Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neuroscience ; 531: 12-23, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37661016

ABSTRACT

Fluvastatin (FLV), the first synthetically derived 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, is a potent inhibitor of cholesterol biosynthesis. While its primary mechanism of action is to reduce cholesterol levels, there is some evidence suggesting that it may also have effects on K+ channels. However, the overall effects of fluvastatin on ionic currents are not yet well understood. The whole-cell clamp recordings were applied to evaluate the ionic currents and action potentials of cells. Here, we have demonstrated that FLV can effectively inhibit the amplitude of erg-mediated K+ current (IK(erg)) in pituitary tumor (GH3) cells, with an IC50 of approximately 3.2 µM. In the presence of FLV, the midpoint in the activation curve of IK(erg) was distinctly shifted to a less negative potential by 10 mV, with minimal modification of the gating charge. However, the magnitude of hyperpolarization-activated cation current (Ih) elicited by long-lasting membrane hyperpolarization was progressively decreased, with an IC50 value of 8.7 µM, upon exposure to FLV. More interestingly, we also found that FLV (5 µM) could regulate the action potential and afterhyperpolarization properties in primary embryonic mouse cortical neurons. Our study presents compelling evidence indicating that FLV has the potential to impact both the amplitude and gating of the ion channels IK(erg) and Ih. We also provide credible evidence suggesting that this drug has the potential to modify the properties of action potentials and the afterhyperpolarization current in electrically excitable cells. However, the assumption that these findings translate to similar in-vivo results remains unclear.


Subject(s)
Neurons , Pituitary Gland , Mice , Animals , Fluvastatin , Neurons/physiology , Cations , Cholesterol
2.
Nutr Neurosci ; 22(12): 867-876, 2019 Dec.
Article in English | MEDLINE | ID: mdl-29642764

ABSTRACT

Objectives: The antiepileptic activity of resveratrol has been revealed in various experimental models of epilepsy. The present study evaluated the effects of resveratrol on the seizures and hyperexcitable neuronal activity associated with activation of N-methyl-d-aspartic acid (NMDA) receptor and inhibition of voltage-gated potassium channels.Methods: The effects of resveratrol on seizure thresholds, excitatory field potentials (EFPs) and action potentials induced by NMDA and 4-aminopyridine (4-AP) were monitored in mice, the mouse cortical slices and rat cortical neurons, respectively.Results: Resveratrol increased the NMDA-induced seizure thresholds and suppressed the frequency of NMDA/glycine-evoked EFPs and action potentials. However, resveratrol lowered the 4-AP-induced thresholds for myoclonic twitch and face and forelimb clonus, yet enhanced the thresholds for running and bouncing clonus and tonic hindlimb extension at the higher dose (50 mg/kg). A similar biphasic response of resveratrol was observed in the frequency of EFPs and action potential firings evoked by 4-AP, with enhancement at lower concentrations, but suppression at higher concentrations.Discussion: These findings suggest that resveratrol might be capable of protecting against the seizure types related to neuronal excitability and progression mediated by NMDA receptor activation, but not suitable for the seizures caused by disturbance of the voltage-dependent potassium channels.


Subject(s)
4-Aminopyridine/pharmacology , Cortical Excitability/drug effects , N-Methylaspartate/pharmacology , Resveratrol/administration & dosage , Seizures/drug therapy , Animals , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Evoked Potentials/drug effects , Female , Male , Mice , Mice, Inbred ICR , Neurons/drug effects , Neurons/physiology , Potassium Channel Blockers , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Seizures/chemically induced
3.
J Biomed Sci ; 23(1): 47, 2016 May 21.
Article in English | MEDLINE | ID: mdl-27209372

