hiPSC-CMs

hiPSC - CM
  • 文章类型: Journal Article
    电气和机械耦合的同步确保了心脏的生理泵功能,但是危及生命的病理可能会危及这种平衡。最近,人类诱导的多能干细胞衍生的心肌细胞(hiPSC-CM)已成为个性化研究的模型,因为它们可以概括人类的疾病特征,如受损的电容量或机械电路中断。这项研究利用了hiPSC-CM的模型,并展示了创新技术来研究电气和机械性能以及由于遗传性心肌病引起的调制。在这项工作中,HiPSC-CM携带Brugada综合征(BRU)或扩张型心肌病(DCM),以双层配置进行组织,以首先验证实验方法,其次模拟生理环境。已采用基于高密度CMOS的微电极阵列(HD-MEA)来研究电活性。此外,通过基于视频的定量评估来研究机械功能,在用β-肾上腺素能激动剂刺激时。本研讨引见了两种实验办法。首先,使用最近开发的光学跟踪器(OPT)和旨在量化心脏运动学的共聚焦无参考牵引力显微镜(cTFM)获得hiPSC-CM层中的高通量机械测量(xy检查)。第二,原子力显微镜(AFM)与流体FM探针,结合xy检查方法,补充了对细胞-细胞机械耦合(xyz-inspection)的三维理解。这种特定的组合代表了一种检测细胞层之间的电气和机械延迟的多技术方法。检查遗传性心肌病后的差异和可能的含义。它不仅可以在所提出的体外模型中检测疾病特征,还可以定量评估其对药物的反应,从而证明了其作为临床和药理学研究的可扩展工具的可行性。
    The synchronization of the electrical and mechanical coupling assures the physiological pump function of the heart, but life-threatening pathologies may jeopardize this equilibrium. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a model for personalized investigation because they can recapitulate human diseased traits, such as compromised electrical capacity or mechanical circuit disruption. This research avails the model of hiPSC-CMs and showcases innovative techniques to study the electrical and mechanical properties as well as their modulation due to inherited cardiomyopathies. In this work, hiPSC-CMs carrying either Brugada syndrome (BRU) or dilated cardiomyopathy (DCM), were organized in a bilayer configuration to first validate the experimental methods and second mimic the physiological environment. High-density CMOS-based microelectrode arrays (HD-MEA) have been employed to study the electrical activity. Furthermore, mechanical function was investigated via quantitative video-based evaluation, upon stimulation with a β-adrenergic agonist. This study introduces two experimental methods. First, high-throughput mechanical measurements in the hiPSC-CM layers (xy-inspection) are obtained using both a recently developed optical tracker (OPT) and confocal reference-free traction force microscopy (cTFM) aimed to quantify cardiac kinematics. Second, atomic force microscopy (AFM) with FluidFM probes, combined with the xy-inspection methods, supplemented a three-dimensional understanding of cell-cell mechanical coupling (xyz-inspection). This particular combination represents a multi-technique approach to detecting electrical and mechanical latency among the cell layers, examining differences and possible implications following inherited cardiomyopathies. It can not only detect disease characteristics in the proposed in vitro model but also quantitatively assess its response to drugs, thereby demonstrating its feasibility as a scalable tool for clinical and pharmacological studies.
