Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Type of study
Publication year range
1.
Neuroreport ; 32(16): 1299-1306, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34605450

ABSTRACT

OBJECTIVES: Iloperidone is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. hERG 3.1, alternatively spliced form of hERG 1A, is considered a potential target for an antipsychotic drug. The present study was designed to study the effects of iloperidone on hERG 1A/3.1 heterotetrameric channels. METHODS: The interactions of iloperidone with hERG 1A/3.1 heterotetrameric channels stably expressed in HEK cells were investigated using the whole-cell patch-clamp technique and western blot analysis. RESULTS: Iloperidone inhibited the hERG 1A/3.1 tail currents at -50 mV in a concentration-dependent manner with an IC50 value of 0.44 µM. The block of hERG 1A/3.1 currents by iloperidone was voltage-dependent and increased over a range of voltage for channel activation. However, the block by iloperidone was voltage-independent at more depolarized potentials where the channels were fully activated. A fast application of iloperidone inhibited the hERG 1A/3.1 current elicited by a 5-s depolarizing pulse to +60 mV to fully inactivate the hERG 1A/3.1 currents. Iloperidone also induced a rapid and reversible inhibition of hERG 1A/3.1 tail currents during repolarization. However, iloperidone had no effect on either hERG 1A or hERG 1A/3.1 channel trafficking to the cell membrane. CONCLUSIONS: Our results indicated that iloperidone concentration-dependently inhibited hERG 1A/3.1 currents by preferentially interacting with the open states of channels, but not by the disruption of membrane trafficking or surface membrane expression of hERG 1A and hERG 1A/3.1 channel proteins.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Isoxazoles/pharmacology , Piperidines/pharmacology , Potassium Channel Blockers/pharmacology , Biotinylation/drug effects , HEK293 Cells , Humans
2.
Nanoscale ; 13(11): 5868-5874, 2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33724290

ABSTRACT

We prepared a poly(pyrrole : biphenyldisulfonic acid : pyrrole (Py:BPDSA:Py)) nanocomposite of molybdenum disulfide (MoS2), P(Py:BPDSA:Py)-MoS2, with high crystallinity. The composite is synthesized by oxidative polymerization of Py:BPDSA:Py as a two-monomer-connected precursor (TMCP) linked by ionic bonding on a molybdenum disulfide (MoS2) monolayer. The chemical, structural and morphological characterization of this composite is confirmed by Raman spectroscopy, FT-IR, X-ray photoelectron spectroscopy (XPS), electron energy loss spectroscopy (EELS), and scanning electron microscopy (SEM). The crystal structure is analysed by X-ray diffraction (XRD) and high-voltage electron microscopy (HVEM), which shows a face-centered cubic (FCC) crystal structure for the composite. Nitrogen adsorption-desorption isotherms show an improved specific surface area (91.3 m2 g-1). The electrochemical properties of the composite with a unique crystal structure and a large specific surface area are analysed through cyclic voltammetry (CV), which shows a specific capacitance of 681 F g-1 demonstrating that the composite can be used as an efficient electrode active material for electrochemical energy storage systems.

3.
Korean J Physiol Pharmacol ; 24(6): 545-553, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33093275

ABSTRACT

Aripiprazole is a quinolinone derivative approved as an atypical antipsychotic drug for the treatment of schizophrenia and bipolar disorder. It acts as with partial agonist activities at the dopamine D2 receptors. Although it is known to be relatively safe for patients with cardiac ailments, less is known about the effect of aripiprazole on voltage-gated ion channels such as transient A-type K+ channels, which are important for the repolarization of cardiac and neuronal action potentials. Here, we investigated the effects of aripiprazole on Kv1.4 currents expressed in HEK293 cells using a whole-cell patch-clamp technique. Aripiprazole blocked Kv1.4 channels in a concentration-dependent manner with an IC50 value of 4.4 µM and a Hill coefficient of 2.5. Aripiprazole also accelerated the activation (time-to-peak) and inactivation kinetics. Aripiprazole induced a voltage-dependent (δ = 0.17) inhibition, which was use-dependent with successive pulses on Kv1.4 currents without altering the time course of recovery from inactivation. Dehydroaripiprazole, an active metabolite of aripiprazole, inhibited Kv1.4 with an IC50 value of 6.3 µM (p < 0.05 compared with aripiprazole) with a Hill coefficient of 2.0. Furthermore, aripiprazole inhibited Kv4.3 currents to a similar extent in a concentration-dependent manner with an IC50 value of 4.9 µM and a Hill coefficient of 2.3. Thus, our results indicate that aripiprazole blocked Kv1.4 by preferentially binding to the open state of the channels.

