Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
1.
Heart Rhythm ; 18(1): 98-108, 2021 01.
Article in English | MEDLINE | ID: mdl-32763429

ABSTRACT

BACKGROUND: Concomitant apamin-sensitive small conductance calcium-activated potassium current (IKAS) activation and sodium current inhibition induce J-wave syndrome (JWS) in rabbit hearts. Sudden death in JWS occurs predominantly in men at night when parasympathetic tone is strong. OBJECTIVE: The purpose of this study was to test the hypotheses that acetylcholine (ACh), the parasympathetic transmitter, activates IKAS and causes JWS in the presence of ajmaline. METHODS: We performed optical mapping in Langendorff-perfused rabbit hearts and whole-cell voltage clamp to determine IKAS in isolated ventricular cardiomyocytes. RESULTS: ACh (1 µM) + ajmaline (2 µM) induced J-point elevations in all (6 male and 6 female) hearts from 0.01± 0.01 to 0.31 ± 0.05 mV (P<.001), which were reduced by apamin (specific IKAS inhibitor, 100 nM) to 0.14 ± 0.02 mV (P<.001). More J-point elevation was noted in male than in female hearts (P=.037). Patch clamp studies showed that ACh significantly (P<.001) activated IKAS in isolated male but not in female ventricular myocytes (n=8). Optical mapping studies showed that ACh induced action potential duration (APD) heterogeneity, which was more significant in right than in left ventricles. Apamin in the presence of ACh prolonged both APD at the level of 25% (P<.001) and APD at the level of 80% (P<.001) and attenuated APD heterogeneity. Ajmaline further increased APD heterogeneity induced by ACh. Ventricular arrhythmias were induced in 6 of 6 male and 1 of 6 female hearts (P=.015) in the presence of ACh and ajmaline, which was significantly suppressed by apamin in the former. CONCLUSION: ACh activates ventricular IKAS. ACh and ajmaline induce JWS and facilitate the induction of ventricular arrhythmias more in male than in female ventricles.


Subject(s)
Acetylcholine/pharmacology , Ajmaline/pharmacology , Arrhythmias, Cardiac/drug therapy , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Calcium-Activated/drug effects , Sodium Channels/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Cholinergic Agonists/pharmacology , Disease Models, Animal , Heart Ventricles/drug effects , Heart Ventricles/pathology , Isolated Heart Preparation/methods , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Optical Imaging , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/metabolism , Rabbits , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Sodium Channels/drug effects , Voltage-Gated Sodium Channel Blockers/pharmacology
2.
J Neurophysiol ; 124(3): 962-972, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32816581

ABSTRACT

Previous studies have found GABA in vestibular end organs. However, existence of GABA receptors or possible GABAergic effects on vestibular nerve afferents has not been investigated. The current study was conducted to determine whether activation of GABAB receptors affects calyx afferent terminals in the central region of the cristae of semicircular canals. We used patch-clamp recording in postnatal day 13-18 (P13-P18) Sprague-Dawley rats of either sex. Application of GABAB receptor agonist baclofen inhibited voltage-sensitive potassium currents. This effect was blocked by selective GABAB receptor antagonist CGP 35348. Application of antagonists of small (SK)- and large-conductance potassium (BK) channels almost completely blocked the effects of baclofen. The remaining baclofen effect was blocked by cadmium chloride, suggesting that it could be due to inhibition of voltage-gated calcium channels. Furthermore, baclofen had no effect in the absence of calcium in the extracellular fluid. Inhibition of potassium currents by GABAB activation resulted in an excitatory effect on calyx terminal action potential firing. While in the control condition calyces could only fire a single action potential during step depolarizations, in the presence of baclofen they fired continuously during steps and a few even showed repetitive discharge. We also found a decrease in threshold for action potential generation and a decrease in first-spike latency during step depolarization. These results provide the first evidence for the presence of GABAB receptors on calyx terminals, showing that their activation results in an excitatory effect and that GABA inputs could be used to modulate calyx response properties.NEW & NOTEWORTHY Using in vitro whole cell patch-clamp recordings from calyx terminals in the vestibular end organs, we show that activation of GABAB receptors result in an excitatory effect, with decreased spike-frequency adaptation and shortened first-spike latencies. Our results suggest that these effects are mediated through inhibition of calcium-sensitive potassium channels.


