Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Cells ; 12(24)2023 12 15.
Article in English | MEDLINE | ID: mdl-38132169

ABSTRACT

Atrial fibrillation (AF), characterised by irregular high-frequency contractions of the atria of the heart, is of increasing clinical importance. The reasons are the increasing prevalence and thromboembolic complications caused by AF. So-called atrial remodelling is characterised, among other things, by atrial dilatation and fibrotic remodelling. As a result, AF is self-sustaining and forms a procoagulant state. But hypercoagulation not only appears to be the consequence of AF. Coagulation factors can exert influence on cells via protease-activated receptors (PAR) and thereby the procoagulation state could contribute to the development and maintenance of AF. In this work, the influence of FXa on Heart Like-1 (HL-1) cells, which are murine adult atrial cardiomyocytes (immortalized), was investigated. PAR1, PAR2, and PAR4 expression was detected. After incubations with FXa (5-50 nM; 4-24 h) or PAR1- and PAR2-agonists (20 µM; 4-24 h), no changes occurred in PAR expression or in the inflammatory signalling cascade. There were no time- or concentration-dependent changes in the phosphorylation of the MAP kinases ERK1/2 or the p65 subunit of NF-κB. In addition, there was no change in the mRNA expression of the cell adhesion molecules (ICAM-1, VCAM-1, fibronectin). Thus, FXa has no direct PAR-dependent effects on HL-1 cells. Future studies should investigate the influence of FXa on human cardiomyocytes or on other cardiac cell types like fibroblasts.


Subject(s)
Atrial Fibrillation , Factor Xa , Animals , Mice , Factor Xa/metabolism , NF-kappa B/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Signal Transduction
2.
Biomed Pharmacother ; 162: 114577, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37001181

ABSTRACT

AIMS: Atrial fibrillation (AF) has been associated with altered expression of the transcription factor Pitx2c and a high incidence of calcium release-induced afterdepolarizations. However, the relationship between Pitx2c expression and defective calcium homeostasis remains unclear and we here aimed to determine how Pitx2c expression affects calcium release from the sarcoplasmic reticulum (SR) and its impact on electrical activity in isolated atrial myocytes. METHODS: To address this issue, we applied confocal calcium imaging and patch-clamp techniques to atrial myocytes isolated from a mouse model with conditional atrial-specific deletion of Pitx2c. RESULTS: Our findings demonstrate that heterozygous deletion of Pitx2c doubles the calcium spark frequency, increases the frequency of sparks/site 1.5-fold, the calcium spark decay constant from 36 to 42 ms and the wave frequency from none to 3.2 min-1. Additionally, the cell capacitance increased by 30% and both the SR calcium load and the transient inward current (ITI) frequency were doubled. Furthermore, the fraction of cells with spontaneous action potentials increased from none to 44%. These effects of Pitx2c deficiency were comparable in right and left atrial myocytes, and homozygous deletion of Pitx2c did not induce any further effects on sparks, SR calcium load, ITI frequency or spontaneous action potentials. CONCLUSION: Our findings demonstrate that heterozygous Pitx2c deletion induces defects in calcium homeostasis and electrical activity that mimic derangements observed in right atrial myocytes from patients with AF and suggest that Pitx2c deficiency confers cellular electrophysiological hallmarks of AF to isolated atrial myocytes.


Subject(s)
Atrial Fibrillation , Animals , Mice , Atrial Fibrillation/genetics , Calcium/metabolism , Homozygote , Sequence Deletion , Myocytes, Cardiac/metabolism
3.
Biochem Biophys Res Commun ; 638: 36-42, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36436340

