Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Heart Rhythm ; 21(2): 184-196, 2024 Feb.
Article En | MEDLINE | ID: mdl-37924963

BACKGROUND: More than a hundred genetic loci have been associated with atrial fibrillation (AF). But the exact mechanism remains unclear and the treatment needs to be improved. OBJECTIVE: This study aimed to investigate the mechanism and potential treatment of NPPA mutation-associated AF. METHODS: Nppa knock-in (KI, p.I137T) rats were generated, and cardiac function was evaluated. Blood pressure was recorded using a tail-cuff system. The expression levels were measured using real-time polymerase chain reaction, enzyme-linked immunosorbent assay or Western blot analysis, and RNA-sequence analysis. Programmed electrical stimulation, patch clamp, and multielectrode array were used to record the electrophysical characteristics. RESULTS: Mutant rats displayed downregulated expression of atrial natriuretic peptide but elevated blood pressure and enlarged left atrial end-diastolic diameter. Further, gene topology analysis suggested that the majority of differently expressed genes in Nppa KI rats were related to inflammation, electrical remodeling, and structural remodeling. The expression levels of C-C chemokine ligand 5 and galectin-3 involved in remodeling were higher, while there were declined levels of Nav1.5, Cav1.2, and connexin 40. AF was more easily induced in KI rats. Electrical remodeling included abbreviated action potentials, effective refractory period, increased late sodium current, and reduced calcium current, giving rise to conduction abnormalities. These electrophysiological changes could be reversed by the late sodium current blocker ranolazine and the Nav1.8 blocker A-803467. CONCLUSION: Our findings suggest that structural remodeling related to inflammation and fibrosis and electrical remodeling involved in late sodium current underly the major effects of the Nppa (p.I137T) variant to induce AF, which can be attenuated by the late sodium current blocker and Nav1.8 blocker.


Atrial Fibrillation , Atrial Remodeling , Procainamide , Animals , Rats , Action Potentials/physiology , Atrial Fibrillation/drug therapy , Atrial Fibrillation/genetics , Atrial Natriuretic Factor , Atrial Remodeling/physiology , Heart Atria , Inflammation/metabolism , Mutation , Myocytes, Cardiac/metabolism , Procainamide/analogs & derivatives , Sodium/metabolism
2.
Biochem Biophys Res Commun ; 691: 149322, 2024 Jan 08.
Article En | MEDLINE | ID: mdl-38039833

BACKGROUND: Bupleurum (Bup), is a traditional effective medicine to treat colds and fevers in clinics. Multiple studies have demonstrated that Bup exhibites various biological activities, including cardioprotective effects, anti-inflammatory, anticancer, antipyretic, antimicrobial, and antiviral effects, etc. Currently, the effects of Bup on cardiac electrophysiology have not been reported yet. METHODS: Electrocardiogram recordings were used to investigate the effects of Bup on aconitine-induced arrhythmias. Patch-clamp techniques were used to explore the effects of Bup on APs and ion currents. RESULTS: Bup reduced the incidence of ventricular fibrillation (VF) and delayed the onset time of ventricular tachycardia (VT) in mice. Additionally, Bup (40 mg/mL) suppressed DADs induced by high-Ca2+ and shortened action potential duration at 50 % completion of repolarization (APD50) and action potential duration at 90 % completion of repolarization (APD90) to 60.89 % ± 8.40 % and 68.94 % ± 3.24 % of the control, respectively. Moreover, Bup inhibited L-type calcium currents (ICa.L) in a dose-dependent manner, with an IC50 value of 25.36 mg/mL. Furthermore, Bup affected the gated kinetics of L-type calcium channels by slowing down steady-state activation, accelerating the steady-state inactivation, and delaying the inactivation-recovery process. However, Bup had no effects on the Transient sodium current (INa.T), ATX II-increased late sodium current (INa.L), transient outward current (Ito), delayed rectifier potassium current (IK), or inward rectifier potassium current (IK1). CONCLUSION: Bup is an antiarrhythmic agent that may exert its antiarrhythmic effects by inhibiting L-type calcium channels.


Bupleurum , Calcium Channels, L-Type , Mice , Animals , Bupleurum/metabolism , Myocytes, Cardiac/metabolism , Anti-Arrhythmia Agents/adverse effects , Arrhythmias, Cardiac , Sodium/metabolism , Potassium/pharmacology , Action Potentials
3.
J Electrocardiol ; 80: 69-80, 2023.
Article En | MEDLINE | ID: mdl-37262953

INTRODUCTION: Naringin, a flavonoid extracted from citrus plants, has a variety of biological effects. Studies have shown that increasing the consumption of flavonoid-rich foods can reduce the incidence of cardiac arrhythmia. Naringin has been reported to have beneficial cardiovascular effects and thus can be used to prevent cardiovascular diseases, but the electrophysiological mechanism through which it prevents arrhythmias has not been elucidated. This study was conducted to investigate the effect of naringin on the transmembrane ion channel currents in mouse ventricular myocytes and the antiarrhythmic effect of this compound on Langendorff-perfused mouse hearts. METHODS: Action potentials (APs) and ionic currents were recorded in isolated ventricular myocytes using the whole-cell patch-clamp technique. Anemone toxin II (ATX II) and CaCl2 were used to induce early afterdepolarizations (EADs) and delayed afterdepolarizations (DADs), respectively. Electrocardiogram (ECG) recordings were conducted in Langendorff-perfused mouse hearts with a BL-420F biological signal acquisition and analysis system. RESULTS: At the cellular level, naringin shortened the action potential duration (APD) of ventricular myocytes and decreased the maximum depolarization velocity (Vmax) of APs.Naringin inhibited the L-type calcium current (ICa.L) and ATX II enhanced the late sodium current (INa.L) in a concentration-dependent manner with IC50 values of 508.5 µmol/L (n = 9) and 311.6 µmol/L (n = 10), respectively. In addition, naringin also inhibited the peak sodium current (INa·P) and delayed the rectifier potassium current (IK) and the transient outward potassium current (Ito). Moreover, naringin reduced ATX II-induced APD prolongation and EADs and had a significant inhibitory effect on CaCl2-induced DADs as well. At the organ level, naringin reduced the incidence of ventricular tachycardia (VT) and ventricular fibrillation (VF) induced by ATX II and shortened the duration of both in isolated hearts. CONCLUSION: Naringin can inhibit the occurrence of EADs and DADs at the cellular level; furthermore, it can inhibit INa.L, ICa.L, INa·P, IK, and Ito in ventricular myocytes. Naringin also inhibits arrhythmias induced by ATX II in hearts. By investigating naringin with this electrophysiological method for the first time, we determined that this flavonoid may be a multichannel blocker with antiarrhythmic effects.


