Action potential duration

动作电位持续时间
  • 文章类型: Journal Article
    我们探索了钠-葡萄糖共转运蛋白2抑制剂empagliflozin在横行主动脉缩窄(TAC)后对完整的实验性肥大小鼠心脏的生理作用。术后药物(2-6周)激发导致晚期Na+电流减少,和增加磷酸化(p-)CaMK-II和Nav1.5,但不是总(t)-CaMK-II,和Na+/Ca2+交换表达,确认以前的心肌细胞水平报告。它挽救了TAC引起的超声心动图射血分数和缩短分数的减少,和舒张前后壁增厚。Langendorff灌注心脏的双电压和Ca2光学作图表明,依帕格列净在80%恢复时(APD80)挽救了TAC诱导的动作电位持续时间增加,恢复80%时的Ca2+瞬态峰值信号和持续时间(CaTD80),在常规10Hz刺激期间达到峰值Ca2+(TTP100)和Ca2+衰变常数(Decay30-90)的倍数,和Ca2+瞬时交替循环长度缩短。异丙肾上腺素在假手术和仅TAC心脏中缩短了APD80,在所有组中缩短CaTD80和Decay30-90,但保留TTP100和Ca2瞬时交替。所有组显示相似的APD80,而仅TAC的心脏显示更大的CaTD80,异丙肾上腺素攻击后的异质性。Empagliflozin消除或减少了室性心动过速和室性早搏以及相关的折返传导模式,在异丙肾上腺素激发的TAC手术心脏中,连续爆发起搏发作。Empagliflozin从而挽救TAC诱导的心室肥厚和收缩功能,Ca2+稳态,和完整心脏的致心律失常变化。
    We explored physiological effects of the sodium-glucose co-transporter-2 inhibitor empagliflozin on intact experimentally hypertrophic murine hearts following transverse aortic constriction (TAC). Postoperative drug (2-6 weeks) challenge resulted in reduced late Na+ currents, and increased phosphorylated (p-)CaMK-II and Nav1.5 but not total (t)-CaMK-II, and Na+/Ca2+ exchanger expression, confirming previous cardiomyocyte-level reports. It rescued TAC-induced reductions in echocardiographic ejection fraction and fractional shortening, and diastolic anterior and posterior wall thickening. Dual voltage- and Ca2+-optical mapping of Langendorff-perfused hearts demonstrated that empagliflozin rescued TAC-induced increases in action potential durations at 80% recovery (APD80), Ca2+ transient peak signals and durations at 80% recovery (CaTD80), times to peak Ca2+ (TTP100) and Ca2+ decay constants (Decay30-90) during regular 10-Hz stimulation, and Ca2+ transient alternans with shortening cycle length. Isoproterenol shortened APD80 in sham-operated and TAC-only hearts, shortening CaTD80 and Decay30-90 but sparing TTP100 and Ca2+ transient alternans in all groups. All groups showed similar APD80, and TAC-only hearts showed greater CaTD80, heterogeneities following isoproterenol challenge. Empagliflozin abolished or reduced ventricular tachycardia and premature ventricular contractions and associated re-entrant conduction patterns, in isoproterenol-challenged TAC-operated hearts following successive burst pacing episodes. Empagliflozin thus rescues TAC-induced ventricular hypertrophy and systolic functional, Ca2+ homeostatic, and pro-arrhythmogenic changes in intact hearts.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    计算机建模提供了补充和加速心脏安全性测试的机会。通过计算机建模,计算模拟方法用于预测电生理相互作用和药物对关键生理过程的药理作用。O'Hara-Rudy's模型用于预测对不同离子通道抑制水平对心脏动作电位持续时间(APD)的反应,已知该持续时间与QT间期直接相关。从模型中得出30%60%和90%抑制的APD数据,以描绘可能的室性心律失常情况以及每个离子通道对模型的边际贡献。计算心外膜的动作电位值,心肌,和心内膜细胞,用动作电位曲线建模。这项研究评估了心脏离子通道抑制数据组合,以在进行综合体外致心律失常试验(CiPA)中规定的心律失常效应的计算机模拟时考虑。不出所料,我们的数据强调了延迟整流钾通道(IKr)作为对APD延长影响最大的通道的重要性.瞬时外向钾通道(Ito)抑制对APD的影响最小,而内向整流(IK1)和延迟整流钾通道(IKs)的慢速成分也具有有限的APD作用。相比之下,快速钠通道(INa)和/或L型钙通道(ICa)抑制的作用导致大量APD改变,支持使用有限数量的心脏离子通道(包括IKr)的输入进行计算机模拟建模的药理学相关性,INa,还有ICa,至少在药物开发的早期阶段。