    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
    这项工作介绍了第一个心房特异性计算机人类诱导多能干细胞衍生的心肌细胞(hiPSC-CMs)模型,基于一组特定表型的IKur,IKCa和IK1膜电流。该模型建立在一些合著者最近发表的新颖的体外实验数据上,以模拟成熟的心房样hiPSC-CM的起搏动作电位。该模型由一个刚性常微分方程组组成,取决于几个参数,已通过自动优化技术进行调整,以紧密匹配选定的实验生物标志物。新模型有效地模拟了电子体外hiPSC-CM的成熟过程,从不稳定的去极化膜舒张电位过渡到稳定的超极化静息电位,并在无节奏的条件下表现出自发的射击活动。此外,我们的模型准确地反映了不同周期长度下的实验速率依赖性数据,并显示了对特定电流阻滞剂的预期反应.这种心房特异性计算机模型为心脏干细胞的电生理研究及其在药物评估和心房颤动治疗中的应用提供了一种新颖的计算工具。
    This work introduces the first atrial-specific in-silico human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) model, based on a set of phenotype-specific IKur,IKCa and IK1 membrane currents. This model is built on novel in-vitro experimental data recently published by some of the co-authors to simulate the paced action potential of matured atrial-like hiPSC-CMs. The model consists of a system of stiff ordinary differential equations depending on several parameters, which have been tuned by automatic optimization techniques to closely match selected experimental biomarkers. The new model effectively simulates the electronic in-vitro hiPSC-CMs maturation process, transitioning from an unstable depolarized membrane diastolic potential to a stable hyperpolarized resting potential, and exhibits spontaneous firing activity in unpaced conditions. Moreover, our model accurately reflects the experimental rate dependence data at different cycle length and demonstrates the expected response to a specific current blocker. This atrial-specific in-silico model provides a novel computational tool for electrophysiological studies of cardiac stem cells and their applications to drug evaluation and atrial fibrillation treatment.
    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
    源自人类诱导的多能干细胞(hiPSC-CM)的心肌细胞提供了一个有吸引力的平台来评估心血管相关事件的机制以及开发和测试用于心脏病的新药。这项工作的重点是比较两种hiPSC-CM分化方案:基于Wnt/β-catenin途径的时间调制的GiWi方法和市售的PSC心肌细胞分化试剂盒。我们强调需要优化几个参数,如细胞密度或小分子浓度(CHIR-99021,IWR-1)以获得功能性hiPSC-CM。两种方案都产生相似的分化效率;因此,特定程序的选择可能取决于实验者的偏好。
    Cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) offer an attractive platform to evaluate the mechanisms of cardiovascular-related incidents and to develop and test new drugs for heart diseases. This work focuses on the comparison of two hiPSC-CM differentiation protocols: the GiWi method based on temporal modulation of the Wnt/β-catenin pathway and the commercially available PSC Cardiomyocyte Differentiation Kit. We underlined the need to optimize several parameters such as cell density or small molecule concentration (CHIR-99021, IWR-1) to obtain functional hiPSC-CMs. Both protocols yield a similar differentiation efficiency; therefore, the choice of a particular procedure may depend on the preferences of the experimenter.
    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
    利用hiPSC与CRISPR/Cas9基因编辑组合创建疾病模型能够实现对差异药理学反应的机械见解。这允许将功效和安全性发现从健康状态转化为疾病状态,并且提供了在药物发现期间更快预测临床结果的手段。钙处理障碍,包括2型ryanodine受体(RYR2)的表达水平降低,与心脏功能障碍有关;在这里,我们创建了RYR2缺乏的人心肌细胞模型,该模型模拟了心力衰竭的某些方面。缺乏RYR2的心肌细胞对L型通道钙抑制剂表现出不同的药理学反应。表型和蛋白质组表征揭示了新的分子见解,包括CSRP3,SLMAP,和代谢变化,包括戊糖磷酸途径的上调和对氧化还原改变的敏感性增加。这种基因工程化的RYR2缺乏的体外心血管模型支持在钙处理和代谢功能障碍的背景下的药理学反应研究,从而使药物反应从健康状态转化为扰动的细胞状态。
    Creation of disease models utilizing hiPSCs in combination with CRISPR/Cas9 gene editing enable mechanistic insights into differential pharmacological responses. This allows translation of efficacy and safety findings from a healthy to a diseased state and provides a means to predict clinical outcome sooner during drug discovery. Calcium handling disturbances including reduced expression levels of the type 2 ryanodine receptor (RYR2) are linked to cardiac dysfunction; here we have created a RYR2 deficient human cardiomyocyte model that mimics some aspects of heart failure. RYR2 deficient cardiomyocytes show differential pharmacological responses to L-type channel calcium inhibitors. Phenotypic and proteomic characterization reveal novel molecular insights with altered expression of structural proteins including CSRP3, SLMAP, and metabolic changes including upregulation of the pentose phosphate pathway and increased sensitivity to redox alterations. This genetically engineered in vitro cardiovascular model of RYR2 deficiency supports the study of pharmacological responses in the context of calcium handling and metabolic dysfunction enabling translation of drug responses from healthy to perturbed cellular states.
