Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
1.
J Med Chem ; 65(1): 303-322, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34962403

ABSTRACT

A series of modified N-cyclohexyl-2-(3,5-dimethyl-1H-pyrazol-1-yl)-6-methylpyrimidin-4-amine (CyPPA) analogues were synthesized by replacing the cyclohexane moiety with different 4-substituted cyclohexane rings, tyrosine analogues, or mono- and dihalophenyl rings and were subsequently studied for their potentiation of KCa2 channel activity. Among the N-benzene-N-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-4-pyrimidinamine derivatives, halogen decoration at positions 2 and 5 of benzene-substituted 4-pyrimidineamine in compound 2q conferred a ∼10-fold higher potency, while halogen substitution at positions 3 and 4 of benzene-substituted 4-pyrimidineamine in compound 2o conferred a ∼7-fold higher potency on potentiating KCa2.2a channels, compared to that of the parent template CyPPA. Both compounds retained the KCa2.2a/KCa2.3 subtype selectivity. Based on the initial evaluation, compounds 2o and 2q were selected for testing in an electrophysiological model of spinocerebellar ataxia type 2 (SCA2). Both compounds were able to normalize the abnormal firing of Purkinje cells in cerebellar slices from SCA2 mice, suggesting the potential therapeutic usefulness of these compounds for treating symptoms of ataxia.


Subject(s)
Cerebellum , Membrane Transport Modulators , Potassium Channels, Calcium-Activated , Purkinje Cells , Pyrimidines , Spinocerebellar Ataxias , Animals , Female , Male , Mice , Cerebellum/drug effects , Disease Models, Animal , Ion Channel Gating , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/pharmacology , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/metabolism , Purkinje Cells/drug effects , Pyrimidines/chemistry , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology , Structure-Activity Relationship
2.
Eur J Pharmacol ; 887: 173482, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32795513

ABSTRACT

Zileuton (Zyflo®) is regarded to be an inhibitor of 5-lipoxygenase. Although its effect on Ca2+-activated K+ currents has been reported, its overall ionic effects on neurons are uncertain. In whole-cell current recordings, zileuton increased the amplitude of Ca2+-activated K+ currents with an EC50 of 3.2 µM in pituitary GH3 lactotrophs. Furthermore, zileuton decreased the amplitudes of both delayed-rectifier K+ current (IK(DR)) and M-type K+ current (IK(M)). Conversely, no modification of hyperpolarization-activated cation current (Ih) was demonstrated in its presence of zileuton, although the subsequent addition of cilobradine effectively suppressed the current. In inside-out current recordings, the addition of zileuton to the bath increased the probability of large-conductance Ca2+-activated K+ (BKCa) channels; however, the subsequent addition of GAL-021 effectively reversed the stimulation of channel activity. The kinetic analyses showed an evident shortening in the slow component of mean closed time of BKCa channels in the presence of zileuton, with minimal change in mean open time or that in the fast component of mean closed time. The elevation of BKCa channels caused by zileuton was also observed in hippocampal mHippoE-14 neurons, without any modification of single-channel amplitude. In conclusion, except for its suppression of 5-lipoxygenase, our results indicate that zileuton does not exclusively act on BKCa channels, and its inhibitory effects on IK(DR) and IK(M) may combine to exert strong influence on the functional activities of electrically excitable cells in vivo.


Subject(s)
Delayed Rectifier Potassium Channels/antagonists & inhibitors , Hydroxyurea/analogs & derivatives , Lipoxygenase Inhibitors/pharmacology , Potassium Channels, Calcium-Activated/agonists , Animals , Arachidonate 5-Lipoxygenase/physiology , Cell Line , Delayed Rectifier Potassium Channels/physiology , Dose-Response Relationship, Drug , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Hydroxyurea/pharmacology , Mice , Potassium Channels, Calcium-Activated/physiology
3.
Int J Biochem Cell Biol ; 123: 105748, 2020 06.
Article in English | MEDLINE | ID: mdl-32353429

