechol pharmacology, Biological Transport, Active, Chlorides metabolism, Colitis chemically induced, Colon drug effects, Colon metabolism, Dextran Sulfate, Enzyme Activation, Intestinal Mucosa drug effects, Intestinal Mucosa metabolism, Ion Channel Gating, Ion Transport, Male, Mice, Mice, Inbred BALB C, Potassium Channels, Calcium-Activated agonists, Protein Kinase C metabolism, Protein Subunits biosynthesis, Protein Subunits genetics, RNA, Messenger biosynthesis, Receptor, Muscarinic M3 agonists, Receptor, Muscarinic M3 biosynthesis, Receptor, Muscarinic M3 genetics, Sodium-Potassium-Exchanging ATPase biosynthesis, Sodium-Potassium-Exchanging ATPase genetics, Tetradecanoylphorbol Acetate pharmacology, Calcium physiology, Colitis metabolism, Potassium Channels, Calcium-Activated physiology
- Abstract
Background and Purpose: Epithelial surface hydration is critical for proper gut function. However, colonic tissues from individuals with inflammatory bowel disease or animals with colitis are hyporesponsive to Cl(-) secretagogues. The Cl(-) secretory responses to the muscarinic receptor agonist bethanechol are virtually absent in colons of mice with dextran sodium sulphate (DSS)-induced colitis. Our aim was to define the mechanism underlying this cholinergic hyporesponsiveness., Experimental Approach: Colitis was induced by 4% DSS water, given orally. Epithelial ion transport was measured in Ussing chambers. Colonic crypts were isolated and processed for mRNA expression via RT-PCR and protein expression via immunoblotting and immunolocalization., Key Results: Expression of muscarinic M(3) receptors in colonic epithelium was not decreased during colitis. Short-circuit current (I(SC)) responses to other Ca(2+)-dependent secretagogues (histamine, thapsigargin, cyclopiazonic acid and calcium ionophore) were either absent or severely attenuated in colonic tissue from DSS-treated mice. mRNA levels of several ion transport molecules (a Ca(2+)-regulated Cl(-) channel, the intermediate-conductance Ca(2+)-activated K(+) channel, the cystic fibrosis transmembrane conductance regulator, the Na(+)/K(+)-ATPase pump or the Na(+)/K(+)/2Cl(-) co-transporter) were not reduced in colonic crypts from DSS-treated mice. However, protein expression of Na(+)/K(+)-ATPase alpha1 subunits was decreased twofold during colitis. Activation of Ca(2+)-activated K(+) channels increased I(SC) significantly less in DSS colons compared with control, as did the protein kinase C activator, phorbol 12-myristate 13-acetate., Conclusions and Implications: Decreased Na(+)/K(+)-ATPase expression probably contributes to overall epithelial hyporesponsiveness during colitis, while dysfunctional K(+) channels may account, at least partially, for lack of epithelial secretory responses to Ca(2+)-mediated secretagogues.
- Published
- 2009
- Full Text
- View/download PDF
12. A novel opener of large-conductance Ca2+ -activated K+ (BK) channel reduces ischemic injury in rat cardiac myocytes by activating mitochondrial K(Ca) channel.
- Author
-
Sakamoto K, Ohya S, Muraki K, and Imaizumi Y
- Subjects
- Animals, Calcium metabolism, Cell Survival drug effects, In Vitro Techniques, Mitochondria, Heart drug effects, Mitochondrial Membranes drug effects, Mitochondrial Membranes metabolism, Myocytes, Cardiac drug effects, Oxidation-Reduction, Patch-Clamp Techniques, Rats, Abietanes pharmacology, Mitochondria, Heart pathology, Myocardial Ischemia pathology, Myocardial Ischemia prevention & control, Myocytes, Cardiac pathology, Potassium Channels, Calcium-Activated agonists
- Abstract
It has been suggested that a new type of large-conductance Ca(2+)-activated K(+) (BK) channel is distributed in the inner mitochondrial membrane (mitoK(Ca) channel) and that its opening may attenuate ischemic cardiac injury. We examined effects of 12,14-dichlorodehydroabietic acid (diCl-DHAA), a novel BK-channel opener, on rat cardiac myocytes and mitochondria. Application of diCl-DHAA concentration-dependently reduced Ca(2+) overload in isolated mitochondria, activated mitoK(Ca) channels in inside-out patches of mitochondrial membrane, facilitated flavoprotein-oxidization in myocytes, and increased cellular viability under simulated ischemia. In conclusion, diCl-DHAA directly opens mitoK(Ca) channels, prevents Ca(2+) influx into matrix, and reduces ischemic injury in cardiac myocytes.
- Published
- 2008
- Full Text
- View/download PDF
13. Calcium-activated potassium channels mediated blood-brain tumor barrier opening in a rat metastatic brain tumor model.
