Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Biol Pharm Bull ; 46(10): 1394-1402, 2023.
Article in English | MEDLINE | ID: mdl-37779040

ABSTRACT

Dimenhydrinate, an H1 receptor antagonist, is generally used for the prevention and treatment of nausea and vomiting. However, cardiac arrhythmias have been reported to be associated with the overdose of histamine H1 receptor antagonists, indicating the probable effect of antihistamines on ion channels. By using a two-microelectrode voltage clamp, we have herein studied the electrophysiological effects of dimenhydrinate on the human Kv1.5 channel in the Xenopus oocyte expression system. Dimenhydrinate acutely and reversibly suppressed the amplitudes of the peak and the steady-state current, within 6 min. The inhibitory effect of dimenhydrinate on the peak and the steady-state Kv1.5 currents increased progressively from -10 to +50 mV. At each test voltage, the drug suppressed both the peak and the steady-state currents to a similar extent. When the oocytes were stimulated at the rates of 5- and 30-s intervals, dimenhydrinate-induced a use-dependent blockade of the human Kv1.5 channel. Dimenhydrinate expedited the timecourse of the Kv1.5 channel activation more effectively than the timecourse of its inactivation. However, the activation and inactivation curves of the channel were not altered by the H1 receptor antagonist. In conclusion, we found that dimenhydrinate inhibits the human Kv1.5 channel by changing the channel's activation mode, thereby possibly increasing the possibility of triggering cardiac arrhythmias and affecting atrial fibrillation.


Subject(s)
Dimenhydrinate , Humans , Dimenhydrinate/metabolism , Dimenhydrinate/pharmacology , Electrophysiological Phenomena , Histamine H1 Antagonists/pharmacology , Oocytes/metabolism , Potassium Channel Blockers/pharmacology
2.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3149-3161, 2023 11.
Article in English | MEDLINE | ID: mdl-37166464

ABSTRACT

Ifenprodil has been known to reduce cardiac contractility and cerebral vasodilation by antagonizing α1-adrenergic and N-methyl D-aspartate receptor-mediated intracellular signals. This study aimed to investigate the direct effect of ifenprodil on the human voltage-gated Kv1.5 channel (hKv1.5) by using a Xenopus oocyte expression system and a two-microelectrode voltage clamp technique. The amplitudes of hKv1.5 currents, including peak and steady state, were suppressed in a concentration-dependent manner (IC50; 43.1 and 35.5 µM, respectively) after 6 min of ifenprodil treatment. However, these effects were ~ 80% reversed by washout, suggesting that ifenprodil directly inhibited the hKv1.5 independent of membrane receptors or intracellular signals. The inhibition rate of steady state showed voltage dependence, wherein the rates increased according to test voltage depolarization. Ifenprodil reduced the time constants of hKv1.5 inactivation but has higher effects on activation. hKv1.5 inhibition by ifenprodil showed use dependency because the drug more rapidly reduced the current at the higher activation frequencies, and subsequent reduction in frequency after high activation frequency caused a partial channel block relief. Therefore, ifenprodil directly blocked the hKv1.5 in an open state and accelerated the time course of the channel inactivation, which provided a biophysical mechanism for the hKv1.5 blocking effects of ifenprodil.


Subject(s)
N-Methylaspartate , Piperidines , Humans , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate , Adrenergic alpha-1 Receptor Antagonists , Kv1.5 Potassium Channel , Potassium Channel Blockers/pharmacology
3.
Eur J Pharmacol ; 912: 174567, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34662565