    In silico modeling offers an opportunity to supplement and accelerate cardiac safety testing. With in silico modeling, computational simulation methods are used to predict electrophysiological interactions and pharmacological effects of novel drugs on critical physiological processes. The O\'Hara-Rudy\'s model was developed to predict the response to different ion channel inhibition levels on cardiac action potential duration (APD) which is known to directly correlate with the QT interval. APD data at 30% 60% and 90% inhibition were derived from the model to delineate possible ventricular arrhythmia scenarios and the marginal contribution of each ion channel to the model. Action potential values were calculated for epicardial, myocardial, and endocardial cells, with action potential curve modeling. This study assessed cardiac ion channel inhibition data combinations to consider when undertaking in silico modeling of proarrhythmic effects as stipulated in the Comprehensive in Vitro Proarrhythmia Assay (CiPA). As expected, our data highlight the importance of the delayed rectifier potassium channel (IKr) as the most impactful channel for APD prolongation. The impact of the transient outward potassium channel (Ito) inhibition on APD was minimal while the inward rectifier (IK1) and slow component of the delayed rectifier potassium channel (IKs) also had limited APD effects. In contrast, the contribution of fast sodium channel (INa) and/or L-type calcium channel (ICa) inhibition resulted in substantial APD alterations supporting the pharmacological relevance of in silico modeling using input from a limited number of cardiac ion channels including IKr, INa, and ICa, at least at an early stage of drug development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    全基因组关联研究(GWAS)报道了RING指E3泛素蛋白连接酶rififylin(RFFL)的单核苷酸多态性与人类QT间期变异性之间的相关性(Newton-Cheh等。,2009).以前,我们已经表明,RFFL下调成年兔心室心肌细胞(ARbCM)中的人样ether-a-go-go相关基因(hERG)钾通道和相应的快速激活延迟整流钾电流(IKr)的表达和功能.在这里,我们报告RFFL也影响瞬态外向电流(Ito),但以一种奇特的方式。ARbCM中的RFFL过表达显着降低了其快速成分的贡献(Ito,f)从35%到21%,并增加其慢成分的贡献(伊藤,s)从65%到79%。自从伊藤,在兔中主要通过Kv4.3进行,我们研究了RFFL对HEK293A细胞中表达的Kv4.3的影响。我们发现RFFL过表达降低了Kv4.3的表达和相应的Ito,f在存在或不存在其辅助亚基KChIP2的情况下以RING结构域依赖性方式。另一方面,在表达Kv1.4的HEK细胞中RFFL过表达导致Kv1.4表达水平和Ito,s,类似地,以依赖于RING域的方式。我们的生理详细的兔心室肌细胞计算模型表明,RFFL过表达对Ito的阴阳效应,f,还有伊藤,s影响动作电位波形的1相,并且除了抑制IKr外还略微减少其持续时间。因此,RFFL通过不同地影响各种钾通道和心脏动作电位持续时间(APD)的多种蛋白质的泛素化来修饰心脏复极化储备。