4.
Nanoscale ; 11(33): 15641-15646, 2019 Sep 07.
Article in English | MEDLINE | ID: mdl-31408081

ABSTRACT

Synthesis of a two-dimensional (2D) highly crystalline composite, P(Py:BPDSA:Py)-GO, from the growth of a close-packed polymer crystal, P(Py:BPDSA:Py), on graphene oxide (GO) sheets via in situ polymerization of two-monomer-connected precursors (TMCPs, Py:BPDSA:Py), in which two pyrrole (Py) molecules are linked through a connector (4,4'-biphenyldisulfonic acid) (BPDSA), is reported. When the TMCP is polymerized on GO, it leads to an exceptionally ordered structure determined by X-ray diffraction (XRD) and high-resolution transmission electron microscopy (HRTEM) studies. X-ray crystallography of the composite shows crystalline peaks with d spacings in the [100] direction. Transmission electron microscopy (TEM) analysis indicates that the composite has a face-centered cubic (FCC) crystal structure. Raman spectroscopy, Fourier transform infrared spectroscopy (FT-IR), and X-ray photoelectron spectroscopy (XPS) show that this composite with a well-defined nanostructure was successfully synthesized. Nitrogen adsorption-desorption isotherms show that this composite, P(Py:BPDSA:Py)-GO, has an improved specific surface area (71 m2 g-1) compared to that of P(Py:BPDSA:Py) (3.1 m2 g-1). The electrochemical properties of the composite studied by cyclic voltammetry indicates a specific capacitance of 480 F g-1 without an additional conducting material such as carbon black, suggesting its use as a pseudocapacitor.

5.
Eur J Pharmacol ; 854: 92-100, 2019 Jul 05.
Article in English | MEDLINE | ID: mdl-30954564

ABSTRACT

Cariprazine is a novel atypical antipsychotic drug that is widely used for the treatment of schizophrenia and bipolar mania/mixed disorder. We used the whole-cell patch-clamp technique to investigate the effects of cariprazine on hERG channels that are stably expressed in HEK cells. Cariprazine inhibited the hERG 1A and hERG 1A/3.1 tail currents at -50 mV in a concentration-dependent manner with IC50 values of 4.1 and 12.2 µM, respectively. The block of hERG 1A currents by cariprazine was voltage-dependent, and increased over a range of voltage for channel activation. Cariprazine shifted the steady-state inactivation curve of the hERG 1A currents in a hyperpolarizing direction and produced a use-dependent block. A fast application of cariprazine inhibited the hERG 1A currents elicited by a 5 s depolarizing pulse to +60 mV to fully inactivate the hERG 1A currents. During a repolarizing pulse wherein the hERG 1A current was deactivated slowly, cariprazine rapidly and reversibly blocked the open state of the hERG 1A current. However, cariprazine did not affect hERG 1A and hERG 1A/3.1 channel trafficking to the cell membrane. Our results indicated that cariprazine concentration-dependently inhibited hERG 1A and hERG 1A/3.1 currents by preferentially interacting with the open states of the hERG 1A channel, but not by the disruption of hERG 1A and hERG 1A/3.1 channel protein trafficking. Our study examined cariprazine's mechanism of action provides a biophysical profile that is necessary to assess the potential therapeutic effects of this drug.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Piperazines/pharmacology , Potassium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Protein Multimerization/drug effects , Protein Structure, Quaternary
6.
Neurosci Lett ; 664: 66-73, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29133173