ABSTRACT

Paroxetine is one of the most effective selective serotonin reuptake inhibitors used to treat depressive and panic disorders that reduce the viability of human T lymphocytes, in which Kv1.3 channels are highly expressed. We examined whether paroxetine could modulate human Kv1.3 channels acutely and directly with the aim of understanding the biophysical effects and the underlying mechanisms of the drug. Kv1.3 channel proteins were expressed in Xenopus oocytes. Paroxetine rapidly inhibited the steady-state current and peak current of these channels within 6 min in a concentration-dependent manner; IC50s were 26.3 µM and 53.9 µM, respectively, and these effects were partially reversed by washout, which excluded the possibility of genomic regulation. At the same test voltage, paroxetine blockade of the steady-state currents was higher than that of the peak currents, and the inhibition of the steady-state current increased relative to the degree of depolarization. Paroxetine decreased the inactivation time constant in a concentration-dependent manner, but it did not affect the activation time constant, which resulted in the acceleration of intrinsic inactivation without changing ultrarapid activation. Blockade of Kv1.3 channels by paroxetine exhibited more rapid inhibition at higher activation frequencies showing the use-dependency of the blockade. Overall, these results show that paroxetine directly suppresses human Kv1.3 channels in an open state and accelerates the process of steady-state inactivation; thus, we have revealed a biophysical mechanism for possible acute immunosuppressive effects of paroxetine.


Subject(s)
Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/metabolism , Paroxetine/pharmacology , Potassium Channel Blockers/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Humans , Oocytes/drug effects , Oocytes/metabolism , Xenopus laevis
4.
Toxicol Appl Pharmacol ; 411: 115365, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33316272

ABSTRACT

Polychlorinated biphenyls (PCBs) are persistent and serious organic pollutants and can theoretically form 209 congeners. PCBs can be divided into two categories: dioxin-like (DL) and non-DL (NDL). NDL-PCBs, which lack aryl hydrocarbon receptor affinity, have been shown to perturb the functions of Jurkat T cells, cerebellar granule cells, and uterine cells. Kv1.3 and Kv1.5 channels are important in immune and heart functions, respectively. We investigated the acute effects of 2,2',6-trichlorinated biphenyl (PCB19), an NDL-PCB, on the currents of human Kv1.3 and Kv1.5 channels. PCB19 acutely blocked the Kv1.3 peak currents concentration-dependently with an IC50 of ~2 µM, without changing the steady-state current. The PCB19-induced inhibition of the Kv1.3 peak current occurred rapidly and voltage-independently, and the effect was irreversible, excluding the possibility of genomic regulation. PCB19 increased the time constants of both activation and inactivation of Kv1.3 channels, resulting in the slowing down of both ultra-rapid activation and intrinsic inactivation. However, PCB19 failed to alter the steady-state curves of activation and inactivation. Regarding the Kv1.5 channel, PCB19 affected neither the peak current nor the steady-state current at the same concentrations tested in the Kv1.3 experiments, showing selective inhibition of PCB19 on the Kv1.3 than the Kv1.5. The presented data indicate that PCB19 could acutely affect the human Kv1.3 channel through a non-genomic mechanism, possibly causing toxic effects on various human physiological functions related to the Kv1.3 channel, such as immune and neural systems.


Subject(s)
Environmental Pollutants/toxicity , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/drug effects , Polychlorinated Biphenyls/toxicity , Potassium Channel Blockers/toxicity , Animals , Dose-Response Relationship, Drug , Female , Humans , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Membrane Potentials , Oocytes , Time Factors , Xenopus laevis
5.
Mol Cell Endocrinol ; 494: 110488, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31207272

ABSTRACT

Hydrocortisone exerts adverse effects on various organs, including the heart. This study investigated the still unclear effects of hydrocortisone on electrophysiological and biochemical aspects of cardiac excitation-contraction coupling. In guinea pigs' hearts, hydrocortisone administration reduced the QT interval of ECG and the action potential duration (APD). In guinea pig ventricular myocytes, hydrocortisone reduced contraction and Ca2+ transient amplitudes. These reductions and the effects on APD were prevented by pretreatment with the protein kinase C (PKC) inhibitor staurosporine. In an overexpression system of Xenopus oocytes, hydrocortisone increased hERG K+ currents and reduced Kv1.5 K+ currents; these effects were negated by pretreatment with staurosporine. Western blot analysis revealed dose- and time-dependent changes in PKCα/ßII, PKCε, and PKCγ phosphorylation by hydrocortisone in guinea pig ventricular myocytes. Therefore, hydrocortisone can acutely affect cardiac excitation-contraction coupling, including ion channel activity, APD, ECG, Ca2+ transients, and contraction, possibly via biochemical changes in PKC.