Flavanones , Myocytes, Cardiac , Mice , Animals , Calcium Chloride/pharmacology , Electrocardiography , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/prevention & control , Flavanones/pharmacology , Action Potentials , Sodium/pharmacology , Potassium
4.
JCI Insight ; 4(7)2019 04 04.
Article En | MEDLINE | ID: mdl-30779710

Iron deficiency is present in ~50% of heart failure (HF) patients. Large multicenter trials have shown that treatment of iron deficiency with i.v. iron benefits HF patients, but the underlying mechanisms are not known. To investigate the actions of iron deficiency on the heart, mice were fed an iron-depleted diet, and some received i.v. ferric carboxymaltose (FCM), an iron supplementation used clinically. Iron-deficient animals became anemic and had reduced ventricular ejection fraction measured by magnetic resonance imaging. Ca2+ signaling, a pathway linked to the contractile deficit in failing hearts, was also significantly affected. Ventricular myocytes isolated from iron-deficient animals produced smaller Ca2+ transients from an elevated diastolic baseline but had unchanged sarcoplasmic reticulum (SR) Ca2+ load, trigger L-type Ca2+ current, or cytoplasmic Ca2+ buffering. Reduced fractional release from the SR was due to downregulated RyR2 channels, detected at protein and message levels. The constancy of diastolic SR Ca2+ load is explained by reduced RyR2 permeability in combination with right-shifted SERCA activity due to dephosphorylation of its regulator phospholamban. Supplementing iron levels with FCM restored normal Ca2+ signaling and ejection fraction. Thus, 2 Ca2+-handling proteins previously implicated in HF become functionally impaired in iron-deficiency anemia, but their activity is rescued by i.v. iron supplementation.


Anemia, Iron-Deficiency/pathology , Heart Failure/etiology , Myocardial Contraction , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Administration, Intravenous , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/drug therapy , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Down-Regulation , Ferric Compounds/administration & dosage , Heart Failure/diagnosis , Heart Failure/prevention & control , Humans , Iron/blood , Magnetic Resonance Imaging , Male , Maltose/administration & dosage , Maltose/analogs & derivatives , Mice , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac , Primary Cell Culture , Sarcoplasmic Reticulum/pathology , Stroke Volume
5.
Sci Rep ; 9(1): 20425, 2019 12 31.
Article En | MEDLINE | ID: mdl-31892729

Ginsenoside Rb1 exerts its pharmacological action by regulating sodium, potassium and calcium ion channels in the membranes of nerve cells. These ion channels are also present in cardiomyocytes, but no studies have been reported to date regarding the effects of Rb1 on cardiac sodium currents (INa), L-type calcium currents (ICaL) and action potentials (APs). Additionally, the antiarrhythmic potential of Rb1 has not been assessed. In this study, we used a whole-cell patch clamp technique to assess the effect of Rb1 on these ion channels. The results showed that Rb1 inhibited INa and ICaL, reduced the action potential amplitude (APA) and maximum upstroke velocity (Vmax), and shortened the action potential duration (APD) in a concentration-dependent manner but had no effect on the inward rectifier potassium current (IK1), delayed rectifier potassium current (IK) or resting membrane potential (RMP). We also designed a pathological model at the cellular and organ level to verify the role of Rb1. The results showed that Rb1 abolished high calcium-induced delayed afterdepolarizations (DADs), depressed the increase in intracellular calcium ([Ca2+]i), relieved calcium overload and protected cardiomyocytes. Rb1 can also reduce the occurrence of ventricular premature beats (VPBs) and ventricular tachycardia (VT) in ischemia-reperfusion (I-R) injury.


Action Potentials/drug effects , Anti-Arrhythmia Agents/pharmacology , Calcium Channels/metabolism , Ginsenosides/pharmacology , Myocytes, Cardiac/drug effects , Sodium Channels/metabolism , Animals , Calcium/metabolism , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rabbits , Sodium/metabolism
6.
Pacing Clin Electrophysiol ; 40(12): 1412-1425, 2017 Dec.
Article En | MEDLINE | ID: mdl-28972668

BACKGROUND: An increase in the late sodium current (INaL ) causes intracellular Na+ overload and subsequently intracellular Ca2+ ([Ca2+ ]i ) overload via the stimulated reverse Na+ -Ca2+ exchange (NCX). Wenxin Keli (WXKL) is an effective antiarrhythmic Chinese herb extract, but the underlying mechanisms are unclear. METHODS AND RESULTS: The INaL , NCX current (INCX ), L-type Ca2+ current (ICaL ), and action potentials were recorded using the whole-cell patch-clamp technique in rabbit ventricular myocytes. Myocyte [Ca2+ ]i transients were measured using a dual excitation fluorescence photomultiplier system. WXKL decreased the enhanced INaL , reverse INCX , diastolic [Ca2+ ]i , and the amplitude of Ca2+ transients induced by sea anemone toxin II (ATX II, a specific INaL channel opener) in a concentration-dependent manner. Hypoxia increased INaL , INCX , and diastolic [Ca2+ ]i , and decreased amplitude of [Ca2+ ]i transients. Hypoxia-reoxygenation aggravated these changes and induced spontaneous [Ca2+ ]i transients and hypercontraction in 86% cells (6/7). The application of WXKL during hypoxia or reoxygenation periods decreased the increased INaL , INCX , and diastolic [Ca2+ ]i , and prevented those events in 82% cells (9/11) under hypoxia-reoxygenation conditions. WXKL also inhibited the ICaL in a dose-dependent manner. Furthermore, WXKL shortened the action potential duration and completely abolished ATX II-induced early afterdepolarizations from 9/9 to /9. In isolated heart electrocardiogram recordings, WXKL inhibited ischemia-reperfusion induced ventricular premature beats and tachycardia. CONCLUSIONS: WXKL attenuated [Ca2+ ]i overload induced by hypoxia-reoxygenation in ventricular myocytes through inhibiting INaL and ICaL and prevents arrhythmias. This could, at least partly, contribute to the antiarrhythmic effects of WXKL.


