Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Front Cardiovasc Med ; 11: 1374881, 2024.
Article in English | MEDLINE | ID: mdl-39045008

ABSTRACT

Background: Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) show tremendous promise for cardiac regeneration following myocardial infarction (MI), but their transplantation gives rise to transient ventricular tachycardia (VT) in large-animal MI models, representing a major hurdle to translation. Our group previously reported that these arrhythmias arise from a focal mechanism whereby graft tissue functions as an ectopic pacemaker; therefore, we hypothesized that hPSC-CMs engineered with a dominant negative form of the pacemaker ion channel HCN4 (dnHCN4) would exhibit reduced automaticity and arrhythmogenic risk following transplantation. Methods: We used CRISPR/Cas9-mediated gene-editing to create transgenic dnHCN4 hPSC-CMs, and their electrophysiological behavior was evaluated in vitro by patch-clamp recordings and optical mapping. Next, we transplanted WT and homozygous dnHCN4 hPSC-CMs in a pig MI model and compared post-transplantation outcomes including the incidence of spontaneous arrhythmias and graft structure by immunohistochemistry. Results: In vitro dnHCN4 hPSC-CMs exhibited significantly reduced automaticity and pacemaker funny current (I f ) density relative to wildtype (WT) cardiomyocytes. Following transplantation with either dnHCN4 or WT hPSC-CMs, all recipient hearts showed transmural infarct scar that was partially remuscularized by scattered islands of human myocardium. However, in contrast to our hypothesis, both dnHCN4 and WT hPSC-CM recipients exhibited frequent episodes of ventricular tachycardia (VT). Conclusions: While genetic silencing of the pacemaker ion channel HCN4 suppresses the automaticity of hPSC-CMs in vitro, this intervention is insufficient to reduce VT risk post-transplantation in the pig MI model, implying more complex mechanism(s) are operational in vivo.

2.
Elife ; 112022 03 22.
Article in English | MEDLINE | ID: mdl-35315774

ABSTRACT

Tongmai Yangxin (TMYX) is a complex compound of the Traditional Chinese Medicine (TCM) used to treat several cardiac rhythm disorders; however, no information regarding its mechanism of action is available. In this study we provide a detailed characterization of the effects of TMYX on the electrical activity of pacemaker cells and unravel its mechanism of action. Single-cell electrophysiology revealed that TMYX elicits a reversible and dose-dependent (2/6 mg/ml) slowing of spontaneous action potentials rate (-20.8/-50.2%) by a selective reduction of the diastolic phase (-50.1/-76.0%). This action is mediated by a negative shift of the If activation curve (-6.7/-11.9 mV) and is caused by a reduction of the cyclic adenosine monophosphate (cAMP)-induced stimulation of pacemaker channels. We provide evidence that TMYX acts by directly antagonizing the cAMP-induced allosteric modulation of the pacemaker channels. Noticeably, this mechanism functionally resembles the pharmacological actions of muscarinic stimulation or ß-blockers, but it does not require generalized changes in cytoplasmic cAMP levels thus ensuring a selective action on rate. In agreement with a competitive inhibition mechanism, TMYX exerts its maximal antagonistic action at submaximal cAMP concentrations and then progressively becomes less effective thus ensuring a full contribution of If to pacemaker rate during high metabolic demand and sympathetic stimulation.


Subject(s)
Cyclic AMP , Second Messenger Systems , Action Potentials , Animals , China , Cyclic AMP/metabolism , Myocytes, Cardiac/metabolism , Rabbits
3.
Prog Biophys Mol Biol ; 166: 22-28, 2021 11.
Article in English | MEDLINE | ID: mdl-32853595

ABSTRACT

This article reviews work over the past three decades that is related to the contribution of the pacemaker current, If, to basal and autonomically regulated spontaneous rate in the sinoatrial node. It also addresses how the actions of the pacemaker current relate to those of Ca homeostasis with respect to basal and autonomically regulated rhythm. In this regard, it explores the relative contributions of Ca-sensitive and Ca-insensitive isoforms of adenylyl cyclase to sinoatrial node automaticity. The latter studies include previously unpublished work making use of mice in which both the type 1 and type 8 Ca-sensitive adenylyl cyclase isoforms were knocked out. These studies indicate that the pacemaker current and the L-type Ca current are distinctly influenced by Ca-sensitive and insensitive adenylyl cyclase isoforms.