Subject(s)
Action Potentials/drug effects , Calcium/metabolism , Electrocardiography , Heart/physiology , Hydrocortisone/pharmacology , Intracellular Space/metabolism , Myocardial Contraction/drug effects , Protein Kinase C/metabolism , Animals , Diastole/drug effects , Ether-A-Go-Go Potassium Channels/metabolism , Guinea Pigs , Heart/diagnostic imaging , Heart/drug effects , Heart Ventricles/cytology , Ion Channel Gating/drug effects , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oocytes/drug effects , Oocytes/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Staurosporine/pharmacology , Time Factors , Xenopus laevis
6.
Naunyn Schmiedebergs Arch Pharmacol ; 390(12): 1221-1228, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28865055

ABSTRACT

PCB19, a 2,2',6-trichlorinated biphenyl, is one of many non-dioxin-like polychlorinated biphenyls (NDL-PCBs), which are ubiquitous pollutants. NDL-PCBs affect cytosolic Ca2+ signaling by promoting Ca2+ release from ryanodine receptor-sensitive Ca2+ pools and inhibiting store-operated Ca2+ entry (SOCE) from the extracellular space. However, NDL-PCB-mediated SOCE inhibition has only been demonstrated in PC12 cells, in which SOCE is thought to be mainly mediated by TRPC family channels. Here, we investigated the effect of PCB19 on SOCE using human embryonic kidney 293 (HEK293) cells, human leukemia T cell line Jurkat-T cells and human promyelocytoma HL-60 cells which are the cell lines that are previously demonstrated to mediate the most common form of SOCE solely by the intrinsic Orai channels. PCB19 reduced thapsigargin-induced Ca2+ influx after Ca2+ pool depletion in HEK293 cells. SOCEs in HEK293, Jurkat T, HL-60 and PC12 cells showed distinct sensitivities to SOCE inhibitors such as Gd3+ and ML-9; however, PCB19 also showed a common effect of inhibiting SOCEs in all cell lines. PCB19-mediated SOCE inhibition was confirmed by demonstrating the ability of PCB19 to inhibit the SOCE current but not the TRPM7 current. These results imply that PCB19 inhibits not only TRPC-mediated SOCE as in PC12 cells but also Orai-mediated SOCE as in many other cells including HEK293, Jurkat T and HL-60 cells.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , ORAI1 Protein/drug effects , Polychlorinated Biphenyls/pharmacology , Animals , Azepines/pharmacology , Gadolinium/pharmacology , HEK293 Cells , HL-60 Cells , Humans , Jurkat Cells , PC12 Cells , Rats , TRPM Cation Channels/drug effects , Thapsigargin/antagonists & inhibitors , Thapsigargin/pharmacology
7.
Eur J Oral Sci ; 125(1): 18-27, 2017 02.
Article in English | MEDLINE | ID: mdl-28032657

ABSTRACT

Bradykinin is an important peptide modulator that affects the function of neurons and immune cells. However, there is no evidence of the bradykinin receptors and their functions in human salivary glands. Here we have identified and characterized bradykinin receptors on human submandibular gland cells. Both bradykinin B1 and B2 receptors are expressed on human submandibular gland cells, A253 cells, and HSG cells. Bradykinin increased the intracellular Ca2+ concentration ([Ca2+ ]i ) in a concentration-dependent manner. Interestingly, a specific agonist of the B1 receptor did not have any effect on [Ca2+ ]i in HSG cells, whereas specific agonists of the B2 receptor had a Ca2+ mobilizing effect. Furthermore, application of the B1 receptor antagonist, R715, did not alter the bradykinin-mediated increase in cytosolic Ca2+ , whereas the B2 receptor antagonist, HOE140, showed a strong inhibitory effect, which implies that bradykinin B2 receptors are functional in modulating the concentration of cytosolic Ca2+ . Bradykinin did not affect a carbachol-induced rise of [Ca2+ ]i and did not modulate translocation of aquaporin-5. However, bradykinin did promote the expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), implying the role of bradykinin in salivary gland inflammation. These data suggest that bradykinin receptors are involved in Ca2+ signaling in human submandibular gland cells and serve a unique role, which is separate from that of other salivary gland G protein-coupled receptors.