Anti-Arrhythmia Agents/pharmacology , Calcium/metabolism , Drugs, Chinese Herbal/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Sodium/metabolism , Animals , Cell Hypoxia/physiology , Female , Male , Rabbits
7.
Exp Physiol ; 102(7): 818-834, 2017 07 01.
Article En | MEDLINE | ID: mdl-28436159

NEW FINDINGS: What is the central question of this study? Hypoxia-induced increase in late sodium current (INa,L ) is associated with conditions causing cellular Ca2+ overload and contributes to arrhythmogenesis in the ventricular myocardium. The INa,L is an important drug target. We investigated intracellular signal transduction pathways involved in modulation of INa,L during hypoxia. What is the main finding and its importance? Hypoxia caused increases in INa,L , reverse Na+ -Ca2+ exchange current and diastolic [Ca2+ ], which were attenuated by inhibitors of Ca2+ -calmodulin-dependent protein kinase II (CaMKII) and protein kinase C and by a Ca2+ chelator. The findings suggest that CaMKII, protein kinase C and Ca2+ all participate in mediation of the effect of hypoxia to increase INa,L . Hypoxia leads to augmentation of the late sodium current (INa,L ) and cellular Na+ loading, increased reverse Na+ -Ca2+ exchange current (reverse INCX ) and intracellular Ca2+ loading in rabbit ventricular myocytes. The purpose of this study was to determine the intracellular signal transduction pathways involved in the modulation of INa,L during hypoxia in ventricular myocytes. Whole-cell and cell-attached patch-clamp techniques were used to record INa,L , and the whole-cell mode was also used to record reverse INCX and to study intercellular signal transduction mechanisms that mediate the increased INa,L . Dual excitation fluorescence photomultiplier systems were used to record the calcium transient in ventricular myocytes. Hypoxia caused increases of INa,L and reverse INCX . These increases were attenuated by KN-93 (an inhibitor of Ca2+ -calmodulin-dependent protein kinase II), bisindolylmaleimide VI (BIM; an inhibitor of protein kinase C) and BAPTA AM (a Ca2+ chelator). KN-93, BIM and BAPTA AM had no effect on INa,L in normoxia. In studies of KN-93, hypoxia alone increased the density of INa,L from -0.31 ± 0.02 to -0.66 ± 0.03 pA pF-1 (n = 6, P < 0.01 versus control) and the density of reverse INCX from 1.02 ± 0.06 to 1.91 ± 0.20 pA pF-1 (n = 7, P < 0.01 versus control) in rabbit ventricular myocytes. In the presence of 1 µm KN-93, the densities of INa,L and reverse INCX during hypoxia were significantly attenuated to -0.44 ± 0.03 (n = 6, P < 0.01 versus hypoxia) and 1.36 ± 0.15 pA pF-1 (n = 7, P < 0.01 versus hypoxia), respectively. In studies of BIM, hypoxia increased INa,L from -0.30 ± 0.03 to -0.60 ± 0.03 pA pF-1 (n = 6, P < 0.01 versus control) and reverse INCX from 0.91 ± 0.10 to 1.71 ± 0.27 pA pF-1 (n = 6, P < 0.01 versus control). In the presence of 1 µm BIM, the densities of INa,L and reverse INCX during hypoxia were significantly attenuated to -0.48 ± 0.02 (n = 6, P < 0.01 versus hypoxia) and 1.33 ± 0.21 pA pF-1 (n = 6, P < 0.01 versus hypoxia), respectively. In studies of BAPTA AM, hypoxia increased INa,L from -0.26 ± 0.04 to -0.63 ± 0.05 pA pF-1 (n = 6, P < 0.01 versus control) and reverse INCX from 0.86 ± 0.09 to 1.68 ± 0.35 pA pF-1 (n = 6, P < 0.01 versus control). The effects of hypoxia on INa,L and reverse INCX were significantly attenuated in the presence of 1 mm BAPTA AM to -0.39 ± 0.02 (n = 6, P < 0.01 versus hypoxia) and 1.12 ± 0.27 pA pF-1 (n = 6, P < 0.01 versus hypoxia), respectively. Results of single-channel studies showed that hypoxia apparently increased the mean open probability and mean open time of sodium channels. These effects were inhibited by either 1 µm KN-93 or 1 mm BAPTA AM. The suppressant effects of drug interventions were reversed upon washout. In addition, KN-93, BIM and BAPTA AM also reversed the hypoxia-enhanced diastolic Ca2+ concentration and the attenuated amplitude of the [Ca2+ ]i transient, maximal velocities of Ca2+ increase and Ca2+ decay. In summary, the findings suggest that Ca2+ -calmodulin-dependent protein kinase II, protein kinase C and Ca2+ all participate in mediation of the effect of hypoxia to increase INa,L .


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Sodium/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cell Hypoxia , Heart Ventricles/metabolism , Indoles/pharmacology , Maleimides/pharmacology , Patch-Clamp Techniques/methods , Rabbits , Sodium Channels/metabolism
8.
Pharmacology ; 99(5-6): 226-235, 2017.
Article En | MEDLINE | ID: mdl-28132058

AIMS: To investigate the effects of ketamine on human hyperpolarization-activated cyclic nucleotide-gated (hHCN) 1, 2, 4 channel currents expressed in Xenopus oocytes and spontaneous action potentials (APs) of rabbit sinoatrial node (SAN). METHODS: The 2-electrode voltage clamp and standard microelectrode techniques were respectively applied to record hHCN channels currents expressed in Xenopus oocytes and APs of SAN separated from rabbit heart. RESULTS: Ketamine (1-625 µmol/L) blocked hHCN1, 2, and 4 currents with IC50 of 67.0, 89.1, and 84.0 µmol/L, respectively, in a concentration-dependent manner. The currents were rapidly blocked by ketamine and partially recovered after washout. The steady-state activation curves of hHCN1, 2, and 4 currents demonstrated a concentration-dependent shift to the left and the rates of activation were significantly decelerated. But ketamine blocked hHCN channels in a voltage-independence and non-use-dependent manner, and did not modify the voltage dependence of activation and reversal potentials. Furthermore, ketamine suppressed phase-4 spontaneous depolarization rate in isolated rabbit SAN and decreased the beat rates in a concentration-dependent manner. CONCLUSION: Ketamine could inhibit hHCN channels expressed in Xenopus oocytes in a concentration-dependent manner as a close-state blocker and decrease beat rates of isolated rabbit SAN. This study may provide novel insights into other unexplained actions of ketamine.