ABSTRACT

Population aging, as well as the handling of age-associated diseases, is a worldwide increasing concern. Among them, Alzheimer's disease stands out as the major cause of dementia culminating in full dependence on other people for basic functions. However, despite numerous efforts, in the last decades, there was no new approved therapeutic drug for the treatment of the disease. Calcium-activated potassium channels have emerged as a potential tool for neuronal protection by modulating intracellular calcium signaling. Their subcellular localization is determinant of their functional effects. When located on the plasma membrane of neuronal cells, they can modulate synaptic function, while their activation at the inner mitochondrial membrane has a neuroprotective potential via the attenuation of mitochondrial reactive oxygen species in conditions of oxidative stress. Here we review the dual role of these channels in the aging phenotype and Alzheimer's disease pathology and discuss their potential use as a therapeutic tool.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Inflammation/metabolism , Mitochondria/metabolism , Neurons/metabolism , Potassium Channels, Calcium-Activated/metabolism , Aging/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Death/genetics , Humans , Memory/drug effects , Neurons/drug effects , Neurons/physiology , Oxidative Stress/genetics , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Reactive Oxygen Species/metabolism
4.
Pharmacol Res ; 151: 104539, 2020 01.
Article in English | MEDLINE | ID: mdl-31707036

ABSTRACT

Aging represents an independent risk factor for the development of cardiovascular disease, and is associated with complex structural and functional alterations in the vasculature, such as endothelial dysfunction. Small- and intermediate-conductance, Ca2+-activated K+ channels (KCa2.3 and KCa3.1, respectively) are prominently expressed in the vascular endothelium, and pharmacological activators of these channels induce robust vasodilation upon acute exposure in isolated arteries and intact animals. However, the effects of prolonged in vivo administration of such compounds are unknown. In our study, we hypothesized that such treatment would ameliorate aging-related cardiovascular deficits. Aged (∼18 months) male Sprague Dawley rats were treated daily with either vehicle or the KCa channel activator SKA-31 (10 mg/kg, intraperitoneal injection; n = 6/group) for 8 weeks, followed by echocardiography, arterial pressure myography, immune cell and plasma cytokine characterization, and tissue histology. Our results show that SKA-31 administration improved endothelium-dependent vasodilation, reduced agonist-induced vascular contractility, and prevented the aging-associated declines in cardiac ejection fraction, stroke volume and fractional shortening, and further improved the expression of endothelial KCa channels and associated cell signalling components to levels similar to those observed in young male rats (∼5 months at end of study). SKA-31 administration did not promote pro-inflammatory changes in either T cell populations or plasma cytokines/chemokines, and we observed no overt tissue histopathology in heart, kidney, aorta, brain, liver and spleen. SKA-31 treatment in young rats had little to no effect on vascular reactivity, select protein expression, tissue histology, plasma cytokines/chemokines or immune cell properties. Collectively, these data demonstrate that administration of the KCa channel activator SKA-31 improved aging-related cardiovascular function, without adversely affecting the immune system or promoting tissue toxicity.


Subject(s)
Aging , Arterial Pressure/drug effects , Benzothiazoles/pharmacology , Heart/drug effects , Potassium Channels, Calcium-Activated/agonists , Aging/drug effects , Animals , Cells, Cultured , Heart/physiology , Male , Potassium Channels, Calcium-Activated/metabolism , Rats, Sprague-Dawley , Stroke Volume/drug effects , Vasodilation/drug effects
5.
J Cardiovasc Pharmacol ; 72(2): 106-111, 2018 08.
Article in English | MEDLINE | ID: mdl-29787401

ABSTRACT

Activation of melatonin receptors induces cardioprotection. Mitochondrial potassium channels (mKCa and mKATP) are involved in the signaling cascade of preconditioning. The melatonin receptor agonist ramelteon is an approved oral medication for treatment of insomnia, but nothing is known about possible cardioprotective properties. We investigated whether (1) ramelteon induces cardioprotection mediated by the melatonin receptor; (2) this effect is concentration-dependent; and (3) mKCa and/or mKATP channels are critically involved in ramelteon-induced cardioprotection. Hearts of male Wistar rats were randomized and placed on a Langendorff system, perfused with Krebs-Henseleit buffer at a constant pressure of 80 mm Hg. All hearts were subjected to 33 minutes of global ischemia and 60 minutes of reperfusion. Before, ischemic hearts were perfused with different concentrations of ramelteon (0.01-5 µM) for determination of a concentration-effect curve. In subsequent experiments, the lowest protective concentration of ramelteon was administered together with paxilline (mKCa channel inhibitor) and 5-hydroxydecanoate (mKATP channel inhibitor). To determine whether the reduction of ischemia and reperfusion injury by ramelteon is mediated by melatonin receptor, we combined ramelteon with luzindole, a melatonin receptor antagonist. Infarct size was determined by triphenyltetrazolium chloride staining. In control animals, infarct size was 58% ± 6%. Ramelteon in a concentration of 0.03 µM reduced infarct size to 28% ± 4% (P < 0.0001 vs. Con). A lower concentration of ramelteon did not initiate cardioprotection, and higher concentrations did not further decrease infarct size. Paxilline, 5-hydroxydecanoate, and luzindole completely blocked the ramelteon-induced cardioprotection. This study shows for the first time that (1) ramelteon induces cardioprotection through melatonin receptor; (2) the effect is not concentration-dependent; and (3) activation of mKCa and mKATP channels is involved.