Subject(s)
Cytokines/metabolism , Receptors, Bradykinin/metabolism , Salivary Glands/metabolism , Adult , Aged , Aged, 80 and over , Aquaporin 5/metabolism , Blotting, Western , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Calcium/metabolism , Carbachol/pharmacology , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Female , Fluorescent Antibody Technique , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Salivary Glands/cytology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
8.
PLoS One ; 11(3): e0150921, 2016.
Article in English | MEDLINE | ID: mdl-26963511

ABSTRACT

Polychlorinated biphenyls (PCBs) are ubiquitous pollutants which accumulate in the food chain. Recently, several molecular mechanisms by which non-dioxin-like (NDL) PCBs mediate neurodevelopmental and neurobehavioral toxicity have been elucidated. However, although the G-protein coupled receptor (GPCR) is a significant target for neurobehavioral disturbance, our understanding of the effects of PCBs on GPCR signaling remains unclear. In this study, we investigated the effects of NDL-PCBs on GPCR-mediated Ca2+ signaling in PC12 cells. We found that ortho-substituted 2,2',6-trichlorinated biphenyl (PCB19) caused a rapid decline in the Ca2+ signaling of bradykinin, a typical Gq- and phospholipase Cß-coupled GPCR, without any effect on its inositol 1,4,5-trisphosphate production. PCB19 reduced thapsigargin-induced sustained cytosolic Ca2+ levels, suggesting that PCB19 inhibits SOCE. The abilities of other NDL-PCBs to inhibit store-operated Ca2+ entry (SOCE) were also examined and found to be of similar potencies to that of PCB19. PCB19 also showed a manner equivalent to that of known SOCE inhibitors. PCB19-mediated SOCE inhibition was confirmed by demonstrating the ability of PCB19 to inhibit the SOCE current and thapsigargin-induced Mn2+ influx. These results imply that one of the molecular mechanism by which NDL-PCBs cause neurobehavioral disturbances involves NDL-PCB-mediated inhibition of SOCE, thereby interfering with GPCR-mediated Ca2+ signaling.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium/metabolism , Polychlorinated Biphenyls/toxicity , Receptors, G-Protein-Coupled/metabolism , Thapsigargin/pharmacology , Animals , Manganese/metabolism , PC12 Cells , Rats
9.
Technol Health Care ; 24 Suppl 1: S11-6, 2015.
Article in English | MEDLINE | ID: mdl-26684562

ABSTRACT

This study measured epidermal and dermal temperatures under different cryogen spray cooling (CSC) conditions to determine the optimum cooling conditions for skin rejuvenation. For this purpose, CSC conditions were applied before a laser transmission for varying spurt times of 50, 150, and 200 ms with delay times of 150 and 200 ms. A long-pulsed 1,064 nm Nd:YAG laser irradiated the skin surface of a pig with a condition of fluence of 26 J/cm2 and a spot diameter of 8 mm. The pulse duration was set to 30 ms during all experiments. This study found that all employed CSC conditions significantly decreased internal-external skin temperatures. Moreover, skin temperatures were influenced more by variations in spurt time of CSC compared with the delay times. Based on these experimental results, two spurt times were selected as the optimum CSC conditions for skin rejuvenation: 50 ms with delay time of 150 and 200 ms and 150 ms with a delay time of 150 and 200 ms.