ABSTRACT

We aimed to investigate the effect of apocynin (APO) on delayed afterdepolarizations (DADs) in rat atrial myocytes and the underlying mechanisms. Rat atrial myocytes were isolated by a Langendorff perfusion apparatus. DADs were induced by isoproterenol (ISO). Action potentials (APs) and ion currents were recorded by the whole-cell clamp technique. The fluorescent indicator fluo-4 was used to visualize intracellular Ca2+ transients, and western blotting was used to measure the expression of related proteins. The incidence of DADs in rat atrial myocytes increased significantly after ISO treatment, leading to an increased incidence of triggered activity (TA). The incidence of ISO-induced DADs and TA were reduced by 100.0 µM APO from 48.89% to 25.56% and 17.78% to 5.56%, respectively. In the range of 3.0 µM-300.0 µM, the effect of APO was concentration dependent, with a half maximal inhibitory concentration (IC50) of 120.1 µM and a Hill coefficient of 1.063. APO reversed the increase in transient inward current (Iti) and Na+/Ca2+-exchange current (INCX) densities induced by ISO in atrial myocytes. The frequency of spontaneous Ca2+ transients in atrial myocytes was reduced by 100.0 µM APO. Compared with ISO, APO downregulated the expression of NOX2 and increased the phosphorylation of PLNSer16 and the sarcoplasmic reticulum Ca2+-ATPase-2a (SERCA2a) level; however, it had little effect on ryanodine-receptor channel type-2 (RyR2). These findings showed that APO may block Iti and INCX and reduce intracellular Ca2+ levels in rat atrial myocytes, thus reducing the incidence of ISO-induced DADs and TA.


Subject(s)
Atrial Fibrillation , Rats , Animals , Isoproterenol/pharmacology , Atrial Fibrillation/metabolism , Myocytes, Cardiac/metabolism , Action Potentials , Calcium/metabolism , Sarcoplasmic Reticulum/metabolism
4.
Cells ; 11(19)2022 10 08.
Article in English | MEDLINE | ID: mdl-36231118

ABSTRACT

Arterial hypertension affects ≈ 1 billion people worldwide. It is associated with increased morbidity and mortality and responsible for millions of deaths each year. Hypertension mediates damage of target organs including the heart. In addition to eliciting left ventricular hypertrophy, dysfunction and heart failure, hypertension also causes left atrial remodeling that may culminate in atrial contractile dysfunction and atrial fibrillation. Here, we will summarize data on the various aspects of left atrial remodeling in (essential) hypertension gathered from studies on patients with hypertension and from spontaneously hypertensive rats, an animal model that closely mimics cardiac remodeling in human hypertension. Analyzing the timeline of remodeling processes, i.e., distinguishing between alterations occurring in prehypertension, in early hypertension and during advanced hypertensive heart disease, we will derive the potential mechanisms underlying left atrial remodeling in (essential) hypertension. Finally, we will discuss the consequences of these remodeling processes for atrial and ventricular function. The data imply that left atrial remodeling is multifactorial, starts early in hypertension and is an important contributor to the progression of hypertensive heart disease, including the development of atrial fibrillation and heart failure.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Heart Failure , Hypertension , Animals , Heart Atria , Humans , Hypertension/complications , Myocardium , Rats , Rats, Inbred SHR
5.
Biomolecules ; 12(8)2022 08 15.
Article in English | MEDLINE | ID: mdl-36009014

ABSTRACT

Phenylacetylglutamine (PAGln), a gut microbiota (GM)-derived metabolite, is associated with cardiovascular disease. Studies have shown that disordered GM participated in the progression of atrial fibrillation (AF), but the relationship between PAGln and AF is unclear. This study investigated the characteristics of PAGln in AF patients and its impact on atrial myocytes. Based on our previous metagenomic data, the relative abundance of porA, a critical bacterial enzyme for PAGln synthesis, exhibited an increased tendency in AF. In an independent cohort consisting of 42 controls without AF and 92 AF patients, plasma PAGln levels were higher in AF patients than in controls (p < 0.001) by immunoassay. Notably, PAGln exerted a predictive potential of AF with an AUC of 0.774 (p < 0.001), and a predictive model constructed based on the PAGln and Taiwan AF score further improved the predictive potential. Furthermore, a positive correlation was determined between PAGln and LA diameter. Subsequently, the effect of PAGln intervention was examined on HL-1 cells in vitro, revealing that PAGln increased apoptosis, reactive oxygen species (ROS) production, CaMKII and RyR2 activation and decreased cell viability. In conclusion, increased PAGln was associated with AF, and PAGln might contribute to the AF pathogenesis by promoting oxidative stress and apoptosis in atrial myocytes.