- Author
-
Hu J, Yuan X, Ko MK, Yin D, Sacapano MR, Wang X, Konda BM, Espinoza A, Prosolovich K, Ong JM, Irvin D, and Black KL
- Subjects
- Animals, Brain Neoplasms metabolism, Coculture Techniques, Disease Models, Animal, Endothelial Cells cytology, Endothelial Cells metabolism, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms pathology, Permeability drug effects, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated antagonists & inhibitors, Potassium Channels, Calcium-Activated genetics, Potentiometry, Protein Transport, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Receptor, Bradykinin B2 metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Blood-Brain Barrier pathology, Brain Neoplasms pathology, Brain Neoplasms secondary, Potassium Channels, Calcium-Activated metabolism
- Abstract
Background: The blood-brain tumor barrier (BTB) impedes the delivery of therapeutic agents to brain tumors. While adequate delivery of drugs occurs in systemic tumors, the BTB limits delivery of anti-tumor agents into brain metastases., Results: In this study, we examined the function and regulation of calcium-activated potassium (KCa) channels in a rat metastatic brain tumor model. We showed that intravenous infusion of NS1619, a KCa channel agonist, and bradykinin selectively enhanced BTB permeability in brain tumors, but not in normal brain. Iberiotoxin, a KCa channel antagonist, significantly attenuated NS1619-induced BTB permeability increase. We found KCa channels and bradykinin type 2 receptors (B2R) expressed in cultured human metastatic brain tumor cells (CRL-5904, non-small cell lung cancer, metastasized to brain), human brain microvessel endothelial cells (HBMEC) and human lung cancer brain metastasis tissues. Potentiometric assays demonstrated the activity of KCa channels in metastatic brain tumor cells and HBMEC. Furthermore, we detected higher expression of KCa channels in the metastatic brain tumor tissue and tumor capillary endothelia as compared to normal brain tissue. Co-culture of metastatic brain tumor cells and brain microvessel endothelial cells showed an upregulation of KCa channels, which may contribute to the overexpression of KCa channels in tumor microvessels and selectivity of BTB opening., Conclusion: These findings suggest that KCa channels in metastatic brain tumors may serve as an effective target for biochemical modulation of BTB permeability to enhance selective delivery of chemotherapeutic drugs to metastatic brain tumors.
- Published
- 2007
- Full Text
- View/download PDF
14. (+/-)-Naringenin as large conductance Ca(2+)-activated K+ (BKCa) channel opener in vascular smooth muscle cells.
- Author
-
Saponara S, Testai L, Iozzi D, Martinotti E, Martelli A, Chericoni S, Sgaragli G, Fusi F, and Calderone V
- Subjects
- Animals, Arteries cytology, Arteries drug effects, Arteries metabolism, Calcium metabolism, Cell Separation, Chelating Agents pharmacology, Egtazic Acid analogs & derivatives, Egtazic Acid pharmacology, Electrophysiology, In Vitro Techniques, Kinetics, Male, Membrane Potentials drug effects, Muscle Relaxation drug effects, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular drug effects, Myocytes, Smooth Muscle drug effects, Patch-Clamp Techniques, Rats, Rats, Sprague-Dawley, Vasodilation drug effects, Flavanones pharmacology, Muscle, Smooth, Vascular metabolism, Potassium Channels, Calcium-Activated agonists
- Abstract
Unlabelled: BACKGROUND AND PURPOSE. The aim of this study was to investigate, in vascular smooth muscle cells, the mechanical and electrophysiological effects of (+/-)-naringenin., Experimental Approach: Aorta ring preparations and single tail artery myocytes were employed for functional and patch-clamp experiments, respectively., Key Results: (+/-)-Naringenin induced concentration-dependent relaxation in endothelium-denuded rat aortic rings pre-contracted with either 20 mM KCl or noradrenaline (pIC(50) values of 4.74 and 4.68, respectively). Tetraethylammonium, iberiotoxin, 4-aminopyridine and 60 mM KCl antagonised (+/-)-naringenin-induced vasorelaxation, while glibenclamide did not produce any significant antagonism. Naringin [(+/-)-naringenin 7-beta-neohesperidoside] caused a concentration-dependent relaxation of rings pre-contracted with 20 mM KCl, although its potency and efficacy were significantly lower than those of (+/-)-naringenin. In rat tail artery myocytes, (+/-)-naringenin increased large conductance Ca(2+)-activated K(+) (BK(Ca)) currents in a concentration-dependent manner; this stimulation was iberiotoxin-sensitive and fully reversible upon drug wash-out. (+/-)-Naringenin accelerated the activation kinetics of BK(Ca) current, shifted, by 22 mV, the voltage dependence of the activation curve to more negative potentials, and decreased the slope of activation. (+/-)-Naringenin-induced stimulation of BK(Ca) current was insensitive either to changes in the intracellular Ca(2+) concentration or to the presence, in the pipette solution, of the fast Ca(2+) chelator BAPTA. However, such stimulation was diminished when the K(+) gradient across the membrane was reduced., Conclusions and Implications: The vasorelaxant effect of the naturally-occurring flavonoid (+/-)-naringenin on endothelium-denuded vessels was due to the activation of BK(Ca) channels in myocytes.