Subject(s)
Cardiovascular Agents/pharmacology , Indenes/pharmacology , Mitochondria, Heart/drug effects , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Potassium Channels, Calcium-Activated/agonists , Potassium Channels/agonists , Receptors, Melatonin/agonists , Animals , Hemodynamics/drug effects , Isolated Heart Preparation , Male , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Potassium Channels/metabolism , Potassium Channels, Calcium-Activated/metabolism , Rats, Wistar , Receptors, Melatonin/metabolism , Signal Transduction/drug effects , Ventricular Function, Left/drug effects
6.
Mol Cell Biochem ; 445(1-2): 187-194, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29305679

ABSTRACT

We have recently found that diabetes is associated with the inactivation of the calcium-activated potassium channels (KCa) in endothelial cells, which may contribute to endothelial dysfunction in diabetic patients at baseline. In the current study, we further investigated the effects of diabetes on coronary arteriolar responses to the small (SK) and intermediate (IK) KCa opener NS309 in diabetic and non-diabetic patients and correlated that data with the changes in the SK/IK protein expression/distribution in the setting of cardioplegic ischemia and reperfusion (CP) and cardiopulmonary bypass (CPB). Coronary arterioles from the harvested right atrial tissue samples from diabetic and non-diabetic patients (n = 8/group) undergoing cardiac surgery were dissected pre- and post-CP/CPB. The in vitro relaxation response of pre-contracted arterioles was examined in the presence of the selective SK/IK opener NS309 (10-9-10-5 M). The protein expression/localization of KCa channels in the harvested atrial tissue samples, coronary microvessels, and primary cultured human coronary endothelial cells were assayed by Western blotting and immunohistochemistry. The relaxation response to NS309 post-CP/CPB was significantly decreased in diabetic and non-diabetic groups compared to their pre-CP/CPB responses, respectively (P < 0.05). Furthermore, this decrease was greater in the diabetic group than that of the non-diabetic group (P < 0.05). There were no significant differences in the total protein expression/distribution of SK/IK in the human myocardium, coronary microvessels or coronary endothelial cells between diabetic and non-diabetic groups or between pre- and post-CP/CPB (P > 0.05). Our results suggest that diabetes further inactivates SK/IK channels of coronary microvasculature early after CP/CPB and cardiac surgery. The lack of diabetic changes in SK/IK protein abundances in the setting of CP/CPB suggests that the effect is post-translational. This alteration may contribute to post-operative endothelial dysfunction in the diabetic patients early after CP/CPB and cardiac surgery.


Subject(s)
Arterioles/drug effects , Cardiopulmonary Bypass , Coronary Vessels/drug effects , Diabetes Mellitus/physiopathology , Heart Arrest, Induced , Potassium Channels, Calcium-Activated/agonists , Aged , Arterioles/metabolism , Arterioles/pathology , Blotting, Western , Case-Control Studies , Coronary Vessels/metabolism , Coronary Vessels/pathology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Heart Atria/metabolism , Humans , Male , Middle Aged , Muscle Proteins/metabolism , Potassium Channels, Calcium-Activated/metabolism
8.
J Cardiovasc Pharmacol ; 70(5): 314-328, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28777255