ABSTRACT

Quetiapine is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. It is mainly metabolized by a cytochrome P450 system in the liver. Norquetiapine is a major active metabolite in humans with a pharmacological profile that differs distinctly from that of quetiapine. We used the whole-cell patch-clamp technique to investigate the effects of norquetiapine on hERG channels that are stably expressed in HEK cells. Quetiapine and norquetiapine inhibited the hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 8.3 and 10.8µM, respectively, which suggested equal potency. The block of hERG currents by norquetiapine was voltage-dependent with a steep increase over a range of voltages for channel activation. However, at more depolarized potentials where the channels were fully activated, the block by norquetiapine was voltage-independent. The steady-state inactivation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of norquetiapine. Norquetiapine did not produce a use-dependent block. A fast application of norquetiapine inhibited the hERG current elicited by a 5s depolarizing pulse to +60mV, which fully inactivated the hERG currents, suggesting an inactivated-state block. During a repolarizing pulse wherein the hERG current was slowly deactivated, albeit remaining in an open state, a fast application of norquetiapine rapidly and reversibly inhibited the open state of the hERG current. Our results indicated that quetiapine and norquetiapine had equal potency in inhibiting hERG tail currents. Norquetiapine inhibited the hERG current by preferentially interacting with the open and/or inactivated states of the channels.


Subject(s)
Cloning, Molecular , Dibenzothiazepines/pharmacology , ERG1 Potassium Channel/antagonists & inhibitors , ERG1 Potassium Channel/physiology , Quetiapine Fumarate/pharmacology , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Dibenzothiazepines/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Quetiapine Fumarate/metabolism
7.
Nanoscale ; 9(19): 6545-6550, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28470300

ABSTRACT

Considerable research approaches have focused on improving the crystallinity of conducting polymers to enhance the electrical conductivity. However, it is difficult to control the arrangement of polymer chains without the use of expensive and complex methods because of the intrinsic morphology of polymers. Herein, we report a one-step in situ process to produce controlled molecular-scale ordered polyaniline (PANI) films by coordination crosslinking with Zn ions using solvent-vapor thermal annealing (SVTA). The resulting PANI film crosslinked by Zn coordination has a face-centered cubic structure at the molecular scale, which was confirmed by high-voltage electron microscopy. The in situ coordination crosslinking produced a new class of molecular ordering in the PANI films and drastically enhanced their conductivity, showing their potential for use in various electronic and energy devices.

8.
Naunyn Schmiedebergs Arch Pharmacol ; 390(6): 633-642, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28265686

ABSTRACT

Risperidone and one of its active metabolites, paliperidone, are widely used for the treatment of schizophrenia. We used a patch-clamp study to investigate the effects of paliperidone on hERG potassium channels expressed in HEK cells. Western blot analyses were used to study the effects of risperidone and paliperidone on hERG and hERG 3.1 isoform channel trafficking. Risperidone and paliperidone inhibited the hERG tail currents in a concentration-dependent manner with IC50 values of 0.16 and 0.57 µM, respectively. The block of hERG currents by paliperidone was voltage-dependent, increasing over a range of voltages for channel activation. A fast application of paliperidone inhibited the hERG current elicited by a 5-s depolarizing pulse to +60 mV to fully inactivate the hERG currents, suggesting an inactivated state block. A fast application of paliperidone during repolarization reversibly inhibited the hERG tail currents in a concentration-dependent manner with a IC50 value of 1.26 µM. Kinetic analysis of paliperidone interaction with the open state of the hERG channels showed that the rate constants of association (k +1) and dissociation (k -1) for paliperidone were 0.45 µM-1 s-1 and 1.07 s-1, respectively. Paliperidone shifted the steady-state inactivation curve of the hERG currents in a hyperpolarizing direction and also produced a use-dependent block. Risperidone and paliperidone had no effect on hERG and hERG 3.1 channel trafficking to the cell membrane. Our results indicated that paliperidone inhibited the hERG current by preferentially interacting with the open and inactivated states of the channel, but not by disruption of hERG channel protein trafficking.