- Published
- 2006
- Full Text
- View/download PDF
15. Regulatory effect of sulphatides on BKCa channels.
- Author
-
Chi S and Qi Z
- Subjects
- Algorithms, Animals, CHO Cells, Cricetinae, DNA Transposable Elements, Dose-Response Relationship, Drug, Electrophysiology, Ion Channel Gating physiology, Membrane Potentials physiology, Patch-Clamp Techniques, Potassium Channels, Calcium-Activated genetics, Potassium Channels, Calcium-Activated metabolism, Potassium Channels, Calcium-Activated agonists, Sulfoglycosphingolipids pharmacology
- Abstract
Background and Purpose: Sulphatides are sulphated glycosphingolipids expressed on the surface of many cell types, particularly neurones. Changes in sulphatide species or content have been associated with epilepsy and Alzheimer's disease. As the large conductance, calcium sensitive K(+) channel (BK(Ca)) are modulated by membrane lipids, the aim of the study was to explore possible effects of sulphatides on BK(Ca) channels., Experimental Approach: Using patch-clamp techniques, we studied effects of exogenous sulphatides on BK(Ca) channels expressed in Chinese hamster ovary cells., Key Results: Sulphatides reversibly increased the whole-cell current and the single channel open probability of BK(Ca) channels dose-dependently. The EC(50) value on the channel at +10 mV was 1.6 microM and the Hill coefficient was 2.5. In inside-out patches, sulphatides increased the single channel open probability from both intra- and extra-cellular faces of the membrane, but more effectively with external application. Furthermore, activation of the channels by sulphatides was independent of intracellular Ca(2+) concentration. Sulphatides also shifted the activation curve of the channels to less positive membrane potentials. Mutant BK(Ca) channels lacking a 59 aminoacid region important for amphipath activation (STREX) were less activated by the sulphatides., Conclusions and Implications: Sulphatides are novel activators of BK(Ca) channels, independent of intracellular Ca(2+) or other signalling molecules but partly dependent on the STREX sequence of the channel protein. As changes of sulphatide content are associated with neuronal dysfunction, as in epilepsy and Alzheimer's disease, our results imply that these effects of sulphatides may play important pathophysiological roles in regulation of BK(Ca) channels.
- Published
- 2006
- Full Text
- View/download PDF
16. KCa+ channels contribute to exercise-induced coronary vasodilation in swine.
- Author
-
Merkus D, Sorop O, Houweling B, Hoogteijling BA, and Duncker DJ
- Subjects
- Adenosine Triphosphate metabolism, Animals, Benzopyrans pharmacology, Blood Gas Analysis, Dihydropyridines pharmacology, Dose-Response Relationship, Drug, Exercise Test, Female, Glyburide pharmacology, Hypoglycemic Agents pharmacology, Male, Oxygen Consumption drug effects, Oxygen Consumption physiology, Physical Conditioning, Animal, Potassium Channel Blockers pharmacology, Potassium Channels drug effects, Potassium Channels physiology, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated drug effects, Sus scrofa, Tetraethylammonium pharmacology, Time Factors, Coronary Vessels physiology, Physical Exertion physiology, Potassium Channels, Calcium-Activated physiology, Vasodilation physiology
- Abstract
Coronary blood flow is controlled via several vasoactive mediators that exert their effect on coronary resistance vessel tone through activation of K(+) channels in vascular smooth muscle. Because Ca(2+)-activated K(+) (K(Ca)(+)) channels are the predominant K(+) channels in the coronary vasculature, we hypothesized that K(Ca)(+) channel activation contributes to exercise-induced coronary vasodilation. In view of previous observations that ATP-sensitive K(+) (K(ATP)(+)) channels contribute, in particular, to resting coronary resistance vessel tone, we additionally investigated the integrated control of coronary tone by K(Ca)(+) and K(ATP)(+) channels. For this purpose, the effect of K(Ca)(+) blockade with tetraethylammonium (TEA, 20 mg/kg iv) on coronary vasomotor tone was assessed in the absence and presence of K(ATP)(+) channel blockade with glibenclamide (3 mg/kg iv) in chronically instrumented swine at rest and during treadmill exercise. During exercise, myocardial O(2) delivery increased commensurately with the increase in myocardial O(2) consumption, so that myocardial O(2) extraction and coronary venous Po(2) (Pcv(O(2))) were maintained constant. TEA (in a dose that had no effect on K(ATP)(+) channels) had a small effect on the myocardial O(2) balance at rest and blunted the exercise-induced increase in myocardial O(2) delivery, resulting in a progressive decrease of Pcv(O(2)) with increasing exercise intensity. Conversely, at rest glibenclamide caused a marked decrease in Pcv(O(2)) that waned at higher exercise levels. Combined K(Ca)(+) and K(ATP)(+) channel blockade resulted in coronary vasoconstriction at rest that was similar to that caused by glibenclamide alone and that was maintained during exercise, suggesting that K(Ca)(+) and K(ATP)(+) channels act in a linear additive fashion. In conclusion, K(Ca)(+) channel activation contributes to the metabolic coronary vasodilation that occurs during exercise. Furthermore, in swine K(Ca)(+) and K(ATP)(+) channels contribute to coronary resistance vessel control in a linear additive fashion.