Subject(s)
Action Potentials/physiology , GABA-B Receptor Agonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Hair Cells, Vestibular/physiology , Potassium Channels, Calcium-Activated/metabolism , Presynaptic Terminals/physiology , Receptors, GABA-B/metabolism , Semicircular Canals/physiology , Action Potentials/drug effects , Animals , Baclofen/pharmacology , Cadmium Chloride/pharmacology , Female , Hair Cells, Vestibular/drug effects , Male , Organophosphorus Compounds/pharmacology , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/drug effects , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects , Semicircular Canals/drug effects
3.
Int J Biochem Cell Biol ; 125: 105792, 2020 08.
Article in English | MEDLINE | ID: mdl-32574707

ABSTRACT

In this review, we describe key signaling pathways regulating potassium channels present in the inner mitochondrial membrane. The signaling cascades covered here include phosphorylation, redox reactions, modulation by calcium ions and nucleotides. The following types of potassium channels have been identified in the inner mitochondrial membrane of various tissues: ATP-sensitive, Ca2+-activated, voltage-gated and two-pore domain potassium channels. The direct roles of these channels involve regulation of mitochondrial respiration, membrane potential and synthesis of reactive oxygen species (ROS). Changes in channel activity lead to diverse pro-life and pro-death responses in different cell types. Hence, characterizing the signaling pathways regulating mitochondrial potassium channels will facilitate understanding the physiological role of these proteins. Additionally, we describe in this paper certain regulatory mechanisms, which are unique to mitochondrial potassium channels.


Subject(s)
Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Potassium Channels/metabolism , Signal Transduction/genetics , Adenosine Triphosphate , Animals , Calcium/metabolism , Humans , Oxidation-Reduction , Potassium Channels/drug effects , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/metabolism , Potassium Channels, Tandem Pore Domain/drug effects , Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels, Voltage-Gated/drug effects , Potassium Channels, Voltage-Gated/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
4.
Am J Physiol Heart Circ Physiol ; 317(2): H357-H363, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31199187

ABSTRACT

Elevated plasma aldosterone (Aldo) levels are associated with greater risk of cardiac ischemic events and cardiovascular mortality. Adenosine-mediated coronary vasodilation is a critical cardioprotective mechanism during ischemia; however, whether this response is impaired by increased Aldo is unclear. We hypothesized that chronic Aldo impairs coronary adenosine-mediated vasodilation via downregulation of vascular K+ channels. Male C57BL/6J mice were treated with vehicle (Con) or subpressor Aldo for 4 wk. Coronary artery function, assessed by wire myography, revealed Aldo-induced reductions in vasodilation to adenosine and the endothelium-dependent vasodilator acetylcholine but not to the nitric oxide donor sodium nitroprusside. Coronary vasoconstriction to endothelin-1 and the thromboxane A2 mimetic U-46619 was unchanged by Aldo. Additional mechanistic studies revealed impaired adenosine A2A, not A2B, receptor-dependent vasodilation by Aldo with a tendency for Aldo-induced reduction of coronary A2A gene expression. Adenylate cyclase inhibition attenuated coronary adenosine dilation but did not eliminate group differences, and adenosine-stimulated vascular cAMP production was similar between Con and Aldo mice. Similarly, blockade of inward rectifier K+ channels reduced but did not eliminate group differences in adenosine dilation whereas group differences were eliminated by blockade of Ca2+-activated K+ (KCa) channels that blunted and abrogated adenosine and A2A-dependent dilation, respectively. Gene expression of several coronary KCa channels was reduced by Aldo. Together, these data demonstrate Aldo-induced impairment of adenosine-mediated coronary vasodilation involving blunted A2A-KCa-dependent vasodilation, independent of blood pressure, providing important insights into the link between plasma Aldo and cardiac mortality and rationale for aldosterone antagonist use to preserve coronary microvascular function.NEW & NOTEWORTHY Increased plasma aldosterone levels are associated with worsened cardiac outcomes in diverse patient groups by unclear mechanisms. We identified that, in male mice, elevated aldosterone impairs coronary adenosine-mediated vasodilation, an important cardioprotective mechanism. This aldosterone-induced impairment involves reduced adenosine A2A, not A2B, receptor-dependent vasodilation associated with downregulation of coronary KCa channels and does not involve altered adenylate cyclase/cAMP signaling. Importantly, this effect of aldosterone occurred independent of changes in coronary vasoconstrictor responsiveness and blood pressure.


Subject(s)
Adenosine/pharmacology , Aldosterone/pharmacology , Coronary Vessels/drug effects , Potassium Channels, Calcium-Activated/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Coronary Vessels/metabolism , Cyclic AMP/metabolism , Down-Regulation , Male , Mice, Inbred C57BL , Potassium Channels, Calcium-Activated/genetics , Potassium Channels, Calcium-Activated/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Signal Transduction
5.
Brain Res ; 1711: 1-6, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30629942