Subject(s)
Antipsychotic Agents/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Paliperidone Palmitate/pharmacology , Risperidone/pharmacology , Antipsychotic Agents/administration & dosage , Cell Membrane/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inhibitory Concentration 50 , Paliperidone Palmitate/administration & dosage , Patch-Clamp Techniques , Risperidone/administration & dosage
9.
Korean J Physiol Pharmacol ; 21(1): 75-82, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28066143

ABSTRACT

The effects of acepromazine on human ether-à-go-go-related gene (hERG) potassium channels were investigated using whole-cell voltage-clamp technique in human embryonic kidney (HEK293) cells transfected with hERG. The hERG currents were recorded with or without acepromazine, and the steady-state and peak tail currents were analyzed for the evaluating the drug effects. Acepromazine inhibited the hERG currents in a concentration-dependent manner with an IC50 value of 1.5 µM and Hill coefficient of 1.1. Acepromazine blocked hERG currents in a voltage-dependent manner between -40 and +10 mV. Before and after application of acepromazine, the half activation potentials of hERG currents changed to hyperpolarizing direction. Acepromazine blocked both the steady-state hERG currents by depolarizing pulse and the peak tail currents by repolarizing pulse; however, the extent of blocking by acepromazine in the repolarizing pulse was more profound than that in the depolarizing pulse, indicating that acepromazine has a high affinity for the open state of the channels, with a relatively lower affinity for the closed state of hERG channels. A fast application of acepromazine during the tail currents inhibited the open state of hERG channels in a concentration-dependent. The steady-state inactivation of hERG currents shifted to the hyperpolarized direction by acepromazine. These results suggest that acepromazine inhibits the hERG channels probably by an open- and inactivated-channel blocking mechanism. Regarding to the fact that the hERG channels are the potential target of drug-induced long QT syndrome, our results suggest that acepromazine can possibly induce a cardiac arrhythmia through the inhibition of hERG channels.

10.
Oncotarget ; 7(42): 67857-67867, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27708218

ABSTRACT

Cerebral edema from the disruption of the blood-brain barrier (BBB) after cerebral ischemia is a major cause of morbidity and mortality as well as a common event in patients with stroke. Caveolins (Cavs) are thought to regulate BBB functions. Here, we report for the first time that Cav-1 overexpression (OE) decreased brain edema from BBB disruption following ischemic insult. Edema volumes and Cav-1 expression levels were measured following photothrombosis and middle cerebral artery occlusion (MCAO). Endothelial cells that were transduced with a Cav-1 lentiviral expression vector were transplanted into rats. BBB permeability was quantified with Evans blue extravasation. Edema volume was determined from measures of the extravasation area, brain water content, and average fluorescence intensity after Cy5.5 injections. Tight junction (TJ) protein expression was measured with immunoblotting. Cav-1 expression levels and vasogenic brain edema correlated strongly after ischemic insult. Cav-1 expression and BBB disruption peaked 3 d after the MCAO. In addition, intravenous administration of endothelial cells expressing Cav-1 effectively increased the Cav-1 levels 3 d after the MCAO ischemic insult. Importantly, Cav-1 OE ameliorated the vasogenic edema by inhibiting the degradation of TJ protein expression in the acute phase of ischemic stroke. These results suggested that Cav-1 OE protected the integrity of the BBB mainly by preventing the degradation of TJ proteins in rats. These findings need to be confirmed in a clinical setting in human subjects.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Caveolin 1/metabolism , Tight Junctions/metabolism , Animals , Brain/blood supply , Brain/metabolism , Brain Edema/etiology , Brain Ischemia/complications , Capillary Permeability , Immunoblotting , Infarction, Middle Cerebral Artery/complications , Male , Rats, Sprague-Dawley
11.
Nat Commun ; 7: 12803, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27640812