Subject(s)
Pacemaker, Artificial , Sinoatrial Node , Action Potentials , Adenylyl Cyclases , Animals , Calcium , Mice , Protein Isoforms
4.
Front Physiol ; 11: 587040, 2020.
Article in English | MEDLINE | ID: mdl-33240105

ABSTRACT

It has long been known that heart rate is regulated by the autonomic nervous system. Recently, we demonstrated that the pacemaker current, I f , is regulated by phosphoinositide 3-kinase (PI3K) signaling independently of the autonomic nervous system. Inhibition of PI3K in sinus node (SN) myocytes shifts the activation of I f by almost 16 mV in the negative direction. I f in the SN is predominantly mediated by two members of the HCN gene family, HCN4 and HCN1. Purkinje fibers also possess I f and are an important secondary pacemaker in the heart. In contrast to the SN, they express HCN2 and HCN4, while ventricular myocytes, which do not normally pace, express HCN2 alone. In the current work, we investigated PI3K regulation of HCN2 expressed in HEK293 cells. Treatment with the PI3K inhibitor PI-103 caused a negative shift in the activation voltage and a dramatic reduction in the magnitude of the HCN2 current. Similar changes were also seen in cells treated with an inhibitor of the protein kinase Akt, a downstream effector of PI3K. The effects of PI-103 were reversed by perfusion of cells with phosphatidylinositol 3,4,5-trisphosphate (the second messenger produced by PI3K) or active Akt protein. We identified serine 861 in mouse HCN2 as a putative Akt phosphorylation site. Mutation of S861 to alanine mimicked the effects of Akt inhibition on voltage dependence and current magnitude. In addition, the Akt inhibitor had no effect on the mutant channel. These results suggest that Akt phosphorylation of mHCN2 S861 accounts for virtually all of the observed actions of PI3K signaling on the HCN2 current. Unexpectedly, Akt inhibition had no effect on I f in SN myocytes. This result raises the possibility that diverse PI3K signaling pathways differentially regulate HCN-induced currents in different tissues, depending on the isoforms expressed.

5.
Pharmacol Rep ; 72(6): 1706-1716, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32451735

ABSTRACT

BACKGROUND: In this study, we investigated the effect of forskolin (FSK, a selective adenylate cyclase agonist) on the automatic diastolic depolarization of sinus node cells (SNC) with hypoxia/reoxygenation (H/R) injury. METHODS: The SNC of the newborn rat was randomly assigned into the control group, the H/R (H/R injury) group, or the H/R + FSK (H/R injury + FSK treatment) group. Patch-clamp was performed to record the action potential and electrophysiological changes. The cellular distribution of intracellular calcium concentration was analyzed by fluorescence staining. RESULTS: Compared with the control cells, spontaneous pulsation frequency (SPF) and diastolic depolarization rate (DDR) of H/R cells were reduced from 244.3 ± 10.6 times/min and 108.7 ± 7.8 mV/s to 130.5 ± 7.6 times/min and 53.4 ± 6.5 mV/s, respectively. FSK significantly increased SPF and DDR of H/R cells to 208.3 ± 8.3 times/min and 93.2 ± 8.9 mV/s (n = 15, both p < 0.01), respectively. H/R reduced the current densities of If, ICa,T and inward INCX, which were significantly increased by 10 µM FSK treatment (n = 15, p < 0.01). Furthermore, reduced expression of HCN4 and NCX1.1 channel protein were significantly increased by FSK. Inhibitor studies showed that both SQ22536 (a selective adenylate cyclase inhibitor) and H89 (a selective protein kinases A [PKA] inhibitor) blocked the effects of FSK on SPF and DDR. CONCLUSIONS: H/R causes pacemaker dysfunction in newborn rat sinoatrial node cells leading to divergence of the DD and the slow of spontaneous APs, which change can be dramatically reversed by FSK through increasing INCX and If current in H/R injury.


Subject(s)
Action Potentials/drug effects , Calcium/metabolism , Colforsin/pharmacology , Sinoatrial Node/drug effects , Adenylyl Cyclases/drug effects , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Cell Hypoxia/drug effects , Cells, Cultured , Female , Male , Rats , Rats, Wistar , Sinoatrial Node/metabolism
6.
Neuroscience ; 433: 230-240, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31982470

ABSTRACT

OBJECTIVE: The spontaneous action potential of isolated sinoatrial node (SAN) cells is regulated by a coupled-clock system of two clocks: the calcium clock and membrane clock. However, it remains unclear whether calcium clock inhibitors have a direct effect on the membrane clock. The purpose of this study was to investigate the direct effect of cyclopiazonic acid (CPA), a selective calcium clock inhibitor, on the function of the membrane clock of SAN cells. METHODS: at SAN cells were isolated by trypsinization and identified based on morphology and electrophysiology. If and HCN currents were recorded via patch clamp technique. The expression of the HCN channel protein was determined by Western blotting analysis. RESULTS: The diastolic depolarization rate of spontaneous action potentials and the current densities of If were reduced by exposure to 10 µM CPA. The inhibitory effect of CPA was concentration-dependent with an IC50 value of 16.3 µM and a Hill coefficient of 0.98. The effect of CPA on If current was also time-dependent, and the If current amplitude was partially restored after washout. Furthermore, the steady-state activation curve of the If current was shifted to a negative potential, indicating that channel activation slowed down. Finally, the protein expression of HCN4 in HEK293 cells was markedly downregulated by CPA. CONCLUSIONS: These results indicate that the direct inhibition effect of CPA on the If current in SAN cells is both concentration- and time-dependent. The underlying mechanisms may involve slowing down steady-state activation and the downregulation of pacemaker channel protein expression.