Action Potentials/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Ketamine/pharmacology , Sinoatrial Node/drug effects , Animals , Dose-Response Relationship, Drug , Humans , Oocytes , Rabbits , Sinoatrial Node/metabolism , Transfection , Xenopus laevis
9.
Acta Pharmacol Sin ; 37(11): 1432-1441, 2016 Nov.
Article En | MEDLINE | ID: mdl-27569391

AIM: The augmentation of late sodium current (INa.L) not only causes intracellular Na+ accumulation, which results in intracellular Ca2+ overload via the reverse mode of the Na+/Ca2+ exchange current (reverse-INCX), but also prolongs APD and induces early afterdepolarizations (EAD), which can lead to arrhythmia and cardiac dysfunction. Thus, the inhibition of INa.L is considered to be a potential way for therapeutic intervention in ischemia and heart failure. In this study we investigated the effects of tolterodine (Tol), a competitive muscarinic receptor antagonist, on normal and veratridine (Ver)-augmented INa.L, reverse-INCX and APD in isolated rabbit ventricular myocytes, which might contribute to its cardioprotective activity. METHODS: Rabbit ventricular myocytes were prepared. The INa.L and reverse-INCX were recorded in voltage clamp mode, whereas action potentials and Ver-induced early afterdepolarizations (EADs) were recorded in current clamp mode. Drugs were applied via superfusion. RESULTS: Tol (3-120 nmol/L) concentration-dependently inhibited the normal and Ver-augmented INa.L with IC50 values of 32.08 nmol/L and 42.47 nmol/L, respectively. Atropine (100 µmol/L) did not affect the inhibitory effects of Tol (30 nmol/L) on Ver-augmented INa.L. In contrast, much high concentrations of Tol was needed to inhibit the transient sodium current (INa.T) with an IC50 value of 183.03 µmol/L. In addition, Tol (30 nmol/L) significantly shifted the inactivation curve of INa.T toward a more depolarizing membrane potential without affecting its activation characteristics. Moreover, Tol (30 nmol/L) significantly decreased Ver-augmented reverse-INCX. Tol (30 nmol/L) increased the action potential duration (APD) by 16% under the basal conditions. Ver (20 µmol/L) considerably extended the APD and evoked EADs in 18/24 cells (75%). In the presence of Ver, Tol (30 nmol/L) markedly decreased the APD and eliminated EADs (0/24 cells). CONCLUSION: Tol inhibits normal and Ver-augmented INaL and decreases Ver-augmented reverse-INCX. In addition, Tol reverses the prolongation of the APD and eliminates the EADs induced by Ver, thus prevents Ver-induced arrhythmia.


Anti-Arrhythmia Agents/pharmacology , Muscarinic Antagonists/pharmacology , Myocytes, Cardiac/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Sodium-Calcium Exchanger/metabolism , Tolterodine Tartrate/pharmacology , Veratridine/pharmacology , Action Potentials , Animals , Female , Heart Ventricles/cytology , In Vitro Techniques , Male , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Rabbits
10.
Acta Pharmacol Sin ; 36(11): 1327-36, 2015 Nov.
Article En | MEDLINE | ID: mdl-26456586

AIM: Intracellular Ca(2+) ([Ca(2+)]i) overload occurs in myocardial ischemia. An increase in the late sodium current (INaL) causes intracellular Na(+) overload and subsequently [Ca(2+)]i overload via the reverse-mode sodium-calcium exchanger (NCX). Thus, inhibition of INaL is a potential therapeutic target for cardiac diseases associated with [Ca(2+)]i overload. The aim of this study was to investigate the effects of ketamine on Na(+)-dependent Ca(2+) overload in ventricular myocytes in vitro. METHODS: Ventricular myocytes were enzymatically isolated from hearts of rabbits. INaL, NCX current (INCX) and L-type Ca(2+) current (ICaL) were recorded using whole-cell patch-clamp technique. Myocyte shortening and [Ca(2+)]i transients were measured simultaneously using a video-based edge detection and dual excitation fluorescence photomultiplier system. RESULTS: Ketamine (20, 40, 80 µmol/L) inhibited INaL in a concentration-dependent manner. In the presence of sea anemone toxin II (ATX, 30 nmol/L), INaL was augmented by more than 3-fold, while ketamine concentration-dependently suppressed the ATX-augmented INaL. Ketamine (40 µmol/L) also significantly suppressed hypoxia or H2O2-induced enhancement of INaL. Furthermore, ketamine concentration-dependently attenuated ATX-induced enhancement of reverse-mode INCX. In addition, ketamine (40 µmol/L) inhibited ICaL by 33.4%. In the presence of ATX (3 nmol/L), the rate and amplitude of cell shortening and relaxation, the diastolic [Ca(2+)]i, and the rate and amplitude of [Ca(2+)]i rise and decay were significantly increased, which were reverted to control levels by tetrodotoxin (TTX, 2 µmol/L) or by ketamine (40 µmol/L). CONCLUSION: Ketamine protects isolated rabbit ventricular myocytes against [Ca(2+)]i overload by inhibiting INaL and ICaL.


Calcium/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Myocytes, Cardiac/drug effects , Sodium-Calcium Exchanger/metabolism , Sodium/metabolism , Animals , Cell Hypoxia/drug effects , Cells, Cultured , Female , Hydrogen Peroxide/metabolism , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rabbits
11.
Exp Physiol ; 100(4): 399-409, 2015 Apr 01.
Article En | MEDLINE | ID: mdl-25641541

NEW FINDINGS: What is the central question of this study? What are the effects of protein kinase C (PKC) and Ca(2+) -calmodulin-dependent protein kinase II (CaMKII) on late sodium current (INaL ), reverse Na(+) -Ca(2+) exchange current (reverse INCX ) or intracellular Ca(2+) levels changed by ouabain? What is the main finding and its importance? Ouabain, even at low concentrations (0.5-8.0 µm), can increase INaL and reverse INCX , and these effects may contribute to the effect of the glycoside to increase Ca(2+) transients and contractility. Both PKC and CaMKII activities may mediate or modulate these processes. It has been reported that the cardiac glycoside ouabain can increase the late sodium current (INaL ), as well as the diastolic intracellular calcium concentration and contractile shortening. Whether an increase of INaL participates in a pathway that can mediate the positive inotropic response to ouabain is unknown. We therefore determined the effects of ouabain on INaL , reverse Na(+) -Ca(2+) exchange current (reverse INCX ), intracellular Ca(2+) ([Ca(2+) ]i ) levels and contractile shortening in rabbit isolated ventricular myocytes. Ouabain (0.1-8 µm) markedly increased INaL and reverse INCX in a concentration-dependent manner, with significant effects at concentrations as low as 0.5 and 1 µm. These effects of ouabain were suppressed by the INaL inhibitors TTX and ranolazine, the protein kinase C inhibitor bisindolylmaleimide and the Ca(2+) -calmodulin-dependent protein kinase II inhibitor KN-93. The enhancement by 0.5 µm ouabain of ventricular myocyte contractility and intracellular Ca(2+) transients was suppressed by 2.0 µm TTX. We conclude that ouabain, even at low concentrations (0.5-8.0 µm), can increase INaL and reverse INCX , and these effects may contribute to the effect of the glycoside to increase Ca(2+) transients and contractility. Both protein kinase C and Ca(2+) -calmodulin-dependent protein kinase II activities may mediate or modulate these processes.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Ouabain/administration & dosage , Protein Kinase C/metabolism , Sodium/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cardiotonic Agents/administration & dosage , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Heart Ventricles/cytology , Heart Ventricles/metabolism , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Rabbits
12.
Am J Physiol Heart Circ Physiol ; 306(3): H455-61, 2014 Feb.
Article En | MEDLINE | ID: mdl-24322614

An increase of cardiac late sodium current (INa.L) is arrhythmogenic in atrial and ventricular tissues, but the densities of INa.L and thus the potential relative contributions of this current to sodium ion (Na(+)) influx and arrhythmogenesis in atria and ventricles are unclear. In this study, whole-cell and cell-attached patch-clamp techniques were used to measure INa.L in rabbit left atrial and ventricular myocytes under identical conditions. The density of INa.L was 67% greater in left atrial (0.50 ± 0.09 pA/pF, n = 20) than in left ventricular cells (0.30 ± 0.07 pA/pF, n = 27, P < 0.01) when elicited by step pulses from -120 to -20 mV at a rate of 0.2 Hz. Similar results were obtained using step pulses from -90 to -20 mV. Anemone toxin II (ATX II) increased INa.L with an EC50 value of 14 ± 2 nM and a Hill slope of 1.4 ± 0.1 (n = 9) in atrial myocytes and with an EC50 of 21 ± 5 nM and a Hill slope of 1.2 ± 0.1 (n = 12) in ventricular myocytes. Na(+) channel open probability (but not mean open time) was greater in atrial than in ventricular cells in the absence and presence of ATX II. The INa.L inhibitor ranolazine (3, 6, and 9 µM) reduced INa.L more in atrial than ventricular myocytes in the presence of 40 nM ATX II. In summary, rabbit left atrial myocytes have a greater density of INa.L and higher sensitivities to ATX II and ranolazine than rabbit left ventricular myocytes.


Acetanilides/pharmacology , Cnidarian Venoms/pharmacology , Heart Atria/cytology , Heart Ventricles/cytology , Myocytes, Cardiac/physiology , Piperazines/pharmacology , Sodium Channel Blockers/pharmacology , Sodium/metabolism , Action Potentials , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organ Specificity , Rabbits , Ranolazine
13.
J Pharm Pharmacol ; 65(9): 1321-8, 2013 Sep.
Article En | MEDLINE | ID: mdl-23927470

OBJECTIVES: The inhibition of the cardiac rapid delayed rectifier potassium current (IKr ) and its cloned equivalent human ether-a-go-go-related gene (hERG) channel illustrate QT interval prolonging effects of a wide range of clinically used drugs. In this study, the direct interaction of the intravenous anaesthetic ketamine with wild-type (WT) and mutation hERG currents (IhERG ) was investigated. METHODS: The hERG channel (WT, Y652A and F656A) was expressed in Xenopus oocytes and studied using standard two-microelectrode voltage-clamp techniques. KEY FINDINGS: WT hERG is blocked in a concentration-dependent manner with IC50 = 12.05 ± 1.38 µm by ketamine, and the steady-state inactivation curves are shifted to more negative potentials (about -27 mV). The mutation to Ala of Y652 and F656 located on the S6 domain attenuate IhERG blockade by ketamine, and produced approximately 9-fold and 2.5-fold increases in IC50 compared with that of WT hERG channel, respectively. CONCLUSIONS: Ketamine blocks WT IhERG expressed in Xenopus oocytes in a concentration-dependent manner and predominantly interacts with the open hERG channels. The interaction of ketamine with hERG channel may involve the aromatic residues Tyr652 and Phe656.


Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ketamine/pharmacology , Potassium Channel Blockers/pharmacology , Amino Acids/metabolism , Animals , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/genetics , Humans , Inhibitory Concentration 50 , Membrane Potentials/drug effects , Mutation , Oocytes/drug effects , Oocytes/metabolism , Xenopus
14.
PLoS One ; 7(12): e51358, 2012.
Article En | MEDLINE | ID: mdl-23272101

BACKGROUND/AIMS: Resveratrol has been demonstrated to be protective in the cardiovascular system. The aim of this study was to assess the effects of resveratrol on hydrogen peroxide (H(2)O(2))-induced increase in late sodium current (I(Na.L)) which augmented the reverse Na(+)-Ca(2+) exchanger current (I(NCX)), and the diastolic intracellular Ca(2+) concentration in ventricular myocytes. METHODS: I(Na.L), I(NCX,) L-type Ca(2+) current (I(Ca.L)) and intracellular Ca(2+) properties were determined using whole-cell patch-clamp techniques and dual-excitation fluorescence photomultiplier system (IonOptix), respectively, in rabbit ventricular myocytes. RESULTS: Resveratrol (10, 20, 40 and 80 µM) decreased I(Na.L) in myocytes both in the absence and presence of H(2)O(2) (300 µM) in a concentration dependent manner. Ranolazine (3-9 µM) and tetrodotoxin (TTX, 4 µM), I(Na.L) inhibitors, decreased I(Na.L) in cardiomyocytes in the presence of 300 µM H(2)O(2). H(2)O(2) (300 µM) increased the reverse I(NCX) and this increase was significantly attenuated by either 20 µM resveratrol or 4 µM ranolazine or 4 µM TTX. In addition, 10 µM resveratrol and 2 µM TTX significantly depressed the increase by 150 µM H(2)O(2) of the diastolic intracellular Ca(2+) fura-2 fluorescence intensity (FFI), fura-fluorescence intensity change (△FFI), maximal velocity of intracellular Ca(2+) transient rise and decay. As expected, 2 µM TTX had no effect on I(Ca.L). CONCLUSION: Resveratrol protects the cardiomyocytes by inhibiting the H(2)O(2)-induced augmentation of I(Na.L.)and may contribute to the reduction of ischemia-induced lethal arrhythmias.


Calcium/metabolism , Heart Ventricles/metabolism , Hydrogen Peroxide/metabolism , Muscle Cells/metabolism , Sodium/metabolism , Stilbenes/pharmacology , Acetanilides/pharmacology , Animals , Antioxidants/pharmacology , Arrhythmias, Cardiac/pathology , Diastole , Dose-Response Relationship, Drug , Electrophysiology/methods , Female , Ischemia , Male , Muscle Cells/cytology , Patch-Clamp Techniques , Piperazines/pharmacology , Rabbits , Ranolazine , Resveratrol , Temperature , Tetrodotoxin/pharmacology
15.
J Ethnopharmacol ; 144(3): 555-61, 2012 Dec 18.
Article En | MEDLINE | ID: mdl-23058991

ETHNOPHARMACOLOGICAL RELEVANCE: Ligustrazine, the biologically active ingredient isolated from a popular Chinese medicinal plant, Ligusticum chuanxiong Hort. (Umbelliferae), has been used effectively to treat ischemic heart diseases, cerebrovascular and thrombotic vascular diseases since the 1970s. MATERIALS AND METHODS: At present, the effect of ligustrazine on L-type calcium current (I(Ca-L)) of ventricular myocytes remains controversial. In this study, we use the whole-cell patch-clamp techniques and video-based edge detection and dual excitation fluorescence photomultiplier systems to study the effects of ligustrazine on I(Ca-L), and calcium transient and contractility in rabbit ventricular myocytes in the absence and presence of isoprenaline (ISO). RESULTS: Ligustrazine (5 µM) in low concentration did not affect I(Ca-L) (P>0.05), higher concentrations of this drug (10, 20, 40, 80 µM) inhibited I(Ca-L) in a concentration-dependent manner and reduced I(Ca-L) by 9.6 ± 2.9%, 21.0 ± 4.3%, 33.9 ± 4.3%, and 51.6 ± 7.3%, respectively. Under normal conditions, ligustrazine (40 µΜ) reduced baseline of fura-2 fluorescence intensities (FFI, 340/380 ratio), namely diastolic calcium concentration, changes in FFI (ΔFFI, 340/380 ratio) and maximal velocity of Ca(2+) rise and decay (340/380 ratio/ms) by 6.3%, 26.1%, 25.2%, and 26.5%, and decreased sarcomere peak shorting (PS) and maximal velocity of shorting and relengthening by 36.4%, 31.9%, and 25.0%, respectively. Similarly, ligustrazine (40 µM) reduced baseline FFI, ΔFFI, and maximal velocity of Ca(2+) rise and decay by 14.1%, 51.1%, 35.2%, and 41.1%, and reduced sarcomere PS and maximal velocity of shorting and relengthening by 38.6%, 50.0% and 39.1%, respectively, in the presence of ISO. CONCLUSIONS: Ligustrazine not only significantly inhibits I(Ca-L) in a concentration-dependent manner but also suppressed calcium transient and contraction in the absence and presence of ISO.


Calcium Channel Blockers/pharmacology , Heart Ventricles/drug effects , Myocytes, Cardiac/drug effects , Pyrazines/pharmacology , Animals , Calcium/physiology , Calcium Channels, L-Type/physiology , Female , Heart Ventricles/cytology , Isoproterenol/pharmacology , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/physiology , Rabbits , Sympathomimetics/pharmacology , Ventricular Function/drug effects
16.
J Cardiovasc Pharmacol ; 60(4): 357-66, 2012 Oct.
Article En | MEDLINE | ID: mdl-23064241

Many studies indicate that an increase in late sodium current (I(Na.L)) of cardiomyocytes causes intracellular Na overload and subsequently raises the reverse Na/Ca exchanger current (INCX), ultimately resulting in intracellular Ca overload. Therefore, using drugs to inhibit the increased INa.L under various pathological conditions can lower intracellular Ca overload. This study was intended to explore the effect of sophocarpine (SOP) on the increase in INa.L, INCX, calcium transient and contraction in rabbit ventricular myocytes induced by Anemonia sulcata toxin II (ATX II), an opener of sodium channel, with the application of whole-cell patch-clamp techniques, the video-based motion edge detection system, and the intracellular calcium concentration determination system. The results indicate that tetrodotoxin (TTX, 4 µM ) obviously decreased INa.L and INCX enlarged by ATX II (30 nM), and SOP (20, 40, and 80 µM) also inhibited both the parameters concentration dependently in rabbit ventricular myocytes. However, transient sodium current remained unaffected by the above-mentioned concentrations of ATX II, TTX, and SOP. In addition, SOP also reversed diastolic calcium concentration, calcium transient amplitude, and ventricular muscle contractility augmented by ATX II. Its effects were similar to those of TTX, a specific inhibitor of the sodium channel. In conclusion, SOP inhibits INa.L, INCX, diastolic Ca concentration, and contractility in rabbit ventricular myocytes, which suggests that relief of intracellular Ca overload through inhibiting INa.L is likely to become a new therapeutic mechanism of SOP against arrhythmia and myocyte damage associated with intracellular Ca overload.