    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
    钠电流(INa)减少发生在许多获得性和遗传性疾病的背景下,并且与心脏传导减慢和心律失常风险增加有关。钠通道阻断剂美西律已显示可恢复突变钠通道向膜的运输。然而,这些研究大多在异源表达系统中使用高浓度的美西律进行。此外,在未患病的心肌细胞环境中对INa的慢性影响仍然未知.在本文中,我们研究了治疗剂量的美西律对INa的慢性和急性影响,以及健康个体诱导多能干细胞衍生心肌细胞(hiPSC-CMs)的动作电位(AP)特征.将对照hiPSC-CM与10μM美西律或媒介物一起孵育48小时。美西律被洗掉后,进行膜片钳分析和免疫细胞化学实验.将hiPSC-CM与美西律一起孵育48小时(然后洗出)可诱导〜75%的INa峰值显着增加,在(in)激活的电压依赖性上没有任何明显的变化。这伴随着AP上冲程速度的显着增加,不改变其他AP参数。免疫细胞化学实验显示,与美西律孵育48小时后,膜Nav1.5荧光显着增加。hiPSC-CM急性再暴露于10µM美西律导致AP持续时间小幅但显著增加,在没有AP上升速度变化的情况下,峰值INa密度,或(in)激活的INa电压依赖性。重要的是,急性再次给药不会抵消美西律长期孵育引起的INa峰值密度的增加和由此产生的AP上冲程速度的增加.总之,长期服用临床相关浓度的美西律增加了非患病hiPSC-CM的INa密度,可能是通过增强钠通道的膜运输。我们的发现将美西律确定为增强和/或恢复INa和心脏传导的潜在治疗策略。
    A sodium current (INa) reduction occurs in the setting of many acquired and inherited conditions and is associated with cardiac conduction slowing and increased arrhythmia risks. The sodium channel blocker mexiletine has been shown to restore the trafficking of mutant sodium channels to the membrane. However, these studies were mostly performed in heterologous expression systems using high mexiletine concentrations. Moreover, the chronic effects on INa in a non-diseased cardiomyocyte environment remain unknown. In this paper, we investigated the chronic and acute effects of a therapeutic dose of mexiletine on INa and the action potential (AP) characteristics in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) of a healthy individual. Control hiPSC-CMs were incubated for 48 h with 10 µM mexiletine or vehicle. Following the wash-out of mexiletine, patch clamp analysis and immunocytochemistry experiments were performed. The incubation of hiPSC-CMs for 48 h with mexiletine (followed by wash-out) induced a significant increase in peak INa of ~75%, without any significant change in the voltage dependence of (in)activation. This was accompanied by a significant increase in AP upstroke velocity, without changes in other AP parameters. The immunocytochemistry experiments showed a significant increase in membrane Nav1.5 fluorescence following a 48 h incubation with mexiletine. The acute re-exposure of hiPSC-CMs to 10 µM mexiletine resulted in a small but significant increase in AP duration, without changes in AP upstroke velocity, peak INa density, or the INa voltage dependence of (in)activation. Importantly, the increase in the peak INa density and resulting AP upstroke velocity induced by chronic mexiletine incubation was not counteracted by the acute re-administration of the drug. In conclusion, the chronic administration of a clinically relevant concentration of mexiletine increases INa density in non-diseased hiPSC-CMs, likely by enhancing the membrane trafficking of sodium channels. Our findings identify mexiletine as a potential therapeutic strategy to enhance and/or restore INa and cardiac conduction.