ABSTRACT

BACKGROUND: Resveratrol, a phytoalexin found in grapes and red wine, exhibits diverse pharmacological activities. However, relatively little is known about whether resveratrol modulates the ion channels in cortical neurons. The large-conductance calcium-activated potassium channels (BKCa) and voltage-gated sodium channels were expressed in cortical neurons and play important roles in regulation of neuronal excitability. The present study aimed to determine the effects of resveratrol on BKCa currents and voltage-gated sodium currents in cortical neurons. RESULTS: Resveratrol concentration-dependently increased the current amplitude and the opening activity of BKCa channels, but suppressed the amplitude of voltage-gated sodium currents. Similar to the BKCa channel opener NS1619, resveratrol decreased the firing rate of action potentials. In addition, the enhancing effects of BKCa channel blockers tetraethylammonium (TEA) and paxilline on action potential firing were sensitive to resveratrol. Our results indicated that the attenuation of action potential firing rate by resveratrol might be mediated through opening the BKCa channels and closing the voltage-gated sodium channels. CONCLUSIONS: As BKCa channels and sodium channels are critical molecular determinants for seizure generation, our findings suggest that regulation of these two channels in cortical neurons probably makes a considerable contribution to the antiseizure activity of resveratrol.


Subject(s)
Action Potentials/drug effects , Cerebral Cortex/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Stilbenes/pharmacology , Voltage-Gated Sodium Channels/metabolism , Animals , Benzimidazoles/pharmacology , Cell Line , Rats , Rats, Sprague-Dawley , Resveratrol
4.
Stem Cells Transl Med ; 3(6): 713-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24736401

ABSTRACT

Recent advances in somatic cell reprogramming have highlighted the plasticity of the somatic epigenome, particularly through demonstrations of direct lineage reprogramming of adult mouse and human fibroblasts to induced pluripotent stem cells (iPSCs) and induced neurons (iNs) under defined conditions. However, human cells appear to be less plastic and have a higher epigenetic hurdle for reprogramming to both iPSCs and iNs. Here, we show that SH2B adaptor protein 1ß (SH2B1) can enhance neurite outgrowth of iNs reprogrammed from human fibroblasts as early as day 14, when combined with miR124 and transcription factors BRN2 and MYT1L (IBM) under defined conditions. These SH2B1-enhanced iNs (S-IBM) showed canonical neuronal morphology, and expressed multiple neuronal markers, such as TuJ1, NeuN, and synapsin, and functional proteins for neurotransmitter release, such as GABA, vGluT2, and tyrosine hydroxylase. Importantly, SH2B1 accelerated mature process of functional neurons and exhibited action potentials as early as day 14; without SH2B1, the IBM iNs do not exhibit action potentials until day 21. Our data demonstrate that SH2B1 can enhance neurite outgrowth and accelerate the maturation of human iNs under defined conditions. This approach will facilitate the application of iNs in regenerative medicine and in vitro disease modeling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neurites/metabolism , Neurogenesis , Neurons/metabolism , Regenerative Medicine/methods , Action Potentials , Adaptor Proteins, Signal Transducing/genetics , Biomarkers/metabolism , Cell Shape , Cells, Cultured , Cellular Reprogramming , Genotype , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Phenotype , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
5.
Toxicol Lett ; 223(2): 139-45, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24012886

ABSTRACT

Methamphetamine (MA), a highly abused amphetamine-like psychostimulant, has surged in popularity worldwide in the last decade. Repeated MA exposure has been shown to affect the alternative splice variant expression of large conductance Ca(2+)-activated K(+) (BK) channels. It remains unclear whether MA affects BK channel activity. The present study investigated the effects of MA on BK channels in NG108-15 mouse neuroblastoma×rat glioma hybrid cells using whole-cell and cell-attached patch clamp techniques. In whole-cell recordings, the macroscopic K(+) outward currents were inhibited by MA with an EC50 of 146µM, but not affected by dopamine (DA). It implies that DA is not involved in the effects of MA on K(+) outward currents. In cell-attached patches, MA significantly decreased BK channel activity. Moreover, MA significantly decreased the BK channel opener NS1619-evoked whole-cell K(+) outward currents and BK channel activity. Finally, the effect of MA on membrane potential was examined by current-clamp configuration. MA caused membrane depolarization and application of NS1619 returned the depolarized potential to resting value. These findings suggest that MA might act as an inhibitor of BK channels, and thereby increase the neuronal excitability and enhance neurotransmitter release.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/drug effects , Methamphetamine/pharmacology , Amphetamine/pharmacology , Animals , Benzimidazoles/pharmacology , Calcium/metabolism , Cell Line, Tumor , Dopamine/pharmacology , Glyburide/pharmacology , Hybrid Cells , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Potentials/drug effects , Mice , Patch-Clamp Techniques , Potassium/metabolism , Rats
6.
J Nat Prod ; 74(4): 580-4, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21391660

