Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
2.
Int Immunopharmacol ; 74: 105706, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31254955

ABSTRACT

Linalool is a natural product present in fruits and aromatic plants with biological activities. Researchers have reported that the inhalation of linalool exerts anti-inflammatory activities. In this study, we examined the therapeutic effects of linalool on airway inflammation and mucus overproduction in mice with allergic asthma. Oral administration of linalool significantly inhibited the levels of eosinophil numbers, Th2 cytokines and immunoglobulin E (IgE) caused by ovalbumin (OVA) exposure. Linalool exerted preventive effects against the influx of inflammatory cells and mucus hypersecretion in the lung tissues. Linalool also dose-dependently decreased the levels of inducible nitric oxide synthase (iNOS) expression and protein kinase B (AKT) activation in the lung tissues. Linalool effectively downregulated the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) caused by OVA exposure. Furthermore, linalool exerted inhibitory effect on OVA-induced airway hyperresponsiveness (AHR). In the in vitro study, the increased secretion of MCP-1 was attenuated with linalool treatment in lipopolysaccharide (LPS)-stimulated H292 airway epithelial cells. In conclusion, linalool effectively exerts a protective role in OVA-induced airway inflammation and mucus hypersecretion, and its protective effects are closely related to the downregulation of inflammatory mediators and MAPKs/NF-κB signaling.


Subject(s)
Acyclic Monoterpenes/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Hypersensitivity/drug therapy , Lung/metabolism , Respiratory Hypersensitivity/drug therapy , Th2 Cells/immunology , Administration, Oral , Allergens/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Immunoglobulin E/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Ovalbumin/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
3.
Int J Mol Med ; 43(4): 1830-1838, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816433

ABSTRACT

Physalis peruviana L. (PP) is well known for its various properties, including its antioxidant property. In our previous study, the protective effects of PP against cigarette smoke­induced airway inflammation were confirmed. The purpose of the present study was to evaluate the anti­inflammatory effect of PP against ovalbumin (OVA)­induced airway inflammation. Treatment with PP inhibited the numbers of eosinophils and the levels of inflammatory cytokines, including interleukin (IL)­4, IL­5 and IL­13, in the bronchoalveolar lavage fluid (BALF) of animal models with OVA­induced allergic asthma. PP also significantly decreased the production of total immunoglobulin E in the serum. Lung sections stained with hematoxylin and eosin revealed that the influx of inflammatory cells was decreased in the lungs of mice treated with PP compared with cells in the OVA group. The increased expression levels of monocyte chemoattractant protein­1 (MCP­1) and T cell marker KEN­5 were also reduced following PP treatment in the lung tissues compared with those in the OVA group. The PAS staining results showed that PP attenuated the overproduction of mucus in the lung. Additionally, western blot analysis revealed that PP significantly downregulated the activation of nuclear factor­κB/p38 mitogen­activated protein kinase/c­Jun N­terminal kinase, and upregulated the expression of heme oxgenase­1 in the lungs. In an in vitro experiment, PP effectively reduced the levels of LPS­stimulated MCP­1 in a concentration­dependent manner. Taken together, these results indicate that PP has considerable potential in the treatment of allergic asthma.


Subject(s)
Inflammation/drug therapy , Lung/pathology , NF-kappa B/metabolism , Physalis/chemistry , Plant Extracts/therapeutic use , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Chemokine CCL2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Enzyme Activation/drug effects , Eosinophils/drug effects , Eosinophils/pathology , Female , Heme Oxygenase (Decyclizing)/metabolism , Immunoglobulin E/blood , Inflammation/blood , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mucus/metabolism , Ovalbumin , Plant Extracts/pharmacology , RAW 264.7 Cells , p38 Mitogen-Activated Protein Kinases/metabolism
4.
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
5.
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
6.
J Pharmacol Sci ; 123(1): 67-77, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-24005046