    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
    人类诱导的多能干细胞衍生的心肌细胞(hiPSC-CM)经常用于临床前心脏毒性测试,并且仍然是根据综合体外致心律失常试验(CiPA)确认基于模型的药物作用预测的重要工具。尽管hiPSC-CM提供了相当大的好处,围绕实验可重复性的担忧已经出现。我们研究了时间变化和实验参数对hiPSC-CM电生理的影响。培养iCell心肌细胞2,并使用微电极阵列(MEA)系统获得生物信号(1-14天)。连续记录显示,在20分钟的平衡期间,跳动率增加了22.6%,场电位持续时间(FPD)减少了7.7%。还观察到整个多孔板的位置特定差异,外排的iCell心肌细胞2比内排的每分钟(BPM)快8.8次。心脏终点也受细胞培养时间的影响;从2-14天开始,搏动率降低(-12.7BPM),FPD加长(+257ms),和尖峰幅度增加(+3.3mV)。细胞培养时间(4-10天)也影响心肌细胞药物反应性(E-4031,硝苯地平,异丙肾上腺素)。qRT-PCR结果表明,心脏指标的每日变化可能与培养物中hiPSC-CM的持续成熟(2-30天)有关。还使用第二细胞系(Cor.4U)重复每日实验。总的来说,我们的研究强调了在进行hiPSC-CMMEA研究时需要考虑和解决的多种变异性来源.为了提高可重复性和数据解释,基于MEA的研究应建立标准化的方案并报告关键的实验条件(例如,细胞系,文化时间,平衡时间,电刺激设置,原始数据值)。
    Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are frequently used for preclinical cardiotoxicity testing and remain an important tool for confirming model-based predictions of drug effects in accordance with the comprehensive in vitro proarrhythmia assay (CiPA). Despite the considerable benefits hiPSC-CMs provide, concerns surrounding experimental reproducibility have emerged. We investigated the effects of temporal changes and experimental parameters on hiPSC-CM electrophysiology. iCell cardiomyocytes2 were cultured and biosignals were acquired using a microelectrode array (MEA) system (2-14 days). Continuous recordings revealed a 22.6% increase in the beating rate and 7.7% decrease in the field potential duration (FPD) during a 20-min equilibration period. Location-specific differences across a multiwell plate were also observed, with iCell cardiomyocytes2 in the outer rows beating 8.8 beats/min faster than the inner rows. Cardiac endpoints were also impacted by cell culture duration; from 2 to 14 days, the beating rate decreased (-12.7 beats/min), FPD lengthened (+257 ms), and spike amplitude increased (+3.3 mV). Cell culture duration (4-10 days) also impacted cardiomyocyte drug responsiveness (E-4031, nifedipine, isoproterenol). qRT-PCR results suggest that daily variations in cardiac metrics may be linked to the continued maturation of hiPSC-CMs in culture (2-30 days). Daily experiments were also repeated using a second cell line (Cor.4U). Collectively, our study highlights multiple sources of variability to consider and address when performing hiPSC-CM MEA studies. To improve reproducibility and data interpretation, MEA-based studies should establish a standardized protocol and report key experimental conditions (e.g., cell line, culture time, equilibration time, electrical stimulation settings, and raw data values).NEW & NOTEWORTHY We demonstrate that iCell cardiomyocytes2 electrophysiology measurements are impacted by deviations in experimental techniques including electrical stimulation protocols, equilibration time, well-to-well variability, and length of hiPSC-CM culture. Furthermore, our results indicate that hiPSC-CM drug responsiveness changes within the first 2 wk following defrost.
    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:2019年冠状病毒病(COVID-19)引起的急性心脏损伤会增加死亡率。由COVID-19引起的急性心脏损伤需要了解严重的急性呼吸综合征冠状病毒2(SARS-CoV-2)如何直接感染心肌细胞。这项研究通过在转录组水平上使用SARS-CoV-2感染人类诱导多能干细胞来源的心肌细胞(hiPSC-CM)的模型,为相关研究提供了坚实的基础,强调本研究与相关研究的相关性。先前已通过生物信息学研究了hiPSC-CM中的SARS-CoV-2感染,而没有提出完整的分子生物学过程。我们为hiPSC-CM中SARS-CoV-2感染的完整分子生物学过程提供了独特的生物信息学观点。
    方法:为了验证RNA-seq数据集,我们使用GSE184715和GSE150392进行分析研究,GSE193722用于细胞水平的验证,和GSE169241用于心脏组织样本的验证。使用GeneCards和MsigDB数据库来寻找与表型相关的基因。除了差异表达分析和主成分分析(PCA),我们还进行了蛋白质-蛋白质相互作用(PPI)分析,功能富集分析,集线器基因分析,上游转录因子预测,和药物预测。
    结果:差异表达基因(DEGs)分为四类:心肌细胞细胞骨架蛋白抑制,原癌基因激活和炎症,线粒体功能障碍,胞浆生理功能。每个枢纽基因都显示出良好的诊断预测,这在其他数据集中得到了很好的验证。抑制的生物学功能包括心肌细胞细胞骨架蛋白,三磷酸腺苷(ATP)合成和电子传递链(ETC),葡萄糖代谢,氨基酸代谢,脂肪酸代谢,丙酮酸代谢,柠檬酸循环,核酸代谢,复制,转录,翻译,泛素化,自噬,和细胞运输。原癌基因,炎症,核因子-κB(NF-κB)途径,干扰素信号被激活,以及炎症因子。病毒感染激活多种途径,包括干扰素途径,原癌基因和线粒体氧化应激,同时抑制心肌细胞骨架蛋白和能量代谢。感染限制了细胞内的合成和代谢,以及线粒体能量合成的原料。线粒体功能障碍和能量异常最终是由原癌基因激活和SARS-CoV-2感染引起的。干扰素途径的激活,原癌基因上调,和线粒体氧化应激引起炎症反应并导致心肌细胞收缩减弱。复制,转录,翻译,泛素化,自噬,细胞运输是生理上下降的功能之一。
    结论:SARS-CoV-2在hiPSC-CM中的感染基本上是通过线粒体功能障碍介导的。针对线粒体功能障碍的治疗性干预措施可以缓解与SARS-CoV-2感染相关的心血管并发症。
    BACKGROUND: Acute cardiac injury caused by coronavirus disease 2019 (COVID-19) increases mortality. Acute cardiac injury caused by COVID-19 requires understanding how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly infects cardiomyocytes. This study provides a solid foundation for related studies by using a model of SARS-CoV-2 infection in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) at the transcriptome level, highlighting the relevance of this study to related studies. SARS-CoV-2 infection in hiPSC-CMs has previously been studied by bioinformatics without presenting the full molecular biological process. We present a unique bioinformatics view of the complete molecular biological process of SARS-CoV-2 infection in hiPSC-CMs.
    METHODS: To validate the RNA-seq datasets, we used GSE184715 and GSE150392 for the analytical studies, GSE193722 for validation at the cellular level, and GSE169241 for validation in heart tissue samples. GeneCards and MsigDB databases were used to find genes associated with the phenotype. In addition to differential expression analysis and principal component analysis (PCA), we also performed protein-protein interaction (PPI) analysis, functional enrichment analysis, hub gene analysis, upstream transcription factor prediction, and drug prediction.
    RESULTS: Differentially expressed genes (DEGs) were classified into four categories: cardiomyocyte cytoskeletal protein inhibition, proto-oncogene activation and inflammation, mitochondrial dysfunction, and intracellular cytoplasmic physiological function. Each of the hub genes showed good diagnostic prediction, which was well validated in other datasets. Inhibited biological functions included cardiomyocyte cytoskeletal proteins, adenosine triphosphate (ATP) synthesis and electron transport chain (ETC), glucose metabolism, amino acid metabolism, fatty acid metabolism, pyruvate metabolism, citric acid cycle, nucleic acid metabolism, replication, transcription, translation, ubiquitination, autophagy, and cellular transport. Proto-oncogenes, inflammation, nuclear factor-kappaB (NF-κB) pathways, and interferon signaling were activated, as well as inflammatory factors. Viral infection activates multiple pathways, including the interferon pathway, proto-oncogenes and mitochondrial oxidative stress, while inhibiting cardiomyocyte backbone proteins and energy metabolism. Infection limits intracellular synthesis and metabolism, as well as the raw materials for mitochondrial energy synthesis. Mitochondrial dysfunction and energy abnormalities are ultimately caused by proto-oncogene activation and SARS-CoV-2 infection. Activation of the interferon pathway, proto-oncogene up-regulation, and mitochondrial oxidative stress cause the inflammatory response and lead to diminished cardiomyocyte contraction. Replication, transcription, translation, ubiquitination, autophagy, and cellular transport are among the functions that decline physiologically.
    CONCLUSIONS: SARS-CoV-2 infection in hiPSC-CMs is fundamentally mediated via mitochondrial dysfunction. Therapeutic interventions targeting mitochondrial dysfunction may alleviate the cardiovascular complications associated with SARS-CoV-2 infection.
    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
    心肌细胞成熟是心脏发育的最后阶段,心肌细胞成熟异常会导致严重的心脏病。CXXC锌指蛋白1(Cfp1),多谱系细胞发育的关键表观遗传因子,其对心肌细胞成熟的影响仍未得到充分研究。本研究探讨了Cfp1在这方面的作用和机制。心肌细胞特异性Cfp1敲除(Cfp1-cKO)小鼠在出生后4周内死亡。来自Cfp1-cKO小鼠的心肌细胞表现出抑制的成熟表型,以结构为特征,新陈代谢,收缩,和细胞周期异常。相比之下,心肌细胞特异性Cfp1转基因(Cfp1-TG)小鼠和过表达Cfp1的人诱导多能干细胞衍生的心肌细胞(hiPSC-CM)表现出更成熟的表型。机械上,Cfp1的缺乏导致组蛋白H3(H3K4me3)修饰的赖氨酸4的三甲基化减少,伴随着异位H3K4me3的形成。此外,Cfp1缺失降低了成年基因中H3K4me3修饰的水平,并增加了胎儿基因中H3K4me3修饰的水平。总的来说,Cfp1通过调节组蛋白H3K4me3修饰来调节对心肌细胞成熟至关重要的基因的表达,从而错综复杂地影响成熟过程。这项研究暗示Cfp1是调节心肌细胞成熟的重要分子,它的功能障碍与心脏病密切相关。
    Cardiomyocyte maturation is the final stage of heart development, and abnormal cardiomyocyte maturation will lead to serious heart diseases. CXXC zinc finger protein 1 (Cfp1), a key epigenetic factor in multi-lineage cell development, remains underexplored in its influence on cardiomyocyte maturation. This study investigates the role and mechanisms of Cfp1 in this context. Cardiomyocyte-specific Cfp1 knockout (Cfp1-cKO) mice died within 4 weeks of birth. Cardiomyocytes derived from Cfp1-cKO mice showed an inhibited maturation phenotype, characterized by structural, metabolic, contractile, and cell cycle abnormalities. In contrast, cardiomyocyte-specific Cfp1 transgenic (Cfp1-TG) mice and human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) overexpressing Cfp1 displayed a more mature phenotype. Mechanistically, deficiency of Cfp1 led to a reduction in trimethylation on lysine 4 of histone H3 (H3K4me3) modification, accompanied by the formation of ectopic H3K4me3. Furthermore, Cfp1 deletion decreased the level of H3K4me3 modification in adult genes and increased the level of H3K4me3 modification in fetal genes. Collectively, Cfp1 modulates the expression of genes crucial to cardiomyocyte maturation by regulating histone H3K4me3 modification, thereby intricately influencing the maturation process. This study implicates Cfp1 as an important molecule regulating cardiomyocyte maturation, with its dysfunction strongly linked to cardiac disease.
    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
    我们的研究是为了调查钙粘蛋白-5(CDH5)血管内皮细胞粘附糖蛋白,可以促进人诱导多能干细胞(hiPSCs)分化为窦房结样起搏细胞(SANLPCs),根据先前的发现,丝素蛋白水凝胶通过激活CDH5诱导大鼠静止心肌细胞直接转化为起搏器细胞。在这项研究中,在心肌细胞分化期间,在第5天至第7天用CDH5(40ng/mL)处理分化的hiPSC。本研究的结果表明,CDH5刺激起搏器特异性标志物的表达,同时抑制与工作心肌细胞相关的标志物,导致SANLPC在hiPSC衍生的心肌细胞(hiPSC-CM)群体中的比例增加。此外,CDH5诱导的典型电生理特征类似于hiPSC-CM中的心脏起搏器细胞。进一步的机理研究表明,CDH5诱导的hiPSC向SANLPCs的富集分化被β-catenin抑制剂iCRT14部分逆转。因此,根据上述发现,由此可以推断,CDH5对β-catenin的调控在促进hiPSCs向SANLPCs的富集分化中起着至关重要的作用,为即将进行的研究中生物起搏器的构建提供了一条新的途径。
    Our study was conducted to investigate whether cadherin-5 (CDH5), a vascular endothelial cell adhesion glycoprotein, could facilitate the differentiation of human induced pluripotent stem cells (hiPSCs) into sinoatrial node-like pacemaker cells (SANLPCs), following previous findings of silk-fibroin hydrogel-induced direct conversion of quiescent cardiomyocytes into pacemaker cells in rats through the activation of CDH5. In this study, the differentiating hiPSCs were treated with CDH5 (40 ng/mL) between Day 5 and 7 during cardiomyocytes differentiation. The findings in the present study demonstrated that CDH5 stimulated the expression of pacemaker-specific markers while suppressing markers associated with working cardiomyocytes, resulting in an increased proportion of SANLPCs among hiPSCs-derived cardiomyocytes (hiPSC-CMs) population. Moreover, CDH5 induced typical electrophysiological characteristics resembling cardiac pacemaker cells in hiPSC-CMs. Further mechanistic investigations revealed that the enriched differentiation of hiPSCs into SANLPCs induced by CDH5 was partially reversed by iCRT14, an inhibitor of β-catenin. Therefore, based on the aforementioned findings, it could be inferred that the regulation of β-catenin by CDH5 played a crucial role in promoting the enriched differentiation of hiPSCs into SANLPCs, which presents a novel avenue for the construction of biological pacemakers in forthcoming research.
    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
    外显子跳跃疗法是Duchenne肌营养不良症(DMD)的一种有前途的治疗策略,这是由编码肌营养不良蛋白的DMD基因的功能丧失突变引起的,导致进行性心肌病.外显子3-9的框内缺失(Δ3-9),表现出非常轻微的临床表型,是通过靶向肌动蛋白结合域1(ABD1)进行外显子跳跃的潜在靶向阅读框;然而,这种方法对DMD心肌病的疗效尚不确定.在这项研究中,我们比较了三种表达Δ3-9,移码Δ3-7或完整DMD的等基因人类诱导多能干细胞衍生的心肌细胞(hiPSC-CM)。RNA测序显示,Δ3-9与野生型样品之间的机械转导相关基因的表达模式相似。此外,我们观察到Δ3-9和野生型hiPSC-CM之间的电生理特性相似;Δ3-7hiPSC-CM显示出电生理改变,CaMKII激活加速。始终如一,Δ3-9hiPSC-CM表达大量内部截短的肌营养不良蛋白,导致维持F-肌动蛋白结合和结蛋白保留。靶向外显子8的反义寡核苷酸有效诱导跳跃外显子8-9以恢复Δ3-7hiPSC-CM中的功能性肌养蛋白和电生理参数,使细胞特性更接近Δ3-9hiPSC-CM。总的来说,针对ABD1的外显子跳跃将阅读框转换为Δ3-9可能成为DMD心肌病的有希望的治疗方法。
    Exon-skipping therapy is a promising treatment strategy for Duchenne muscular dystrophy (DMD), which is caused by loss-of-function mutations in the DMD gene encoding dystrophin, leading to progressive cardiomyopathy. In-frame deletion of exons 3-9 (Δ3-9), manifesting a very mild clinical phenotype, is a potential targeted reading frame for exon-skipping by targeting actin-binding domain 1 (ABD1); however, the efficacy of this approach for DMD cardiomyopathy remains uncertain. In this study, we compared three isogenic human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) expressing Δ3-9, frameshifting Δ3-7, or intact DMD. RNA sequencing revealed a resemblance in the expression patterns of mechano-transduction-related genes between Δ3-9 and wild-type samples. Furthermore, we observed similar electrophysiological properties between Δ3-9 and wild-type hiPSC-CMs; Δ3-7 hiPSC-CMs showed electrophysiological alterations with accelerated CaMKII activation. Consistently, Δ3-9 hiPSC-CMs expressed substantial internally truncated dystrophin protein, resulting in maintaining F-actin binding and desmin retention. Antisense oligonucleotides targeting exon 8 efficiently induced skipping exons 8-9 to restore functional dystrophin and electrophysiological parameters in Δ3-7 hiPSC-CMs, bringing the cell characteristics closer to those of Δ3-9 hiPSC-CMs. Collectively, exon-skipping targeting ABD1 to convert the reading frame to Δ3-9 may become a promising therapy for DMD cardiomyopathy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号