ABSTRACT

Diosgenin (3ß-hydroxy-5-spirostene, 1), a plant-derived sapogenin, is used as a dietary supplement. However, the biological effects of 1 related to viral replication remain unexplored. In this study, the effects of 1 on hepatitis C virus (HCV) replication were evaluated. Based on a reporter-based HCV subgenomic replicon system, 1 was found to inhibit HCV replication at low micromolar concentrations. The EC(50) (concentration at which 50% of HCV replication is inhibited) of 1 was 3.8 µM. No cellular toxicity was observed at this concentration. Diosgenin (1) also significantly reduced the levels of viral RNA and viral proteins as evaluated by quantitative real-time reverse transcriptase PCR and Western blot analysis, respectively. In addition, in an alternative HCV antiviral system more closely aligned to all steps involved in the HCV infection and life cycle, 1 totally abolished HCV replication at 20 µM. Moreover, 1 reduced the phosphorylation of signal transducer and activator of transcription 3. A combination of 1 and interferon-α exerted an additive effect on the resultant anti-HCV activity.


Subject(s)
Antiviral Agents/pharmacology , Diosgenin/pharmacology , Hepacivirus/drug effects , Sapogenins/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Dietary Supplements/analysis , Diosgenin/chemistry , Diosgenin/isolation & purification , Hepacivirus/genetics , Humans , Molecular Structure , RNA, Viral/analysis , RNA, Viral/drug effects , Sapogenins/chemistry , Sapogenins/isolation & purification , Viral Nonstructural Proteins/drug effects
7.
Naunyn Schmiedebergs Arch Pharmacol ; 379(2): 127-35, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18818902

ABSTRACT

The hyperpolarization-activated cation current (I (h)) in rat pituitary lactotrophs (GH(3) cells) was characterized. Tramadol-induced block of this current was investigated. Effects of various related compounds on I (h) in GH(3) cells were also compared. Tramadol caused a time- and concentration-dependent reduction in the amplitude of I (h) with an IC(50) value of 13.6 microM. ZD7288 (30 microM), CsCl (2 mM), and propofol (30 microM) were effective in suppressing the amplitude of I (h). 2',5'-dideoxyadenosine (100 microM) suppressed I (h), while sp-cAMPS (100 microM) had no effect on it. Tramadol (10 microM) shifted the activation curve of I (h) to a more negative potential by approximately -20 mV, although no change in the slope factor was observed. Under current-clamp configuration, tramadol (10 microM) could reduce the firing frequency of action potentials. Intracellular Ca(2+) measurements revealed its ability to reduce spontaneous Ca(2+) oscillations in GH(3) cells. The results suggests that during cell exposure to tramadol used at clinically relevant concentration, the tramadol-mediated inhibition of I (h) could be direct and mediated via a non-opioid mechanism and would be one of the ionic mechanisms underlying reduced cell excitability.


Subject(s)
Analgesics, Opioid/pharmacology , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Lactotrophs/drug effects , Tramadol/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cations , Cell Line , Cesium/pharmacology , Chlorides/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dideoxyadenosine/analogs & derivatives , Dideoxyadenosine/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Lactotrophs/metabolism , Patch-Clamp Techniques , Potassium Channels/metabolism , Propofol/pharmacology , Pyrimidines/pharmacology , Rats , Thionucleotides/pharmacology
8.
Mol Pharmacol ; 74(6): 1696-704, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18809671