    Genome-wide association studies have reported a correlation between a SNP of the RING finger E3 ubiquitin protein ligase rififylin (RFFL) and QT interval variability in humans (Newton-Cheh et al., 2009). Previously, we have shown that RFFL downregulates expression and function of the human-like ether-a-go-go-related gene potassium channel and corresponding rapidly activating delayed rectifier potassium current (IKr) in adult rabbit ventricular cardiomyocytes. Here, we report that RFFL also affects the transient outward current (Ito), but in a peculiar way. RFFL overexpression in adult rabbit ventricular cardiomyocytes significantly decreases the contribution of its fast component (Ito,f) from 35% to 21% and increases the contribution of its slow component (Ito,s) from 65% to 79%. Since Ito,f in rabbits is mainly conducted by Kv4.3, we investigated the effect of RFFL on Kv4.3 expressed in HEK293A cells. We found that RFFL overexpression reduced Kv4.3 expression and corresponding Ito,f in a RING domain-dependent manner in the presence or absence of its accessory subunit Kv channel-interacting protein 2. On the other hand, RFFL overexpression in Kv1.4-expressing HEK cells leads to an increase in both Kv1.4 expression level and Ito,s, similarly in a RING domain-dependent manner. Our physiologically detailed rabbit ventricular myocyte computational model shows that these yin and yang effects of RFFL overexpression on Ito,f, and Ito,s affect phase 1 of the action potential waveform and slightly decrease its duration in addition to suppressing IKr. Thus, RFFL modifies cardiac repolarization reserve via ubiquitination of multiple proteins that differently affect various potassium channels and cardiac action potential duration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:短QT综合征3型(SQTS3)是一种罕见的致心律失常疾病,由编码内向整流钾通道Kir2.1的基因KCNJ2的功能获得突变引起。我们使用多学科方法,并研究了在QT间期和阵发性房颤患者中发现的新生突变Kir2.1E299V的体内模型中的致心律失常机制。
    结果:我们使用静脉内腺相关病毒介导的基因转移来产生小鼠模型,并证实了Kir2.1WT或Kir2.1E299V的心脏特异性表达。心电图上,Kir2.1E299V小鼠概括了患者的QT间期缩短和心房特异性心律失常。在Kir2.1E299V小鼠中PR间隔也显著较短。膜片钳在心房和心室Kir2.1E299V心肌细胞中显示出极短的动作电位,这是由于缺乏向内整流,并且在电压为-80mV时IK1升高。相对于Kir2.1WT,心房Kir2.1E299V心肌细胞在电压为负至-80mV时斜率电导显著降低.在确认更高比例的异四聚体Kir2后。在心房中包含Kir2.2亚基的x通道,计算机模拟3D模拟预测了Kir2.1E299V-Kir2.2WT通道中多胺阻滞的心房特异性损伤和孔径减小。在心室心肌细胞中,突变通过在超极化方向上改变INa激活和失活来增加兴奋性,保护心室不发生心律失常.此外,来自Kir2.1E299V小鼠的Purkinje肌细胞表现出明显高于Kir2.1WT的INa密度,解释PR间隔中的缩写。
    结论:第一个心脏特异性SQTS3的体内小鼠模型概括了具有Kir2.1E299V突变的患者的电生理表型。Kir2.1E299V消除了两个心腔的整流,但通过增加浦肯野纤维网络和心室的兴奋性来防止室性心律失常。因此,主要的心律失常可能是由于Kir2.1E299V-Kir2.2WT异四聚体缺乏向内矫正和心房特异性孔径减小所致的室上性心律失常.
    OBJECTIVE: Short QT syndrome type 3 (SQTS3) is a rare arrhythmogenic disease caused by gain-of-function mutations in KCNJ2, the gene coding the inward rectifier potassium channel Kir2.1. We used a multidisciplinary approach and investigated arrhythmogenic mechanisms in an in-vivo model of de-novo mutation Kir2.1E299V identified in a patient presenting an extremely abbreviated QT interval and paroxysmal atrial fibrillation.