ABSTRACT

Hypotension and low cerebral perfusion pressure are associated with low cerebral blood flow, cerebral ischemia, and poor outcomes after traumatic brain injury (TBI). Cerebral autoregulation is impaired after TBI, contributing to poor outcome. In prior studies, ERK mitogen activated protein kinase (MAPK) and ET-1 had been observed to be upregulated and contribute to impairment of cerebral autoregulation and histopathology after fluid percussion brain injury (FPI). Activation of ATP and Calcium sensitive (Katp and Kca) channels produce cerebrovasodilation and contribute to autoregulation, both impaired after TBI. Upregulation of ERK MAPK and endothelin-1 (ET-1) produces K channel function impairment after CNS injury. Inhaled nitric oxide (iNO) has recently been observed to prevent impairment of cerebral autoregulation and hippocampal CA1 and CA3 neuronal cell necrosis after FPI via block of upregulation of ERK MAPK and ET-1. We presently investigated whether iNO prevented impairment of Katp and Kca-mediated cerebrovasodilation after FPI in pigs equipped with a closed cranial window. Results show that pial artery dilation in response to the Katp agonist cromakalim, the Kca agonist NS1619, PGE2 and the NO releaser sodium nitroprusside (SNP) were blocked by FPI, but such impairment was prevented by iNO administered at 2 h post injury. Protection lasted for at least 1 h after iNO administration was stopped. Using vasodilaton as an index of function, these data indicate that iNO prevents impairment of cerebral autoregulation and limits histopathology after TBI through protection of K channel function via blockade of ERK MAPK and ET-1.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Homeostasis/drug effects , Nitric Oxide/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Brain Injuries, Traumatic/pathology , Calcium/metabolism , Calcium Channels/metabolism , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Endothelin-1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Male , Nitric Oxide/metabolism , Potassium Channels, Calcium-Activated/drug effects , Swine , Vasodilation/drug effects , Vasodilator Agents/pharmacology
6.
Biol Res ; 51(1): 38, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30290763

ABSTRACT

BACKGROUND: Hydrogen sulfide has been shown to improve the quality of oocytes destined for in vitro fertilization. Although hydrogen sulfide is capable of modulating ion channel activity in somatic cells, the role of hydrogen sulfide in gametes and embryos remains unknown. Our observations confirmed the hypothesis that the KATP and L-type Ca2+ ion channels play roles in porcine oocyte ageing and revealed a plausible contribution of hydrogen sulfide to the modulation of ion channel activity. RESULTS: We confirmed the benefits of the activation and suppression of the KATP and L-type Ca2+ ion channels, respectively, for the preservation of oocyte quality. CONCLUSIONS: Our experiments identified hydrogen sulfide as promoting the desired ion channel activity, with the capacity to protect porcine oocytes against cell death. Further experiments are needed to determine the exact mechanism of hydrogen sulfide in gametes and embryos.


Subject(s)
Calcium Channels/physiology , Cellular Senescence/physiology , Hydrogen Sulfide/pharmacology , Oocytes/drug effects , Potassium Channels, Calcium-Activated/physiology , Adenosine Triphosphate , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Female , Minoxidil/pharmacology , Oocytes/metabolism , Phenotype , Potassium Channels, Calcium-Activated/drug effects , Signal Transduction/drug effects , Swine , Verapamil/pharmacology
7.
Neurotoxicol Teratol ; 68: 27-35, 2018.
Article in English | MEDLINE | ID: mdl-29715503

ABSTRACT

Despite serious health effects, volatile industrial products containing toluene are deliberately inhaled for their psychoactive actions, mainly among adolescents and young adults. Chronic toluene inhalation induces multiple alterations at the cellular and behavioral level; however, modifications of neuronal networks associated with the reward system after repeated toluene exposure are not thoroughly characterized. Here we used whole-cell recordings to determine the effects of repeated exposure to toluene (1000, 4000 or 8000 ppm for 30 min, twice a day, for ten days) on the neurophysiological properties of prelimbic layer 5 pyramidal neurons of the medial prefrontal cortex (mPFC) in adolescent male Wistar rats. Neurons from animals repeatedly exposed to toluene showed a concentration-dependent increase in action potential firing discharge. This increase was related to a reduction of the small-conductance calcium-activated potassium current (after-hyperpolarization current, IAHP) that controls the firing frequency of neurons. Likewise, toluene altered the kinetics of the action potential. The hyperexcitability seen in toluene-exposed animals was also associated with an increase in the glutamatergic spontaneous synaptic activity converging on mPFC neurons. In summary, repeated toluene exposure enhances the excitability of prelimbic layer 5 pyramidal neurons of the mPFC in adolescent rats.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Potassium Channels, Calcium-Activated/drug effects , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Toluene/toxicity , Age Factors , Animals , Behavior, Animal/drug effects , Bicuculline/pharmacology , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/physiology , Kynurenic Acid/pharmacology , Male , Prefrontal Cortex/drug effects , Rats
8.
J Ethnopharmacol ; 220: 16-25, 2018 Jun 28.
Article in English | MEDLINE | ID: mdl-29609011