- Published
- 2006
- Full Text
- View/download PDF
17. Effects of NS1608, a BK(Ca) channel agonist, on the contractility of guinea-pig urinary bladder in vitro.
- Author
-
Mora TC and Suarez-Kurtz G
- Subjects
- 4-Aminopyridine pharmacology, Animals, Apamin pharmacology, Guinea Pigs, In Vitro Techniques, Muscle Contraction physiology, Muscle, Smooth physiology, Peptides pharmacology, Potassium Channel Blockers pharmacology, Potassium Channels, Calcium-Activated antagonists & inhibitors, Ryanodine pharmacology, Urinary Bladder drug effects, Urinary Bladder physiology, Muscle Contraction drug effects, Muscle, Smooth drug effects, Phenylurea Compounds pharmacology, Potassium Channels, Calcium-Activated agonists
- Abstract
1. The functional effects of NS1608 ((N-(3-(trifluoromethyl)phenyl)-N'-(2-hydroxy-5-chlorophenyl)urea), an opener of the large conductance, Ca2+-activated K+ (BK(Ca)) channel, on the contractility of guinea-pig urinary bladder muscle are described. 2. NS1608 (0.3-30 microM) had no significant effect on the integrated myogenic activity (tension integral) or the electrically evoked twitches of detrusor muscle strips. Possible mechanisms for the discrepancy between the lack of functional effects of NS1608 per se on detrusor contractility and this drug's agonistic effect on BK(Ca) currents in isolated bladder myocytes are discussed. 3. 4-Aminopyridine (1 mM), a blocker of voltage-gated K+ (K(V)) channels, increased the tension integral 2.7-fold, on average. NS1608 (30 microM) counteracted this effect. 4. Apamin (100 nM), a selective blocker of the small conductance, Ca2+-activated K+ (SK(Ca)) channel, increased the tension integral 1.7-fold, on average. This effect was reversed by NS1608 (30 microM). 5. Ryanodine (10 microM), a modulator of the sarcoplasmic reticulum (SR) Ca2+-release channel, increased the tension integral 1.9-fold, on average. This effect was reversed by NS1608 (30 microM). 6. Iberiotoxin (IbTX, 50 nM), a selective blocker of the BK(Ca) channel, caused additional increases in the tension integral of detrusor strips pretreated with apamin or ryanodine and prevented the inhibitory effects of NS1608 (30 microM) in detrusor contractility. 7. The present study shows that blockade of repolarizing currents carried by, respectively apamin- and 4-aminopyridine-sensitive K+ channels unmasks an activation of BK(Ca) in guinea-pig urinary bladder smooth muscle strips.
- Published
- 2005
- Full Text
- View/download PDF
18. Voltage dependence of ATP-dependent K+ current in rat cardiac myocytes is affected by IK1 and IK(ACh).
- Author
-
Wellner-Kienitz MC, Bender K, Rinne A, and Pott L
- Subjects
- ATP-Binding Cassette Transporters agonists, ATP-Binding Cassette Transporters physiology, Animals, CHO Cells, Cells, Cultured, Cricetinae, Intermediate-Conductance Calcium-Activated Potassium Channels, Membrane Potentials drug effects, Membrane Potentials physiology, Myocytes, Cardiac drug effects, Pinacidil pharmacology, Potassium Channels agonists, Potassium Channels physiology, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated physiology, Potassium Channels, Inwardly Rectifying agonists, Rats, Receptors, Drug agonists, Receptors, Drug physiology, Sulfonylurea Receptors, Acetylcholine pharmacology, Adenosine Triphosphate physiology, Myocytes, Cardiac physiology, Potassium Channels, Inwardly Rectifying physiology
- Abstract
In this study we have investigated the voltage dependence of ATP-dependent K+ current (I(K(ATP))) in atrial and ventricular myocytes from hearts of adult rats and in CHO cells expressing Kir6.2 and SUR2A. The current-voltage relation of 2,4-dinitrophenole (DNP) -induced I(K(ATP)) in atrial myocytes and expressed current in CHO cells was linear in a voltage range between 0 and -100 mV. In ventricular myocytes, the background current-voltage relation of which is dominated by a large constitutive inward rectifier (I(K1)), the slope conductance of I(K(ATP)) was reduced at membrane potentials negative to E(K) (around -50 mV), resulting in an outwardly rectifying I-V relation. Overexpression of Kir2.1 by adenoviral gene transfer, a subunit contributing to I(K1) channels, in atrial myocytes resulted in a large I(K1)-like background current. The I-V relation of I(K(ATP)) in these cells showed a reduced slope conductance negative to E(K) similar to ventricular myocytes. In atrial myocytes with an increased background inward-rectifier current through Kir3.1/Kir3.4 channels (I(K(ACh))), irreversibly activated by intracellular loading with GTP-gamma-S, the I-V relation of I(K(ATP)) showed a reduced slope negative to E(K), as in ventricular myocytes and atrial myocytes overexpressing Kir2.1. It is concluded that the voltage dependencies of membrane currents are not only dependent on the molecular composition of the charge-carrying channel complexes but can be affected by the activity of other ion channel species. We suggest that the interference between inward I(K(ATP)) and other inward rectifier currents in cardiac myocytes reflects steady-state changes in K+ driving force due to inward K+ current.