Subject(s)
Atrial Fibrillation , Gastrointestinal Microbiome , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Humans , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
6.
Cells ; 11(14)2022 07 21.
Article in English | MEDLINE | ID: mdl-35883704

ABSTRACT

Protein kinase A (PKA) is a key nodal signaling molecule that regulates a wide range of cellular functions in the cytosol and mitochondria. The distribution of A-kinase anchoring proteins that tether PKA, the local interaction with degradation molecules, and regulation by Ca2+, may lead to distinct spatiotemporal cAMP/PKA signaling in these compartments. In this work, FRET-based sensors were used to investigate PKA signaling in the cytosol, outer mitochondrial membrane (OMM), and mitochondrial matrix (MM) and its crosstalk with Ca2+ in response to electrical stimulation of cultured rabbit atrial cells. A gradual decrease in PKA activity eliminating the ability of the atrial cells to respond to physiological electrical stimulation, was observed upon treatment of cells with H-89. Chelation of intracellular Ca2+ by BAPTA reduced PKA activity and diminished its response to forskolin, an AC stimulator. Under basal conditions, PKA activity in response to forskolin was lower in the OMM compared to the cytosol and MM. In response to electrical stimulation in the presence of ISO, distinct compartmentalization of PKA activity was observed, with higher activity in the cytosol and MM than in the OMM. Thus, distinct Ca2+-dependent spatiotemporal cAMP/PKA signaling exists in atrial cells, likely mediating its excitation and mitochondrial function.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Myocytes, Cardiac , Animals , Colforsin , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , Rabbits
7.
Int J Mol Sci ; 23(15)2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35897638

ABSTRACT

In the atria, the rapid delayed rectifier channel (IKr) is a critical contributor to repolarization. In lipotoxic atria, increased activity of the serine/threonine mammalian target of rapamycin (mTOR) may remodel IKr and predispose patients to arrhythmias. To investigate whether mTOR produced defects in IKr channel function (protein expression and gating mechanisms), electrophysiology and biochemical assays in HEK293 cells stably expressing hERG1a/1b, and adult guinea pig atrial myocytes were used. Feeding with the saturated fatty acid palmitic acid high-fat diet (HFD) was used to induce lipotoxicity. Lipotoxicity-challenged HEK293 cells displayed an increased density of hERG1a/1b currents due to a targeted and significant increase in hERG1b protein expression. Furthermore, lipotoxicity significantly slowed the hERG1a/1b inactivation kinetics, while the activation and deactivation remained essentially unchanged. mTOR complex 1 (mTORC1) inhibition with rapamycin (RAP) reversed the increase in hERG1a/1b density and inactivation. Compared to lipotoxic myocytes, RAP-treated cells displayed action potential durations (APDs) and IKr densities similar to those of controls. HFD feeding triggered arrhythmogenic changes (increased the IKr density and shortened the APD) in the atria, but this was not observed in low-fat-fed controls. The data are the first to show the modulation of IKr by mTORC1, possibly through the remodeling of hERG1b, in lipotoxic atrial myocytes. These results offer mechanistic insights with implications for targeted therapeutic options for the therapy of acquired supraventricular arrhythmias in obesity and associated pathologies.


Subject(s)
Arrhythmias, Cardiac , Myocytes, Cardiac , Action Potentials , Animals , Arrhythmias, Cardiac/metabolism , Guinea Pigs , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocytes, Cardiac/metabolism , TOR Serine-Threonine Kinases/metabolism
8.
Mol Biol Rep ; 49(7): 5939-5952, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35368226