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lippia origanoides H.B.K. is an aromatic species used in folk medicine to treat respiratory diseases, including asthma. AIM OF THE STUDY: The aim of this work was to evaluate the relaxing potential and mechanism of action of the L. origanoides (LOO) essential oil in isolated guinea-pig trachea. MATERIALS AND METHODS: Leaves from L. origanoides were collected at experimental fields under organic cultivation, at the Forest Garden of Universidade Estadual de Feira de Santana. Essential oil was extracted by hydrodistillation, analyzed by GC/FID and GC/MS and the volatile constituents were identified. Spasmolytic activity and relaxant mechanism of LOO were assayed in isolated guinea-pig trachea contracted with histamine, carbachol or hyperpolarizing KCl. RESULTS: Chemical analysis revealed the presence of carvacrol (53.89%) as major constituent. LOO relaxed isolated guinea-pig trachea pre-contracted with KCl 60 mM [EC50 = 30.02 µg/mL], histamine 1 µM [EC50 = 9.28 µg/mL] or carbachol 1 µM [EC50 = 51.80 µg/mL]. The pre-incubation of glibenclamide, CsCl, propranolol, indomethacin, hexamethonium, aminophylline or L-NAME in histamine-induced contractions did not alter significantly the relaxant effect of LOO. However, the presence of 4-aminopyridine, tetraethylammonium or methylene blue reduced LOO effect, while the presence of dexamethasone or atropine potentialized the LOO relaxant effect. LOO pre-incubation inhibited carbachol-evoked contractions, with this effect potentialized in the presence of sodium nitroprusside and blocked in the presence of ODQ. CONCLUSIONS: The relaxant mechanism of LOO on the tracheal smooth muscle possibly involves stimulating of soluble guanylyl cyclase with consequent activation of the voltage-gated and Ca2+-activated K+ channels.


Subject(s)
Lippia/chemistry , Muscle, Smooth/drug effects , Oils, Volatile/pharmacology , Trachea/drug effects , Animals , Female , Gas Chromatography-Mass Spectrometry , Guinea Pigs , Male , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/metabolism , Oils, Volatile/isolation & purification , Parasympatholytics/isolation & purification , Parasympatholytics/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/metabolism , Potassium Channels, Voltage-Gated/drug effects , Potassium Channels, Voltage-Gated/metabolism , Soluble Guanylyl Cyclase/metabolism , Trachea/metabolism
9.
Biol. Res ; 51: 38, 2018. graf
Article in English | LILACS | ID: biblio-1038781

ABSTRACT

BACKGROUND: Hydrogen sulfide has been shown to improve the quality of oocytes destined for in vitro fertilization. Although hydrogen sulfide is capable of modulating ion channel activity in somatic cells, the role of hydrogen sulfide in gametes and embryos remains unknown. Our observations confirmed the hypothesis that the KATP and L-type Ca2+ ion channels play roles in porcine oocyte ageing and revealed a plausible contribution of hydrogen sulfide to the modulation of ion channel activity. RESULTS: We confirmed the benefits of the activation and suppression of the KATP and L-type Ca2+ ion channels, respectively, for the preservation of oocyte quality. CONCLUSIONS: Our experiments identified hydrogen sulfide as promoting the desired ion channel activity, with the capacity to protect porcine oocytes against cell death. Further experiments are needed to determine the exact mechanism of hydrogen sulfide in gametes and embryos.


Subject(s)
Animals , Female , Oocytes/drug effects , Calcium Channels/physiology , Cellular Senescence/physiology , Potassium Channels, Calcium-Activated/physiology , Hydrogen Sulfide/pharmacology , Oocytes/metabolism , Phenotype , Swine , Calcium Channel Blockers/pharmacology , Verapamil/pharmacology , Calcium Channels/drug effects , Signal Transduction/drug effects , Adenosine Triphosphate , Potassium Channels, Calcium-Activated/drug effects , Minoxidil/pharmacology
10.
J Pharmacol Sci ; 133(4): 276-279, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28302447

ABSTRACT

Membrane hyperpolarization is suggested to be a trigger for skeletal muscle differentiation. We investigated whether DCEBIO, an opener of the small/intermediate conductance Ca2+ activated K+ (SKCa/IKCa) channels, increase myogenic differentiation in C2C12 skeletal myoblasts. DCEBIO significantly increased myotube formation, protein expression level of myosin heavy chain II, and mRNA expression level of myogenin in C2C12 myoblasts cultured in differentiation medium. DCEBIO induced myotube formation and hyperpolarization were reduced by the IKCa channel blocker TRAM-34, but not by the SKCa channel blocker apamin. These findings show that DCEBIO increases myogenic differentiation by activating IKCa channels.


Subject(s)
Benzimidazoles/pharmacology , Cell Differentiation/drug effects , Myoblasts/cytology , Potassium Channels, Calcium-Activated/drug effects , Apamin/pharmacology , Cells, Cultured , Gene Expression/drug effects , Humans , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/cytology , Myoblasts/metabolism , Myogenin/genetics , Myogenin/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Pyrazoles/pharmacology , RNA, Messenger/metabolism
11.
J Neurosci ; 36(42): 10759-10768, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798131