ABSTRACT

1,3-Bis-[2-hydroxy-5-(trifluoromethyl)phenyl]urea (NS1643) is reported to be an activator of human ether-à-go-go-related gene current. However, it remains unknown whether it has any effects on other types of ion channels. The effects of NS1643 on ion currents and membrane potential were investigated in this study. NS1643 stimulated Ca(2+)-activated K(+) current [I(K(Ca))] in a concentration-dependent manner with an EC(50) value of 1.8 microM in pituitary tumor (GH(3)) cells. In inside-out recordings, this compound applied to the intracellular side of the detached channels stimulated large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels with no change in single-channel conductance. It shifted the activation curve of BK(Ca) channels to less depolarized voltages without altering the gating charge of the channels. NS1643-stimulated channel activity depended on intracellular Ca(2+), and mean closed time during exposure to NS1643 was reduced. NS1643 (3 microM) had little or no effect on peak amplitude of ether-à-go-go-related gene-mediated K(+) current evoked by membrane hyperpolarization, although it increased the amplitude of late-sustained components of K(+) inward current, which was suppressed by paxilline but not by azimilide. NS1643 (3 microM) had no effect on L-type Ca(2+) current. This compound reduced repetitive firing of action potentials, and further application of paxilline attenuated its decrease in firing rate. In addition, NS1643 enhanced BK(Ca)-channel activity in human embryonic kidney 293T cells expressing alpha-hSlo. In summary, we clearly show that NS1643 interacts directly with the BK(Ca) channel to increase the amplitude of I(K(Ca)) in pituitary tumor (GH(3)) cells. The alpha-subunit of the channel may be a target for the action of this small compound.


Subject(s)
Cresols/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/agonists , Phenylurea Compounds/pharmacology , Action Potentials , Animals , Calcium/metabolism , Cell Line, Tumor , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/agonists , Humans , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Patch-Clamp Techniques , Pituitary Neoplasms , Rats
9.
Toxicol Sci ; 106(2): 454-63, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18779382

ABSTRACT

Aconitine (ACO), a highly toxic diterpenoid alkaloid, is recognized to have effects on cardiac voltage-gated Na(+) channels. However, it remains unknown whether it has any effects on K(+) currents. The effects of ACO on ion currents in differentiated clonal cardiac (H9c2) cells and in cultured neonatal rat ventricular myocytes were investigated in this study. In H9c2 cells, ACO suppressed ultrarapid-delayed rectifier K(+) current (I(Kur)) in a time- and concentration-dependent fashion. The IC(50) value for ACO-induced inhibition of I(Kur) was 1.4 microM. ACO could accelerate the inactivation of I(Kur) with no change in the activation time constant of this current. Steady-state inactivation curve of I(Kur) during exposure to ACO could be demonstrated. Recovery from block by ACO was fitted by a single-exponential function. The inhibition of I(Kur) by ACO could still be observed in H9c2 cells preincubated with ruthenium red (30 microM). Intracellular dialysis with ACO (30 microM) had no effects on I(Kur). I(Kur) elicited by simulated action potential (AP) waveforms was sensitive to block by ACO. Single-cell Ca(2+) imaging revealed that ACO (10 microM) alone did not affect intracellular Ca(2+) in H9c2 cells. In cultured neonatal rat ventricular myocytes, ACO also blocked I(Kur) and prolonged AP along with appearance of early afterdepolarizations. Multielectrode recordings on neonatal rat ventricular tissues also suggested that ACO-induced electrocardiographic changes could be associated with inhibition of I(Kur). This study provides the evidence that ACO can produce a depressant action on I(Kur) in cardiac myocytes.


Subject(s)
Aconitine/toxicity , Heart Ventricles/drug effects , Potassium Channel Blockers/toxicity , Potassium Channels/drug effects , Animals , Animals, Newborn , Cell Differentiation , Cell Line , Electrodes , Heart Ventricles/cytology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Ruthenium Red/pharmacology
10.
Eur J Pharmacol ; 590(1-3): 93-8, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18582866