Subject(s)
Sinoatrial Node , Action Potentials , Calcium , HEK293 Cells , Humans , Indoles/pharmacology
7.
Front Pharmacol ; 9: 551, 2018.
Article in English | MEDLINE | ID: mdl-29899698

ABSTRACT

Objective: We investigated the role of astragaloside in the treatment of sick sinus syndrome (SSS). Methods: Neonatal New Zealand rabbits were selected for the study. Rabbit sinoatrial node (SAN) cells were isolated by the method of dual enzymatic digestion and differential adherence. The injury model was prepared through simulated ischemia and reperfusion (I/R), and changes in the pacemaker current (If) were recorded using the whole-cell patch-clamp technique. The proteins F-actin and vinculin were examined between various groups of SAN cells using a microplate reader and laser scanning confocal microscopy. The mRNA level and protein expression of hyperpolarization-activated cyclic nucleotide gated potassium channel 4 (HCN4) were assessed by q-PCR and western blot method. Results: The peak current density of If was decreased to -19.64 ± 2.14 pA/pF in SAN cells after simulated I/R, and the difference was highly significant (P < 0.01). Following simulated I/R, 100, 200, or 300 µmol L-1 astragaloside was added to the extracellular solution of SAN cells; the peak current density of the If increased to -30.43 ± 1.98, -34.83 ± 1.6, and -52.72 ± 1.7 pA/pF, respectively (P < 0.01). Adding 100 µmol L-1 astragaloside to normal SAN cells also led to an enhanced peak current density of the If (P < 0.05). In a concentration-dependent manner, especially at 300 µmol/L, astragaloside was capable of increasing the expression of HCN4 and protecting the structural stability of F-actin and vinculin in the damaged SAN cells. Conclusion: We estimated that astragaloside could shorten the action potential duration 20 (APD20) and APD50 in damaged SAN cells of neonatal rabbits, thereby increasing the expression of HCN4 and the If current density in damaged SAN cells of neonatal rabbits in a voltage-dependent manner, accelerating the steady-state activation of the If channels, and protecting damaged cytoskeleton.

8.
Heart Vessels ; 33(7): 802-819, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29349559

ABSTRACT

We found that a female infant presenting with left bundle branch block and left ventricular noncompaction carries uninvestigated gene mutations HCN4(G811E), SCN5A(L1988R), DMD(S2384Y), and EMD(R203H). Here, we explored the possible pathogenicity of HCN4(G811E), which results in a G811E substitution in hyperpolarization-activated cyclic nucleotide-gated channel 4, the main subunit of the cardiac pacemaker channel. Voltage-clamp measurements in a heterologous expression system of HEK293T cells showed that HCN4(G811E) slightly reduced whole-cell HCN4 channel conductance, whereas it did not affect the gating kinetics, unitary conductance, or cAMP-dependent modulation of voltage-dependence. Immunocytochemistry and immunoblot analysis showed that the G811E mutation did not impair the membrane trafficking of the channel subunit in the heterologous expression system. These findings indicate that HCN4(G811E) may not be a monogenic factor to cause the cardiac disorders.


Subject(s)
Bradycardia/genetics , Bundle-Branch Block/genetics , Heart Defects, Congenital/genetics , Heart Ventricles/abnormalities , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Muscle Proteins/genetics , Mutation , Potassium Channels/genetics , Bradycardia/diagnosis , Bradycardia/etiology , Bundle-Branch Block/complications , Bundle-Branch Block/diagnosis , DNA Mutational Analysis , Echocardiography, Doppler, Color , Female , HEK293 Cells , Heart Defects, Congenital/complications , Heart Defects, Congenital/diagnosis , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Immunoblotting , Immunohistochemistry , Infant, Newborn , Muscle Proteins/metabolism , Potassium Channels/metabolism , Sinoatrial Node/metabolism , Sinoatrial Node/pathology
9.
Vasc Health Risk Manag ; 12: 453-470, 2016.
Article in English | MEDLINE | ID: mdl-27895488