ABSTRACT

BACKGROUND: Gout is usually found in patients with atrial fibrillation (AF). K+ efflux is a common trigger of NLRP3 inflammasome activation which is involved in the pathogenesis of AF. We investigated the role of the K+ channel Kv1.5 in monosodium urate crystal (MSU)-induced activation of the NLRP3 inflammasome and electrical remodeling in mouse and human macrophages J774.1 and THP-1, and mouse atrial myocytes HL-1. METHODS AND RESULTS: Macrophages, primed with lipopolysaccharide (LPS), were stimulated by MSU. HL-1 cells were incubated with the conditioned medium (CM) from MSU-stimulated macrophages. Western blot, ELISA and patch clamp were used. MSU induced caspase-1 expression in LPS-primed J774.1 cells and IL-1ß secretion, suggesting NLRP3 inflammasome activation. A selective Kv1.5 inhibitor, diphenyl phosphine oxide-1 (DPO-1), and siRNAs against Kv1.5 suppressed the levels of caspase-1 and IL-1ß. MSU reduced intracellular K+ concentration which was prevented by DPO-1 and siRNAs against Kv1.5. MSU increased expression of Hsp70, and Kv1.5 on the plasma membrane. siRNAs against Hsp70 were suppressed but heat shock increased the expression of Hsp70, caspase-1, IL-1ß, and Kv1.5 in MSU-stimulated J774.1 cells. The CM from MSU-stimulated macrophages enhanced the expression of caspase-1, IL-1ß and Kv1.5 with increased Kv1.5-mediated currents that shortened action potential duration in HL-1 cells. These responses were abolished by DPO-1 and a siRNA against Kv1.5. CONCLUSIONS: Kv1.5 regulates MSU-induced activation of NLRP3 inflammasome in macrophages. MSUrelated activation of NLRP3 inflammasome and electrical remodeling in HL-1 cells are via macrophages. Kv1.5 may have therapeutic value for diseases related to gout-induced activation of the NLRP3 inflammsome, including AF.


Subject(s)
Atrial Remodeling , Gout , Kv1.5 Potassium Channel/metabolism , Animals , Caspase 1/metabolism , Gout/drug therapy , Gout/metabolism , Gout/pathology , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Uric Acid/metabolism , Uric Acid/pharmacology
9.
Methods Mol Biol ; 2483: 195-204, 2022.
Article in English | MEDLINE | ID: mdl-35286677

ABSTRACT

The ubiquitous second messengers' 3',5'-cyclic adenosine monophosphate (cAMP ) and 3',5'-cyclic guanosine monophosphate (cGMP) are crucial in regulating cardiomyocyte function, as well as pathological processes, by acting in distinct subcellular microdomains and thus controlling excitation-contraction coupling. Spatio-temporal intracellular dynamics of cyclic nucleotides can be measured in living cells using fluorescence resonance energy transfer (FRET ) by transducing isolated cells with genetically encoded biosensors. While FRET experiments have been regularly performed in cardiomyocytes from different animal models, human-based translational experiments are very challenging due to the difficulty to culture and transduce adult human cardiomyocytes. Here, we describe a technique for obtaining human atrial and ventricular myocytes which allows to keep them alive in culture long enough to transduce them and visualize cAMP and cGMP in physiological and pathological human settings.


Subject(s)
Myocytes, Cardiac , Nucleotides, Cyclic , Animals , Cyclic AMP , Cyclic GMP , Fluorescence Resonance Energy Transfer/methods , Humans
10.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638854

ABSTRACT

Atrial fibrillation (AF) is the most common type of cardiac arrhythmia, affecting more than 33 million people worldwide. Despite important advances in therapy, AF's incidence remains high, and treatment often results in recurrence of the arrhythmia. A better understanding of the cellular and molecular changes that (1) trigger AF and (2) occur after the onset of AF will help to identify novel therapeutic targets. Over the past 20 years, a large body of research has shown that intracellular Ca2+ handling is dramatically altered in AF. While some of these changes are arrhythmogenic, other changes counteract cellular arrhythmogenic mechanisms (Calcium Signaling Silencing). The intracellular Na+ concentration ([Na+])i is a key regulator of intracellular Ca2+ handling in cardiac myocytes. Despite its importance in the regulation of intracellular Ca2+ handling, little is known about [Na+]i, its regulation, and how it might be changed in AF. Previous work suggests that there might be increases in the late component of the atrial Na+ current (INa,L) in AF, suggesting that [Na+]i levels might be high in AF. Indeed, a pharmacological blockade of INa,L has been suggested as a treatment for AF. Here, we review calcium signaling silencing and changes in intracellular Na+ homeostasis during AF. We summarize the proposed arrhythmogenic mechanisms associated with increases in INa,L during AF and discuss the evidence from clinical trials that have tested the pharmacological INa,L blocker ranolazine in the treatment of AF.