Subject(s)
Aerosol Propellants/administration & dosage , Epidermis/physiology , Plasma Skin Regeneration/methods , Skin Temperature/physiology , Animals , Cryotherapy/methods , Swine
10.
Technol Health Care ; 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26409534

ABSTRACT

This study measured epidermal and dermal temperatures under different cryogen spray cooling (CSC) conditions to determine the optimum cooling conditions for skin rejuvenation. For this purpose, CSC conditions were applied before a laser transmission for varying spurt times of 50, 150, and 200 ms with delay times of 150 and 200 ms. A long-pulsed 1,064 nm Nd:YAG laser irradiated the skin surface of a pig with a condition of fluence of 26 J/cm2 and a spot diameter of 8 mm. The pulse duration was set to 30 ms during all experiments. This study found that all employed CSC conditions significantly decreased internal-external skin temperatures. Moreover, skin temperatures were influenced more by variations in spurt time of CSC compared with the delay times. Based on these experimental results, two spurt times were selected as the optimum CSC conditions for skin rejuvenation: 50 ms with delay time of 150 and 200 ms and 150 ms with a delay time of 150 and 200 ms.

11.
Nature ; 519(7544): 472-6, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25799991

ABSTRACT

Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP, nitric oxide signalling is often depressed by heart disease. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cardiomegaly/enzymology , Cardiomegaly/metabolism , Cyclic GMP/metabolism , Nitric Oxide , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/deficiency , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Aortic Valve Stenosis/complications , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Cells/enzymology , Myocardium/enzymology , Natriuretic Peptides/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Pressure , Signal Transduction/drug effects , Stress, Physiological , Up-Regulation
12.
Naunyn Schmiedebergs Arch Pharmacol ; 388(6): 653-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25743574

ABSTRACT

Glucocorticoids are hormones released in response to stress that are involved in various physiological processes including immune functions. One immune-modulating mechanism is achieved by the Kv1.3 voltage-dependent potassium channel, which is expressed highly in lymphocytes including effector memory T lymphocytes (TEM). Although glucocorticoids are known to inhibit Kv1.3 function, the detailed inhibitory mechanism is not yet fully understood. Here we studied the rapid non-genomic effects of cortisone and hydrocortisone on the human Kv1.3 channel expressed in Xenopus oocytes. Both cortisone and hydrocortisone reduced the amplitude of the Kv1.3 channel current in a concentration-dependent manner. Both cortisone and hydrocortisone rapidly and irreversibly inhibited Kv1.3 currents, eliminating the possibility of genomic regulation. Inhibition rate was stable relative to the degree of depolarization. Kinetically, cortisone altered the activating gate of Kv1.3 and hydrocortisone interacted with this channel in an open state. These results suggest that cortisone and hydrocortisone inhibit Kv1.3 currents via a non-genomic mechanism, providing a mechanism for the immunosuppressive effects of glucocorticoids.


Subject(s)
Cortisone/pharmacology , Hydrocortisone/pharmacology , Kv1.3 Potassium Channel/physiology , Potassium Channel Blockers/pharmacology , Animals , Humans , Kv1.3 Potassium Channel/genetics , Membrane Potentials/drug effects , Oocytes/drug effects , Oocytes/physiology , Xenopus
13.
Biochem Biophys Res Commun ; 459(4): 604-9, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25753203

ABSTRACT

Calcineurin is a calcium/calmodulin-dependent phosphatase that has been implicated in T cell activation through the induction of nuclear factors of activated T cells (NFAT). We have previously suggested that endogenous regulator of calcineurin (RCAN1, also known as DSCR1) is targeted by protein kinase A (PKA) for the control of calcineurin activity. In the present study, we characterized the PKA-mediated phosphorylation site in RCAN1 by mass spectrometric analysis and revealed that PKA directly phosphorylated RCAN1 at the Ser 93. PKA-induced phosphorylation and the increase in the half-life of the RCAN1 protein were prevented by the substitution of Ser 93 with Ala (S93A). Furthermore, the PKA-mediated phosphorylation of RCAN1 at Ser 93 potentiated the inhibition of calcineurin-dependent pro-inflammatory cytokine gene expression by RCAN1. Our results suggest the presence of a novel phosphorylation site in RCAN1 and that its phosphorylation influences calcineurin-dependent inflammatory target gene expression.