ABSTRACT

General anesthetic propofol (2,6-bis(isopropyl)-phenol) possess a chemical structure unrelated to other anesthetic drugs. It has been known to block a variety of ion currents. This study is designed to determine the effect of this drug on ion currents in differentiated H9c2 cardiac cells. The effects of propofol, an intravenous anesthetic agent with a distinct chemical structure, on ion currents of differentiated clonal cardiac (H9c2) cells were investigated in this study. Propofol (10-300 microM) suppressed the amplitude of delayed rectifier K(+) current (I(K(DR))) in a concentration-dependent manner with an IC(50) value of 36 microM. This compound reduced activation time constant and increased current inactivation, although no voltage dependency of propofol-induced block of I(K(DR)) can demonstrated. Neither diazoxide, pinacidil, nor caffeic acid phenethyl ester had any effect on propofol-induced block of I(K(DR)). Propofol (30 microM) had no effect on erg-mediated K(+) current in these cells; however, it suppressed L-type Ca(2+) current (I(Ca,L)) of cardiac and skeletal types to a similar extent. Intracellular dialysis with propofol (100 microM) had no effects on I(K(DR)) or I(Ca,L). Numerical simulations of I(K(DR)) based on a Markovian model reproduce the experimental results and show that propofol-induced blockade of I(K(DR)) is associated with an decrease in forward rate of the activation process and an increase in transitional rate into the inactivated state. Propofol can suppress I(K(DR)) in differentiated H9c2 cardiac cells in a concentration- and state-dependent manner. These effects can significantly contribute its action on functional activity of heart cells.


Subject(s)
Anesthetics, Intravenous/pharmacology , Calcium Channels, L-Type/drug effects , Delayed Rectifier Potassium Channels/antagonists & inhibitors , Heart/drug effects , Potassium Channel Blockers/pharmacology , Propofol/pharmacology , Action Potentials/drug effects , Animals , Apamin/pharmacology , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Glyburide/pharmacology , Heart/physiology , Rats
11.
Neuropharmacology ; 54(6): 912-23, 2008 May.
Article in English | MEDLINE | ID: mdl-18336846

ABSTRACT

The effects of aconitine (ACO), a highly toxic alkaloid, on ion currents in differentiated NG108-15 neuronal cells were investigated in this study. ACO (0.3-30 microM) suppressed the amplitude of delayed rectifier K+ current (I K(DR)) in a concentration-dependent manner with an IC50 value of 3.1 microM. The presence of ACO enhanced the rate and extent of I K(DR) inactivation, although it had no effect on the initial activation phase of I K(DR). It could shift the inactivation curve of I K(DR) to a hyperpolarized potential with no change in the slope factor. Cumulative inactivation for I K(DR) was also enhanced by ACO. Orphenadrine (30 microM) or methyllycaconitine (30 microM) slightly suppressed I K(DR) without modifying current decay. ACO (10 microM) had an inhibitory effect on voltage-dependent Na+ current (I Na). Under current-clamp recordings, ACO increased the firing and widening of action potentials in these cells. With the aid of the minimal binding scheme, the ACO actions on I K(DR) was quantitatively provided with a dissociation constant of 0.6 microM. A modeled cell was designed to duplicate its inhibitory effect on spontaneous pacemaking. ACO also blocked I K(DR) in neuroblastoma SH-SY5Y cells. Taken together, the experimental data and simulations show that ACO can block delayed rectifier K+ channels of neurons in a concentration- and state-dependent manner. Changes in action potentials induced by ACO in neurons in vivo can be explained mainly by its blocking actions on I K(DR) and I Na.


Subject(s)
Aconitine/pharmacology , Delayed Rectifier Potassium Channels/drug effects , Neurons/metabolism , Potassium Channel Blockers , Action Potentials/drug effects , Algorithms , Cell Differentiation/drug effects , Cell Line , Computer Simulation , Data Interpretation, Statistical , Electrophysiology , Humans , Kinetics , Neurons/drug effects , Patch-Clamp Techniques
12.
Life Sci ; 82(1-2): 11-20, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18068197