Subject(s)
Atrial Fibrillation/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Homeostasis/physiology , Myocytes, Cardiac/metabolism , Sodium/metabolism , Animals , Atrial Fibrillation/physiopathology , Humans , Myocytes, Cardiac/cytology , Sarcolemma/metabolism , Sodium-Calcium Exchanger/metabolism
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 41(3): 406-411, 2021 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-33849832

ABSTRACT

OBJECTIVE: To investigate the injury types of atrial myocytes induced by heat exposure and the effect of amiodarone on heat-induced injuries in atrial myocytes. OBJECTIVE: The optimal temperature for heat exposure and optimal concentration of amiodarone were determined by measuring the cell viability exposed to different temperatures and different concentrations of amiodarone. Heat exposure of HL1 atrial myocytes was conducted using a water bath, and the effect of amiodarone on cell viability was assessed with MTS method; cell apoptosis was detected using flow cytometry, and the levels of IL-1ß, IL-6, TNF-α, SOD and MDA were detected with enzyme-linked immunosorbent assay (ELISA). OBJECTIVE: Compared with the blank control cells, the cells exposed to a temperature of 52 ℃ showed a significantly decreased survival rate and a lowered SOD activity (P < 0.001) with increased IL-1ß and MDA levels (P < 0.01) and markedly increased apoptosis rate and IL-6 level (P < 0.001). Compared with the heat exposure group, amiodarone resulted in significantly decreased survival rate of the atrial myocytes (P < 0.01), obviously decreased SOD activity (P < 0.05), and increased cell apoptosis rate (P < 0.05) and IL-1ß, IL-6, MDA and TNF-α levels (P < 0.01 or 0.001). OBJECTIVE: Heat exposure induces apoptosis, inflammation and oxidative stress in mouse HL1 atrial myocytes, and amiodarone can enhance the effects of heat exposure to aggravate the cell injuries.


Subject(s)
Amiodarone , Hot Temperature , Amiodarone/pharmacology , Animals , Apoptosis , Inflammation , Mice , Oxidative Stress
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1014298

ABSTRACT

Aim To explore type 1 diabetes mice and the advance glycation end products (AGE) involved in electrical remodeling of atrial myocytes. Methods The diabetic mouse model was induced by intraperitoneal injection of STZ; action potential duration, and the current density of I

13.
Chinese Pharmacological Bulletin ; (12): 180-186, 2021.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1014313

ABSTRACT

Aim To explore the role of cotranscriptional activator p300 in regulating the electrical remodeling of atrial myocytes in aging mouse, which resulted in atrial fibrillation. Methods The left atrial appendage tissues of 5 , 13 and 18monthold C57BL/6 mice were collected respectively. Western blot was used to detect the protein expression levels of p300, L type calcium channel (Cavl. 2) and aging related protein p53/p21. Acute enzymatic hydrolysis was used to isolate single atrial myocytes, and the wholecell patchclamp technique was used to detect the Ltype calcium current (I

14.
Heart Rhythm O2 ; 1(3): 206-214, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32864638

ABSTRACT

BACKGROUND: Atrial-ventricular differences in voltage-gated Na+ currents might be exploited for atrial-selective antiarrhythmic drug action for the suppression of atrial fibrillation without risk of ventricular tachyarrhythmia. Eleclazine (GS-6615) is a putative antiarrhythmic drug with properties similar to the prototypical atrial-selective Na+ channel blocker ranolazine that has been shown to be safe and well tolerated in patients. OBJECTIVE: The present study investigated atrial-ventricular differences in the biophysical properties and inhibition by eleclazine of voltage-gated Na+ currents. METHODS: The fast and late components of whole-cell voltage-gated Na+ currents (respectively, I Na and I NaL) were recorded at room temperature (∼22°C) from rat isolated atrial and ventricular myocytes. RESULTS: Atrial I Na activated at command potentials ∼5.5 mV more negative and inactivated at conditioning potentials ∼7 mV more negative than ventricular I Na. There was no difference between atrial and ventricular myocytes in the eleclazine inhibition of I NaL activated by 3 nM ATX-II (IC50s ∼200 nM). Eleclazine (10 µM) inhibited I Na in atrial and ventricular myocytes in a use-dependent manner consistent with preferential activated state block. Eleclazine produced voltage-dependent instantaneous inhibition in atrial and ventricular myocytes; it caused a negative shift in voltage of half-maximal inactivation and slowed the recovery of I Na from inactivation in both cell types. CONCLUSIONS: Differences exist between rat atrial and ventricular myocytes in the biophysical properties of I Na. The more negative voltage dependence of I Na activation/inactivation in atrial myocytes underlies differences between the 2 cell types in the voltage dependence of instantaneous inhibition by eleclazine. Eleclazine warrants further investigation as an atrial-selective antiarrhythmic drug.