- Published
- 2004
- Full Text
- View/download PDF
19. Stimulatory actions of caffeic acid phenethyl ester, a known inhibitor of NF-kappaB activation, on Ca2+-activated K+ current in pituitary GH3 cells.
- Author
-
Lin MW, Yang SR, Huang MH, and Wu SN
- Subjects
- Amidines pharmacology, Animals, Caffeic Acids chemistry, Calcium chemistry, Calcium metabolism, Calcium Channel Blockers pharmacology, Calcium Channels, L-Type drug effects, Calcium Channels, L-Type metabolism, Cell Line, Tumor, Electric Conductivity, Ion Channel Gating drug effects, Ion Channel Gating physiology, Masoprocol chemistry, Masoprocol pharmacology, Membrane Potentials drug effects, Membrane Potentials physiology, Patch-Clamp Techniques methods, Phenylethyl Alcohol chemistry, Pituitary Gland cytology, Potassium Channels, Calcium-Activated physiology, Rats, Riluzole pharmacology, tert-Butylhydroperoxide pharmacology, Caffeic Acids pharmacology, NF-kappa B antagonists & inhibitors, Phenylethyl Alcohol analogs & derivatives, Phenylethyl Alcohol pharmacology, Pituitary Gland drug effects, Pituitary Gland physiology, Potassium Channels, Calcium-Activated agonists
- Abstract
Caffeic acid phenethyl ester (CAPE), a phenolic antioxidant derived from the propolis of honeybee hives, is known to be an inhibitor of activation of nuclear transcript factor NF-kappaB. Its effects on ion currents have been investigated in pituitary GH(3) cells. This compound increased Ca(2+)-activated K(+) current (I(K(Ca))) in a concentration-dependent manner with an EC(50) value of 14 +/- 2 microm. However, the magnitude of CAPE-induced stimulation of I(K(Ca)) was attenuated in GH(3) cells preincubated with 2,2'-azo-bis-(2-amidinopropane) hydrochloride (100 microm) or t-butyl hydroperoxide (1 mm). CAPE (50 microm) slightly suppressed voltage-dependent L-type Ca(2+) current. In inside-out configuration, CAPE (20 microm) applied to the intracellular face of the detached patch enhanced the activity of large conductance Ca(2+)-activated K(+) (BK(Ca)) channels with no modification in single-channel conductance. After BK(Ca) channel activity was increased by CAPE (20 microm), subsequent application of nordihydroguaiaretic acid (20 microm) did not further increase the channel activity. CAPE-stimulated channel activity was dependent on membrane potential. CAPE could also increase Ca(2+) sensitivity of BK(Ca) channels in these cells. Its increase in the open probability could primarily involve a decrease in the mean closed time. In current-clamp conditions, CAPE hyperpolarized the membrane potential and reduced the firing of action potentials. The stimulatory effects on these channels may partly contribute to the underlying mechanisms through which this compound influences the functional activities of neurons or neuroendocrine cells. Caution has to be used in attributing its response in the activation of NF-kappaB.
- Published
- 2004
- Full Text
- View/download PDF
20. Ligand-dependent activation of Slo family channels is defined by interchangeable cytosolic domains.
- Author
-
Xia XM, Zhang X, and Lingle CJ
- Subjects
- Animals, Calcium metabolism, Hydrogen-Ion Concentration, Ion Channel Gating, Large-Conductance Calcium-Activated Potassium Channel alpha Subunits, Large-Conductance Calcium-Activated Potassium Channels, Mice, Oocytes metabolism, Patch-Clamp Techniques, Potassium Channels, Calcium-Activated physiology, Protein Structure, Tertiary, Xenopus, Cytosol metabolism, Potassium Channels, Calcium-Activated agonists
- Abstract
Large-conductance Ca2+- and voltage-regulated K+ channels (Slo1 BK-type) are controlled by two physiological stimuli, membrane voltage and cytosolic Ca2+. Regulation by voltage is similar to that in voltage-dependent K+ channels, arising from positively charged amino acids primarily within the S4 transmembrane helices. The basis for regulation by Ca2+ remains controversial. One viewpoint suggests that the extensive cytosolic C terminus contains the Ca2+ regulatory machinery, whereas another suggests that the pore-forming module contains the Ca2+-sensing elements. To address this issue, we take advantage of another Slo family member, the pH-regulated homolog Slo3. We reason that if the ligand-sensing apparatus is uniquely associated with a particular domain (either the pore or the cytosolic domain), exchange of those domains between Slo1 and Slo3 should result in exchange of ligand dependence in association with the key domain. The results show that the Slo3 cytosolic module confers pH-dependent regulation on the Slo1 pore module, whereas the Slo1 cytosolic module confers Ca2+-dependent regulation on the Slo3 pore module. Thus, ligand-specific regulation is defined by interchangeable cytosolic regulatory modules.
