Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Type of study
Publication year range
1.
Heart Rhythm ; 21(2): 184-196, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37924963

ABSTRACT

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.


Subject(s)
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.
Acta Pharmacol Sin ; 37(11): 1432-1441, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27569391

ABSTRACT

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.


Subject(s)
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
3.
Acta Pharmacol Sin ; 36(11): 1327-36, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26456586

ABSTRACT

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.


Subject(s)
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
4.
J Cardiovasc Pharmacol ; 60(4): 357-66, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23064241

ABSTRACT

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.


Subject(s)
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
5.
Sheng Li Xue Bao ; 64(4): 433-43, 2012 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-22907304

ABSTRACT

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.


Subject(s)
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
6.
Acta Pharmacol Sin ; 31(11): 1438-46, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20953210

ABSTRACT

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.


Subject(s)
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
7.
Sheng Li Xue Bao ; 59(2): 233-9, 2007 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-17437049

ABSTRACT

Whole-cell and cell-attached patch-clamp techniques were used to record the changes of persistent sodium current (I(Na.P)) in ventricular myocytes of guinea pig to investigate the effect of low extracellular pH on I(Na.P) and its mechanism. The results showed that low extracellular pH (7.0, 6.8 and 6.5) obviously increased the amplitude of whole-cell I(Na.P) in a [H(+)] concentration-dependent manner. Under the condition of extracellular pH 6.5, I(Na.P) was markedly augmented from control (pH 7.4) value of (0.347+/-0.067) pA/pF to (0.817+/- 0.137) pA/pF (P<0.01, n=6), whereas the reducing agent dithiothreitiol (DTT, 1 mmol/L) reversed the increased IN(Na.P) from (0.817+/-0.137) pA/pF to (0.233+/-0.078) pA/pF (P<0.01 vs pH 6.5, n=6). Decreasing extracellular pH to 6.5 also increased the persistent sodium channel activity in cell-attached patches. The mean open probability and mean open time were increased from control value of 0.021+/-0.007 and (0.899+/-0.074) ms to 0.205+/-0.023 and (1.593+/-0.158) ms, respectively (both P<0.01, n=6), and such enhancement was reversed by application of 1 mmol/L DTT [to 0.019+/-0.005 and (0.868+/-0.190) ms, both P<0.01 vs pH 6.5, n=6]. Furthermore, protein kinase C (PKC) inhibitor bisindolylmaleimide (BIM, 5 micromol/L) reduced the enhanced mean open probability and mean open time at pH 6.5 from 0.214+/-0.024 and (1.634+/-0.137) ms to 0.025+/-0.006 and (0.914+/-0.070) ms, respectively (both P<0.01 vs pH 6.5, n=6). The results demonstrate that low extracellular pH markedly increases I(Na.P) in guinea pig ventricular myocytes, in which activation of PKC may be involved.


Subject(s)
Extracellular Space/chemistry , Myocytes, Cardiac/physiology , Sodium Channels/physiology , Animals , Culture Media/chemistry , Female , Guinea Pigs , Heart Ventricles/cytology , Hydrogen-Ion Concentration , Male , Membrane Potentials/physiology , Myocytes, Cardiac/cytology , Patch-Clamp Techniques , Sodium/metabolism
8.
J Membr Biol ; 211(2): 81-7, 2006.
Article in English | MEDLINE | ID: mdl-17041784

ABSTRACT

As an important in vivo antioxidant, vitamin C is commonly used clinically to alleviate hypoxia-induced heart symptoms. To approach the protective mechanisms of vitamin C on hearts during hypoxia, we investigated the electrophysiological effects of vitamin C (1 mM: , pretreated before hypoxia) on Na(+) currents (including transient and persistent Na(+) currents) in guinea pig ventricular myocytes during hypoxia by the whole-cell and single-channel patch-clamp techniques. Whole-cell recordings showed that the mean current density of I (NaT) in the hypoxia group decreased from the control value of 40.2142 +/- 1.7735 to 27.1663 +/- 1.8441 pA/pF and current density of I (NaP) increased from 0.3987 +/- 0.0474 to 1.1854 +/- 01994 pA/pF (n = 9, P < 0.05 vs. control) at 15 min. However, when vitamin C was administered before hypoxia as pretreatment, I (NaT )and I (NaP )varied moderately (mean current density of I (NaT) decreasing from 41.6038 +/- 2.9762 to 34.6341 +/- 1.9651 pA/pF and current density of I (NaP) increasing from 0.3843 +/- 0.0636 to 0.6734 +/- 0.1057 pA/pF; n = 9, P < 0.05 vs. hypoxia group). Single-channel recordings (cell-patched) showed that the mean open probability and open time of I (NaP) increased significantly in both groups at hypoxia 15 min. However, the increased current values of the hypoxia group were still marked at hypoxia 15 min (n = 9, P < 0.05 vs. vitamin C + hypoxia group). Our results indicate that vitamin C can attenuate the disturbed effects of hypoxia on Na(+) currents (I (NaT) and I (NaP)) of cardiac myocytes in guinea pigs effectively.


Subject(s)
Ascorbic Acid/pharmacology , Myocytes, Cardiac/drug effects , Sodium Channels/physiology , Animals , Antioxidants/pharmacology , Cell Hypoxia , Cells, Cultured , Female , Guinea Pigs , Heart Ventricles/cytology , Male , Membrane Potentials/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Sodium/metabolism , Time Factors
9.
Acta Pharmacol Sin ; 26(7): 828-34, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15960889

ABSTRACT

AIM: To study the effect of hydrogen peroxide (H2O2) on persistent sodium current (I(Na.P)) in guinea pig ventricular myocytes. METHODS: The whole-cell, cell-attached, and inside-out patch-clamp techniques were applied on isolated ventricular myocytes from guinea pig. RESULTS: H2O2 (0.1 mmol/L, 0.5 mmol/L and 1.0 mmol/L) increased the amplitude of whole-cell I(Na.P) in a concentration-dependent manner, and glutathione (GSH 1 mmol/L) reversed the increased I(Na.P). H2O2 (1 mmol/L) increased persistent sodium channel activity in cell-attached and inside-out patches. The mean open probability was increased from control values of 0.015+/-0.004 and 0.012+/-0.003 to 0.106+/-0.011 and 0.136+/-0.010, respectively (P< 0.01 vs control). They were then decreased to 0.039+/-0.024 and 0.027+/-0.006, respectively, after the addition of 1 mmol/L GSH (P<0.01 vs H2O2). The time when open probability began to increase and reached a maximum was shorter in inside-out patches than that in cell-attached patches (4.8+/-1.0 min vs 11.5+/-3.9 min, P<0.01; 9.6+/-1.6 min vs 18.7+/-4.7 min, P<0.01). CONCLUSION: H2O2 increased the I(Na.P) of guinea pig ventricular myocytes in a concentration-dependent manner, possibly by directly oxidating the cell membrane.


Subject(s)
Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/physiology , Sodium Channels/drug effects , Animals , Dose-Response Relationship, Drug , Female , Glutathione/pharmacology , Guinea Pigs , Heart Ventricles , Hydrogen Peroxide/administration & dosage , Lidocaine/pharmacology , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...