ABSTRACT

Azelastine is a second generation histamine H1-receptor antagonist used as an anti-asthmatic and anti-allergic drug that can induce QT prolongation and torsades de pointes. We investigated the acute effects of azelastine on human ether-a-go-go-related gene (hERG) channels, action potential duration (APD), and L-type (I(Ca,L)) and T-type Ca²âº current (I(Ca,T)) to determine the electrophysiological basis for its proarrhythmic potential. Azelastine increased the APD at 90% of repolarization concentration dependently, with an IC50 of 1.08 nM in guinea-pig ventricular myocytes. We examined the effects of azelastine on the hERG channels expressed in Xenopus oocytes and HEK293 cells using two-microelectrode voltage-clamp and patch-clamp techniques. Azelastine induced a concentration-dependent decrease of the hERG current amplitude at the end of the voltage steps and tail currents. The IC50 for the azelastine-induced block of the hERG currents expressed in HEK293 cells was 11.43 nM, while the drug inhibited I(Ca,L) and I(Ca,T) with IC50 values of 7.60 and 26.21 µM, respectively. The S6 domain mutations, Y652A partially attenuated and F656A abolished hERG current block. These results suggest that azelastine is a potent blocker of hERG channels rather than I(Ca,L) or I(Ca,T), providing molecular mechanisms for the arrhythmogenic side effects during the clinical administration of azelastine.


Subject(s)
Action Potentials/drug effects , Arrhythmias, Cardiac/chemically induced , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Electrophysiological Phenomena/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Histamine H1 Antagonists/adverse effects , Histamine H1 Antagonists/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Phthalazines/adverse effects , Phthalazines/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Guinea Pigs , HEK293 Cells , Heart Ventricles/cytology , Humans , Oocytes , Patch-Clamp Techniques , Protein Structure, Tertiary , Xenopus laevis
7.
Vascul Pharmacol ; 59(3-4): 90-5, 2013.
Article in English | MEDLINE | ID: mdl-23876554

ABSTRACT

The effect of efonidipine, a commercially available antihypertensive drug and Ca(2+) channel inhibitor, on voltage-dependent K(+) (Kv) channels was studied in freshly isolated rabbit coronary arterial smooth muscle cells using the whole-cell patch clamp technique. The amplitude of Kv current was decreased by application of efonidipine in a dose-dependent manner, with IC50 of 0.26µM and a Hill coefficient of 0.91, which suggests 1:1 binding stoichiometry. Efonidipine did not affect voltage-dependent activation of the Kv channel, but shifted the inactivation curve by -8.87mV. The inhibitory effect of efonidipine was not significantly changed by depletion of extracellular Ca(2+) or intracellular ATP, which indicated no involvement of the Ca(2+) channel or intracellular protein kinase-dependent cascades. We conclude that efonidipine dose-dependently inhibits Kv current in a phosphorylation- and Ca(2+) channel-independent manner.


Subject(s)
Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Nitrophenols/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/drug effects , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacology , Calcium Channel Blockers/administration & dosage , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Dihydropyridines/administration & dosage , Dose-Response Relationship, Drug , Inhibitory Concentration 50 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitrophenols/administration & dosage , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacology , Patch-Clamp Techniques , Phosphorylation , Potassium Channel Blockers/administration & dosage , Potassium Channels, Voltage-Gated/metabolism , Rabbits
8.
Eur J Pharmacol ; 702(1-3): 165-73, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23395964

ABSTRACT

Fluphenazine is a potent antipsychotic drug that can increase action potential duration and induce QT prolongation in several animal models and in humans. As the block of cardiac human ether-a-go-go-related gene (hERG) channels is one of the leading causes of acquired long QT syndrome, we investigated the acute effects of fluphenazine on hERG channels to determine the electrophysiological basis for its proarrhythmic potential. Fluphenazine at concentrations of 0.1-1.0 µM increased the action potential duration at 90% of repolarization (APD90) and action potential duration at 50% of repolarization (APD50) in 5 min when action potentials were elicited under current-clamp conditions in guinea pig ventricular myocytes. We examined the effects of fluphenazine on hERG channels expressed in Xenopus oocytes and HEK293 cells using two-microelectrode voltage-clamp and patch-clamp techniques. The IC50 for the fluphenazine-induced block of hERG currents in HEK293 cells at 36 °C was 0.102 µM at +20 mV. Fluphenazine-induced a concentration-dependent decrease of the current amplitude at the end of the voltage steps and hERG tail currents. The fluphenazine-dependent hERG block in Xenopus oocytes increased progressively relative to the degree of depolarization. Fluphenazine affected the channels in the activated and inactivated states but not in the closed states, and the S6 domain mutation from tyrosine to alanine at amino acid 652 (Y652A) attenuated the hERG current block. These results suggest that the antipsychotic drug fluphenazine is a potent blocker of hERG channels, providing a molecular mechanism for the drug-induced arrhythmogenic side effects.