- Published
- 2004
- Full Text
- View/download PDF
21. Localized IP3-evoked Ca2+ release activates a K+ current in primary vagal sensory neurons.
- Author
-
Hoesch RE, Weinreich D, and Kao JP
- Subjects
- Animals, Calcium Signaling drug effects, Calibration, Chelating Agents pharmacology, Egtazic Acid pharmacology, Electrophysiology, Extracellular Space drug effects, Extracellular Space physiology, In Vitro Techniques, Indicators and Reagents, Kinetics, Male, Membrane Potentials physiology, Neurons, Afferent drug effects, Patch-Clamp Techniques, Photolysis, Rabbits, Vagus Nerve cytology, Vagus Nerve drug effects, Calcium metabolism, Egtazic Acid analogs & derivatives, Inositol 1,4,5-Trisphosphate pharmacology, Neurons, Afferent physiology, Potassium Channels, Calcium-Activated agonists, Vagus Nerve physiology
- Abstract
Electrophysiological and microfluorimetric techniques were used to determine whether intracellular photorelease of caged IP(3), and the consequent release of Ca(2+), could trigger a Ca(2+)-activated K(+) current (I(IP3)). Photorelease of caged IP(3) evoked an I(IP3) that averaged 2.36 +/- 0.35 (SE) pA/pF in 24 of 28 rabbit primary vagal sensory neurons (nodose ganglion neurons, NGNs) voltage-clamped at -50 mV. I(IP3) was abolished by intracellular BAPTA (2 mM), a Ca(2+) chelator. Changing the K(+) equilibrium potential by increasing extracellular K(+) ion concentration caused a predicted Nernstian shift in the reversal potential of I(IP3). These results indicated that I(IP3) was a Ca(2+)-dependent K(+) current. I(IP3) was unaffected by three common antagonists of Ca(2+)-activated K(+) currents: bath-applied iberiotoxin (50 nM) or apamin (100 nM), and intracellular 8-Br-cAMP (100 microM) included in the patch pipette. We have previously demonstrated that both IP(3)-evoked Ca(2+) release and Ca(2+)-induced Ca(2+) release (CICR) are co-expressed in NGNs and that CICR can trigger a Ca(2+)-activated K(+) current. In the present study, using caffeine, a CICR agonist, to selectively attenuate intracellular Ca(2+) stores, we showed that IP(3)-evoked Ca(2+) release occurs independently of CICR, but interestingly, that a component of I(IP3) requires CICR. These data suggest that IP(3)-evoked Ca(2+) release activates a K(+) current that is pharmacologically distinct from other Ca(2+)-activated K(+) currents in NGNs. We describe several models that explain our results based on Ca(2+) signaling microdomains in NGNs.
- Published
- 2004
- Full Text
- View/download PDF
22. Theophylline attenuates Ca2+ sensitivity and modulates BK channels in porcine tracheal smooth muscle.
- Author
-
Ise S, Nishimura J, Hirano K, Hara N, and Kanaide H
- Subjects
- Animals, Calcium pharmacology, Dose-Response Relationship, Drug, In Vitro Techniques, Large-Conductance Calcium-Activated Potassium Channels, Muscle Contraction drug effects, Muscle Contraction physiology, Muscle, Smooth metabolism, Potassium Channels, Calcium-Activated agonists, Swine, Trachea metabolism, Calcium metabolism, Muscle, Smooth drug effects, Potassium Channels, Calcium-Activated metabolism, Theophylline pharmacology, Trachea drug effects
- Abstract
Theophylline, a nonselective phosphodiesterase inhibitor, has long been regarded as a major bronchodilator in the treatment of human asthma. Using front-surface fluorometry with fura-2 and alpha-toxin permeabilization, the effects of theophylline on intracellular Ca2+ concentration ([Ca2+]i), tension development and Ca2+ sensitivity of the contractile apparatus were investigated in porcine tracheal smooth muscle strips. Application of theophylline induced a relaxation without a significant decrease in [Ca2+]i when strips were precontracted by 40 mm K+ depolarization, while theophylline significantly decreased both [Ca2+]i and tension induced by carbachol. The effects of theophylline on the increases in [Ca2+]i and tension induced by carbachol were significantly inhibited by iberiotoxin, an inhibitor of large-conductance Ca2+-activated K+ channels. In the absence of extracellular Ca2+, theophylline significantly attenuated carbachol-induced transient increases in tension development, while it did not affect carbachol-induced transient increase in [Ca2+]i. The [Ca2+]i-force relationship, which was determined by cumulative applications of extracellular Ca2+ (0-5 mm) during 40 mm K+ depolarization, was significantly shifted to the right by theophylline. In alpha-toxin permeabilized strips, theophylline significantly increased the EC50 value of [Ca2+]i for contraction and enhanced the effect of cAMP, but not of cGMP. These results indicate that theophylline induces relaxation of the porcine tracheal smooth muscle through an activation of BK channels, and a resultant decrease in [Ca2+]i and an attenuation of Ca2+ sensitivity, presumably through the action of cAMP.