Subject(s)
Calcineurin/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Amino Acid Sequence , DNA-Binding Proteins , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Molecular Sequence Data , Muscle Proteins/chemistry , Phosphorylation
14.
Eur J Pharmacol ; 746: 158-66, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25449034

ABSTRACT

Glucocorticoids are the primary hormones that respond to stress and protect organisms from dangerous situations. The glucocorticoids hydrocortisone and its dormant form, cortisone, affect the cardiovascular system with changes such as increased blood pressure and cardioprotection. Kv1.5 channels play a critical role in the maintenance of cellular membrane potential and are widely expressed in pancreatic ß-cells, neurons, myocytes, and smooth muscle cells of the pulmonary vasculature. We examined the electrophysiological effects of both cortisone and hydrocortisone on human Kv1.5 channels expressed in Xenopus oocytes using a two-microelectrode voltage clamp technique. Both cortisone and hydrocortisone rapidly and irreversibly suppressed the amplitude of Kv1.5 channel current with IC50 values of 50.2±4.2µM and 33.4±3.2µM, respectively, while sustained the current trace shape of Kv1.5 current. The inhibitory effect of cortisone on Kv1.5 decreased progressively from -10mV to +30mV, while hydrocortisone׳s inhibition of the channel did not change across the same voltage range. Both cortisone and hydrocortisone blocked Kv1.5 channel currents in a non-use-dependent manner and neither altered the channel׳s steady-state activation or inactivation curves. These results show that cortisone and hydrocortisone inhibited Kv1.5 channel currents differently, and that Kv1.5 channels were more sensitive to hydrocortisone than to cortisone.


Subject(s)
Cortisone/pharmacology , Electrophysiological Phenomena/drug effects , Hydrocortisone/pharmacology , Kv1.5 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Humans , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Oocytes/metabolism , Time Factors , Xenopus laevis/genetics
15.
J Neurochem ; 132(3): 276-85, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25418874

ABSTRACT

Methyl-ß-cyclodextrin (MßCD) is a reagent that depletes cholesterol and disrupts lipid rafts, a type of cholesterol-enriched cell membrane microdomain. Lipid rafts are essential for neuronal functions such as synaptic transmission and plasticity, which are sensitive to even low doses of MßCD. However, how MßCD changes synaptic function, such as N-methyl-d-aspartate receptor (NMDA-R) activity, remains unclear. We monitored changes in synaptic transmission and plasticity after disrupting lipid rafts with MßCD. At low concentrations (0.5 mg/mL), MßCD decreased basal synaptic transmission and miniature excitatory post-synaptic current without changing NMDA-R-mediated synaptic transmission and the paired-pulse facilitation ratio. Interestingly, low doses of MßCD failed to deplete cholesterol or affect α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA-R) and NMDA-R levels, while clearly reducing GluA1 levels selectively in the synaptosomal fraction. Low doses of MßCD decreased the inhibitory effects of NASPM, an inhibitor for GluA2-lacking AMPA-R. MßCD successfully decreased NMDA-R-mediated long-term potentiation but did not affect the formation of either NMDA-R-mediated or group I metabotropic glutamate receptor-dependent long-term depression. MßCD inhibited de-depression without affecting de-potentiation. These results suggest that MßCD regulates GluA1-dependent synaptic potentiation but not synaptic depression in a cholesterol-independent manner.


Subject(s)
Receptors, AMPA/physiology , Synapses/drug effects , beta-Cyclodextrins/pharmacology , Animals , Cholesterol/metabolism , In Vitro Techniques , Male , Membrane Microdomains/drug effects , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/drug effects , Synaptosomes/drug effects
16.
Toxicon ; 92: 157-65, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25448388