Subject(s)
Action Potentials/drug effects , Antipsychotic Agents/administration & dosage , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Fluphenazine/administration & dosage , Potassium Channel Blockers/administration & dosage , Animals , Ether-A-Go-Go Potassium Channels/physiology , Guinea Pigs , HEK293 Cells , Humans , Molecular Docking Simulation , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Oocytes/drug effects , Oocytes/physiology , Xenopus laevis
9.
Toxicol Appl Pharmacol ; 262(1): 60-9, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22676973

ABSTRACT

Polychlorinated biphenyls (PCBs) have been known as serious persistent organic pollutants (POPs), causing developmental delays and motor dysfunction. We have investigated the effects of two PCB congeners, 3,3',4,4'-tetrachlorobiphenyl (PCB 77) and 3,3',4,4',5-pentachlorobiphenyl (PCB 126) on ECG, action potential, and the rapidly activating delayed rectifier K+ current (I(Kr)) of guinea pigs' hearts, and hERG K+ current expressed in Xenopus oocytes. PCB 126 shortened the corrected QT interval (QTc) of ECG and decreased the action potential duration at 90% (APD(90)), and 50% of repolarization (APD50) (P<0.05) without changing the action potential duration at 20% (APD20). PCB 77 decreased APD20 (P<0.05) without affecting QTc, APD90, and APD50. The PCB 126 increased the I(Kr) in guinea-pig ventricular myocytes held at 36°C and hERG K+ current amplitude at the end of the voltage steps in voltage-dependent mode (P<0.05); however, PCB 77 did not change the hERG K+ current amplitude. The PCB 77 increased the diastolic Ca²âº and decreased Ca²âº transient amplitude (P<0.05), however PCB 126 did not change. The results suggest that PCB 126 shortened the QTc and decreased the APD90 possibly by increasing I(Kr), while PCB 77 decreased the APD20 possibly by other modulation related with intracellular Ca²âº. The present data indicate that the environmental toxicants, PCBs, can acutely affect cardiac electrophysiology including ECG, action potential, intracellular Ca²âº, and channel activity, resulting in toxic effects on the cardiac function in view of the possible accumulation of the PCBs in human body.


Subject(s)
Action Potentials/drug effects , Delayed Rectifier Potassium Channels/drug effects , Ether-A-Go-Go Potassium Channels/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Calcium/metabolism , Delayed Rectifier Potassium Channels/metabolism , ERG1 Potassium Channel , Electrocardiography , Environmental Pollutants/toxicity , Ether-A-Go-Go Potassium Channels/metabolism , Guinea Pigs , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oocytes , Xenopus laevis
10.
Biochem Biophys Res Commun ; 394(3): 536-41, 2010 Apr 09.
Article in English | MEDLINE | ID: mdl-20211602

ABSTRACT

Desipramine is a tricyclic antidepressant for psychiatric disorders that can induce QT prolongation, which may lead to torsades de pointes. Since blockade of cardiac human ether-a-go-go-related gene (hERG) channels is an important cause of acquired long QT syndrome, we investigated the acute effects of desipramine on hERG channels to determine the electrophysiological basis for its pro-arrhythmic potential. We examined the effects of desipramine on the hERG channels expressed in Xenopus oocytes using two-microelectrode voltage-clamp techniques. Desipramine-induced concentration-dependent decreases in the current amplitude at the end of the voltage steps and hERG tail currents. The IC(50) for desipramine needed to block the hERG current in Xenopus oocytes decreased progressively relative to the degree of depolarization. Desipramine affected the channels in the activated and inactivated states but not in the closed states. The S6 domain mutations, Tyr-652 located in the S6 domain of the hERG channel reduced the potency of the channel block by desipramine more than a mutation of Phe-656 in the same region. These results suggest that desipramine is a blocker of the hERG channels, providing a molecular mechanism for the arrhythmogenic side effects during the clinical administration of desipramine.


Subject(s)
Antidepressive Agents, Tricyclic/adverse effects , Desipramine/adverse effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Long QT Syndrome/chemically induced , Animals , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Humans , Inhibitory Concentration 50 , Mutation , Oocytes , Phenylalanine/genetics , Protein Structure, Tertiary/genetics , Tyrosine/genetics , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...