ABSTRACT

The design of crystalline polymers is intellectually stimulating and synthetically challenging, especially when the polymerization of any monomer occurs in a linear dimension. Such linear growth often leads to entropically driven chain entanglements and thus is detrimental to attempts to realize the full potential of conjugated molecular structures. Here we report the polymerization of two-monomer-connected precursors (TMCPs) in which two pyrrole units are linked through a connector, yielding highly crystalline polymers. The simultaneous growth of the TMCP results in a close-packed crystal in polypyrrole (PPy) at the molecular scale with either a hexagonal close-packed or face-centred cubic structure, as confirmed by high-voltage electron microscopy, and the structure that formed could be controlled by simply changing the connector. The electrical conductivity of the TMCP-based PPy is almost 35 times that of single-monomer-based PPy, demonstrating its promise for application in diverse fields.

12.
Stroke ; 47(5): 1336-43, 2016 05.
Article in English | MEDLINE | ID: mdl-27012742

ABSTRACT

BACKGROUND AND PURPOSE: Most patients with cerebral infarction die of brain edema because of the breakdown of the blood-brain barrier (BBB) in ischemic tissue. Caveolins (a group of proteins) are key modulators of vascular permeability; however, a direct role of caveolin-1 (Cav-1) in the regulation of BBB permeability during ischemic injury has yet to be identified. METHODS: Cav-1 expression was measured by immunoblotting after photothrombotic ischemia. A direct functional role of Cav-1 in cerebral edema and BBB permeability during cerebral ischemia was investigated by genetic manipulation (gene disruption and re-expression) of Cav-1 protein expression in mice. RESULTS: There was a significant correlation between the extent of BBB disruption and the Cav-1 expression. In Cav-1-deficient (Cav-1(-/-)) mice, the extent of BBB disruption after cerebral ischemia was increased compared with wild-type (Cav-1(+/+)) mice, whereas the increase in cerebral edema volume was ameliorated by lentiviral-mediated re-expression of Cav-1. Furthermore, Cav-1(-/-) mice had significantly higher degradation of tight junction proteins and proteolytic activity of matrix metalloproteinase than Cav-1(+/+) mice. Conversely, re-expression of Cav-1 in Cav-1(-/-) mice restored tight junction protein expression and reduced matrix metalloproteinase proteolytic activity. CONCLUSIONS: These results indicate that Cav-1 is a critical determinant of BBB permeability. Strategies for regulating Cav-1 represent a novel therapeutic approach to controlling BBB disruption and subsequent neurological deterioration during cerebral ischemia.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Brain Ischemia/metabolism , Caveolin 1/metabolism , Animals , Caveolin 1/genetics , Disease Models, Animal , Gene Expression Regulation , Mice , Mice, Knockout
13.
Neurosci Lett ; 609: 97-102, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26484507

ABSTRACT

Olanzapine is widely used in the treatment of schizophrenia and related psychoses. We investigated the effects of olanzapine on human ether-a-go-go related gene (hERG) channels stably expressed in human embryonic kidney (HEK) cells using the whole-cell patch-clamp technique. Olanzapine inhibited hERG tail currents at -50mV in a concentration-dependent manner with an IC50 value of 8.0µM and a Hill coefficient of 0.9. The voltage-dependent inhibition of the hERG currents by olanzapine increased steeply in the voltage range of channel activation. Olanzapine also shifted the steady-state activation curve of the hERG currents in a hyperpolarizing direction. At more depolarized potentials where the channels were fully activated (between 0 and +50mV), the olanzapine inhibition was voltage-independent. The steady-state inactivation curve of the hERG currents was shifted in the hyperpolarizing direction in the presence of olanzapine. A fast application of olanzapine induced a reversible inhibition of the hERG tail currents during repolarization in a concentration-dependent manner with an IC50 value of 11.9µM, suggesting an open-channel block. Olanzapine also decreased the hERG current elicited by a 5s depolarizing pulse to +60mV to inactivate the hERG currents, suggesting an inhibition of the activated (open and/or inactivated) states of the channels. These results indicated that olanzapine inhibited the hERG current by preferentially interacting with the activated states of the channel.


Subject(s)
Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Antipsychotic Agents/adverse effects , Benzodiazepines/adverse effects , ERG1 Potassium Channel , HEK293 Cells , Humans , Olanzapine , Patch-Clamp Techniques
14.
Eur J Pharmacol ; 752: 1-7, 2015 Apr 05.
Article in English | MEDLINE | ID: mdl-25680947

ABSTRACT

The effects of tamoxifen, and its active metabolite endoxifen (4-hydroxy-N-desmethyl-tamoxifen), on hERG currents stably expressed in HEK cells were investigated using the whole-cell patch-clamp technique and an immunoblot assay. Tamoxifen and endoxifen inhibited hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 1.2 and 1.6µM, respectively. The steady-state activation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of endoxifen. The voltage-dependent inhibition of hERG currents by endoxifen increased steeply in the voltage range of channel activation. The inhibition by endoxifen displayed a shallow voltage dependence (δ=0.18) in the full activation voltage range. A fast application of endoxifen induced a reversible block of hERG tail currents during repolarization in a concentration-dependent manner, which suggested an interaction with the open state of the channel. Endoxifen also decreased the hERG current elicited by a 5s depolarizing pulse to +60mV to inactivate the hERG currents, suggesting an interaction with the activated (open and/or inactivated) states of the channels. Tamoxifen and endoxifen inhibited the hERG channel protein trafficking to the plasma membrane in a concentration-dependent manner with endoxifen being more potent than tamoxifen. These results indicated that tamoxifen and endoxifen inhibited the hERG current by direct channel blockage and by the disruption of channel trafficking to the plasma membrane in a concentration-dependent manner. A therapeutic concentration of endoxifen inhibited the hERG current by preferentially interacting with the activated (open and/or inactivated) states of the channel.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Tamoxifen/analogs & derivatives , Tamoxifen/metabolism , Cloning, Molecular , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Tamoxifen/pharmacology
15.
Pflugers Arch ; 467(8): 1663-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25231973

ABSTRACT

Raloxifene is widely used for the treatment and prevention of postmenopausal osteoporosis. We examined the effects of raloxifene on the Kv4.3 currents expressed in Chinese hamster ovary (CHO) cells using the whole-cell patch-clamp technique and on the long-term modulation of Kv4.3 messenger RNA (mRNA) by real-time PCR analysis. Raloxifene decreased the Kv4.3 currents with an IC50 of 2.0 µM and accelerated the inactivation and activation kinetics in a concentration-dependent manner. The inhibitory effects of raloxifene on Kv4.3 were time-dependent: the association and dissociation rate constants for raloxifene were 9.5 µM(-1) s(-1) and 23.0 s(-1), respectively. The inhibition by raloxifene was voltage-dependent (δ = 0.13). Raloxifene shifted the steady-state inactivation curves in a hyperpolarizing direction and accelerated the closed-state inactivation of Kv4.3. Raloxifene slowed the time course of recovery from inactivation, thus producing a use-dependent inhibition of Kv4.3. ß-Estradiol and tamoxifen had little effect on Kv4.3. A preincubation of ICI 182,780, an estrogen receptor antagonist, for 1 h had no effect on the inhibitory effect of raloxifene on Kv4.3. The metabolites of raloxifene, raloxifene-4'-glucuronide and raloxifene-6'-glucuronide, had little or no effect on Kv4.3. Coexpression of KChIP2 subunits did not alter the drug potency and steady-state inactivation of Kv4.3 channels. Long-term exposure to raloxifene (24 h) significantly decreased the expression level of Kv4.3 mRNA. This effect was not abolished by the coincubation with ICI 182,780. Raloxifene inhibited Kv4.3 channels by interacting with their open state during depolarization and with the closed state at subthreshold potentials. This effect was not mediated via an estrogen receptor.


Subject(s)
Bone Density Conservation Agents/pharmacology , Potassium Channel Blockers/pharmacology , Raloxifene Hydrochloride/pharmacology , Receptors, Estrogen , Shal Potassium Channels/antagonists & inhibitors , Animals , CHO Cells , Cloning, Molecular , Cricetulus , Dose-Response Relationship, Drug , Down-Regulation , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Ion Channel Gating/drug effects , Kinetics , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Membrane Potentials , Patch-Clamp Techniques , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Tamoxifen/pharmacology , Transfection
16.
Brain Res ; 1597: 77-85, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25498859

ABSTRACT

Donepezil is a potent, selective inhibitor of acetylcholinesterase, which is used for the treatment of Alzheimer's disease. Whole-cell patch-clamp technique and Western blot analyses were used to study the effects of donepezil on the human ether-a-go-go-related gene (hERG) channel. Donepezil inhibited the tail current of the hERG in a concentration-dependent manner with an IC50 of 1.3 µM. The metabolites of donepezil, 6-ODD and 5-ODD, inhibited the hERG currents in a similar concentration-dependent manner; the IC50 values were 1.0 and 1.5 µM, respectively. A fast drug perfusion system demonstrated that donepezil interacted with both the open and inactivated states of the hERG. A fast application of donepezil during the tail currents inhibited the open state of the hERG in a concentration-dependent manner with an IC50 of 2.7 µM. Kinetic analysis of donepezil in an open state of the hERG yielded blocking and unblocking rate constants of 0.54 µM(-1)s(-1) and 1.82 s(-1), respectively. The block of the hERG by donepezil was voltage-dependent with a steep increase across the voltage range of channel activation. Donepezil caused a reduction in the hERG channel protein trafficking to the plasma membrane at low concentration, but decreased the channel protein expression at higher concentrations. These results suggest that donepezil inhibited the hERG at a supratherapeutic concentration, and that it did so by preferentially binding to the activated (open and/or inactivated) states of the channels and by inhibiting the trafficking and expression of the hERG channel protein in the plasma membrane.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Indans/pharmacology , Piperidines/pharmacology , Blotting, Western , Cell Membrane/drug effects , Cell Membrane/physiology , Donepezil , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Fluoxetine/pharmacology , HEK293 Cells , Humans , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Selective Serotonin Reuptake Inhibitors/pharmacology
17.
Eur J Pharmacol ; 740: 1-8, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-24998874

ABSTRACT

Haloperidol is commonly used in clinical practice to treat acute and chronic psychosis, but it also has been associated with adverse cardiovascular events. We investigated the effects of haloperidol on Kv4.3 currents stably expressed in CHO cells using a whole-cell patch-clamp technique. Haloperidol did not significantly inhibit the peak amplitude of Kv4.3, but accelerated the decay rate of inactivation of Kv4.3 in a concentration-dependent manner. Thus, the effects of haloperidol on Kv4.3 were estimated from the integral of the Kv4.3 currents during the depolarization pulse. The Kv4.3 was decreased by haloperidol in a concentration-dependent manner with an IC50 value of 3.6 µM. Haloperidol accelerated the decay rate of Kv4.3 inactivation and activation kinetics in a concentration-dependent manner, thereby decreasing the time-to-peak. Haloperidol shifted the voltage dependence of the steady-state activation and inactivation of Kv4.3 in a hyperpolarizing direction. Haloperidol also caused an acceleration of the closed-state inactivation of Kv4.3. Haloperidol produced a use-dependent block of Kv4.3, which was accompanied by a slowing of recovery from the inactivation of Kv4.3. These results suggest that haloperidol blocks Kv4.3 by both interacting with the open state of Kv4.3 channels during depolarization and accelerating the closed-state inactivation at subthreshold membrane potentials.