    RESULTS: We used intravenous adeno-associated virus-mediated gene transfer to generate mouse models, and confirmed cardiac-specific expression of Kir2.1WT or Kir2.1E299V. On ECG, the Kir2.1E299V mouse recapitulated the QT interval shortening and the atrial-specific arrhythmia of the patient. The PR interval was also significantly shorter in Kir2.1E299V mice. Patch-clamping showed extremely abbreviated action potentials in both atrial and ventricular Kir2.1E299V cardiomyocytes due to a lack of inward-going rectification and increased IK1 at voltages positive to -80 mV. Relative to Kir2.1WT, atrial Kir2.1E299V cardiomyocytes had a significantly reduced slope conductance at voltages negative to -80 mV. After confirming a higher proportion of heterotetrameric Kir2.x channels containing Kir2.2 subunits in the atria, in-silico 3D simulations predicted an atrial-specific impairment of polyamine block and reduced pore diameter in the Kir2.1E299V-Kir2.2WT channel. In ventricular cardiomyocytes, the mutation increased excitability by shifting INa activation and inactivation in the hyperpolarizing direction, which protected the ventricle against arrhythmia. Moreover, Purkinje myocytes from Kir2.1E299V mice manifested substantially higher INa density than Kir2.1WT, explaining the abbreviation in the PR interval.
    CONCLUSIONS: The first in-vivo mouse model of cardiac-specific SQTS3 recapitulates the electrophysiological phenotype of a patient with the Kir2.1E299V mutation. Kir2.1E299V eliminates rectification in both cardiac chambers but protects against ventricular arrhythmias by increasing excitability in both Purkinje-fiber network and ventricles. Consequently, the predominant arrhythmias are supraventricular likely due to the lack of inward rectification and atrial-specific reduced pore diameter of the Kir2.1E299V-Kir2.2WT heterotetramer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    细胞外钾浓度可能会改变缺血心肌边界区(BZ)的电生理特性。我们评估了[K]变化下整个缺血-正常边界的去极化和复极化特征。在急性缺血/再灌注模型中,对26只大鼠([K]2.3-6.4mM)进行了64导联心外膜标测。[K+]<4.7mM(低钾正常)的动物有一个ST段抬高和激活延迟的缺血区,具有ST段抬高且无激活延迟的BZ,和一个没有电生理异常的正常区域。[K]>4.7mM(正常高钾)的动物仅具有缺血区和正常区,没有过渡区。在线性回归分析中,激活-复极化间隔和局部传导速度与[K]成反比,并调整了心肌区域。与正常高钾动物相比,低正常动物的再灌注收缩负荷(ESB)更大。两组之间室性心动过速/纤颤的发生率没有差异。在膜片钳实验中,低氧在5.4mM时缩短了动作电位持续时间,但在1.3mM[K]时则没有。IK(ATP)电流在1.3mM低于在5.4mM的[K+]。我们得出的结论是,低正常[K]中的BZ形成与IK(ATP)对缺氧的反应减弱和再灌注ESB增加有关。
    Extracellular potassium concentration might modify electrophysiological properties in the border zone of ischemic myocardium. We evaluated the depolarization and repolarization characteristics across the ischemic-normal border under [K+] variation. Sixty-four-lead epicardial mapping was performed in 26 rats ([K+] 2.3-6.4 mM) in a model of acute ischemia/reperfusion. The animals with [K+] < 4.7 mM (low-normal potassium) had an ischemic zone with ST-segment elevation and activation delay, a border zone with ST-segment elevation and no activation delay, and a normal zone without electrophysiological abnormalities. The animals with [K+] >4.7 mM (normal-high potassium) had only the ischemic and normal zones and no transitional area. Activation-repolarization intervals and local conduction velocities were inversely associated with [K+] in linear regression analysis with adjustment for the zone of myocardium. The reperfusion extrasystolic burden (ESB) was greater in the low-normal as compared to normal-high potassium animals. Ventricular tachycardia/fibrillation incidence did not differ between the groups. In patch-clamp experiments, hypoxia shortened action potential duration at 5.4 mM but not at 1.3 mM of [K+]. IK(ATP) current was lower at 1.3 mM than at 5.4 mM of [K+]. We conclude that the border zone formation in low-normal [K+] was associated with attenuation of IK(ATP) response to hypoxia and increased reperfusion ESB.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心肌梗死(MI)后恶性室性心律失常(VA)主要由心肌电生理重构引起。Brahma相关基因1(BRG1)是ATPase催化亚基,属于染色质重塑复合物家族,称为开关/蔗糖非发酵染色质(SWI/SNF)。BRG1已被报道为分子伴侣,与各种转录因子或蛋白质相互作用以调节心脏疾病中的转录。在这项研究中,我们研究了BRG1在缺血性梗死后离子通道重塑和VA中的潜在作用。通过结扎左冠状动脉前降支(LAD)建立心肌梗死(MI)小鼠,监测心电图(ECG)。使用心外膜光学电压标测在Langendorff灌注的心脏中表征MI小鼠心脏的心外膜传导。在从小鼠分离的单个心室心肌细胞中进行膜片钳分析。我们表明,在MI后的第一周,边界区的BRG1表达逐渐增加。通过尾静脉注射AAV9-BRG1-shRNA对BRG1的心脏特异性缺失显着改善了MI小鼠对电诱导的VA的敏感性并缩短了QTc间隔。BRG1敲除显着提高了MI小鼠心脏的传导速度(CV)并逆转了延长的动作电位持续时间。此外,BRG1敲低改善了Na电流(INa)和瞬时外向钾电流(Ito)的密度降低,以及在MI小鼠心脏边界区和缺氧处理的新生小鼠心室心肌细胞中Nav1.5和Kv4.3的表达。我们发现MI增加了BRG1,T细胞因子4(TCF4)和β-catenin之间的结合,形成转录复合物,抑制SCN5A和KCND3的转录活性,从而影响MI后VA的发生率。
    Malignant ventricular arrhythmia (VA) after myocardial infarction (MI) is mainly caused by myocardial electrophysiological remodeling. Brahma-related gene 1 (BRG1) is an ATPase catalytic subunit that belongs to a family of chromatin remodeling complexes called Switch/Sucrose Non-Fermentable Chromatin (SWI/SNF). BRG1 has been reported as a molecular chaperone, interacting with various transcription factors or proteins to regulate transcription in cardiac diseases. In this study, we investigated the potential role of BRG1 in ion channel remodeling and VA after ischemic infarction. Myocardial infarction (MI) mice were established by ligating the left anterior descending (LAD) coronary artery, and electrocardiogram (ECG) was monitored. Epicardial conduction of MI mouse heart was characterized in Langendorff-perfused hearts using epicardial optical voltage mapping. Patch-clamping analysis was conducted in single ventricular cardiomyocytes isolated from the mice. We showed that BRG1 expression in the border zone was progressively increased in the first week following MI. Cardiac-specific deletion of BRG1 by tail vein injection of AAV9-BRG1-shRNA significantly ameliorated susceptibility to electrical-induced VA and shortened QTc intervals in MI mice. BRG1 knockdown significantly enhanced conduction velocity (CV) and reversed the prolonged action potential duration in MI mouse heart. Moreover, BRG1 knockdown improved the decreased densities of Na+ current (INa) and transient outward potassium current (Ito), as well as the expression of Nav1.5 and Kv4.3 in the border zone of MI mouse hearts and in hypoxia-treated neonatal mouse ventricular cardiomyocytes. We revealed that MI increased the binding among BRG1, T-cell factor 4 (TCF4) and β-catenin, forming a transcription complex, which suppressed the transcription activity of SCN5A and KCND3, thereby influencing the incidence of VA post-MI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:除QT亚间期测量外,评估T波对称性可以提供与心律失常发生相关的心室复极异常的新的独立数据。然而,T波对称性变化的原因尚不完全清楚。计算机研究表明,更对称的T波与较短的动作电位持续时间(APD)和较大的心室复极离散度(DOR)有关。本模拟的目的是研究T波对称性与动作电位(AP)形状之间的关联。
    方法:使用形状为心室壁段的细胞自动机模型来模拟心电图,和两个生物物理详细的心室AP模型-兔子和人类。对称比(SR)被计算为T波开始-峰与峰-末端面积比。APD的个体效应和综合效应,对SR上的DOR和AP形状进行了模拟。为了研究AP形状的影响,模拟了从三角形到矩形的不同AP。
    结果:模拟表明,AP形状以及APD和DOR对T波对称性有很大贡献。在所有模拟中,具有平坦阶段3(三角形)的AP产生非对称T波(SR≥1.5),除了最短的APD范围。具有快速阶段3(矩形)的AP在短APD和长APD上都与更对称的T波(SR≤=1)相关。
    结论:SR标记与标准ECG参数(QT间期,Tpeak-Tend间隔)可能有助于识别心律失常的三角AP形状。
    Assessing T-wave symmetry in addition to QT subintervals measurements can provide novel independent data about ventricular repolarization abnormalities linked with arrhythmogenesis. However, the causes of the changes of T-wave symmetry are not completely understood. In silico studies showed that the more symmetrical T-waves were associated with shorter action potential duration (APD) and larger dispersion of ventricular repolarization (DOR). The aim of present simulation was to study the association between T-wave symmetry and action potential (AP) shape.
    ECGs were simulated using a cellular automata model shaped as a ventricular wall segment, and two biophysically-detailed models of ventricular AP - the rabbit and the human. The symmetry ratio (SR) was calculated as a T-wave onset-peak to peak-end area ratio. The individual and combined effects of APD, DOR and AP shape on SR were simulated. To study the effect of AP shape, different APs from triangulated to rectangular were simulated.
    The simulations showed that AP shape along with APD and DOR contributes much to T-wave symmetry. APs with a flat phase 3 (triangulated) produced asymmetrical T-waves (SR ≥ 1.5) in all simulations, except the shortest APD range. APs with a rapid phase 3 (rectangular) were associated with more symmetrical T-waves (SR ≤ =1) both at the short and the long APDs.
    SR marker in combination with the standard ECG parameters (QT interval, Tpeak-Tend interval) may be useful to identify the proarrhythmic triangulated AP shape.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:室性心动过速(VT)的电生理标测繁琐且可重复性差。对成像的衬底分析不能明确显示VT电路。
    目的:本研究试图引入一种基于计算机断层扫描的模型个性化方法,允许在临床兼容的时间范围内模拟梗死后室性心动过速。
    方法:在10例患者中(年龄65±11岁,9名男性)适用于室性心动过速后消融术,计算机断层扫描得出的壁厚图与25个电解剖图(窦性心律,节奏,和VT)。分析了壁厚与电生理特性(激活-恢复间隔)之间的关系。然后使用壁厚参数化快速且易于处理的器官尺度波传播模型。从多个地点模拟起搏方案,以在计算机上测试VT诱导。将计算机上的VT与临床上绘制的VT回路进行了比较。
    结果:临床,在9例患者中可以用详细的地图诱导6种不同的VT。所提出的模型允许快速模拟(中位数:6min/起搏部位)。来自100个不同部位/患者的稳定起搏(600毫秒)的模拟从未触发任何心律失常。应用S1-S2或S1-S2-S3诱导方案允许在临床上可诱导的10名患者中的9名进行计算机模拟VT的诱导。临床上不可诱导的患者也是计算机上不可诱导的。总共模拟了42种不同的VT(每位患者4.2±2)。六个计算机上的VT与临床上绘制的VT电路相匹配。
    结论:所提出的框架允许在几个小时内进行个性化模拟。9名患者中有6名,模拟显示了与心内记录匹配的折返模式。
    Electrophysiological mapping of ventricular tachycardia (VT) is tedious and poorly reproducible. Substrate analysis on imaging cannot explicitly display VT circuits.
    This study sought to introduce a computed tomography-based model personalization approach, allowing for the simulation of postinfarction VT in a clinically compatible time frame.
    In 10 patients (age 65 ± 11 years, 9 male) referred for post-VT ablation, computed tomography-derived wall thickness maps were registered to 25 electroanatomical maps (sinus rhythm, paced, and VT). The relationship between wall thickness and electrophysiological characteristics (activation-recovery interval) was analyzed. Wall thickness was then employed to parameterize a fast and tractable organ-scale wave propagation model. Pacing protocols were simulated from multiple sites to test VT induction in silico. In silico VTs were compared to VT circuits mapped clinically.
    Clinically, 6 different VTs could be induced with detailed maps in 9 patients. The proposed model allowed for fast simulation (median: 6 min/pacing site). Simulations of steady pacing (600 milliseconds) from 100 different sites/patient never triggered any arrhythmia. Applying S1-S2 or S1-S2-S3 induction schemes allowed for the induction of in silico VTs in the 9 of 10 patients who were clinically inducible. The patient who was not inducible clinically was also noninducible in silico. A total of 42 different VTs were simulated (4.2 ± 2 per patient). Six in silico VTs matched a VT circuit mapped clinically.
    The proposed framework allows for personalized simulations in a matter of hours. In 6 of 9 patients, simulations show re-entrant patterns matching intracardiac recordings.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心室复极显示显著的性别特异性,与所研究的物种无关,女性在心电图中与较长的QT间隔有关。从临床的角度来看,女性患药物诱发的尖端扭转和症状性长QT综合征的风险更大.这里,我们提出了一种光学作图(OM)方法来揭示小鼠心脏切片制备中的性别特异性动作电位(AP)异质性。与雄性小鼠相比,雌性小鼠的左心室心外膜复极化时间更长,相互,更多可变AP持续时间(APD),产生不太突出的透壁APD梯度。通过将OM与数学建模相结合,我们建议IKto发挥重要作用,f和IKur在AP中扩大在女性中。其他跨膜电流,包括INAL,仅对基础APD有轻微影响。和许多心脏病理生理学一样,增加[Ca2+]i会带来心律失常的风险,以性别选择性方式评估AP形态对L型钙通道(LTCC)激活增强的反应.在药理学LTCC激活后,雌性小鼠与雄性小鼠的APD及其变异均显着增加,我们假设这是由于基于数学建模的性别特异性INaL表达。总之,我们证明了左心室心外膜的复极化更加延迟,水平的LV透壁APD梯度,女性与男性相比,心外膜APD对Ca2流入的反应更为明显。数学建模量化了在正常和病理生理条件下选定的离子电流对性别特异性AP形态的相对贡献。
    Ventricular repolarization shows notable sex-specificity, with female sex being associated with longer QT-intervals in electrocardiography irrespective of the species studied. From a clinical point of view, women are at a greater risk for drug-induced torsade de pointes and symptomatic long-QT syndrome. Here, we present an optical mapping (OM) approach to reveal sex-specific action potential (AP) heterogeneity in a slice preparation of mouse hearts. Left ventricular epicardial repolarization in female versus male mice shows longer and, interindividually, more variable AP duration (APD), yielding a less prominent transmural APD gradient. By combining OM with mathematical modeling, we suggest a significant role of IKto,f and IKur in AP broadening in females. Other transmembrane currents, including INaL , only marginally affect basal APD. As in many cardiac pathophysiologies, increasing [Ca2+ ]i poses a risk for arrhythmia, the response of AP morphology to enhanced activation of L-type calcium channels (LTCC) was assessed in a sex-selective manner. Both APD and its variation increased significantly more in female versus male mice after pharmacological LTCC activation, which we hypothesize to be due to sex-specific INaL expression based on mathematical modeling. Altogether, we demonstrate a more delayed repolarization of LV epicardium, a leveled LV transmural APD gradient, and a more pronounced epicardial APD response to Ca2+ influx in females versus males. Mathematical modeling quantifies the relative contributions of selected ionic currents to sex-specific AP morphology under normal and pathophysiological conditions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号