ABSTRACT

Riluzole is known to be of therapeutic use in the management of amyotrophic lateral sclerosis. In this study, we investigated the effects of riluzole on ion currents in cultured differentiated human skeletal muscle cells (dHSkMCs). Western blotting revealed the protein expression of alpha-subunits for both large-conductance Ca2+-activated K+ (BK(Ca)) channel and Na+ channel (Na(v)1.5) in these cells. Riluzole could reduce the frequency of spontaneous beating in dHSkMCs. In whole-cell configuration, riluzole suppressed voltage-gated Na+ current (I(Na)) in a concentration-dependent manner with an IC50 value of 2.3 microM. Riluzole (10 microM) also effectively increased Ca2+-activated K+ current (I(K(Ca))) which could be reversed by iberiotoxin (200 nM) and paxilline (1 microM), but not by apamin (200 nM). In inside-out patches, when applied to the inside of the cell membrane, riluzole (10 microM) increased BK(Ca)-channel activity with a decrease in mean closed time. Simulation studies also unraveled that both decreased conductance of I(Na) and increased conductance of I(K(Ca)) utilized to mimic riluzole actions in skeletal muscle cells could combine to decrease the amplitude of action potentials and increase the repolarization of action potentials. Taken together, inhibition of I(Na) and stimulation of BK(Ca)-channel activity caused by this drug are partly, if not entirely, responsible for its muscle relaxant actions in clinical setting.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Muscle, Skeletal/drug effects , Riluzole/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Action Potentials/drug effects , Cells, Cultured , Computer Simulation , Dose-Response Relationship, Drug , Electrophysiology , Humans , Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/biosynthesis , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , NAV1.5 Voltage-Gated Sodium Channel , Peptides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/biosynthesis
13.
Pflugers Arch ; 455(4): 687-99, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17701422

ABSTRACT

Di-8-ANEPPS (4-{2-[6-(dibutylamino)-2-naphthalenyl]-ethenyl}-1-(3-sulfopropyl)pyridinium inner salt) has been used as a fast-response voltage-sensitive styrylpyridinium probe. However, little is known regarding the mechanism of di-8-ANEPPS actions on ion currents. In this study, the effects of this dye on ion currents were investigated in pituitary GH(3) cells. In whole-cell configuration, di-8-ANEPPS (10 microM) reversibly increased the amplitude of Ca(2+)-activated K(+) current. In inside-out configuration, di-8-ANEPPS (10 microM) applied to the intracellular surface of the membrane caused no change in single-channel conductance; however, it did enhance the activity of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels with an EC(50) value of 7.5 microM. This compound caused a left shift in the activation curve of BK(Ca) channels with no change in the gating charge of these channels. A decrease in mean closed time of the channels was seen in the presence of this dye. In the cell-attached mode, di-8-ANEPPS applied on the extracellular side of the membrane also activated BK(Ca) channels. However, neither voltage-gated K(+) nor ether-à-go-go-related gene (erg)-mediated K(+) currents in GH(3) cells were affected by di-8-APPNES. Under current-clamp configuration, di-8-ANEPPS (10 microM) decreased the firing of action potentials in GH(3) cells. In pancreatic betaTC-6 cells, di-8-APPNES (10 microM) also increased BK(Ca)-channel activity. Taken together, this study suggests that during the exposure to di-8-ANEPPS, the stimulatory effects on BK(Ca) channels could be one of potential mechanisms through which it may affect cell excitability.


Subject(s)
Fluorescent Dyes/pharmacology , Insulinoma/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/agonists , Pituitary Neoplasms/metabolism , Potassium Channels/agonists , Potassium/metabolism , Pyridinium Compounds/pharmacology , Action Potentials , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/metabolism , Ion Channel Gating/drug effects , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Potentials , Mice , Patch-Clamp Techniques , Potassium Channels/metabolism , Potassium Channels, Voltage-Gated/metabolism , Rats
14.
Biochem Pharmacol ; 73(9): 1347-57, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17234159