Alkaloids/pharmacology , Calcium/metabolism , Cnidarian Venoms/pharmacology , Sodium/metabolism , Alkaloids/administration & dosage , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Dose-Response Relationship, Drug , Female , Heart Ventricles/drug effects , Heart Ventricles/pathology , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Rabbits , Sodium Channels/metabolism , Sodium-Calcium Exchanger/metabolism , Tetrodotoxin/pharmacology
17.
Sheng Li Xue Bao ; 64(4): 433-43, 2012 Aug 25.
Article Zh | MEDLINE | ID: mdl-22907304

The objectives of this study were to investigate the effects of veratridine (VER) on persistent sodium current (I(Na.P)), Na(+)/Ca(2+) exchange current (I(NCX)), calcium transients and the action potential (AP) in rabbit ventricular myocytes, and to explore the mechanism in intracellular calcium overload and myocardial contraction enhancement by using whole-cell patch clamp recording technique, visual motion edge detection system, intracellular calcium measurement system and multi-channel physiological signal acquisition and processing system. The results showed that I(Na.P) and reverse I(NCX) in ventricular myocytes were obviously increased after giving 10, 20 µmol/L VER, with the current density of I(Na.P) increasing from (-0.22 ± 0.12) to (-0.61 ± 0.13) and (-2.15 ± 0.14) pA/pF (P < 0.01, n = 10) at -20 mV, and that of reverse I(NCX) increasing from (1.62 ± 0.12) to (2.19 ± 0.09) and (2.58 ± 0.11) pA/pF (P < 0.05, n = 10) at +50 mV. After adding 4 µmol/L tetrodotoxin (TTX), current density of I(Na.P) and reverse I(NCX) returned to (-0.07 ± 0.14) and (1.69 ± 0.15) pA/pF (P < 0.05, n = 10). Another specific blocker of I(Na.P), ranolazine (RAN), could obviously inhibit VER-increased I(Na.P) and reverse I(NCX). After giving 2.5 µmol/L VER, the maximal contraction rate of ventricular myocytes increased from (-0.91 ± 0.29) to (-1.53 ± 0.29) µm/s (P < 0.01, n = 7), the amplitude of contraction increased from (0.10 ± 0.04) to (0.16 ± 0.04) µm (P < 0.05, n = 7), and the baseline of calcium transients (diastolic calcium concentration) increased from (1.21 ± 0.08) to (1.37 ± 0.12) (P < 0.05, n = 7). After adding 2 µmol/L TTX, the maximal contraction rate and amplitude of ventricular myocytes decreased to (-0.86 ± 0.24) µm/s and (0.09 ± 0.03) µm (P < 0.01, n = 7) respectively. And the baseline of calcium transients reduced to (1.17 ± 0.09) (P < 0.05, n = 7). VER (20 µmol/L) could extend action potential duration at 50% repolarization (APD(50)) and at 90% repolarization (APD(90)) in ventricular myocytes from (123.18 ± 23.70) to (271.90 ± 32.81) and from (146.94 ± 24.15) to (429.79 ± 32.04) ms (P < 0.01, n = 6) respectively. Early afterdepolarizations (EADs) appeared in 3 out of the 6 cases. After adding 4 µmol/L TTX, APD(50) and APD(90) were reduced to (99.07 ± 22.81) and (163.84 ± 26.06) ms (P < 0.01, n = 6) respectively, and EADs disappeared accordingly in 3 cases. It could be suggested that: (1) As a specific agonist of the I(Na.P), VER could result in I(Na.P) increase and intracellular Na(+) overload, and subsequently intracellular Ca(2+) overload with the increase of reverse I(NCX). (2) The VER-increased I(Na.P) could further extend the action potential duration (APD) and induce EADs. (3) TTX could restrain the abnormal VER-induced changes of the above-mentioned indexes, indicating that these abnormal changes were caused by the increase of I(Na.P). Based on this study, it is concluded that as the I(Na.P) agonist, VER can enhance reverse I(NCX) by increasing I(Na.P), leading to intracellular Ca(2+) overload and APD abnormal extension.


Action Potentials , Myocytes, Cardiac/drug effects , Sodium-Calcium Exchanger/metabolism , Veratridine/pharmacology , Acetanilides/pharmacology , Animals , Calcium/metabolism , Myocardial Contraction , Myocytes, Cardiac/cytology , Patch-Clamp Techniques , Piperazines/pharmacology , Rabbits , Ranolazine , Tetrodotoxin/pharmacology
18.
Am J Physiol Cell Physiol ; 302(8): C1141-51, 2012 Apr 15.
Article En | MEDLINE | ID: mdl-22189558

An increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) augments late sodium current (I(Na.L)) in cardiomyocytes. This study tests the hypothesis that both Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC) mediate the effect of increased [Ca(2+)](i) to increase I(Na.L). Whole cell and open cell-attached patch clamp techniques were used to record I(Na.L) in rabbit ventricular myocytes dialyzed with solutions containing various concentrations of [Ca(2+)](i). Dialysis of cells with [Ca(2+)](i) from 0.1 to 0.3, 0.6, and 1.0 µM increased I(Na.L) in a concentration-dependent manner from 0.221 ± 0.038 to 0.554 ± 0.045 pA/pF (n = 10, P < 0.01) and was associated with an increase in mean Na(+) channel open probability and prolongation of channel mean open-time (n = 7, P < 0.01). In the presence of 0.6 µM [Ca(2+)](i), KN-93 (10 µM) and bisindolylmaleimide (BIM, 2 µM) decreased I(Na.L) by 45.2 and 54.8%, respectively. The effects of KN-93 and autocamtide-2-related inhibitory peptide II (2 µM) were not different. A combination of KN-93 and BIM completely reversed the increase in I(Na.L) as well as the Ca(2+)-induced changes in Na(+) channel mean open probability and mean open-time induced by 0.6 µM [Ca(2+)](i). Phorbol myristoyl acetate increased I(Na.L) in myocytes dialyzed with 0.1 µM [Ca(2+)](i); the effect was abolished by Gö-6976. In summary, both CaMKII and PKC are involved in [Ca(2+)](i)-mediated augmentation of I(Na.L) in ventricular myocytes. Inhibition of CaMKII and/or PKC pathways may be a therapeutic target to reduce myocardial dysfunction and cardiac arrhythmias caused by calcium overload.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Sodium/metabolism , Animals , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Female , Heart Ventricles/cytology , Heart Ventricles/metabolism , Indoles/pharmacology , Male , Maleimides/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocytes, Cardiac/enzymology , Peptides/pharmacology , Protein Kinase C/antagonists & inhibitors , Rabbits , Sodium Channels/metabolism , Sulfonamides/pharmacology
19.
Circulation ; 123(16): 1713-20, 2011 Apr 26.
Article En | MEDLINE | ID: mdl-21482963

BACKGROUND: The reverse rate dependence (RRD) of actions of I(Kr)-blocking drugs to increase the action potential duration (APD) and beat-to-beat variability of repolarization (BVR) of APD is proarrhythmic. We determined whether inhibition of endogenous, physiological late Na(+) current (late I(Na)) attenuates the RRD and proarrhythmic effect of I(Kr) inhibition. METHODS AND RESULTS: Duration of the monophasic APD (MAPD) was measured from female rabbit hearts paced at cycle lengths from 400 to 2000 milliseconds, and BVR was calculated. In the absence of a drug, duration of monophasic action potential at 90% completion of repolarization (MAPD(90)) and BVR increased as the cycle length was increased from 400 to 2000 milliseconds (n=36 and 26; P<0.01). Both E-4031 (20 nmol/L) and d-sotalol (10 µmol/L) increased MAPD(90) and BVR at all stimulation rates, and the increase was greater at slower than at faster pacing rates (n=19, 11, 12 and 7, respectively; P<0.01). Tetrodotoxin (1 µmol/L) and ranolazine significantly attenuated the RRD of MAPD(90,) reduced BVR (P<0.01), and abolished torsade de pointes in hearts treated with either 20 nmol/L E-4031 or 10 µmol/L d-sotalol. Endogenous late I(Na) in cardiomyocytes stimulated at cycle lengths from 500 to 4000 milliseconds was greater at slower than at faster stimulation rates, and rapidly decreased during the first several beats at faster but not at slower rates (n=8; P<0.01). In a computational model, simulated RRD of APD caused by E-4031 and d-sotalol was attenuated when late I(Na) was inhibited. CONCLUSION: Endogenous late I(Na) contributes to the RRD of I(Kr) inhibitor-induced increases in APD and BVR and to bradycardia-related ventricular arrhythmias.


Action Potentials/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Sodium Channels/physiology , Torsades de Pointes/physiopathology , Acetanilides/pharmacology , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Bradycardia/drug therapy , Bradycardia/physiopathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Heart Rate/drug effects , Heart Rate/physiology , Long QT Syndrome/drug therapy , Long QT Syndrome/physiopathology , Models, Cardiovascular , Myocardial Contraction/drug effects , Patch-Clamp Techniques , Piperazines/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Rabbits , Ranolazine , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Torsades de Pointes/drug therapy
20.
Acta Pharmacol Sin ; 31(11): 1438-46, 2010 Nov.
Article En | MEDLINE | ID: mdl-20953210

AIM: To study the effects and mechanisms by which hyposmotic challenge modulate function of L-type calcium current (I(Ca,L)) in rat ventricular myocytes. METHODS: The whole-cell patch-clamp techniques were used to record I(Ca,L) in rat ventricular myocytes. RESULTS: Hyposmotic challenge(∼220 mosmol/L) induced biphasic changes of I(Ca,L), a transient increase followed by a sustained decrease. I(Ca,L) increased by 19.1%±6.1% after short exposure (within 3 min) to hyposmotic solution. On the contrary, long hyposmotic challenge (10 min) decreased I(Ca,L) to 78.1%±11.0% of control, caused the inactivation of I(Ca,L), and shifted the steady-state inactivation curve of I(Ca,L) to the right. The decreased I(Ca,L) induced by hyposmotic swelling was reversed by isoproterenol or protein kinase A (PKA) activator foskolin. Hyposmotic swelling also reduced the stimulated I(Ca,L) by isoproterenol or foskolin. PKA inhibitor H-89 abolished swelling-induced transient increase of I(Ca,L), but did not affect the swelling-induced sustained decrease of I(Ca,L). NO donor SNAP and protein kinase G (PKG) inhibitor Rp-8-Br-PET-cGMPS did not interfere with swelling-induced biphasic changes of I(Ca,L). Protein kinase C (PKC) activator PMA decreased I(Ca,L) and hyposmotic solution with PMA reverted the decreased I(Ca,L) by PMA. PKC inhibitor BIM prevented the swelling-induced biphasic changes of I(Ca,L). CONCLUSION: Hyposmotic challenge induced biphasic changes of I(Ca,L), a transient increase followed by a sustained decrease, in rat ventricular myocytes through PKC pathway, but not PKG pathway. PKA system could be responsible for the transient increase of I(Ca,L) during short exposure to hyposmotic solution.


Calcium Channels, L-Type/physiology , Heart Ventricles/cytology , Ion Channel Gating/drug effects , Myocytes, Cardiac/metabolism , Protein Kinase C/metabolism , Animals , Calcium Channels, L-Type/metabolism , Cell Culture Techniques , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Female , Heart Ventricles/drug effects , Heart Ventricles/enzymology , Heart Ventricles/metabolism , Isoproterenol/pharmacology , Membrane Potentials/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Osmolar Concentration , Patch-Clamp Techniques , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction
...