ABSTRACT

Potentiation of excitatory inputs onto dopamine neurons of the ventral tegmental area (VTA) induced by cocaine exposure allows remodeling of the mesocorticolimbic circuitry, which ultimately drives drug-adaptive behavior. This potentiation is mediated by changes in NMDAR and AMPAR subunit composition. It remains unknown how this synaptic plasticity affects the activity of dopamine neurons. Here, using rodents, we demonstrate that a single cocaine injection increases the firing rate and bursting activity of VTA dopamine neurons, and that these increases persist for 7 d. This enhanced activity depends on the insertion of low-conductance, Ca2+-impermeable NMDARs that contain GluN3A. Since such receptors are not capable of activating small-conductance potassium channels, the intrinsic excitability of VTA dopamine neurons increases. Activation of group I mGluRs rescues synaptic plasticity and restores small-conductance calcium-dependent potassium channel function, normalizing the firing activity of dopamine neurons. Our study characterizes a mechanism linking drug-evoked synaptic plasticity to neural activity, revealing novel targets for therapeutic interventions. SIGNIFICANCE STATEMENT: We show that cocaine-evoked synaptic changes onto ventral tegmental area (VTA) dopamine (DA) neurons leads to long-lasting increases in their burst firing. This increase is due to impaired function of Ca2+-activated small-conductance calcium-dependent potassium (SK) channels; SK channels regulate firing of VTA DA neurons, but this regulation was absent after cocaine. Cocaine exposure drives the insertion of GluN3A-containing NMDARs onto VTA DA neurons. These receptors are Ca2+-impermeable, and thus SK channels are not efficiently activated by synaptic activity. In GluN3A knock-out mice, cocaine did not alter SK channel function or VTA DA neuron firing. This study directly links synaptic changes to increased intrinsic excitability of VTA DA neurons after cocaine, and explains how acute cocaine induces long-lasting remodeling of the mesolimbic DA system.


Subject(s)
Calcium/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Ventral Tegmental Area/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Female , Male , Membrane Glycoproteins/metabolism , Membrane Potentials/drug effects , Mice, Knockout , Neuronal Plasticity , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/drug effects , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/cytology
12.
J Neurosci ; 36(40): 10376-10391, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27707972

ABSTRACT

Methamphetamine (METH) is a substrate for the dopamine transporter that increases extracellular dopamine levels by competing with dopamine uptake and increasing reverse transport of dopamine via the transporter. METH has also been shown to alter the excitability of dopamine neurons. The mechanism of METH regulation of the intrinsic firing behaviors of dopamine neurons is less understood. Here we identified an unexpected and unique property of METH on the regulation of firing activity of mouse dopamine neurons. METH produced a transient augmentation of spontaneous spike activity of midbrain dopamine neurons that was followed by a progressive reduction of spontaneous spike activity. Inspection of action potential morphology revealed that METH increased the half-width and produced larger coefficients of variation of the interspike interval, suggesting that METH exposure affected the activity of voltage-dependent potassium channels in these neurons. Since METH has been shown to affect Ca2+ homeostasis, the unexpected findings that METH broadened the action potential and decreased the amplitude of afterhyperpolarization led us to ask whether METH alters the activity of Ca2+-activated potassium (BK) channels. First, we identified BK channels in dopamine neurons by their voltage dependence and their response to a BK channel blocker or opener. While METH suppressed the amplitude of BK channel-mediated unitary currents, the BK channel opener NS1619 attenuated the effects of METH on action potential broadening, afterhyperpolarization repression, and spontaneous spike activity reduction. Live-cell total internal reflection fluorescence microscopy, electrophysiology, and biochemical analysis suggest METH exposure decreased the activity of BK channels by decreasing BK-α subunit levels at the plasma membrane. SIGNIFICANCE STATEMENT: Methamphetamine (METH) competes with dopamine uptake, increases dopamine efflux via the dopamine transporter, and affects the excitability of dopamine neurons. Here, we identified an unexpected property of METH on dopamine neuron firing activity. METH transiently increased the spontaneous spike activity of dopamine neurons followed by a progressive reduction of the spontaneous spike activity. METH broadened the action potentials, increased coefficients of variation of the interspike interval, and decreased the amplitude of afterhyperpolarization, which are consistent with changes in the activity of Ca2+-activated potassium (BK) channels. We found that METH decreased the activity of BK channels by stimulating BK-α subunit trafficking. Thus, METH modulation of dopamine neurotransmission and resulting behavioral responses is, in part, due to METH regulation of BK channel activity.


Subject(s)
Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Methamphetamine/pharmacology , Action Potentials/drug effects , Animals , Benzimidazoles/pharmacology , Calcium Signaling/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Electrophysiological Phenomena/drug effects , HEK293 Cells , Humans , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channels , Mice , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/drug effects
13.
Braz J Med Biol Res ; 49(8)2016 Jul 25.
Article in English | MEDLINE | ID: mdl-27464023

ABSTRACT

A relationship between thyroid hormones and the cardiovascular system has been well established in the literature. The present in vitro study aimed to investigate the mechanisms involved in the vasodilator effect produced by the acute application of 10-8-10-4 M triiodothyronine (T3) to isolated rat aortic rings. Thoracic aortic rings from 80 adult male Wistar rats were isolated and mounted in tissue chambers filled with Krebs-Henseleit bicarbonate buffer in order to analyze the influence of endothelial tissue, inhibitors and blockers on the vascular effect produced by T3. T3 induced a vasorelaxant response in phenylephrine-precontracted rat aortic rings at higher concentrations (10-4.5-10-4.0 M). This outcome was unaffected by 3.1×10-7 M glibenclamide, 10-3 M 4-aminopyridine (4-AP), 10-5 M indomethacin, or 10-5 M cycloheximide. Contrarily, vasorelaxant responses to T3 were significantly (P<0.05) attenuated by endothelium removal or the application of 10-6 M atropine, 10-5 M L-NG-nitroarginine methyl ester (L-NAME), 10-7 M 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), 10-6 M (9S,10R,12R)-2,3,9,10,11,12-Hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i](1,6)benzodiazocine-10-carboxylic acid, methyl ester KT 5823, 10-2 M tetraethylammonium (TEA), or 10-7 M apamin plus 10-7 M charybdotoxin. The results suggest the involvement of endothelial mechanisms in the vasodilator effect produced by the acute in vitro application of T3 to rat aortic rings. Possible mechanisms include the stimulation of muscarinic receptors, activation of the NO-cGMP-PKG pathway, and opening of Ca2+-activated K+ channels.


Subject(s)
Aorta, Thoracic/drug effects , Endothelium, Vascular/drug effects , Triiodothyronine/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Atropine/pharmacology , Dimethyl Sulfoxide/pharmacology , Glyburide/pharmacology , Indomethacin/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Phenylephrine/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Rats, Wistar
14.
J Med Invest ; 63(1-2): 80-4, 2016.
Article in English | MEDLINE | ID: mdl-27040058

ABSTRACT

PURPOSE: Activation of the mitochondrial calcium-activated potassium (mKCa) channel reportedly confers resistance to myocardial ischemic stress. However, the role of the mKCa channel in postconditioning induced by volatile anesthetic remains unclear. METHODS: Male Japanese white rabbits underwent coronary artery occlusion for 30 min followed by reperfusion for 3 h. Volatile anesthetic, isoflurane, was administered at 3 min prior to reperfusion for 5 min. Rabbits were injected with the mKCa channel blocker, iberiotoxin, or the mKCa channel opener, NS1619, at 8 min prior to reperfusion. Myocardial infarct size and the area at risk (AAR) were measured at the end of the experiment. RESULTS: Isoflurane significantly reduced infarct size (23.0 ± 9.8% of the AAR, P<0.05) compared with the control (44.0 ± 9.1%). Iberiotoxin abolished the cardioprotective impact of isoflurane (43.0 ± 11.6%), while iberiotoxin alone exerted no effect on infarct size (45.0 ± 9.5%). NS1619 and isoflurane/NS1619 both significantly reduced infarct size (21.0 ± 10.3% and 19.0 ± 8.8%, respectively, P<0.05 vs control group), but isoflurane/NS1619 showed no additional benefits compared with isoflurane alone. CONCLUSION: These results indicate that activation of the mKCa channel contribute isoflurane-induced postconditioning.


Subject(s)
Ischemic Postconditioning/methods , Isoflurane/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/metabolism , Anesthetics, Inhalation/pharmacology , Animals , Cardiotonic Agents/pharmacology , Male , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Rabbits
15.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1081-7, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053645

ABSTRACT

Nitric oxide (NO) increases cutaneous blood flow; however, the underpinning mechanism(s) remains to be elucidated. We hypothesized that the cutaneous blood flow response during intradermal administration of sodium nitroprusside (SNP, a NO donor) is regulated by calcium-activated potassium (KCa) channels and cyclooxygenase (COX) in young adults. We also hypothesized that these contributions are diminished in older adults given that aging can downregulate KCa channels and reduce COX-derived vasodilator prostanoids. In 10 young (23 ± 5 yr) and 10 older (54 ± 4 yr) adults, cutaneous vascular conductance (CVC) was measured at four forearm skin sites infused with 1) Ringer (Control), 2) 50 mM tetraethylammonium (TEA), a nonspecific KCa channel blocker, 3) 10 mM ketorolac, a nonspecific COX inhibitor, or 4) 50 mM TEA + 10 mM ketorolac via intradermal microdialysis. All skin sites were coinfused with incremental doses of SNP (0.005, 0.05, 0.5, 5, and 50 mM each for 25 min). During SNP administration, CVC was similar at the ketorolac site (0.005-50 mM, all P > 0.05) relative to Control, but lower at the TEA and TEA + ketorolac sites (0.005-0.05 mM, all P < 0.05) in young adults. In older adults, ketorolac increased CVC relative to Control during 0.005-0.05 mM SNP administration (all P < 0.05), but this increase was not observed when TEA was coadministered (all P > 0.05). Furthermore, TEA alone did not modulate CVC during any concentration of SNP administration in older adults (all P > 0.05). We show that during low-dose NO administration (e.g., 0.005-0.05 mM), KCa channels contribute to cutaneous blood flow regulation in young adults; however, in older adults, COX inhibition increases cutaneous blood flow through a KCa channel-dependent mechanism.


Subject(s)
Aging/physiology , Blood Flow Velocity/physiology , Nitric Oxide/administration & dosage , Potassium Channels, Calcium-Activated/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Skin Physiological Phenomena/drug effects , Adult , Aging/drug effects , Blood Flow Velocity/drug effects , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Potassium Channels, Calcium-Activated/drug effects , Prostaglandin-Endoperoxide Synthases/drug effects , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/administration & dosage
16.
Braz. j. med. biol. res ; 49(8): e5304, 2016. graf
Article in English | LILACS | ID: lil-787383

ABSTRACT

A relationship between thyroid hormones and the cardiovascular system has been well established in the literature. The present in vitro study aimed to investigate the mechanisms involved in the vasodilator effect produced by the acute application of 10-8–10-4 M triiodothyronine (T3) to isolated rat aortic rings. Thoracic aortic rings from 80 adult male Wistar rats were isolated and mounted in tissue chambers filled with Krebs-Henseleit bicarbonate buffer in order to analyze the influence of endothelial tissue, inhibitors and blockers on the vascular effect produced by T3. T3 induced a vasorelaxant response in phenylephrine-precontracted rat aortic rings at higher concentrations (10-4.5–10-4.0 M). This outcome was unaffected by 3.1×10-7 M glibenclamide, 10-3 M 4-aminopyridine (4-AP), 10-5 M indomethacin, or 10-5 M cycloheximide. Contrarily, vasorelaxant responses to T3 were significantly (P<0.05) attenuated by endothelium removal or the application of 10-6 M atropine, 10-5 M L-NG-nitroarginine methyl ester (L-NAME), 10-7 M 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), 10-6 M (9S,10R,12R)-2,3,9,10,11,12-Hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3′,2′,1′-kl]pyrrolo[3,4-i](1,6)benzodiazocine-10-carboxylic acid, methyl ester KT 5823, 10-2 M tetraethylammonium (TEA), or 10-7 M apamin plus 10-7 M charybdotoxin. The results suggest the involvement of endothelial mechanisms in the vasodilator effect produced by the acute in vitro application of T3 to rat aortic rings. Possible mechanisms include the stimulation of muscarinic receptors, activation of the NO-cGMP-PKG pathway, and opening of Ca2+-activated K+ channels.


Subject(s)
Animals , Male , Aorta, Thoracic/drug effects , Triiodothyronine/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Endothelium, Vascular/drug effects , Phenylephrine/pharmacology , Atropine/pharmacology , Dimethyl Sulfoxide/pharmacology , Indomethacin/pharmacology , Glyburide/pharmacology , Rats, Wistar , NG-Nitroarginine Methyl Ester/pharmacology , Potassium Channels, Calcium-Activated/drug effects
17.
Cell Physiol Biochem ; 37(2): 747-58, 2015.
Article in English | MEDLINE | ID: mdl-26356265

ABSTRACT

BACKGROUND/AIMS: Vasoconstrictor-induced rhythmic contraction of arteries or veins has been observed both in vivo and in vitro. Many studies have reported that gap junctions, ryanodine receptors, Na+, K+-ATPase and other factors are involved in vasoconstrictor-induced rhythmic contraction in vascular smooth muscle. However, the mechanism is still not completely understood. METHODS: We used vessel tension measurements, intracellular recordings and intracellular Cl- concentration ([Cl-]i) measurements to investigate the mechanism underlying phenylephrine (PE)-induced rhythmic contraction in the mouse aorta. RESULTS: We found that Na+-K+-2Cl- cotransporter 1 (NKCC1) inhibitor bumetanide abolished PE-induced rhythmic contraction. The Cl- channel blockers DIDS and niflumic acid initially augmented the amplitude of PE-induced rhythmic contraction but later inhibited the rhythmic contraction. The large Ca2+-activated K+ channel blocker TEA and iberiotoxin increased the amplitude of PE-induced rhythmic contraction. The voltage-dependent Ca2+ channel blocker, nifedipine, and a Ca2+-free solution abolished PE-induced rhythmic contraction. The inhibitor of ryanodine receptors in the sarcoplasmic reticulum, ryanodine, inhibited PE-induced rhythmic contraction. Moreover, bumetanide hyperpolarized the membrane potential of vascular smooth muscle cells in a resting state or after PE pre-treatment. Bumetanide, niflumic acid, ryanodine, iberiotoxin, nifedipine and Ca2+-free buffer significantly suppressed the PE-induced [Cl-]i increase. CONCLUSION: These data indicate that NKCC1 is involved in the formation of PE-induced rhythmic contraction, and we also provide a method with which to indirectly observe the NKCC1 activity in isolated intact mouse thoracic aortas.


Subject(s)
Aorta, Thoracic/drug effects , Calcium Signaling/drug effects , Phenylephrine/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Vasoconstrictor Agents/pharmacology , Animals , Aorta, Thoracic/metabolism , Bumetanide/pharmacology , Female , Male , Membrane Potentials/drug effects , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Potassium Channels, Calcium-Activated/drug effects
18.
Adv Exp Med Biol ; 860: 61-7, 2015.
Article in English | MEDLINE | ID: mdl-26303468

ABSTRACT

Obesity related pathologies are the health care crisis of our generation. The fat cell derived adipokine leptin has been shown to be a central stimulant of respiration. Very high levels of leptin, however, are associated with the depressed ventilatory phenotype observed in obesity hypoventilation syndrome. Leptin receptors have been identified on carotid body type I cells but how their activation might influence the physiology of these cells is not known.The acute application of leptin evoked calcium signaling responses in isolated type I cells. Cells increased their Fura 2 ratio by 0.074 ± 0.010 ratio units (n = 39, P < 0.001). Leptin also increased the peak membrane currents in 6 of 9 cells increasing the peak macroscopic currents at +10 mV by 61 ± 14 % (p < 0.02). Leptin administered in the presence of the selective BK(Ca) channel inhibitor Paxilline (0.5 µM) failed to increase membrane currents (n = 5). Interestingly, leptin did not significantly alter the resting membrane potential of isolated type I cells (n = 9) and anoxic/acidic depolarizations were unaffected by leptin (n = 7, n = 6).These data suggest that leptin receptors are functional in type I cells but that their acute activation does not alter chemosensory properties. Future studies will use chronic models of leptin dysregulation.


Subject(s)
Calcium/metabolism , Carotid Body/drug effects , Leptin/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Animals , Carotid Body/cytology , Carotid Body/physiology , Potassium Channels, Calcium-Activated/physiology , Rats , Rats, Sprague-Dawley
19.
Neurochem Int ; 90: 28-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26163046

ABSTRACT

Large-conductance Ca(2+)-activated K(+) channels (BK channels) are widely expressed throughout the vertebrate nervous system, and are involved in the regulation of neurotransmitter release and neuronal excitability. Here, the neuroprotective effects of NS11021, a selective and chemically unrelated BK channel activator, and potential molecular mechanism involved have been studied in rat cortical neurons exposed to glutamate in vitro. Pretreatment with NS11021 significantly inhibited the loss of neuronal viability, LDH release and neuronal apoptosis in a dose-dependent manner. All these protective effects were fully antagonized by the BK-channel inhibitor paxilline. NS11021-induced neuroprotection was associated with reduced oxidative stress, as evidenced by decreased reactive oxygen species (ROS) generation, lipid peroxidation and preserved activity of antioxidant enzymes. Moreover, NS11021 significantly attenuated the glutamate-induced endoplasmic reticulum (ER) calcium release and activation of ER stress markers, including glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and caspase-12. Pretreatment with NS11021 also mitigated the mitochondrial membrane potential (MMP) collapse, cytochrome c release, and preserved mitochondrial Ca(2+) buffering capacity and ATP synthesis after glutamate exposure. Taken together, these results suggest that activation of BK channels via NS11021 protects cortical neurons against glutamate-induced excitatory damage, which may be dependent on the inhibition of ER stress and preservation of mitochondrial dysfunction.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Glutamic Acid/metabolism , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Potassium Channels, Calcium-Activated/metabolism , Tetrazoles/pharmacology , Thiourea/analogs & derivatives , Animals , Apoptosis/drug effects , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/physiology , Potassium Channels, Calcium-Activated/drug effects , Rats, Sprague-Dawley , Thiourea/pharmacology
20.
J Thorac Cardiovasc Surg ; 149(4): 1036-40, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25749140

ABSTRACT

OBJECTIVE: Beta-blockers are a heterogeneous class of agents that are used in the treatment of many cardiovascular diseases, especially hypertension and atherosclerosis, and that are commonly prescribed after cardiac surgery. In the present study, the aim is to investigate the vasorelaxant effects of some common beta-adrenoceptor blockers on the human radial artery in vitro, as well as their relaxation mechanisms. METHODS: Radial artery rings sourced from human patients were mounted in an organ bath and tested for changes in isometric tension in relaxation response to labetalol, nebivolol, and propranolol in the presence and absence of NG-nitro-L-arginine methyl ester (3 × 10(-5) mol/L) and tetraethyl ammonium (3 × 10(-4) mol/L). RESULTS: The labetalol (10(-8) to 10(-4) mol/L), nebivolol (10(-8) to 10(-4) mol/L), and propranolol (10(-8) to 10(-4) mol/L) induced concentration-dependent relaxations on the radial artery rings, which had been precontracted with phenylephrine (10(-6) mol/L). The relaxation response induced by labetalol in the isolated radial artery rings was significantly higher when compared with the nebivolol and propranolol samples (P < .05). NG-nitro-L-arginine methyl ester significantly reduced the relaxation of nebivolol (P < .05), and tetraethyl ammonium significantly reduced the relaxation of labetalol, nebivolol, and propranolol (P < .05). CONCLUSIONS: We speculated that the relaxant effect of labetalol, nebivolol, and propranolol was due partly to the Ca(2+)-activated K(+) channels. In addition, the relaxation induced by nebivolol was largely related with nitric oxide release. Nebivolol, and partly propranolol, may provide significant therapeutic benefit, but labetalol can be a good alternative for coronary artery bypass grafting with radial artery use.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Coronary Artery Bypass/methods , Labetalol/pharmacology , Nebivolol/pharmacology , Propranolol/pharmacology , Radial Artery/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/metabolism , Radial Artery/physiology , Radial Artery/surgery
SELECTION OF CITATIONS
SEARCH DETAIL