ABSTRACT

A high resting heart rate (≥70-75 b.p.m.) is a risk factor for patients with heart failure (HF) with reduced ejection fraction (EF), probably in the sense of accelerated atherosclerosis, with an increased morbidity and mortality. Beta-blockers not only reduce heart rate but also have negative inotropic and blood pressure-lowering effects, and therefore, in many patients, they cannot be given in the recommended dose. Ivabradine specifically inhibits the pacemaker current (funny current, If) of the sinoatrial node cells, resulting in therapeutic heart rate lowering without any negative inotropic and blood pressure-lowering effect. According to the European Society of Cardiology guidelines, ivabradine should be considered to reduce the risk of HF hospitalization and cardiovascular death in symptomatic patients with a reduced left ventricular EF ≤35% and sinus rhythm ≥70 b.p.m. despite treatment with an evidence-based dose of beta-blocker or a dose below the recommended dose (recommendation class "IIa" = weight of evidence/opinion is in favor of usefulness/efficacy: "should be considered"; level of evidence "B" = data derived from a single randomized clinical trial or large nonrandomized studies). Using a heart rate cutoff of ≥ 75 b.p.m., as licensed by the European Medicines Agency, treatment with ivabradine 5-7.5 mg b.i.d. reduces cardiovascular mortality by 17%, HF mortality by 39% and HF hospitalization rate by 30%. A high resting heart rate is not only a risk factor in HF with reduced EF but also at least a risk marker in HF with preserved EF, in acute HF and also in special forms of HF. In this review, we discuss the proven role of ivabradine in the validated indication "HF with reduced EF" together with interesting preliminary findings, and the potential role of ivabradine in further, specific forms of HF.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Benzazepines/therapeutic use , Heart Conduction System/drug effects , Heart Failure/drug therapy , Heart Rate/drug effects , Action Potentials , Age Factors , Aged , Aged, 80 and over , Animals , Anti-Arrhythmia Agents/adverse effects , Benzazepines/adverse effects , Biological Clocks/drug effects , Chronic Disease , Cyclic Nucleotide-Gated Cation Channels/drug effects , Cyclic Nucleotide-Gated Cation Channels/metabolism , Drug Therapy, Combination , Heart Conduction System/metabolism , Heart Conduction System/physiopathology , Heart Failure/diagnosis , Heart Failure/mortality , Heart Failure/physiopathology , Humans , Ivabradine , Middle Aged , Patient Selection , Quality of Life , Risk Assessment , Risk Factors , Stroke Volume , Treatment Outcome , Ventricular Function, Left
10.
Int J Cardiol ; 221: 529-36, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27414735

ABSTRACT

BACKGROUND: Gap junction (GJ) dysfunctions predispose cardiac tissues to various arrhythmias. Sinoatrial node (SAN) and pulmonary veins (PVs) are closely related atrial dysrhythmia. This study evaluated whether GJ modifications modulate SAN and PVs electrical activities. METHODS: Conventional microelectrodes were used to record action potentials in isolated rabbit SAN, PVs, and connected PV-SAN tissue preparations before and after heptanol (GJ inhibitor) and PQ1 (GJ enhancer) administration with and without isoproterenol. A whole-cell patch clamp was used to record the electrical activities before and after heptanol in single SAN and PV cardiomyocytes. RESULTS: Heptanol (1, 3, and 10µM) reduced the spontaneous beating rates of isolated SAN preparations but not PVs. Heptanol (10µM) decelerated the SAN leading rhythm in the PV-SAN preparations and induced PV burst firings without (3 of 6, 50%) and with (6 of 6, 100%) isoproterenol (1µM). Heptanol (10µM) also reduced the spontaneous beating rates in single SAN cardiomyocyte, but not PV cardiomyocyte, with a decreased pacemaker current. PQ1 (50 and 500nM) treatment did not change the spontaneous beating rates in isolated SAN and PV preparations. In the connected PV-SAN preparations, PQ1 (500nM) did not induce any PV firing even having additional isoproterenol treatment (1µM). Moreover, PQ1 (500nM) prevented heptanol-induced electrical changes in SAN and PVs preparations. CONCLUSION: GJ dysfunction modulates SAN and PV electrical activity, which may contribute to atrial arrhythmogenesis. GJ enhancer has a therapeutic potential in SAN dysfunction and atrial arrhythmogenesis.