Subject(s)
Antipsychotic Agents/pharmacology , Haloperidol/pharmacology , Potassium Channel Blockers/pharmacology , Shal Potassium Channels/antagonists & inhibitors , Animals , CHO Cells , Cricetulus , Shal Potassium Channels/genetics , Shal Potassium Channels/physiology
18.
Naunyn Schmiedebergs Arch Pharmacol ; 387(1): 23-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24045971

ABSTRACT

Escitalopram, a selective serotonin reuptake inhibitor, is the pharmacologically active S-enantiomer of the racemic mixture of RS-citalopram and is widely used in the treatment of depression. The effects of escitalopram and citalopram on the human ether-a-go-go-related gene (hERG) channels expressed in human embryonic kidney cells were investigated using voltage-clamp and Western blot analyses. Both drugs blocked hERG currents in a concentration-dependent manner with an IC50 value of 2.6 µM for escitalopram and an IC50 value of 3.2 µM for citalopram. The blocking of hERG by escitalopram was voltage-dependent, with a steep increase across the voltage range of channel activation. However, voltage independence was observed over the full range of activation. The blocking by escitalopram was frequency dependent. A rapid application of escitalopram induced a rapid and reversible blocking of the tail current of hERG. The extent of the blocking by escitalopram during the depolarizing pulse was less than that during the repolarizing pulse, suggesting that escitalopram has a high affinity for the open state of the hERG channel, with a relatively lower affinity for the inactivated state. Both escitalopram and citalopram produced a reduction of hERG channel protein trafficking to the plasma membrane but did not affect the short-term internalization of the hERG channel. These results suggest that escitalopram blocked hERG currents at a supratherapeutic concentration and that it did so by preferentially binding to both the open and the inactivated states of the channels and by inhibiting the trafficking of hERG channel protein to the plasma membrane.


Subject(s)
Citalopram/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/physiology , Potassium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , ERG1 Potassium Channel , HEK293 Cells , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology
19.
FEBS Lett ; 583(15): 2557-64, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19619541

ABSTRACT

Forced changes in the expression of regulator of calcineurin 1 (RCAN1) affects cell growth. This has been linked to the suppression of calcineurin-nuclear factor of activated T cells signaling by RCAN1. Here, we describe a novel role of RCAN1 isoform 4 in proper expression of Ras protein and its signaling. RCAN1 isoform 4 knockdown attenuated growth factor-induced extracellular signal-regulated kinase activation and cell growth; reduced Ras levels and its translation rate; and led to a reduction of eukaryotic initiation factor 4E in the initiation complex and a slight repression of global protein synthesis. Experiments utilizing activity-modified mutants of calcineurin A demonstrated that these effects were calcineurin-independent. Our findings reveal a previously unknown role of RCAN1-4 in protein synthesis, which may be relevant to cell growth.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Protein Isoforms/metabolism , Signal Transduction/physiology , ras Proteins/metabolism , Calcineurin/genetics , Calcineurin/metabolism , Calcineurin Inhibitors , Cell Line , DNA-Binding Proteins , Enzyme Activation , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Protein Isoforms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , ras Proteins/genetics
20.
Korean J Physiol Pharmacol ; 13(6): 483-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20054496

ABSTRACT

Despite the potential importance of the human regulator of calcineurin 1 (RCAN-1) gene in the modulation of cell survival under stress, little is known about its role in death-inducing signal pathways. In this study, we addressed the effects of RCAN1.4 knockdown on cellular susceptibility to apoptosis and the activation of death pathway proteins. Transfection of siRNAs against RCAN1.4 resulted in enhanced Fas- and etoposide-induced apoptosis, which was associated with increased expression and translocation of Bax to mitochondria. Our results suggest that enhanced expression and activation of p53 was responsible for the upregulation of Bax and the increased sensitivity to apoptosis, which could be reversed by p53 knockdown. To explain the observed upregulation of p53, we propose a downregulation of the ubiquitin ligase HDM2, probably translationally. These findings show the importance of appropriate RCAN1.4 expression in the modulation of cell survival and reveal a link between RCAN1.4 and p53.

SELECTION OF CITATIONS
SEARCH DETAIL
...