- Published
- 2003
- Full Text
- View/download PDF
23. EDHF and endothelial potassiun channels: IKCa and SKCa.
- Author
-
Félétou M, Vanhoutte PM, Weston AH, and Edwards G
- Subjects
- Endothelial Cells metabolism, Intermediate-Conductance Calcium-Activated Potassium Channels, Potassium Channels agonists, Potassium Channels metabolism, Potassium Channels, Calcium-Activated agonists, Small-Conductance Calcium-Activated Potassium Channels, Biological Factors pharmacology, Endothelial Cells drug effects, Potassium Channels, Calcium-Activated metabolism
- Published
- 2003
- Full Text
- View/download PDF
24. Differential role of PTK and ERK MAPK in superoxide impairment of K(ATP) and K(Ca) channel cerebrovasodilation.
- Author
-
Ross J and Armstead WM
- Subjects
- Adenosine Triphosphate metabolism, Animals, Animals, Newborn, Benzimidazoles pharmacology, Calcitonin Gene-Related Peptide pharmacology, Cromakalim pharmacology, Enzyme Inhibitors pharmacology, Female, Genistein pharmacology, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System physiology, Male, Pia Mater blood supply, Potassium Channels, Calcium-Activated agonists, Superoxides metabolism, Swine, Tyrphostins pharmacology, Vasodilator Agents pharmacology, Cerebrovascular Circulation physiology, Mitogen-Activated Protein Kinases metabolism, Potassium Channels, Calcium-Activated metabolism, Protein-Tyrosine Kinases metabolism, Vasodilation physiology
- Abstract
Previously, superoxide (O2 -) has been observed to impair pial artery dilation (PAD) to activators of the ATP-sensitive (KATP) and calcium-sensitive (KCa) K+ channels. This study tested the hypothesis that activation of protein tyrosine kinase (PTK) and the ERK isoform of MAPK by O2 - contribute to impairment of KATP and KCa channel PAD. Exposure of the cerebral cortex to a xanthine oxidase O2 --generating system (OX) blunted PAD to cromakalim, a KATP agonist, but preadministration of genistein, a PTK antagonist, or U-0126, an ERK MAPK inhibitor, almost completely prevented such impairment (11 +/- 1 and 22 +/- 1 vs. 3 +/- 1 and 7 +/- 1 vs. 10 +/- 1 and 16 +/- 2% for cromakalim with 10-8 and 10-6 M PAD during control, OX, and OX + genistein conditions). In contrast, neither genistein nor U-0126 robustly protected PAD to NS-1619, a KCa agonist, after OX exposure (11 +/- 1 and 18 +/- 2 vs. 1 +/- 1 and 2 +/- 1 vs. 4 +/- 1 and 6 +/- 1% for 10-8 and 10-6 M NS-1619 during control, OX, and OX + genistein conditions). These data show that PTK and ERK MAPK activation contribute to O2 --induced KATP and KCa channel PAD impairment and suggest a differential greater role for PTK and ERK MAPK in KATP vs. KCa channel PAD impairment.
- Published
- 2003
- Full Text
- View/download PDF
25. Different mechanisms underlying the stimulation of K(Ca) channels by nitric oxide and carbon monoxide.
- Author
-
Wu L, Cao K, Lu Y, and Wang R
- Subjects
- Animals, COS Cells, Calcium metabolism, Cytoplasm metabolism, Dose-Response Relationship, Drug, Male, Nitric Oxide Donors pharmacology, Nitroprusside pharmacology, Oligonucleotides, Antisense pharmacology, Patch-Clamp Techniques, Potassium metabolism, Potassium Channels, Calcium-Activated agonists, Protein Binding, Rats, Rats, Sprague-Dawley, Saponins pharmacology, Time Factors, Transfection, Triterpenes pharmacology, Carbon Monoxide pharmacology, Nitric Oxide pharmacology, Oleanolic Acid analogs & derivatives, Potassium Channels, Calcium-Activated metabolism
- Abstract
The molecular mechanisms underlying the effects of nitric oxide (NO) and carbon monoxide (CO), individually and collectively, on large-conductance calcium-activated K(+) (K(Ca)) channels were investigated in rat vascular smooth muscle cells (SMCs). Both NO and CO increased the activity of native K(Ca) channels. Dehydrosoyasaponin-I, a specific agonist for beta subunit of K(Ca) channels, increased the open probability of native K(Ca) channels only when it was delivered to the cytoplasmic surface of membrane. CO, but not NO, further increased the activity of native K(Ca) channels that had been maximally stimulated by dehydrosoyasaponin-I. After treatment of SMCs with anti-K(Ca),beta subunit antisense oligodeoxynucleotides, the stimulatory effect of NO, but not of CO, on K(Ca) channels was nullified. CO, but not NO, enhanced the K(Ca) current densities of heterologously expressed cloned K(Ca),alpha subunit, showing that the presence of K(Ca),beta subunit is not a necessity for the effect of CO but essential for that of NO. Finally, pretreatment of SMCs with NO abolished the effects of subsequently applied CO or diethyl pyrocarbonate on K(Ca) channels. In summary, the stimulatory effects of CO and NO on K(Ca) channels rely on the specific interactions of these gases with K(Ca),alpha and K(Ca),beta subunits.