Subject(s)
Aminoquinolines/pharmacology , Atrial Fibrillation , Myocytes, Cardiac , Action Potentials/drug effects , Action Potentials/physiology , Animals , Atrial Fibrillation/physiopathology , Atrial Fibrillation/prevention & control , Cardiovascular Agents/pharmacology , Gap Junctions/drug effects , Gap Junctions/physiology , Heart Atria/physiopathology , Heptanol/pharmacology , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Pulmonary Veins/physiopathology , Rabbits , Sinoatrial Node/physiopathology
11.
Pflugers Arch ; 468(4): 609-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26687128

ABSTRACT

Na(+)/Ca(2+) exchanger current (INCX) triggered by spontaneous Ca(2+) release from sarcoplasmic reticulum (SR) has been suggested as one of the cardiac pacemaker mechanisms ("Ca(2+) clock model"). In human embryonic stem cell-derived cardiomyocytes (hESC-CMs) showing spontaneous action potentials (APs), we found that substantial population (35 %) showed regular oscillation of inward currents (SICs) in nystatin-perforated voltage clamp between -40 and 40 mV (-80 ± 10.6 pA, at -20 mV). SICs were similarly observed between nodal, atrial, and ventricular hESC-CMs. Oscillations of [Ca(2+)]i synchronized with SICs were observed under voltage clamp. SICs were eliminated by lowering [Ca(2+)]e, L-type Ca(2+) channel (VOCCL) blocker (nifedipine, 10 µM), ryanodine receptor (RyR) agonist (caffeine, 10 mM), or NCX inhibitor (1 µM SN-6 and 10 µM KB-R7943). Plasma membrane expression of NCX1 was confirmed using immunofluorescence confocal microcopy. Both caffeine and SN-6 slowed the pacemaker potential but did not abolish the AP generation. The inhibitors of funny current (3 µM ivabradine) or voltage-gated K(+) channel currents (1 µM E4031 and 10 µM chromanol-293B) also did not abolish but slowed the pacemaker potential. In a computational model of cardiac pacemaker by Maltsev and Lakatta (2009), after modifying the spatial distribution of RyR, VOCCL, and NCX by using our multiparameter adjust algorithm, we could successfully reproduce spontaneous SR Ca(2+) release and SICs under voltage clamp. It was proposed that, under the membrane depolarization activating VOCCL, oscillatory Ca(2+) releases via RyR induce sharp increases in subsarcolemmal [Ca(2+)]i and inward INCX (SICs). Since the hESC-CMs without SICs still showed spontaneous APs, the putative "Ca(2+) clock" would provide a redundant pacemaker or augmenting mechanism in hESC-CMs.


Subject(s)
Action Potentials , Calcium Signaling , Embryonic Stem Cells/cytology , Myocytes, Cardiac/metabolism , Sodium-Calcium Exchanger/metabolism , Calcium Channels, L-Type/metabolism , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
12.
Clin Exp Pharmacol Physiol ; 41(9): 698-707, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24862298

ABSTRACT

The pacemaker current If conducted by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels plays a critical role in the regulation of cardiac automaticity, with If density increased in hypertrophied ventricular myocytes. Amiodarone, a highly effective anti-arrhythmic agent, blocks human HCN currents and native If under normal conditions. To determine the effects of amiodarone under pathological conditions, we monitored If under after both acute (0.01, 0.1, 1, 10 and 100 µmol/L) and chronic (10 µmol/L) amiodarone treatment in ventricular myocytes from spontaneously hypertensive rats (SHR) with left ventricular hypertrophy using the whole-cell patch-clamp technique. The If current density was significantly greater in SHR ventricular myocytes than in cells from healthy normotensive control Wistar-Kyoto (WKY) rats. Acute application of amiodarone significantly decreased If density in myocytes from both SHR and WKY rats. The inhibition was concentration dependent with an IC50 of 4.9 ± 1.2 and 6.9 ± 1.3 µmol/L in myocytes from SHR and WKY rats, respectively. Amiodarone increased the activation and deactivation times of If in myocytes from SHR, although it did not alter the relationship of voltage-dependent activation and the reversal potential of If in myocytes from SHR. Chronic exposure of myocytes from SHR to amiodarone potently inhibited If and downregulated HCN2 and HCN4, the major channel subtypes underlying native If , at both the mRNA and protein level. These findings indicate that amiodarone inhibits If under hypertrophied conditions through dual mechanisms: (i) direct channel blockade of If currents; and (ii) indirect suppression via negative regulation of HCN channel gene expression. These unique properties of amiodarone may contribute to its anti-arrhythmic properties under pathological conditions.


Subject(s)
Amiodarone/pharmacology , Biological Clocks/drug effects , Electric Conductivity , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Animals , Biological Clocks/physiology , Cell Separation , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Potassium Channels/metabolism , Pyrimidines/pharmacology , Rats , Rats, Inbred SHR
13.
J Mol Cell Cardiol ; 72: 64-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24583250

ABSTRACT

Lower heart rate is associated with better survival in patients with multiple organ dysfunction syndrome (MODS), a disease mostly caused by sepsis. The benefits of heart rate reduction by ivabradine during MODS are currently being investigated in the MODIfY clinical trial. Ivabradine is a selective inhibitor of the pacemaker current If and since If is impaired by lipopolysaccharide (LPS, endotoxin), a trigger of sepsis, we aimed to explore If blocking potency of ivabradine under elevated endotoxin levels in human atrial cardiomyocytes. Treatment of myocytes with S-LPS (containing the lipid A moiety, a core oligosaccharide and an O-polysaccharide chain) but not R595 (an O-chain lacking LPS-form) caused If inhibition under acute and chronic septic conditions. The specific interaction of S-LPS but not R595 to pacemaker channels HCN2 and HCN4 proves the necessity of O-chain for S-LPS-HCN interaction. The efficacy of ivabradine to block If was reduced under septic conditions, an observation that correlated with lower intracellular ivabradine concentrations in S-LPS- but not R595-treated cardiomyocytes. Computational analysis using a sinoatrial pacemaker cell model revealed that despite a reduction of If under septic conditions, ivabradine further decelerated pacemaking activity. This novel finding, i.e. If inhibition by ivabradine under elevated endotoxin levels in vitro, may provide a molecular understanding for the efficacy of this drug on heart rate reduction under septic conditions in vivo, e.g. the MODIfY clinical trial.


Subject(s)
Action Potentials/drug effects , Benzazepines/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Muscle Proteins/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Sinoatrial Node/drug effects , Clinical Trials as Topic , Heart Atria/cytology , Heart Atria/drug effects , Heart Atria/metabolism , Heart Rate/drug effects , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ivabradine , Models, Biological , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Potassium Channels/metabolism , Primary Cell Culture , Sinoatrial Node/cytology , Sinoatrial Node/metabolism
14.
J Mol Cell Cardiol ; 67: 12-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24370890

ABSTRACT

The ability of human pluripotent stem cells (hPSCs) to differentiate into any cell type of the three germ layers makes them a very promising cell source for multiple purposes, including regenerative medicine, drug discovery, and as a model to study disease mechanisms and progression. One of the first specialized cell types to be generated from hPSC was cardiomyocytes (CM), and differentiation protocols have evolved over the years and now allow for robust and large-scale production of hPSC-CM. Still, scientists are struggling to achieve the same, mainly ventricular, phenotype of the hPSC-CM in vitro as their adult counterpart in vivo. In vitro generated cardiomyocytes are generally described as fetal-like rather than adult. In this review, we compare the in vivo development of cardiomyocytes to the in vitro differentiation of hPSC into CM with focus on electrophysiology, structure and contractility. Furthermore, known epigenetic changes underlying the differences between adult human CM and CM differentiated from pluripotent stem cells are described. This should provide the reader with an extensive overview of the current status of human stem cell-derived cardiomyocyte phenotype and function. Additionally, the reader will gain insight into the underlying signaling pathways and mechanisms responsible for cardiomyocyte development.


Subject(s)
Cell Differentiation , Electrophysiological Phenomena , Myocytes, Cardiac/cytology , Culture Techniques , Epigenomics , Heart/embryology , Heart/growth & development , Humans , Pluripotent Stem Cells/cytology , Signal Transduction
15.
Am J Physiol Endocrinol Metab ; 305(11): E1384-97, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24105416

ABSTRACT

Kisspeptin (Kiss1) neurons in the rostral periventricular area of the third ventricle (RP3V) provide excitatory drive to gonadotropin-releasing hormone (GnRH) neurons to control fertility. Using whole cell patch clamp recording and single-cell (sc)RT-PCR techniques targeting Kiss1-CreGFP or tyrosine hydroxylase (TH)-EGFP neurons, we characterized the biophysical properties of these neurons and identified the critical intrinsic properties required for burst firing in 17ß-estradiol (E2)-treated, ovariectomized female mice. One-fourth of the RP3V Kiss1 neurons exhibited spontaneous burst firing. RP3V Kiss1 neurons expressed a hyperpolarization-activated h-current (Ih) and a T-type calcium current (IT), which supported hyperpolarization-induced rebound burst firing. Under voltage clamp conditions, all Kiss1 neurons expressed a kinetically fast Ih that was augmented 3.4-fold by high (LH surge-producing)-E2 treatment. scPCR analysis of Kiss1 neurons revealed abundant expression of the HCN1 channel transcripts. Kiss1 neurons also expressed a Ni(2+)- and TTA-P2-sensitive IT that was augmented sixfold with high-E2 treatment. CaV3.1 mRNA was also highly expressed in these cells. Current clamp analysis revealed that rebound burst firing was induced in RP3V Kiss1 neurons in high-E2-treated animals, and the majority of Kiss1 neurons had a hyperpolarization threshold of -84.7 mV, which corresponded to the V½ for IT de-inactivation. Finally, Kiss1 neurons in the RP3V were hyperpolarized by µ- and κ-opioid and GABAB receptor agonists, suggesting that these pathways also contribute to rebound burst firing. Therefore, Kiss1 neurons in the RP3V express the critical channels and receptors that permit E2-dependent rebound burst firing and provide the biophysical substrate that drives the preovulatory surge of GnRH.


Subject(s)
Estradiol/pharmacology , Kisspeptins/metabolism , Neurons/physiology , Preoptic Area/metabolism , Animals , Female , Follicular Phase/drug effects , Follicular Phase/metabolism , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kisspeptins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Ovariectomy , Preoptic Area/drug effects , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Third Ventricle/drug effects , Third Ventricle/metabolism
16.
Heart Rhythm ; 10(12): 1903-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055949

ABSTRACT

BACKGROUND: Human-induced pluripotent stem cell (h-iPSC)-derived cardiac myocytes are a unique model in which human myocyte function and dysfunction are studied, especially those from patients with genetic disorders. They are also considered a major advance for drug safety testing. However, these cells have considerable unexplored potential limitations when applied to quantitative action potential (AP) analysis. One major factor is spontaneous activity and resulting variability and potentially anomalous behavior of AP parameters. OBJECTIVE: To demonstrate the effect of using an in silico interface on electronically expressed I(K1), a major component lacking in h-iPSC-derived cardiac myocytes. METHODS: An in silico interface was developed to express synthetic I(K1) in cells under whole-cell voltage clamp. RESULTS: Electronic I(K1) expression established a physiological resting potential, eliminated spontaneous activity, reduced spontaneous early and delayed afterdepolarizations, and decreased AP variability. The initiated APs had the classic rapid upstroke and spike and dome morphology consistent with data obtained with freshly isolated human myocytes as well as the readily recognizable repolarization attributes of ventricular and atrial cells. The application of 1 µM of BayK-8644 resulted in anomalous AP shortening in h-iPSC-derived cardiac myocytes. When I(K1) was electronically expressed, BayK-8644 prolonged the AP, which is consistent with the existing results on native cardiac myocytes. CONCLUSIONS: The electronic expression of I(K1) is a simple and robust method to significantly improve the physiological behavior of the AP and electrical profile of h-iPSC-derived cardiac myocytes. Increased stability enables the use of this preparation for a controlled quantitative analysis of AP parameters, for example, drug responsiveness, genetic disorders, and dynamic behavior restitution profiles.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Channels, L-Type/biosynthesis , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Action Potentials , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques
17.
J Cardiovasc Electrophysiol ; 24(9): 1021-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23631727

ABSTRACT

BACKGROUND: Sinus node (SN) dysfunction is observed in some long-QT syndrome (LQTS) patients, but has not been studied as a function of LQTS genotype. LQTS6 involves mutations in the hERG ß-subunit MiRP1, which also interacts with hyperpolarization-activated, cyclic nucleotide gated (HCN) channels-the molecular correlate of SN pacemaker current (If ). An LQTS registry search identified a 55-year male with M54T MiRP1 mutation, history of sinus bradycardia (39-56 bpm), and prolonged QTc. OBJECTIVE: We tested if LQTS6 incorporates sinus bradycardia due to abnormal If . METHODS: We transiently co-transfected neonatal rat ventricular myocytes (to study currents in a myocyte background) with human HCN4 (hHCN4, primary SN isoform) or human HCN2 (hHCN2) and one of the following: empty vector, wild-type hMiRP1 (WT), M54T hMiRP1 (M54T). Current amplitude, voltage dependence, and kinetics were measured by whole cell patch clamp. RESULTS: M54T co-expression decreased HCN4 current density by 80% compared to hHCN4 alone or with WT, and also slowed HCN4 activation at physiologically relevant voltages. Neither WT nor M54T altered HCN4 voltage dependence. A computer simulation predicts that these changes in HCN4 current would decrease rate and be additive with published effects of M54T mutation on hERG kinetics on rate. CONCLUSIONS: We conclude that M54T LQTS6 mutation can cause sinus bradycardia through effects on both hERG and HCN currents. Patients with other LQTS6 mutations should be examined for SN dysfunction, and the effect on HCN current determined.