ABSTRACT

The agonists selective for estrogen receptor (ER)-alpha (4,4',4''-(4-propyl-[(1)H]-pyrazole-1,3,5-triyl) tris-phenol, PPT) and ER-beta (2,3-bis(4-hydroxyphenyl)-propionitrile, DPN) are known to stimulate ER-alpha and ER-beta receptors, respectively. It remains unknown whether these two agents regulate the activity of ion channels via a direct stimulation. In this study, we tested the hypothesis that DPN or PPT stimulates the large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels in cultured human cardiac fibroblasts (HCFs). In whole-cell configuration, depolarizing pulses evoked K(+) outward currents in an outward rectification in HCFs, the amplitude of which was increased in the presence of DPN or PPT. In inside-out patches, the activity of BK(Ca)-channel with a conductance of 167+/-8 pS was observed in these cells. PPT or DPN applied to the intracellular face of the membrane enhanced the activity of BK(Ca) channels with no change in single-channel conductance. DPN and PPT increased BK(Ca)-channel activity with an EC(50) value of 2.3 and 2.6 microM, respectively. The mean closed time of these channels during the exposure to these compounds was reduced with no change in the gating charge of the channels. Intracellular Ca(2+) was not altered by these two compounds. RT-PCR analysis revealed that no change in the transcriptional level of the BK(Ca)-channel alpha-subunit was observed in chronic treatment with these two compounds. PPT- and DPN-stimulated increase in BK(Ca) channels reveal novel pharmacological properties attributable to the activity of these channels, and their increase in BK(Ca) channels activity in HCFs may contribute to cell function.


Subject(s)
Estradiol Congeners/pharmacology , Fibroblasts/drug effects , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Myocardium/cytology , Nitriles/pharmacology , Receptors, Estrogen/agonists , Calcium/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Fibroblasts/metabolism , Fulvestrant , Heart , Humans , RNA, Messenger , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/pharmacology , Time Factors
15.
J Cell Physiol ; 210(3): 655-66, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17133362

ABSTRACT

The effects of rottlerin, a known inhibitor of protein kinase C-delta activation, on ion currents were investigated in pituitary tumor (GH3) cells. Rottlerin (0.3-100 microM) increased the amplitude of Ca2+-activated K+ current (I K(Ca)) in a concentration-dependent manner with an EC50 value of 1.7 microM. In intracellular perfusion with rottlerin (1 microM) or staurosporine (10 microM), phorbol 12-myristate 13-acetate-induced inhibition of I K(Ca) in these cells was abolished. In cell-attached mode, rottlerin applied on the extracellular side of the membrane caused activation of large-conductance Ca2+-activated K+ (BK(Ca)) channels, and a further application of BAPTA-AM (10 microM) to the bath had no effect on rottlerin-stimulated channel activity. When cells were exposed to rottlerin, the activation curve of these channels was shifted to less positive potential with no change in the slope factor. Rottlerin increased BK(Ca)-channel activity in outside-out patches. Its change in kinetic behavior of BK(Ca) channels is primarily due to an increase in mean open time. With the aid of minimal kinetic scheme, a quantitative description of rottlerin stimulation on BK(Ca) channels in GH3 cells was also provided. Under current-clamp configuration, rottlerin (1 microM) decreased the firing of action potentials. I K(Ca) elicited by simulated action potential waveforms was enhanced by this compound. In human cortical HCN-1A cells, rottlerin (1 microM) could also interact with the BK(Ca) channel to stimulate I K(Ca). Therefore, rottlerin may directly activate BK(Ca) channels in neurons or endocrine cells.


Subject(s)
Acetophenones/pharmacology , Adenoma/metabolism , Benzopyrans/pharmacology , Enzyme Inhibitors/pharmacology , Neurons/metabolism , Pituitary Neoplasms/metabolism , Potassium Channels, Calcium-Activated/drug effects , Protein Kinase C-delta/antagonists & inhibitors , Action Potentials/drug effects , Adenoma/pathology , Animals , Carcinogens/pharmacology , Cell Line , Cell Line, Tumor , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Neurons/cytology , Pituitary Neoplasms/pathology , Potassium Channels/drug effects , Potassium Channels/metabolism , Potassium Channels, Calcium-Activated/metabolism , Protein Kinase C-delta/physiology , Rats , Staurosporine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
16.
Planta Med ; 72(5): 430-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16557457