- Published
- 2002
- Full Text
- View/download PDF
26. Mechanisms underlying the activation of large conductance Ca2+-activated K+ channels by nordihydroguaiaretic acid.
- Author
-
Yamamura H, Sakamoto K, Ohya S, Muraki K, and Imaizumi Y
- Subjects
- Animals, Calcium metabolism, Cell Line, Coronary Vessels drug effects, Coronary Vessels physiology, Humans, In Vitro Techniques, Large-Conductance Calcium-Activated Potassium Channels, Membrane Potentials drug effects, Mitochondria drug effects, Mitochondria physiology, Muscle, Smooth, Vascular physiology, Muscle, Smooth, Vascular ultrastructure, Patch-Clamp Techniques, Rats, Swine, Masoprocol pharmacology, Muscle, Smooth, Vascular drug effects, Potassium Channels, Calcium-Activated agonists
- Abstract
The mechanisms underlying the activation of large conductance Ca2+-activated K+ (BK) channel by nordihydroguaiaretic acid (NDGA) were examined in human embryonic kidney (HEK293) cells, where BK channel alpha (BKalpha) or a plus beta1 subunit (BKalphabeta1) was heterologously expressed, and also in freshly isolated porcine coronary arterial smooth muscle cells (PCASMCs). The activity of both BKalpha and BKalphabeta1 channels was increased by 10 microM NDGA in similar manners, indicating the selective action on the a subunit to increase Ca2+ sensitivity. The application of NDGA to PCASMCs induced outward current and hyperpolarization under voltage and current clamp, respectively, in a concentration-dependent manner (> or = 3 microM). These effects were blocked by 100 nM iberiotoxin. Electrical events induced by NDGA (> or = 10 microM) were, unexpectedly, associated with the increase in [Ca2+]i. After the treatment with caffeine and ryanodine, the [Ca2+]i increase by NDGA was markedly reduced and the hyperpolarization by NDGA was attenuated. The Ca2+ release by 10 microM NDGA was preceded by membrane depolarization of mitochondria. These results indicate that BK channel opening by NDGA in PCASMCs is due to the direct action on a subunit and also to Ca2+ release from sarcoplasmic reticulum, presumably via, at least in part, the inhibition of mitochondria respiration.
- Published
- 2002
- Full Text
- View/download PDF
27. Endothelin-Induced cyclooxygenase-dependent superoxide generation contributes to K+ channel functional impairment after brain injury.
- Author
-
Armstead WM
- Subjects
- ATP-Binding Cassette Transporters, Animals, Animals, Newborn, Benzimidazoles pharmacology, Brain Injuries pathology, Calcitonin Gene-Related Peptide pharmacology, Cromakalim pharmacology, Enzyme Activation physiology, Enzyme Induction, Female, KATP Channels, Male, Pia Mater pathology, Potassium Channels agonists, Potassium Channels drug effects, Potassium Channels, Calcium-Activated agonists, Potassium Channels, Calcium-Activated drug effects, Potassium Channels, Inwardly Rectifying, Superoxide Dismutase metabolism, Swine, Vasodilation physiology, Vasodilator Agents pharmacology, Brain Injuries metabolism, Endothelins pharmacology, Potassium Channels physiology, Prostaglandin-Endoperoxide Synthases metabolism, Superoxides metabolism
- Abstract
This study determined if endothelin (ET-1) generates superoxide anion (O2-) in a cyclooxygenase-dependent manner and if such production contributes to impairment of dilation to activators of ATP-sensitive K+ (KATP) and calcium-sensitive K+ (Kca) channels following fluid percussion brain injury (FPI) in newborn pigs equipped with closed cranial windows. Superoxide dismutase (SOD)-inhibitable nitroblue tetrazolium (NBT) reduction was determined as an index of O2- generation. Under non-brain injury conditions, topical ET-1 (10(-10) M, the concentration present in CSF following FPI) increased SOD-inhibitable NBT reduction from 1 +/- 1 to 17 +/- 3 pmol/mm2. Indomethacin, a cyclooxygenase inhibitor, blunted such NBT reduction (1 +/- 1 to 4 +/- 1 pmol/mm2), while the ET-1 antagonist BQ123 blocked NBT reduction. BQ123 and indomethacin also blunted the NBT reduction observed after FPI. Under non-brain injury conditions, ET-1 (10(-10) M) coadministered with the KATP and Kca channel agonists cromakalim and NS1619 (10-8, 10(-6) M) diminished dilation to these K+ channel agonists, while indomethacin partially prevented such impairment (13 +/- 1 and 23 +/- 1 vs. 2 +/- 1 and 6 +/- 1 vs. 6 +/- 1 and 14 +/- 2% for cromakalim in untreated, ET-1, and ET-1 plus indomethacin-treated piglets, respectively). Cromakalim- and NS1619-induced pial artery dilation was attenuated following FPI, while indomethacin or BQ123 preadministration partially prevented such impairment (13 +/- 1 and 23 +/- 1, sham control; 1 +/- 1 and 4 +/- 1, FPI; 8 +/- 1 and 16 +/- 3%, FPI and indomethacin-pretreated for responses to cromakalim 10(-8), 10-6 M, respectively). These data show that ET-1 increased O2- production in a cyclooxygenase-dependent manner and contributed to this production after FPI. These data also show that ET-1 blunted KATP and Kca channel-mediated cerebrovasodilation in a cyclooxygenase dependent manner. These data suggest that ET-1-induced cyclooxygenase-dependent O2- generation contributes to KATP and Kca channel function impairment after FPI.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.