ABSTRACT

Crotamine is a peptide toxin found in the venom of the rattlesnake Crotalus durissus terrificus. Interestingly, crotamine demonstrates promising anticancer, antimicrobial, and antifungal activities. The crotamine peptide can also deliver plasmids into rapidly dividing cells, such as cancer and stem cells, and demonstrates potent analgesic effects. Efficiently producing crotamine in mammalian cells is difficult because it is both cell-permeable and cytotoxic. Prokaryotic expression of this peptide is also difficult to maintain because it does not fold properly in the cytoplasm, resulting in aggregation and in the formation of inclusion bodies. In our current study, we show for the first time that N-terminal fusion with three protein tags-N-utilization substance protein A (NusA), protein disulfide isomerase b'a' domain (PDIb'a'), and maltose-binding protein (MBP)-enables the soluble overexpression of crotamine in the cytoplasm of Escherichia coli. MBP-tagged crotamine was purified using Ni affinity, anion exchange, and MBP chromatography. The tag was cleaved using TEV protease, and the final product was pure on a silver-stained gels. In total, 0.9 mg pure crotamine was obtained from each liter of bacterial culture with endotoxin level approximately 0.15 EU/µg, which is low enough to use in biomedical applications. The identity and intramolecular disulfide bonds were confirmed using MALDI-TOF MS analysis. Purified crotamine inhibited the hKv1.3 channel (but not hKv1.5) in a dose-dependent manner with IC50 value of 67.2 ± 44.7 nM (n = 10), indicating the correct protein folding. The crotamine product fused with MBP at its N-terminus also inhibited the hKv1.3 channel, suggesting that the N-terminus is not involved in the channel binding of the toxin.


Subject(s)
Crotalid Venoms/analysis , Kv1.3 Potassium Channel/antagonists & inhibitors , Maltose-Binding Proteins/metabolism , Crotalid Venoms/isolation & purification , Crotalid Venoms/metabolism , Escherichia coli , Inhibitory Concentration 50 , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Biol Pharm Bull ; 37(9): 1495-504, 2014.
Article in English | MEDLINE | ID: mdl-25177033

ABSTRACT

The effects of paroxetine, a selective serotonin reuptake inhibitor, on human ether-a-go-go-related gene (HERG) channels were investigated using the whole-cell patch-clamp technique. The HERG channels were stably expressed in human embryonic kidney cells. Paroxetine inhibited the peak tail currents of the HERG channel in a concentration-dependent manner, with an IC50 value of 0.45 µM and a Hill coefficient of 0.85. These effects were reversible after wash-out of the drug. The paroxetine-induced inhibition of the HERG channels was voltage-dependent. There was a steep increase in inhibition over the voltage range of the channel opening. Also, a shallow voltage-dependent inhibition was detected over the voltage range in which the channels were fully activated. The fractional electrical distance was estimated to be 0.11. Paroxetine induced a leftward shift in the voltage-dependence of the steady-state activation of the HERG channels. Before and after application of the 1 µM paroxetine, the half-maximum activation was -14.21 mV and -27.04 mV, respectively, with no shift in the slope value. The HERG channel block was not use-dependent. The characteristics of the block were dependent on open and inactivated channel states rather than closed state. Paroxetine had no effect on activation and deactivation kinetics, steady-state inactivation. These results suggest that paroxetine blocks the HERG channels by binding to these channels in the open and inactivated states.


Subject(s)
Antidepressive Agents/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Paroxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/physiology , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Models, Molecular
18.
Brain Res ; 1589: 54-60, 2014 11 17.
Article in English | MEDLINE | ID: mdl-25014275

ABSTRACT

Cyclooxygenase (COX) products and pattern recognition receptors are important modulators of neuroinflammation; however, the role of prostaglandins and toll-like receptor (TLR) signaling and the functional crosstalk between COX modulators remains unclear, especially in astrocytes that closely modulate neuronal functions. Here, we studied the effect of prostaglandins on toll-like receptor 3 (TLR3)-induced cytokine expression in human astroglioma CRT-MG cells. Prostaglandin E2 (PGE2) was shown to increase cytosolic cAMP levels in an EP2 receptor dependent manner. Interestingly, the TLR3 agonist polyinosinic:polycytidylic acid (poly(I:C)) mediated phosphorylation of NF-κB and extracellular stress-related kinase 1/2 (ERK1/2), which significantly decreased following PGE2 treatment. In addition, PGE2 increased the phosphorylation and inactivation of glycogen synthesis kinase-3ß (GSK-3ß), whereas poly(I:C) decreased it. We observed that PGE2 decreased tumor necrosis factor-α (TNF-α) production evoked by poly(I:C), whereas PGE2 potentiated poly(I:C)-triggered interleukin-8 (IL-8) production. These results suggest that prostaglandin modulates the TLR3-mediated cytokine profile in astrocytes via EP2 receptors and regulates the NF-κB, ERK1/2 and GSK-3ß signaling pathways.