Subject(s)
Biological Clocks/genetics , Bradycardia/diagnosis , Bradycardia/genetics , Mutation/genetics , Potassium Channels, Voltage-Gated/genetics , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Down-Regulation/genetics , Humans , Male , Middle Aged , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Rats , Rats, Wistar
18.
J Neurogastroenterol Motil ; 16(3): 265-73, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20680165

ABSTRACT

BACKGROUND/AIMS: Capsaicin (8-methyl-N-vanillyl-6-ninenamide), a compound found in hot peppers, has been reported to have different physiological actions on different cell types. Not much work has been done about the effect of capsaicin on the function of interstitial cells of Cajal (ICC). In the present study, we examined the action of external application of capsaicin on pacemaker activity in the cultured ICC from the small intestine of mouse. METHODS: We investigated the effect of capsaicin on pacemaker currents in cultured ICC from the small intestine of mouse using a whole cell patch-clamp technique and Ca(2+)-imaging analysis. RESULTS: When capsaicin was applied externally to the pacemaker generating ICC, it completely inhibited the pacemaker potential under current-clamp mode (I = 0) and the pacemaker current under voltage-clamp mode at a -70 mV of holding potentials. The effect of capsaicin on pacemaker activity in ICC was shown dose dependently. The effect of capsaicin was not through the transient receptor potential of the vanilloid type 1 (TRPV1) channel as capsazepine did not block the effect of capsaicin. L-NAME, an inhibitor of nitric oxide synthase, also did not block the capsaicin-induced effects. When the action of capsaicin was examined in the intracellular calcium oscillation, it completely abolished the calcium oscillation. CONCLUSIONS: These results prove that the capsaicin has the inhibitory effects on the ICC which is carried out neither through TRPV channel nor the nitric oxide production. Intracellular Ca(2+) was also an important target for actions of capsaicin on ICC.

19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-349805

ABSTRACT

The possibility of rats mesenchymal stem cells (MSCs) modified with murine hyperpolarization-activated cyclic nucleotide-gated 2 (mHCN2) gene as biological pacemakers in vitro was studied.The cultured MSCs were transfected with pIRES2-EGFP plasmid carrying enhanced green fluorescent protein (EGFP) gene and mHCN2 gene.The identification using restriction enzyme and sequencing indicated that the mHCN2 gene was inserted to the pIRES2-EGFP.Green fluorescence could be seen in MSCs after transfection for 24-48 h.The expression of mHCN2 mRNA and protein in the transfected cells was detected by RT-PCR and Western blot,and the quantity of mHCN2 mRNA and protein expression in transfected MSCs was 5.31 times and 7.55 times higher than that of the non-transfected MSCs respectively (P<0.05,P<0.05).IHCN2 was recorded by whole-cell patch clamp method.The effect of Cs+,a specific blocker of pacemaker current,was measured after perfusion by patch clamp.The results of inward current indicated that there was no inward current recording in non-transfected MSCs and a large voltage-dependent inward and Cs+-sensitive current activated on hyperpolarizations presented in the transfected MSCs.IHCN2 was fully activated around -140 mV with an activation threshold of-60 mV.The midpoint (V50) was -95.1±0.9 mV (n=9).The study demonstrates that mHCN2 mRNA and protein can be expressed and the currents of HCN2 channels can be detected in genetically modified MSCs.The gene-modified MSCs present a novel method for pacemaker genes into the heart or other electrical syncytia.

20.
Article in English | WPRIM (Western Pacific) | ID: wpr-103878

ABSTRACT

BACKGROUND/AIMS: Capsaicin (8-methyl-N-vanillyl-6-ninenamide), a compound found in hot peppers, has been reported to have different physiological actions on different cell types. Not much work has been done about the effect of capsaicin on the function of interstitial cells of Cajal (ICC). In the present study, we examined the action of external application of capsaicin on pacemaker activity in the cultured ICC from the small intestine of mouse. METHODS: We investigated the effect of capsaicin on pacemaker currents in cultured ICC from the small intestine of mouse using a whole cell patch-clamp technique and Ca2+-imaging analysis. RESULTS: When capsaicin was applied externally to the pacemaker generating ICC, it completely inhibited the pacemaker potential under current-clamp mode (I = 0) and the pacemaker current under voltage-clamp mode at a -70 mV of holding potentials. The effect of capsaicin on pacemaker activity in ICC was shown dose dependently. The effect of capsaicin was not through the transient receptor potential of the vanilloid type 1 (TRPV1) channel as capsazepine did not block the effect of capsaicin. L-NAME, an inhibitor of nitric oxide synthase, also did not block the capsaicin-induced effects. When the action of capsaicin was examined in the intracellular calcium oscillation, it completely abolished the calcium oscillation. CONCLUSIONS: These results prove that the capsaicin has the inhibitory effects on the ICC which is carried out neither through TRPV channel nor the nitric oxide production. Intracellular Ca2+ was also an important target for actions of capsaicin on ICC.


Subject(s)
Animals , Mice , Calcium Signaling , Capsaicin , Gastrointestinal Motility , Interstitial Cells of Cajal , Intestine, Small , NG-Nitroarginine Methyl Ester , Nitric Oxide , Nitric Oxide Synthase , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL