Myocytes, Cardiac

肌细胞, 心脏
  • 文章类型: Journal Article
    阿霉素(DOX)是一种蒽环类抗癌剂,在治疗实体肿瘤中非常有效。鉴于多柔比星诱导的心脏毒性涉及多种机制,很难确定毒性的精确分子靶标。文献综述的结果表明,天然产物可能对阿霉素诱导的心脏毒性提供心脏保护作用。在体外和体内。然而,需要进一步的验证性研究来证实这一说法。最重要的是将更多的注意力转向复杂的信号网络,这些信号网络对于心肌细胞的存活和功能障碍至关重要。尽管在预防DOX引起的心脏毒性的天然产物的临床前研究中取得了令人鼓舞的进展,这些尚未翻译为临床使用。阻碍开发基于天然产物的心脏保护性佐剂的最重要障碍之一是在人体中缺乏足够的生物利用度。这篇综述概述了关于多柔比星DOX诱导的心脏毒性的最新知识,重点关注天然化合物和草药制剂在预防这种不良反应方面的潜在益处。作为文献搜索引擎,Scopus中的浏览器,PubMed,使用了WebofScience数据库和ClinicalTrials.gov寄存器。
    Doxorubicin (DOX) is an anthracycline anticancer agent that is highly effective in the treatment of solid tumors. Given the multiplicity of mechanisms involved in doxorubicin-induced cardiotoxicity, it is difficult to identify a precise molecular target for toxicity. The findings of a literature review suggest that natural products may offer cardioprotective benefits against doxorubicin-induced cardiotoxicity, both in vitro and in vivo. However, further confirmatory studies are required to substantiate this claim. It is of the utmost importance to direct greater attention towards the intricate signaling networks that are of paramount importance for the survival and dysfunction of cardiomyocytes. Notwithstanding encouraging progress made in preclinical studies of natural products for the prevention of DOX-induced cardiotoxicity, these have not yet been translated for clinical use. One of the most significant obstacles hindering the development of cardioprotective adjuvants based on natural products is the lack of adequate bioavailability in humans. This review presents an overview of current knowledge on doxorubicin DOX-induced cardiotoxicity, with a focus on the potential benefits of natural compounds and herbal preparations in preventing this adverse effect. As literature search engines, the browsers in the Scopus, PubMed, Web of Science databases and the ClinicalTrials.gov register were used.
    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
    原理:细胞外囊泡(EV)被认为在发育和疾病过程中介导细胞间的通讯。然而,细胞间EV转移的生物学见解仍然难以捉摸,也在心里,并且在技术上具有挑战性。这里,我们的目的是研究心肌细胞来源的EV在新生儿心脏中的生物学转移。方法:我们利用CD9作为电动汽车的标志物,并产生了两系心肌细胞特异性EV报告小鼠:Tnnt2-Cre;双联反向CD9/EGFP和αMHC-MerCreMer;双联反向CD9/EGFP。两个小鼠系用于确定发育中的心肌细胞是否在体外和体内将EV转移到其他心脏细胞(非肌细胞和心肌细胞),并研究心肌细胞衍生的EV的细胞间转运途径。结果:在报告小鼠品系和出生后心脏的概念证明中都证实了心肌细胞的遗传标记,EGFP+/MYH1-非肌细胞的一部分牢固地存在,表明体内心肌细胞来源的EV转移。然而,两组直接和间接EGFP+/-心肌细胞共培养结果显示,心肌细胞衍生的EGFP+EV转移需要细胞-细胞接触,且从培养基中摄取EGFP+EV是有限的.当与小鼠巨噬细胞共培养时观察到相同的情况。进一步的机械见解表明,心肌细胞EV转移通过I型隧道纳米管发生。结论:虽然当前的概念假设电动汽车通过分泌物转移到周围环境中,我们的数据显示,在发育中的心脏中,心肌细胞衍生的EV转移通过相邻细胞之间的纳米管发生.这些数据是否是基本的,是否与成人心脏和其他器官有关还有待确定,但它们暗示EV转移的正常发育过程是通过细胞-细胞接触而不是通过细胞外室。
    Rationale: Extracellular vesicles (EVs) are thought to mediate intercellular communication during development and disease. Yet, biological insight to intercellular EV transfer remains elusive, also in the heart, and is technically challenging to demonstrate. Here, we aimed to investigate biological transfer of cardiomyocyte-derived EVs in the neonatal heart. Methods: We exploited CD9 as a marker of EVs, and generated two lines of cardiomyocyte specific EV reporter mice: Tnnt2-Cre; double-floxed inverted CD9/EGFP and αMHC-MerCreMer; double-floxed inverted CD9/EGFP. The two mouse lines were utilized to determine whether developing cardiomyocytes transfer EVs to other cardiac cells (non-myocytes and cardiomyocytes) in vitro and in vivo and investigate the intercellular transport pathway of cardiomyocyte-derived EVs. Results: Genetic tagging of cardiomyocytes was confirmed in both reporter mouse lines and proof of concept in the postnatal heart showed that, a fraction of EGFP+/MYH1- non-myocytes exist firmly demonstrating in vivo cardiomyocyte-derived EV transfer. However, two sets of direct and indirect EGFP +/- cardiac cell co-cultures showed that cardiomyocyte-derived EGFP+ EV transfer requires cell-cell contact and that uptake of EGFP+ EVs from the medium is limited. The same was observed when co-cultiring with mouse macrophages. Further mechanistic insight showed that cardiomyocyte EV transfer occurs through type I tunneling nanotubes. Conclusion: While the current notion assumes that EVs are transferred through secretion to the surroundings, our data show that cardiomyocyte-derived EV transfer in the developing heart occurs through nanotubes between neighboring cells. Whether these data are fundamental and relate to adult hearts and other organs remains to be determined, but they imply that the normal developmental process of EV transfer goes through cell-cell contact rather than through the extracellular compartment.
    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
    原理:心肌梗塞(MI)是一种严重的全球性临床疾病,普遍存在。成年哺乳动物心脏对损伤的反应产生新的心肌细胞(CMs)的能力有限仍然是开发有效疗法的主要障碍。目前的方法集中在通过细胞周期再入诱导现有CM的增殖。然而,这种方法主要提高细胞周期蛋白依赖性激酶6(CDK6)和DNA含量,缺乏适当的胞质分裂,并导致功能失调的双核CMs的形成。胞质分裂依赖于核糖体生物发生(Ribo-bio),核仁素(Ncl)调节的一个关键过程。我们的目标是确定一种促进DNA合成和胞质分裂的新方法。方法:各种技术,包括RNA/蛋白质测序分析,Ribo-Halo,Ribo-disome,流式细胞术,和心脏特异性肿瘤抑制视网膜母细胞瘤-1(Rb1)基因敲除小鼠,用于评估增殖/细胞周期再入和Ribo-bio/胞质分裂的系列信号传导。超声心动图,共焦成像,和组织学用于评估心功能。结果:与对照小鼠相比,分析显示MI小鼠心脏中Rb1的水平显着升高,circASXL1的水平降低。Rb1的缺失仅诱导细胞周期重新进入,同时增强Ribo-生物调节剂Ncl导致胞质分裂。机械上,生物信息学和损失/增益研究发现circASXL1/CDK6/Rb1调节细胞周期重新进入。此外,Ribo-Halo,Ribo-disome和circRNA下拉测定表明circASXL1通过Ncl/Ribo-bio促进胞质分裂。重要的是,来自脐带间充质干细胞(UMSC-Exo)的外泌体能够通过促进细胞周期折返和Ribo-bio/胞质分裂的协调信号来增强心脏功能。通过在UMSC-Exo中沉默circASXL1来减弱这些作用。结论:circASXL1/CDK6/Rb1/细胞周期折返和circASXL1/Ncl/Ribo-bio/胞质分裂的系列信号在心脏修复中起着至关重要的作用。UMSC-Exo通过以circASXL1依赖性方式刺激CM细胞周期折返和胞质分裂来有效修复梗塞心肌。这项研究提供了针对MI的circASXL1信号网络的创新治疗策略,并提供了增强心脏修复的潜在途径。
    Rationale: Myocardial infarction (MI) is a severe global clinical condition with widespread prevalence. The adult mammalian heart\'s limited capacity to generate new cardiomyocytes (CMs) in response to injury remains a primary obstacle in developing effective therapies. Current approaches focus on inducing the proliferation of existing CMs through cell-cycle reentry. However, this method primarily elevates cyclin dependent kinase 6 (CDK6) and DNA content, lacking proper cytokinesis and resulting in the formation of dysfunctional binucleated CMs. Cytokinesis is dependent on ribosome biogenesis (Ribo-bio), a crucial process modulated by nucleolin (Ncl). Our objective was to identify a novel approach that promotes both DNA synthesis and cytokinesis. Methods: Various techniques, including RNA/protein-sequencing analysis, Ribo-Halo, Ribo-disome, flow cytometry, and cardiac-specific tumor-suppressor retinoblastoma-1 (Rb1) knockout mice, were employed to assess the series signaling of proliferation/cell-cycle reentry and Ribo-bio/cytokinesis. Echocardiography, confocal imaging, and histology were utilized to evaluate cardiac function. Results: Analysis revealed significantly elevated levels of Rb1, bur decreased levels of circASXL1 in the hearts of MI mice compared to control mice. Deletion of Rb1 induces solely cell-cycle reentry, while augmenting the Ribo-bio modulator Ncl leads to cytokinesis. Mechanically, bioinformatics and the loss/gain studies uncovered that circASXL1/CDK6/Rb1 regulates cell-cycle reentry. Moreover, Ribo-Halo, Ribo-disome and circRNA pull-down assays demonstrated that circASXL1 promotes cytokinesis through Ncl/Ribo-bio. Importantly, exosomes derived from umbilical cord mesenchymal stem cells (UMSC-Exo) had the ability to enhance cardiac function by facilitating the coordinated signaling of cell-cycle reentry and Ribo-bio/cytokinesis. These effects were attenuated by silencing circASXL1 in UMSC-Exo. Conclusion: The series signaling of circASXL1/CDK6/Rb1/cell-cycle reentry and circASXL1/Ncl/Ribo-bio/cytokinesis plays a crucial role in cardiac repair. UMSC-Exo effectively repairs infarcted myocardium by stimulating CM cell-cycle reentry and cytokinesis in a circASXL1-dependent manner. This study provides innovative therapeutic strategies targeting the circASXL1 signaling network for MI and offering potential avenues for enhanced cardiac repair.
    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
    L型CaV1.2电流(ICa,L)将电激发与心肌细胞收缩联系起来。ICa,严格调节L以控制心输出量。Rad是与Ras相关的,与L型钙通道β亚基(CaVβ)结合以促进ICa抑制的单体蛋白,L.除了Rad核心基序赋予的CaVβ相互作用外,高度保守的RadC端可以在体外指导膜缔合和抑制ICa,L在永生化细胞系中。在这项工作中,我们检验了这样一个假设,即在心肌细胞中,Rad的多碱性C端赋予了t-小管定位,Rad依赖性ICa需要膜靶向,L调节。我们将3xFlag表位引入内源性小鼠Rrad基因的N末端以促进亚细胞定位的分析。将全长3xFlag-Rad(Flag-Rad)小鼠与第二个转基因小鼠模型进行比较,其中3xFlag-Rad的延伸的多碱性C-末端在丙氨酸277处被截短(Flag-RadΔCT)。分离心室心肌细胞用于抗Flag-Rad免疫细胞化学和离体电生理学。全长Flag-Rad显示重复的t-管状模式,而Flag-RadΔCT未能显示膜缔合。ICa,Flag-RadΔCT心肌细胞中的L显示超极化激活中点,最大电导增加。此外,Flag-RadΔCT细胞的电流衰减更快。心肌ICa,RadC端缺失模型中的L表型ICa,响应于β-AR刺激而调制L。机械上,多元RadC末端通过膜缔合赋予CaV1.2调节。干扰Rad膜关联构成了增强心脏功能的特定目标,可作为射血分数降低的心力衰竭的治疗方法。
    L-type CaV1.2 current (ICa,L) links electrical excitation to contraction in cardiac myocytes. ICa,L is tightly regulated to control cardiac output. Rad is a Ras-related, monomeric protein that binds to L-type calcium channel β subunits (CaVβ) to promote inhibition of ICa,L. In addition to CaVβ interaction conferred by the Rad core motif, the highly conserved Rad C-terminus can direct membrane association in vitro and inhibition of ICa,L in immortalized cell lines. In this work, we test the hypothesis that in cardiomyocytes the polybasic C-terminus of Rad confers t-tubular localization, and that membrane targeting is required for Rad-dependent ICa,L regulation. We introduced a 3xFlag epitope to the N-terminus of the endogenous mouse Rrad gene to facilitate analysis of subcellular localization. Full-length 3xFlag-Rad (Flag-Rad) mice were compared with a second transgenic mouse model, in which the extended polybasic C-termini of 3xFlag-Rad was truncated at alanine 277 (Flag-RadΔCT). Ventricular cardiomyocytes were isolated for anti-Flag-Rad immunocytochemistry and ex vivo electrophysiology. Full-length Flag-Rad showed a repeating t-tubular pattern whereas Flag-RadΔCT failed to display membrane association. ICa,L in Flag-RadΔCT cardiomyocytes showed a hyperpolarized activation midpoint and an increase in maximal conductance. Additionally, current decay was faster in Flag-RadΔCT cells. Myocardial ICa,L in a Rad C-terminal deletion model phenocopies ICa,L modulated in response to β-AR stimulation. Mechanistically, the polybasic Rad C-terminus confers CaV1.2 regulation via membrane association. Interfering with Rad membrane association constitutes a specific target for boosting heart function as a treatment for heart failure with reduced ejection fraction.
    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:抗肿瘤药物,包括阿霉素,伊达比星,和表柔比星,已发现由于氧化应激-线粒体功能障碍-铁凋亡(ORMFs)而对心脏产生不利影响,这是对蒽环类抗生素诱导的心脏毒性的贡献。为了更好地理解这种现象,本研究分析了ORMFS基因的时间分辨测量值.
    方法:使用人类3D心脏微组织细胞模型研究了三种蒽环类药物对ORMFs基因的影响。以两种剂量(治疗性和毒性)在14天内收集转录组数据。WGCNA确定了关键模块相关基因,功能富集分析研究了ssGSEA量化的生物过程,如免疫细胞浸润和血管生成。从心力衰竭患者和对照受试者收集活检。收集GSE59672和GSE2965用于验证。分子对接用于鉴定蒽环类抗生素与关键基因的相互作用。
    结果:在体内或体外筛选ORMFs基因。使用WGCNA,六个共表达的基因模块被分组,MEblue成为最重要的模块。获得了8个与蓝色模块相交的关键基因与动态响应基因:CD36,CDH5,CHI3L1,HBA2,HSD11B1,OGN,RPL8和VWF。与对照样品相比,除RPL8外,所有关键基因在体外ANT处理设置中都下调,他们的表达水平随着时间的推移而变化。根据功能分析,关键模块相关基因参与血管生成和免疫系统通路.在所有ANT处理的设置中,ssGSEA显示血管生成评分和免疫细胞活性的显著下调,包括活化的CD4T细胞,未成熟B细胞,记忆B细胞,自然杀伤细胞,1型辅助T细胞,和2型T辅助细胞。分子对接显示RPL8和CHI3L1对蒽环类药物显示出显著的结合亲和力。
    结论:本研究集中于人心脏微组织和ANT治疗患者心脏活检组织中ORMFs基因的动态特征。已经强调,在蒽环类抗生素诱导的心脏毒性的情况下,ORMF基因可能有助于免疫浸润和血管生成。对这些基因的透彻了解可能会导致疾病的诊断和治疗。
    BACKGROUND: Antineoplastic medications, including doxorubicin, idarubicin, and epirubicin, have been found to adversely affect the heart due to oxidative stress - mitochondrial dysfunction - ferroptosis (ORMFs), which act as contributing attributes to anthracycline-induced cardiotoxicity. To better understand this phenomenon, the time-resolved measurements of ORMFS genes were analyzed in this study.
    METHODS: The effect of three anthracycline drugs on ORMFs genes was studied using a human 3D cardiac microtissue cell model. Transcriptome data was collected over 14 days at two doses (therapeutic and toxic). WGCNA identified key module-related genes, and functional enrichment analysis investigated the biological processes quantified by ssGSEA, such as immune cell infiltration and angiogenesis. Biopsies were collected from heart failure patients and control subjects. GSE59672 and GSE2965 were collected for validation. Molecular docking was used to identify anthracyclines\'s interaction with key genes.
    RESULTS: The ORMFs genes were screened in vivo or in vitro. Using WGCNA, six co-expressed gene modules were grouped, with MEblue emerging as the most significant module. Eight key genes intersecting the blue module with the dynamic response genes were obtained: CD36, CDH5, CHI3L1, HBA2, HSD11B1, OGN, RPL8, and VWF. Compared with control samples, all key genes except RPL8 were down-regulated in vitro ANT treatment settings, and their expression levels varied over time. According to functional analyses, the key module-related genes were engaged in angiogenesis and the immune system pathways. In all ANT-treated settings, ssGSEA demonstrated a significant down-regulation of angiogenesis score and immune cell activity, including Activated CD4 T cell, Immature B cell, Memory B cell, Natural killer cell, Type 1 T helper cell, and Type 2 T helper cell. Molecular docking revealed that RPL8 and CHI3L1 show significant binding affinity for anthracyclines.
    CONCLUSIONS: This study focuses on the dynamic characteristics of ORMFs genes in both human cardiac microtissues and cardiac biopsies from ANT-treated patients. It has been highlighted that ORMFs genes may contribute to immune infiltration and angiogenesis in cases of anthracycline-induced cardiotoxicity. A thorough understanding of these genes could potentially lead to improved diagnosis and treatment of the 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
    糖尿病性心肌病(DCM)是由糖尿病引起的慢性疾病,这被认为是一种世界性的具有挑战性的疾病。本研究旨在探讨敲除NACHT的作用和潜在机制。含LRR和PYD结构域的蛋白3(NLRP3),与各种疾病的发生和发展有关的炎症小体,高糖或糖尿病诱导的心肌细胞焦亡和铁凋亡,近年来发现的两种调节非坏死细胞死亡模式。在本研究中,同时进行体内和体外研究。采用55mg/kg腹腔注射链脲佐菌素(STZ)诱导糖尿病大鼠。腹膜内注射MCC950(10mg/kg)后,另一方面,用35mmol/L葡萄糖模拟H9C2心肌细胞的DCM模型,并将NLRP3的短发夹RNA载体转染到细胞中。结果表明,在体内研究,心肌纤维排列松散,表现为炎症细胞浸润,在DM组中,线粒体cr破裂,GSDMD-NT表达显着增加,而xCT和GPX4的蛋白表达显著降低,两者都被MCC950逆转。高糖在体外降低细胞活力和ATP水平,伴随着LDH释放的增加。与单独处理的HG相比,在NLRP3敲低后所有上述指标均逆转。此外,成焦率和铁死亡相关因子的蛋白表达显著降低或升高,与免疫荧光显示的结果一致。此外,在mtROS激动剂鱼藤酮(ROT)治疗后,NLRP3敲低对HG的保护作用被逆转。总之,抑制NLRP3抑制DM诱导的心肌损伤。线粒体ROS的促进消除了敲低NLRP3的保护作用,并诱导了焦凋亡和铁凋亡的发生。这些发现可能为临床糖尿病引起的心肌损伤治疗提供了新的治疗基础机制。
    Diabetic cardiomyopathy (DCM) is a chronic disease caused by diabetes mellitus, which is recognized as a worldwide challenging disease. This study aimed to investigate the role and the potential mechanism of knocking down the NACHT-, LRR- and PYD domains-containing protein 3 (NLRP3), an inflammasome associated with onset and progression of various diseases, on high glucose or diabetes -induced cardiac cells pyroptosis and ferroptosis, two regulated non-necrosis cell death modalities discovered recent years. In the present study, both in vivo and in vitro studies were conducted simultaneously. Diabetic rats were induced by 55 mg/kg intraperitoneal injection of streptozotocin (STZ). Following the intraperitoneal injection of MCC950 (10 mg/kg), On the other hand, the DCM model in H9C2 cardiac cells was simulated with 35 mmol/L glucose and a short hairpin RNA vector of NLRP3 were transfected to cells. The results showed that in vivo study, myocardial fibers were loosely arranged and showed inflammatory cell infiltration, mitochondrial cristae were broken and the GSDMD-NT expression was found notably increased in the DM group, while the protein expressions of xCT and GPX4 was significantly decreased, both of which were reversed by MCC950. High glucose reduced the cell viability and ATP level in vitro, accompanied by an increase in LDH release. All of the above indicators were reversed after NLRP3 knockdown compared with the HG treated alone. Moreover, the protein expressions of pyroptosis- and ferroptosis-related fators were significantly decreased or increased, consistent with the results shown by immunofluorescence. Furthermore, the protective effects of NLRP3 knockdown against HG were reversed following the mtROS agonist rotenone (ROT) treatment. In conclusion, inhibition of NLRP3 suppressed DM-induced myocardial injury. Promotion of mitochondrial ROS abolished the protective effect of knockdown NLRP3, and induced the happening of pyroptosis and ferroptosis. These findings may present a novel therapeutic underlying mechanism for clinical diabetes-induced myocardial injury treatment.
    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:干细胞衍生的细胞外囊泡(EV)是一类具有优异生物相容性的新兴治疗剂,生物活性和促再生能力。基于EV的药物的潜在目标之一是心血管疾病(CVD)。在这项工作中,我们使用了在不同氧气浓度(21、5和3%O2)下培养的人诱导多能干细胞(hiPSCs;hiPPS-EV)衍生的EV来剖析负责心脏保护的分子机制。
    方法:通过超滤结合尺寸排阻色谱(UFSEC)分离电动汽车,然后通过纳米粒子跟踪分析进行表征,原子力显微镜(AFM)和蛋白质印迹方法。液相色谱和串联质谱结合生物信息学分析用于鉴定各种氧气条件下的差异富集蛋白。我们直接比较了这些EV在心肌细胞(CM)损伤的氧-葡萄糖剥夺/复氧(OGD/R)模型中的心脏保护作用。利用先进的分子生物学,荧光显微镜,原子力谱和生物信息学技术,我们研究了参与细胞存活调节的细胞内信号通路,细胞凋亡和抗氧化反应。在用ML385抑制NRF2后的CM中评估EV对NRF2调节的信号传导的直接作用。
    结果:我们证明,与其他测试的氧气条件(常氧;EV-N和低氧3%O2;EV-H3)衍生自5%O2的生理性缺氧(EV-H5)的hiPS-EV对受损CM的细胞保护功能增强。这是由于转移后受体细胞中Akt激酶的磷酸化率较高,调节AMPK活性和减少细胞凋亡。此外,我们提供了使用AFM测量的EV-H5治疗的CM的钙信号改善和持续收缩性的直接证据.机械上,我们的质谱和生物信息学分析显示,EV-H5中的差异富集蛋白与NRF2调节的抗氧化途径相关.在这方面,EV-H5在OGD/R时增加NRF2蛋白的核易位并增强其在CMs中的转录。相比之下,用ML385抑制NRF2消除了电动汽车对CMs的保护作用。
    结论:在这项工作中,与EV-N和EV-H3相比,我们证明了EV-H5具有更好的心脏保护功能.这样的电动汽车在恢复应力CM的氧化还原平衡方面最有效,保持其收缩功能并防止细胞死亡。我们的数据支持生理性缺氧产生的hiPS-EV的潜在用途,作为具有再生特性的无细胞疗法,用于治疗心脏病。
    BACKGROUND: Stem cell-derived extracellular vesicles (EVs) are an emerging class of therapeutics with excellent biocompatibility, bioactivity and pro-regenerative capacity. One of the potential targets for EV-based medicines are cardiovascular diseases (CVD). In this work we used EVs derived from human induced pluripotent stem cells (hiPSCs; hiPS-EVs) cultured under different oxygen concentrations (21, 5 and 3% O2) to dissect the molecular mechanisms responsible for cardioprotection.
    METHODS: EVs were isolated by ultrafiltration combined with size exclusion chromatography (UF + SEC), followed by characterization by nanoparticle tracking analysis, atomic force microscopy (AFM) and Western blot methods. Liquid chromatography and tandem mass spectrometry coupled with bioinformatic analyses were used to identify differentially enriched proteins in various oxygen conditions. We directly compared the cardioprotective effects of these EVs in an oxygen-glucose deprivation/reoxygenation (OGD/R) model of cardiomyocyte (CM) injury. Using advanced molecular biology, fluorescence microscopy, atomic force spectroscopy and bioinformatics techniques, we investigated intracellular signaling pathways involved in the regulation of cell survival, apoptosis and antioxidant response. The direct effect of EVs on NRF2-regulated signaling was evaluated in CMs following NRF2 inhibition with ML385.
    RESULTS: We demonstrate that hiPS-EVs derived from physiological hypoxia at 5% O2 (EV-H5) exert enhanced cytoprotective function towards damaged CMs compared to EVs derived from other tested oxygen conditions (normoxia; EV-N and hypoxia 3% O2; EV-H3). This resulted from higher phosphorylation rates of Akt kinase in the recipient cells after transfer, modulation of AMPK activity and reduced apoptosis. Furthermore, we provide direct evidence for improved calcium signaling and sustained contractility in CMs treated with EV-H5 using AFM measurements. Mechanistically, our mass spectrometry and bioinformatics analyses revealed differentially enriched proteins in EV-H5 associated with the antioxidant pathway regulated by NRF2. In this regard, EV-H5 increased the nuclear translocation of NRF2 protein and enhanced its transcription in CMs upon OGD/R. In contrast, inhibition of NRF2 with ML385 abolished the protective effect of EVs on CMs.
    CONCLUSIONS: In this work, we demonstrate a superior cardioprotective function of EV-H5 compared to EV-N and EV-H3. Such EVs were most effective in restoring redox balance in stressed CMs, preserving their contractile function and preventing cell death. Our data support the potential use of hiPS-EVs derived from physiological hypoxia, as cell-free therapeutics with regenerative properties for the treatment of cardiac diseases.
    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: English Abstract
    目的:探讨miRNA-224-5p在缺氧/复氧(H/R)诱导的H9c2心肌细胞损伤中的调控作用。
    方法:从160例急性心肌梗死患者和80例健康对照(HC)中收集血浆样本,以测量miRNA-224-5p水平和其他生化参数。在培养的H/R损伤的H9c2细胞中,用miR-224-5p模拟物或阴性对照序列转染对细胞活力的影响,丙二醛(MDA)含量,并检测了超氧化物歧化酶2(SOD2)和乳酸脱氢酶(LDH)的活性。进行双荧光素酶报告基因测定以验证miR-224-5p与PTEN之间的靶向关系。生物信息学方法用于分析靶基因的潜在机制。qRT-PCR检测miRNA-224-5p在处理细胞中的表达,PTEN的蛋白质表达,Bcl-2,Bax,caspase-3,SOD2,p-PI3K/PI3K,使用蛋白质印迹法测定p-Akt/Ak和p-FoxO1/FoxO1,用流式细胞仪分析细胞凋亡。
    结果:血糖水平,C反应蛋白,CK,AMI组CK-MB和cTnI明显高于HC组(P<0.05)。miR-224-5p的表达水平在STEMI和NSTEMI患者以及H/R损伤的H9c2细胞中显著降低。H/R损伤后,H9c2细胞的活力随时间而降低。PTEN是miR-224-5p的靶基因,而PI3K/Akt途径是最显著的富集途径。H/R损伤的H9c2细胞显示SOD2活性显著降低,LDH活性和MDA含量增加,细胞凋亡增加,p-PI3K蛋白表达水平降低,p-Akt,p-FoxO1,SOD2和Bcl-2,以及PTEN的表达增加,Bax,和裂解的caspase-3。通过在H/R暴露之前用miR-224-5p模拟物转染细胞,这些变化明显减弱。
    结论:MiR-224-5p过表达可通过PI3K/Akt/FoxO1轴上调抗氧化基因SOD2的表达,从而减轻H/R诱导的H9c2细胞氧化应激,减少细胞凋亡。
    OBJECTIVE: To investigate the regulatory role of miRNA-224-5p in hypoxia/reoxygenation (H/R) -induced H9c2 cardiomyocyte injury.
    METHODS: Plasma samples were collected from 160 patients with acute myocardial infarction and 80 healthy controls(HC) to measure miRNA-224-5p levels and other biochemical parameters. In cultured H9c2 cells with H/R injury, the effects of transfection with miR-224-5p mimics or a negative control sequence on cell viability, malondialdehyde (MDA) content, and superoxide dismutase 2 (SOD2) and lactate dehydrogenase (LDH) activities were tested. Dual luciferase reporter gene assay was performed to verify the targeting relationship between miR-224-5p and PTEN. Bioinformatics methods were used to analyze the potential mechanisms of the target genes. The expression of miRNA-224-5p in the treated cells was detected with qRT-PCR, the protein expressions of PTEN, Bcl-2, Bax, cleaved caspase-3, SOD2, p-PI3K/PI3K, p-Akt/Ak and p-FoxO1/FoxO1 were determined using Western blotting, and cell apoptosis was analysed with flow cytometry.
    RESULTS: The levels of blood glucose, C-reactive protein, CK, CK-MB and cTnI were significantly higher in the AMI group compared with the HC group (P < 0.05). The expression level of miR-224-5p was significantly lowered in patients with STEMI and NSTEMI and in H9c2 cells with H/R injury. The viability of H9c2 cells decreased time-dependently following H/R injury. PTEN was a target gene of miR-224-5p, and the PI3K/Akt pathway was the most significantly enriched pathway. H9c2 cells with H/R injury showed significantly decreased SOD2 activity, increased LDH activity and MDA content, increased cell apoptosis, decreased protein expression levels of p-PI3K, p-Akt, p-FoxO1, SOD2, and Bcl-2, and increased expressions of PTEN, Bax, and cleaved caspase-3. These changes were obviously attenuated by trasnfection of the cells with miR-224-5p mimics prior to H/R exposure.
    CONCLUSIONS: MiR-224-5p overexpression upregulates the expression of the antioxidant gene SOD2 through the PI3K/Akt/FoxO1 axis to relieve H/R-induced oxidative stress and reduce apoptosis of H9c2 cells.
    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
    肥厚型心肌病(HCM)是一种以左心室肥厚和舒张功能障碍为特征的遗传性疾病,并增加心律失常和心力衰竭的风险。一些HCM患者发展为肥厚型心肌病(D-HCM)的扩张期,预后不良;然而,其发病机制尚不清楚,病理模型很少。这项研究建立了来自D-HCM患者的疾病特异性人类诱导多能干细胞(iPSCs),该患者在MYBPC3(c.1377delC)中具有突变,一种常见的HCM致病基因,并使用疾病特异性iPSC衍生的心肌细胞(iPSC-CMs)研究了相关的病理生理机制。我们证实了D-HCM患者来源的iPSC(D-HCMiPSC)中多能标志物的表达和分化成三个胚层的能力。D-HCMiPSC-CM显示心肌肌节结构破坏,线粒体受损数量增加。在D-HCMiPSC-CM中,Ca2+成像显示Ca2+信号异常增加,衰减时间延长。细胞代谢分析显示基础呼吸增加,最大呼吸,D-HCMiPSC-CM中的备用呼吸能力。RNA测序还显示线粒体电子传递系统相关基因的表达增加。D-HCMiPSC-CM显示异常的Ca2+处理和高代谢状态,与以前报道的HCM患者来源的iPSC-CM相似。尽管需要进一步研究,这有望成为D-HCM的有用病理模型.
    Hypertrophic cardiomyopathy (HCM) is an inherited disorder characterized by left ventricular hypertrophy and diastolic dysfunction, and increases the risk of arrhythmias and heart failure. Some patients with HCM develop a dilated phase of hypertrophic cardiomyopathy (D-HCM) and have poor prognosis; however, its pathogenesis is unclear and few pathological models exist. This study established disease-specific human induced pluripotent stem cells (iPSCs) from a patient with D-HCM harboring a mutation in MYBPC3 (c.1377delC), a common causative gene of HCM, and investigated the associated pathophysiological mechanisms using disease-specific iPSC-derived cardiomyocytes (iPSC-CMs). We confirmed the expression of pluripotent markers and the ability to differentiate into three germ layers in D-HCM patient-derived iPSCs (D-HCM iPSCs). D-HCM iPSC-CMs exhibited disrupted myocardial sarcomere structures and an increased number of damaged mitochondria. Ca2+ imaging showed increased abnormal Ca2+ signaling and prolonged decay time in D-HCM iPSC-CMs. Cell metabolic analysis revealed increased basal respiration, maximal respiration, and spare-respiratory capacity in D-HCM iPSC-CMs. RNA sequencing also showed an increased expression of mitochondrial electron transport system-related genes. D-HCM iPSC-CMs showed abnormal Ca2+ handling and hypermetabolic state, similar to that previously reported for HCM patient-derived iPSC-CMs. Although further studies are required, this is expected to be a useful pathological model for D-HCM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号