15.
J Mol Cell Cardiol ; 144: 127-139, 2020 07.
Article in English | MEDLINE | ID: mdl-32445844

ABSTRACT

Ion channel trafficking powerfully influences cardiac electrical activity as it regulates the number of available channels at the plasma membrane. Studies have largely focused on identifying the molecular determinants of the trafficking of the atria-specific KV1.5 channel, the molecular basis of the ultra-rapid delayed rectifier current IKur. Besides, regulated KV1.5 channel recycling upon changes in homeostatic state and mechanical constraints in native cardiomyocytes has been well documented. Here, using cutting-edge imaging in live myocytes, we investigated the dynamics of this channel in the plasma membrane. We demonstrate that the clathrin pathway is a major regulator of the functional expression of KV1.5 channels in atrial myocytes, with the microtubule network as the prominent organizer of KV1.5 transport within the membrane. Both clathrin blockade and microtubule disruption result in channel clusterization with reduced membrane mobility and internalization, whereas disassembly of the actin cytoskeleton does not. Mobile KV1.5 channels are associated with the microtubule plus-end tracking protein EB1 whereas static KV1.5 clusters are associated with stable acetylated microtubules. In human biopsies from patients in atrial fibrillation associated with atrial remodeling, drastic modifications in the trafficking balance occurs together with alteration in microtubule polymerization state resulting in modest reduced endocytosis and increased recycling. Consequently, hallmark of atrial KV1.5 dynamics within the membrane is clathrin- and microtubule- dependent. During atrial remodeling, predominance of anterograde trafficking activity over retrograde trafficking could result in accumulation ok KV1.5 channels in the plasma membrane.


Subject(s)
Clathrin/metabolism , Microtubules/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Multimerization , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Remodeling/genetics , Clathrin/chemistry , Clathrin-Coated Vesicles , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Electrophysiological Phenomena , Heart Atria/metabolism , Humans , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Microtubules/chemistry , Microtubules/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Potassium Channels, Voltage-Gated/chemistry , Rats , Sarcolemma/metabolism , Signal Transduction
16.
Heart Rhythm ; 16(10): 1575-1583, 2019 10.
Article in English | MEDLINE | ID: mdl-30954598

ABSTRACT

BACKGROUND: Our recent study demonstrated that the nonselective cation current mediated by the transient receptor potential canonical 1 (TRPC1) channel is activated by endothelin-1 (ET-1) in human atrial myocytes; however, the related signal molecules involved are unknown. OBJECTIVE: The purpose of this study was to investigate how the TRPC1 channel is regulated by ET-1 and whether it is upregulated in human atria from patients with atrial fibrillation (AF). METHODS: Whole-cell patch technique and molecular biology techniques were used in the study. RESULTS: The ET-1-evoked TRPC1 current was inhibited by the ET-1 type A (ETA) receptor antagonist BQ123 and the ET-1 type B (ETB) receptor antagonist BQ788 as well as the protein kinase C inhibitor chelerythrine. ETA receptor-mediated TRPC1 channel activity was selectively inhibited by the phosphoinositide-3-kinase inhibitor wortmannin, while ETB receptor-mediated TRPC1 activity was inhibited by the phospholipase C inhibitor U73122. The messenger RNAs and proteins of the TRPC1 channel and ETA receptor, but not the ETB receptor, were significantly upregulated in atria from patients with AF. The basal TRPC1 current increased in AF myocytes, and the response to ET-1 was greater in AF myocytes than in sinus rhythm myocytes. ET-1 induced a delayed repolarization in 20% of AF myocytes. CONCLUSION: These results demonstrate for the first time that TRPC1 activation by ET-1 is mediated by protein kinase C through the distinct phospholipids pathways phosphoinositide-3-kinase and phospholipase C and that the TRPC1 channel and ETA receptor are upregulated in AF atria, which are likely involved in atrial electrical remodeling in patients with AF.