ABSTRACT

The effects of diosgenin (3beta-hydroxy-5-spirostene), a plant-derived sapogenin, on ion currents in human cortical neurons (HCN-1A) were investigated. In the whole-cell configuration, diosgenin (0. -30 microM) increased the amplitude of K+ outward current (I(K)). Diosgenin-stimulated I(K) was sensitive to inhibition by paxilline (1 microM), but not by apamin (200 nM) or glibenclamide (10 muM). In the cell-attached configuration, diosgenin applied to the bath increased the activity of large-conductance Ca2+-activated K+ (BK(Ca)) channels without altering single-channel conductance. Diosgenin enhanced BK(Ca)-channel activity with an EC50 value of 25 microM. However, in inside-out patches, diosgenin applied to the intracellular surface had no effect on BK(Ca)-channel activity, while cilostazol or caffeic acid phenethyl ester increased it. As shown with the aid of intracellular Ca2+ measurements, diosgenin elevated intracellular Ca2+ in HCN-1A cells. Western blotting also revealed the presence of the alpha-subunit of BK (Ca) channels in these cells. The sustained stimulation of I(K) arises primarily from the diosgenin-induced Ca2+ influx across the cell membrane. The effect of diosgenin on these channels may affect the functional activity of cortical neurons. Abbreviations. I(K):K+ outward current I(K(Ca)):Ca2+-activated K+ current BK(Ca) channel:Large-conductance Ca2+-activated K+ channel CAPE:caffeic acid phenethyl ester [Ca2+] (i):intracellular Ca2+ concentration I/V relationship:current/voltage relationship K (ATP) channel:ATP-sensitive K+ channel K(Ca) channel:Ca2+-activated K+ channel.


Subject(s)
Calcium Channel Blockers/pharmacology , Dioscorea , Diosgenin/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Phytotherapy , Calcium Channel Blockers/administration & dosage , Cells, Cultured/drug effects , Diosgenin/administration & dosage , Dose-Response Relationship, Drug , Humans , Neurons/drug effects , Sapogenins/administration & dosage , Sapogenins/pharmacology
17.
J Membr Biol ; 213(3): 175-85, 2006.
Article in English | MEDLINE | ID: mdl-17483867

ABSTRACT

Cardiac fibroblasts are involved in the maintenance of myocardial tissue structure. However, little is known about ion currents in human cardiac fibroblasts. It has been recently reported that cardiac fibroblasts can interact electrically with cardiomyocytes through gap junctions. Ca(2+)-activated K(+) currents (I (K[Ca])) of cultured human cardiac fibroblasts were characterized in this study. In whole-cell configuration, depolarizing pulses evoked I (K(Ca)) in an outward rectification in these cells, the amplitude of which was suppressed by paxilline (1 microM: ) or iberiotoxin (200 nM: ). A large-conductance, Ca(2+)-activated K(+) (BK(Ca)) channel with single-channel conductance of 162 +/- 8 pS was also observed in human cardiac fibroblasts. Western blot analysis revealed the presence of alpha-subunit of BK(Ca) channels. The dynamic Luo-Rudy model was applied to predict cell behavior during direct electrical coupling of cardiomyocytes and cardiac fibroblasts. In the simulation, electrically coupled cardiac fibroblasts also exhibited action potential; however, they were electrically inert with no gap-junctional coupling. The simulation predicts that changes in gap junction coupling conductance can influence the configuration of cardiac action potential and cardiomyocyte excitability. I (k(Ca)) can be elicited by simulated action potential waveforms of cardiac fibroblasts when they are electrically coupled to cardiomyocytes. This study demonstrates that a BK(Ca) channel is functionally expressed in human cardiac fibroblasts. The activity of these BK(Ca) channels present in human cardiac fibroblasts may contribute to the functional activities of heart cells through transfer of electrical signals between these two cell types.


Subject(s)
Fibroblasts/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Myocardium/cytology , Myocytes, Cardiac/physiology , Action Potentials , Cells, Cultured , Fibroblasts/metabolism , Humans , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Models, Theoretical , Myoblasts, Cardiac/physiology , Myocardium/metabolism , Patch-Clamp Techniques , Paxillin/pharmacology , Peptides/pharmacology , Potassium Channel Blockers/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...