ABSTRACT

Both big (BKCa) and small (SKCa) conductance Ca-sensitive K channels are present in mammalian cardiac cell mitochondria (m). We used pharmacological agonists and antagonists of BKCa and SKCa channels to examine the importance of endogenous opening of these channels and the relative contribution of either or both of these channels to protect against contractile dysfunction and reduce infarct size after ischemia reperfusion (IR) injury through a mitochondrial protective mechanism. After global cardiac IR injury of ex vivo perfused Guinea pig hearts, we found the following: both agonists NS1619 (for BKCa) and DCEB (for SKCa) improved contractility; BKCa antagonist paxilline (PAX) alone or with SKCa antagonist NS8593 worsened contractility and enhanced infarct size; both antagonists PAX and NS8593 obliterated protection by their respective agonists; BKCa and SKCa antagonists did not block protection afforded by SKCa and BKCa agonists, respectively; and all protective effects by the agonists were blocked by scavenging superoxide anions (O2) with Mn(III) tetrakis (4-benzoic acid) porphyrin (TBAP). Contractile function was inversely associated with global infarct size. In in vivo rats, infusion of NS8593, PAX, or both antagonists enhanced regional infarct size while infusion of either NS1619 or DCEB reduced infarct size. In cardiac mitochondria isolated from ex vivo hearts after IR, combined SKCa and BKCa agonists improved respiratory control index and Ca retention capacity compared with IR alone, whereas the combined antagonists did not alter respiratory control index but worsened Ca retention capacity. Although the differential protective bioenergetics effects of endogenous or exogenous BKCa and SKCa channel opening remain unclear, each channel likely responds to different sensing Ca concentrations and voltage gradients over time during oxidative stress-induced injury to individually or together protect cardiac mitochondria and myocytes.


Subject(s)
Cardiotonic Agents/pharmacology , Mitochondria, Heart/physiology , Myocytes, Cardiac/physiology , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/physiology , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Animals , Benzimidazoles/pharmacology , Female , Guinea Pigs , Male , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
9.
Neurochem Int ; 109: 13-23, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28511953

ABSTRACT

Ca2+-activated K+ (KCa) channels regulate after-hyperpolarization in many types of neurons in the central and peripheral nervous system. Small conductance Ca2+-activated K+ (KCa2/SK) channels, a subfamily of KCa channels, are widely expressed in the nervous system, and in the cardiovascular system. Voltage-independent SK channels are activated by alterations in intracellular Ca2+ ([Ca2+]i) which facilitates the opening of these channels through binding of Ca2+ to calmodulin that is constitutively bound to the SK2 C-terminus. In neurons, SK channels regulate synaptic plasticity and [Ca2+]i homeostasis, and a number of recent studies elaborated on the emerging neuroprotective potential of SK channel activation in conditions of excitotoxicity and cerebral ischemia, as well as endoplasmic reticulum (ER) stress and oxidative cell death. Recently, SK channels were discovered in the inner mitochondrial membrane and in the membrane of the endoplasmic reticulum which sheds new light on the underlying molecular mechanisms and pathways involved in SK channel-mediated protective effects. In this review, we will discuss the protective properties of pharmacological SK channel modulation with particular emphasis on intracellularly located SK channels as potential therapeutic targets in paradigms of neuronal dysfunction.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Nervous System Diseases/metabolism , Potassium Channels, Calcium-Activated/metabolism , Animals , Carbamates/pharmacology , Carbamates/therapeutic use , Cell Membrane/drug effects , Endoplasmic Reticulum/drug effects , Humans , Mitochondria/drug effects , Nervous System Diseases/drug therapy , Neurons/drug effects , Neurons/metabolism , Piperidines/pharmacology , Piperidines/therapeutic use , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors
10.
Int J Cardiol ; 241: 351-357, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28487150

ABSTRACT

BACKGROUND: Opening of mitochondrial calcium-activated potassium channels (BKCa) reduces infarct size after myocardial ischemia/reperfusion injury (I/R). It is unknown if targeting BKCa-channels improves cardiac performance in the long-term after I/R. METHODS: Experiments were conducted in compliance with institutional and national guidelines in C57BL/6 mice (n=7-8/group). Animals were randomized into two groups. Preconditioning was induced by intraperitoneal application of NS1619 (NS, 1µg/g bw) 10min before ischemia, control animals (Con) received the vehicle. All animals underwent 45min of myocardial ischemia and four weeks of reperfusion. Transthoracal Echocardiography (TTE) was conducted one and four weeks after ischemia (TTEW1/TTEW4) and additionally a cardiac MRI was done in week four. At the end of experiments the infarction scar was determined by AZAN staining. RESULTS: TTE revealed that NS1619 improved ejection fraction one week (Con: 36±4%, NS: 45±4%; P<0.05) and four weeks after I/R (Con: 33±11%, NS: 46±8%; P<0.05). Preconditioning with NS1619 reduced end-diastolic volume at both time points (TTEW1: Con: 60±12µl, NS: 45±8µl; TTEW4: Con: 82±31µl, NS: 44±8µl; each P<0.05) and increased fractional shortening after four weeks (TTEW4: Con: 12±6%, NS: 24±8%; P<0.05). MRI-analysis after four weeks confirmed the echocardiographic results. NS1619 increased ejection fraction by 45% (MRI: Con: 29±6%, NS: 42±9%; P<0.05 vs. Con) and reduced end-diastolic and -systolic volume (EDV, ESV) compared to control (MRI: EDV: Con: 110±19µl, NS: 88±16µl; ESV: Con: 79±19µl, NS: 53±18µl; each P<0.05). Preconditioning reduced infarction scar after four weeks by 25% (Con: 12±3%, NS: 9±2%; P<0.05). CONCLUSIONS: Preconditioning by opening of BKCa-channels with NS1619 improves cardiac performance after four weeks of reperfusion and reduces myocardial infarction scar.


Subject(s)
Coronary Occlusion/diagnostic imaging , Coronary Occlusion/physiopathology , Potassium Channels, Calcium-Activated/physiology , Ventricular Function, Left/physiology , Animals , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Coronary Occlusion/drug therapy , Echocardiography/trends , Electrocardiography/trends , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Mice , Mice, Inbred C57BL , Potassium Channels, Calcium-Activated/agonists , Random Allocation , Time Factors , Ventricular Function, Left/drug effects
11.
Eur J Pharmacol ; 765: 375-83, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26362752

ABSTRACT

Fluoxetine, a selective serotonin reuptake inhibitor (SSRI), has effects beyond its antidepressant properties, altering, e.g., mechanisms involved in blood pressure and vasomotor tone control. Although many studies have addressed the acute impact of fluoxetine on the cardiovascular system, there is a paucity of information on the chronic vascular effects of this SSRI. We tested the hypothesis that chronic fluoxetine treatment enhances the vascular reactivity to vasodilator stimuli by increasing nitric oxide (NO) signaling and activation of potassium (K+) channels. Wistar rats were divided into two groups: (I) vehicle (water for 21 days) or (II) chronic fluoxetine (10 mg/kg/day in the drinking water for 21 days). Fluoxetine treatment increased endothelium-dependent and independent vasorelaxation (analyzed by mesenteric resistance arteries reactivity) as well as constitutive NO synthase (NOS) activity, phosphorylation of eNOS at Serine1177 and NO production, determined by western blot and fluorescence. On the other hand, fluoxetine treatment did not alter vascular expression of neuronal and inducible NOS or guanylyl cyclase (GC). Arteries from fluoxetine-treated rats exhibited increased relaxation to pinacidil. Increased acetylcholine vasorelaxation was abolished by a calcium-activated K+ channel (KCa) blocker, but not by an inhibitor of KATP channels. On the other hand, vascular responses to Bay 41-2272 and 8-bromo-cGMP were similar between the groups. In conclusion, chronic fluoxetine treatment increases endothelium-dependent and independent relaxation of mesenteric resistance arteries by mechanisms that involve increased eNOS activity, NO generation, and KCa channels activation. These effects may contribute to the cardiovascular effects associated with chronic fluoxetine treatment.


Subject(s)
Fluoxetine/administration & dosage , Mesenteric Arteries/metabolism , Nitric Oxide/biosynthesis , Potassium Channels, Calcium-Activated/metabolism , Vasoconstriction/physiology , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Male , Mesenteric Arteries/drug effects , Nitric Oxide/agonists , Organ Culture Techniques , Potassium Channels, Calcium-Activated/agonists , Rats , Rats, Wistar , Vasoconstriction/drug effects
12.
J Mol Cell Cardiol ; 72: 364-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24787473

ABSTRACT

Endothelial dysfunction is a common early pathogenic event in patients with type 2 diabetes (T2D) who exhibit cardiovascular disease. In the present study, we have examined the effect of SKA-31, a positive modulator of endothelial Ca(2+)-activated K(+) (KCa) channels, on total coronary flow in isolated hearts from Goto-Kakizaki rats, a non-obese model of T2D exhibiting metabolic syndrome. Total coronary flow and left ventricular developed pressure were monitored simultaneously in isolated, spontaneously beating Langendorff-perfused hearts. Acute administrations of bradykinin (BK) or adenosine (ADO) increased coronary flow, but responses were significantly blunted in diabetic hearts at 10-12 and 18-20weeks of age compared with age-matched Wistar controls, consistent with the presence of endothelial dysfunction. In contrast, SKA-31 dose-dependently (0.01-5µg) increased total coronary flow to comparable levels in both control and diabetic rat hearts at both ages. Flow responses to sodium nitroprusside were not different between control and diabetic hearts, suggesting normal arterial smooth muscle function. Importantly, exposure to a sub-threshold concentration of SKA-31 (i.e. 0.3µM) rescued the impaired BK and ADO-evoked vasodilatory responses in diabetic hearts. Endothelial KCa channel activators may thus help to preserve coronary flow in diabetic myocardium.


Subject(s)
Benzothiazoles/pharmacology , Coronary Circulation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Endothelium, Vascular/drug effects , Heart/drug effects , Potassium Channels, Calcium-Activated/agonists , Adenosine/pharmacology , Age Factors , Animals , Bradykinin/pharmacology , Coronary Vessels/drug effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Heart/physiopathology , Infusion Pumps , Male , Nitroprusside/pharmacology , Organ Culture Techniques , Potassium Channels, Calcium-Activated/metabolism , Rats , Rats, Wistar
13.
PLoS One ; 8(11): e78727, 2013.
Article in English | MEDLINE | ID: mdl-24236040

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.


Subject(s)
Motor Neurons/physiology , Nitric Oxide/physiology , Potassium Channels, Calcium-Activated/metabolism , 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
14.
J Physiol ; 591(20): 5107-23, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23959673

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.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Mesenteric Arteries/metabolism , Muscle Cells/metabolism , Potassium Channels, Calcium-Activated/metabolism , Vasodilation , 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
15.
PLoS One ; 8(7): e69551, 2013.
Article in English | MEDLINE | ID: mdl-23874973

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.


Subject(s)
Cell Survival/drug effects , Potassium Channels, Calcium-Activated/metabolism , Potassium/metabolism , 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
16.
Assay Drug Dev Technol ; 11(3): 163-72, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23198866

ABSTRACT

The intermediate-conductance Ca(2+)-activated K(+) channel (KCa3.1) has been proposed to play many physiological roles, and modulators of KCa3.1 activity are potentially interesting as new drugs. In order to identify new chemical scaffolds, high-throughput screening (HTS) assays are needed. In the current study, we present an HTS assay that has been optimized for the detection of inhibitors as well as activators of KCa3.1 in a combined assay. We used HEK293 cells heterologously expressing KCa3.1 in a fluorescence-based Tl(+) influx assay, where the permeability of potassium channels to Tl(+) is taken advantage of. We found the combined activator-and-inhibitor assay to be robust and insensitive to dimethyl sulfoxide (up to 1%), and conducted an HTS campaign of 217,119 small molecules. In total, 224 confirmed activators and 312 confirmed inhibitors were found, which corresponded to a hit rate of 0.10% and 0.14%, respectively. The confirmed hits were further characterized in a fluorometric imaging plate reader-based concentration-response assay, and selected compounds were subjected to secondary testing in an assay for endogenous KCa3.1 activity using human erythrocytes (red blood cell assay). Although the estimated potencies were slightly higher in the RBC assay, there was an overall good correlation across all clusters. The campaign led to the identification of several chemical series of KCa3.1 activators and inhibitors, comprising already known pharmacophores and new chemical series. One of these were the benzothiazinones that constitute a new class of highly potent KCa3.1 inhibitors, exemplified by 4-{[3-(trifluoromethyl)phenyl]methyl}-2H-1,4-benzothiazin-3(4H)-one (NS6180).


Subject(s)
High-Throughput Screening Assays/methods , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Acetamides/chemical synthesis , Acetamides/pharmacology , Algorithms , Data Interpretation, Statistical , Erythrocytes/chemistry , Erythrocytes/metabolism , Fluorometry , HEK293 Cells , Humans , Inflammatory Bowel Diseases/drug therapy , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Small Molecule Libraries , Thallium/chemistry , Thallium/pharmacokinetics , Thiazines/pharmacology , Trityl Compounds/chemical synthesis , Trityl Compounds/pharmacology
17.
J Neurosci ; 32(44): 15533-46, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23115190

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.


Subject(s)
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 , 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
18.
Behav Brain Res ; 232(1): 37-43, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22487247

ABSTRACT

Repeated stress impacts emotion, and can induce mood and anxiety disorders. These disorders are characterized by imbalance of emotional responses. The amygdala is fundamental in expression of emotion, and is hyperactive in many patients with mood or anxiety disorders. Stress also leads to hyperactivity of the amygdala in humans. In rodent studies, repeated stress causes hyperactivity of the amygdala, and increases fear conditioning behavior that is mediated by the basolateral amygdala (BLA). Calcium-activated potassium (K(Ca)) channels regulate BLA neuronal activity, and evidence suggests reduced small conductance K(Ca) (SK) channel function in male rats exposed to repeated stress. Pharmacological enhancement of SK channels reverses the BLA neuronal hyperexcitability caused by repeated stress. However, it is not known if pharmacological targeting of SK channels can repair the effects of repeated stress on amygdala-dependent behaviors. The purpose of this study was to test whether enhancement of SK channel function reverses the effects of repeated restraint on BLA-dependent auditory fear conditioning. We found that repeated restraint stress increased the expression of cued conditioned fear in male rats. However, 1-Ethyl-2-benzimidazolinone (1-EBIO, 1 or 10 mg/kg) or CyPPA (5 mg/kg) administered 30 min prior to testing of fear expression brought conditioned freezing to control levels, with little impact on fear expression in control handled rats. These results demonstrate that enhancement of SK channel function can reduce the abnormalities of BLA-dependent fear memory caused by repeated stress. Furthermore, this indicates that pharmacological targeting of SK channels may provide a novel target for alleviation of psychiatric symptoms associated with amygdala hyperactivity.


Subject(s)
Fear/drug effects , Fear/psychology , Memory/drug effects , Potassium Channels, Calcium-Activated/agonists , Stress, Psychological/psychology , Acoustic Stimulation , Animals , Benzimidazoles/pharmacology , Calcium Channel Agonists/pharmacology , Conditioning, Psychological/drug effects , Cues , Data Interpretation, Statistical , Electroshock , Handling, Psychological , Male , Membrane Potentials/drug effects , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley
19.
Circ Res ; 110(10): 1311-21, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22492531

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.


Subject(s)
Endothelium, Vascular/physiology , Epigastric Arteries/physiology , Gap Junctions/physiology , Potassium Channels, Calcium-Activated/physiology , Vascular Resistance/physiology , 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
20.
Acta Physiol (Oxf) ; 205(2): 247-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22099068

ABSTRACT

AIM: To investigate the effect of 4,5-dichloro-1,3-diethyl-1,3-dihydro-benzoimidazol-2-one (NS4591), a novel SK/IK channels positive modulator, on human myometrial activity. METHODS: Organ bath studies were performed on myometrial preparations obtained from women undergoing elective caesarean section at term (N = 11) or hysterectomy (N = 11). NS4591 was added cumulatively in the concentration range of 0.3-30 µm. In separate experiments, the effects of pre-incubation of muscle preparation with the SK or IK channel blockers apamin (1 µm) and TRAM34 (10 µm) on the outcomes of NS4591 were evaluated. Simultaneous vehicle controls were performed for all experiments. The effects of drugs were studied on spontaneous contractions. RESULTS: NS4591 exerted an inhibitory effect on myometrial contractions in muscle strips from non-pregnant and pregnant women. The contractility in non-pregnant and pregnant myometrium was reduced to the following values respectively: amplitude 20.65 ± 7.38% (P < 0.001) and 42.85 ± 11.04% (P < 0.05) and area under the curve 11.72 ± 7.39% (P < 0.001) and 34.84 ± 10.50% (P < 0.001) and are reflective of 30 µm NS4591 compared to vehicle control. In non-pregnant tissue, apamin partially reduced the inhibitory effects of NS4591, but we observed relaxation mediated by NS4591 despite pre-incubation with TRAM34. In contrast, in pregnant tissue, neither apamin nor TRAM34 could reverse the relaxatory effects of NS4591. CONCLUSION: Our findings imply that SK/IK channels are present and functional in myometrium from pregnant and non-pregnant women. The SK/IK channel-positive modulator NS4591 exerts relaxation of human myometrium in vitro, and this may have implications for the clinical management of preterm labour.


Subject(s)
Benzimidazoles/pharmacology , Myometrium/drug effects , Potassium Channels, Calcium-Activated/agonists , Uterine Contraction/drug effects , Adult , Cesarean Section , Dose-Response Relationship, Drug , Female , Humans , Hysterectomy , Middle Aged , Myometrium/physiology , Pregnancy , Uterine Contraction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...