Subject(s)
Astrocytoma/metabolism , Cytokines/metabolism , Prostaglandins/metabolism , Toll-Like Receptor 3/metabolism , Animals , Astrocytes/metabolism , Blotting, Western , Cell Line, Tumor , Cerebral Cortex/metabolism , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , NF-kappa B/metabolism , Phosphorylation/physiology , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 3/agonists
19.
Brain Res ; 1557: 34-42, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24560601

ABSTRACT

Melatonin is involved in various neuronal functions such as circadian rhythmicity and thermoregulation. Melatonin has a wide range of pharmacologically effective concentration levels from the nanomolar to millimolar levels. Recently, the antiepileptic effect of high dose melatonin has been the focus of clinical studies; however, its detailed mechanism especially in relation to neurotransmitter release and synaptic transmission remains unclear. We studied the effect of melatonin at high concentrations on the neurotransmitter release by monitoring norepinephrine release in PC12 cells, and excitatory postsynaptic potential in rat hippocampal slices. Melatonin inhibits the 70mM K(+)-induced Ca(2+) increase at millimolar levels without effect on bradykinin-triggered Ca(2+) increase in PC12 cells. Melatonin (1mM) did not affect A2A adenosine receptor-evoked cAMP production, and classical melatonin receptor antagonists did not reverse the melatonin-induced inhibitory effect, suggesting G-protein coupled receptor independency. Melatonin inhibits the 70mM K(+)-induced norepinephrine release at a similar effective concentration range in PC12 cells. We confirmed that melatonin (100µM) inhibits excitatory synaptic transmission of the hippocampal Schaffer collateral pathway with the decrease in basal synaptic transmission and the increase in paired pulse ratio. These results show that melatonin inhibits neurotransmitter release through the blocking of voltage-sensitive Ca(2+) channels and suggest a possible mechanism for the antiepileptic effect of melatonin.


Subject(s)
Calcium Channel Blockers/pharmacology , Hippocampus/drug effects , Melatonin/pharmacology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cyclic AMP/metabolism , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , HEK293 Cells , Hippocampus/physiology , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Norepinephrine/metabolism , PC12 Cells , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/metabolism , Receptors, Melatonin/agonists , Receptors, Melatonin/antagonists & inhibitors , Receptors, Melatonin/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
20.
Proc Natl Acad Sci U S A ; 111(4): 1551-6, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24453217

ABSTRACT

Chronic neurohormonal and mechanical stresses are central features of heart disease. Increasing evidence supports a role for the transient receptor potential canonical channels TRPC3 and TRPC6 in this pathophysiology. Channel expression for both is normally very low but is increased by cardiac disease, and genetic gain- or loss-of-function studies support contributions to hypertrophy and dysfunction. Selective small-molecule inhibitors remain scarce, and none target both channels, which may be useful given the high homology among them and evidence of redundant signaling. Here we tested selective TRPC3/6 antagonists (GSK2332255B and GSK2833503A; IC50, 3-21 nM against TRPC3 and TRPC6) and found dose-dependent blockade of cell hypertrophy signaling triggered by angiotensin II or endothelin-1 in HEK293T cells as well as in neonatal and adult cardiac myocytes. In vivo efficacy in mice and rats was greatly limited by rapid metabolism and high protein binding, although antifibrotic effects with pressure overload were observed. Intriguingly, although gene deletion of TRPC3 or TRPC6 alone did not protect against hypertrophy or dysfunction from pressure overload, combined deletion was protective, supporting the value of dual inhibition. Further development of this pharmaceutical class may yield a useful therapeutic agent for heart disease management.


Subject(s)
Cardiomegaly/genetics , TRPC Cation Channels/antagonists & inhibitors , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Rats , TRPC Cation Channels/genetics , TRPC6 Cation Channel
SELECTION OF CITATIONS
SEARCH DETAIL
...