Subject(s)
Atrial Fibrillation/genetics , Endothelin-1/pharmacology , Protein Kinase C/metabolism , Signal Transduction/genetics , TRPC Cation Channels/genetics , Atrial Fibrillation/physiopathology , Blotting, Western , Cells, Cultured , Heart Atria/metabolism , Humans , Myocytes, Cardiac/metabolism , Real-Time Polymerase Chain Reaction/methods , Reference Values , Sensitivity and Specificity , Up-Regulation/genetics
17.
Cardiovasc J Afr ; 30(2): 79-86, 2019.
Article in English | MEDLINE | ID: mdl-30882133

ABSTRACT

AIM: We aimed to study the effect of allocryptopine (All) on the late sodium current (INa,Late) of atrial myocytes in spontaneously hypertensive rats (SHR). METHODS: The enzyme digestion method was used to separate single atrial myocytes from SHR and Wistar-Kyoto (WKY) rats. INa,Late was recorded using the patch-clamp technique, and the effect of All was evaluated on the current. RESULTS: Compared with WKY rat cells, an increase in the INa,Late current in SHR myocytes was found. After treatment with 30 µM All, the current densities were markedly decreased; the ratio of INa,Late/INa,peak of SHR was reduced by 30 µM All. All reduced INa,Late by alleviating inactivation of the channel and increasing the window current of the sodium channel. Furthermore, INa,Late densities of three SCN5A mutations declined substantially with 30 µM All in a concentration-dependent manner. CONCLUSIONS: The results clearly show that an increase in INa,Late in SHR atrial myocytes was inhibited by All derived from Chinese herbal medicine.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Berberine Alkaloids/pharmacology , Heart Atria/drug effects , Hypertension/drug therapy , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Sodium/metabolism , Action Potentials , Animals , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , HEK293 Cells , Heart Atria/metabolism , Heart Rate , Humans , Hypertension/complications , Hypertension/metabolism , Hypertension/physiopathology , Male , Mutation , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors
18.
Cardiovasc Res ; 115(3): 578-589, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30219899

ABSTRACT

AIMS: Single nucleotide polymorphisms on chromosome 4q25 have been associated with risk of atrial fibrillation (AF) but the exiguous knowledge of the mechanistic links between these risk variants and underlying electrophysiological alterations hampers their clinical utility. Here, we tested the hypothesis that 4q25 risk variants cause alterations in the intracellular calcium homoeostasis that predispose to spontaneous electrical activity. METHODS AND RESULTS: Western blotting, confocal calcium imaging, and patch-clamp techniques were used to identify mechanisms linking the 4q25 risk variants rs2200733T and rs13143308T to defects in the calcium homoeostasis in human atrial myocytes. Our findings revealed that the rs13143308T variant was more frequent in patients with AF and that myocytes from carriers of this variant had a significantly higher density of calcium sparks (14.1 ± 4.5 vs. 3.1 ± 1.3 events/min, P = 0.02), frequency of transient inward currents (ITI) (1.33 ± 0.24 vs. 0.26 ± 0.09 events/min, P < 0.001) and incidence of spontaneous membrane depolarizations (1.22 ± 0.26 vs. 0.56 ± 0.17 events/min, P = 0.001) than myocytes from patients with the normal rs13143308G variant. These alterations were linked to higher sarcoplasmic reticulum calcium loading (10.2 ± 1.4 vs. 7.3 ± 0.5 amol/pF, P = 0.01), SERCA2 expression (1.37 ± 0.13 fold, P = 0.03), and RyR2 phosphorylation at ser2808 (0.67 ± 0.08 vs. 0.47 ± 0.03, P = 0.01) but not at ser2814 (0.28 ± 0.14 vs. 0.31 ± 0.14, P = 0.61) in patients carrying the rs13143308T risk variant. Furthermore, the presence of a risk variant or AF independently increased the ITI frequency and the increase in the ITI frequency observed in carriers of the risk variants was exacerbated in those with AF. By contrast, the presence of a risk variant did not affect the amplitude or properties of the L-type calcium current in patients with or without AF. CONCLUSIONS: Here, we identify the 4q25 variant rs13143308T as a genetic risk marker for AF, specifically associated with excessive calcium release and spontaneous electrical activity linked to increased SERCA2 expression and RyR2 phosphorylation.


Subject(s)
Atrial Fibrillation/genetics , Calcium Signaling/genetics , Calcium/metabolism , Chromosomes, Human, Pair 4 , Heart Atria/metabolism , Myocytes, Cardiac/metabolism , Polymorphism, Single Nucleotide , Action Potentials/genetics , Aged , Atrial Fibrillation/diagnosis , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Case-Control Studies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Heart Atria/physiopathology , Heart Rate/genetics , Homeostasis , Humans , Male , Myocytes, Cardiac/pathology , Phenotype , Phosphorylation , Risk Factors , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
19.
Stem Cell Reports ; 11(6): 1378-1390, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30416051

ABSTRACT

Cardiomyocytes (CMs) generated from human induced pluripotent stem cells (hiPSCs) are under investigation for their suitability as human models in preclinical drug development. Antiarrhythmic drug development focuses on atrial biology for the treatment of atrial fibrillation. Here we used recent retinoic acid-based protocols to generate atrial CMs from hiPSCs and establish right atrial engineered heart tissue (RA-EHT) as a 3D model of human atrium. EHT from standard protocol-derived hiPSC-CMs (Ctrl-EHT) and intact human muscle strips served as comparators. RA-EHT exhibited higher mRNA and protein concentrations of atrial-selective markers, faster contraction kinetics, lower force generation, shorter action potential duration, and higher repolarization fraction than Ctrl-EHTs. In addition, RA-EHTs but not Ctrl-EHTs responded to pharmacological manipulation of atrial-selective potassium currents. RA- and Ctrl-EHTs' behavior reflected differences between human atrial and ventricular muscle preparations. Taken together, RA-EHT is a model of human atrium that may be useful in preclinical drug screening.


Subject(s)
Heart Atria/anatomy & histology , Models, Cardiovascular , Tissue Engineering/methods , 4-Aminopyridine/pharmacology , Action Potentials/drug effects , Biomarkers/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cell Size/drug effects , Gene Expression Regulation/drug effects , Heart Atria/cytology , Humans , Kinetics , Myocardial Contraction/drug effects , Organ Specificity/drug effects , Organ Specificity/genetics , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Receptors, Muscarinic/metabolism , Tretinoin/pharmacology
20.
Cell Physiol Biochem ; 50(6): 2296-2313, 2018.
Article in English | MEDLINE | ID: mdl-30423577

ABSTRACT

BACKGROUND/AIMS: The atrium is exposed to high shear stress during heart failure and valvular diseases. We aimed to understand atrial shear-induced Ca2+ signaling and its underlying mechanisms. METHODS: Pressurized micro-flow was applied to single rat atrial myocytes, and Ca2+ signal, membrane potential, and ATP release were assessed using confocal imaging, patch clamp technique, and luciferin-luciferase assay, respectively. RESULTS: Shear stress (∼16 dyn/cm2) induced global Ca2+ waves (∼0.1 events/s) from the periphery to the center of cells in a transverse direction ("T-wave"; ∼145 µm/s). Pharmacological interventions and simultaneous recording of membrane potential and Ca2+ demonstrated that shear-induced T-waves resulted from action potential (AP)-triggered Ca2+ release from the sarcoplasmic reticulum. T-waves were not sensitive to inhibitors of known shear signaling mechanisms except connexin hemichannels and ATP release. Shear stress caused ATP release from these myocytes (∼1.1x10-17 moles/unit membrane, µm2); ATP release was increased by enhancement of connexin hemichannels and suppressed by inhibition of the hemichannels, but not affected by inhibitors of other ATP release pathways. Blockade of P2X receptor, but not pannexin or the Na+-Ca2+ exchanger, eliminated shear-induced T-wave initiation. CONCLUSION: Our data suggest that shear stress triggers APs and concomitant Ca2+ signaling via activation of P2X receptors by connexin hemichannel-mediated ATP release in atrial myocytes.


Subject(s)
Calcium Signaling , Receptors, Purinergic P2X/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Connexins/metabolism , Male , Membrane Potentials/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NADPH Oxidases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X/chemistry , Sarcoplasmic Reticulum/metabolism , Shear Strength , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism , Tetrodotoxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL