Myocytes, Cardiac

肌细胞, 心脏
  • 文章类型: Journal Article
    COVID-19患者的不良心脏结局,特别是那些已经有心脏病的人,激发基于人类细胞的芯片上器官模型的发展,以概括心脏损伤和功能障碍,并筛选心脏保护性疗法。这里,我们开发了一种芯片心脏模型来研究SARS-CoV-2在健康心肌中的发病机理,该模型由人诱导多能干细胞(iPSC)衍生的心肌细胞和心功能不全模型建立,模仿血管紧张素II(AngII)引起的先前存在的高血压疾病的方面。我们概述了SARS-CoV-2引起的心脏损伤的细胞病变特征,包括逐渐受损的收缩功能和钙处理,凋亡,和肌节混乱。与单独的SARS-CoV-2相比,在AngII治疗的芯片心脏中存在SARS-CoV-2会降低收缩力,并较早地发生收缩功能障碍,并大大增强了炎性细胞因子。为了开发潜在的疗法,我们评估了来自人类iPSC的细胞外囊泡(EV)的心脏保护作用,该作用减轻了收缩力的损害,细胞凋亡减少,减少肌节蛋白的破坏,增强β-氧化基因表达。AngII或EV治疗均不影响病毒载量。我们鉴定了微小RNAmiR-20a-5p和miR-19a-3p作为这些EV的心脏保护作用的潜在介质。
    Adverse cardiac outcomes in COVID-19 patients, particularly those with preexisting cardiac disease, motivate the development of human cell-based organ-on-a-chip models to recapitulate cardiac injury and dysfunction and for screening of cardioprotective therapeutics. Here, we developed a heart-on-a-chip model to study the pathogenesis of SARS-CoV-2 in healthy myocardium established from human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and a cardiac dysfunction model, mimicking aspects of preexisting hypertensive disease induced by angiotensin II (Ang II). We recapitulated cytopathic features of SARS-CoV-2-induced cardiac damage, including progressively impaired contractile function and calcium handling, apoptosis, and sarcomere disarray. SARS-CoV-2 presence in Ang II-treated hearts-on-a-chip decreased contractile force with earlier onset of contractile dysfunction and profoundly enhanced inflammatory cytokines compared to SARS-CoV-2 alone. Toward the development of potential therapeutics, we evaluated the cardioprotective effects of extracellular vesicles (EVs) from human iPSC which alleviated the impairment of contractile force, decreased apoptosis, reduced the disruption of sarcomeric proteins, and enhanced beta-oxidation gene expression. Viral load was not affected by either Ang II or EV treatment. We identified MicroRNAs miR-20a-5p and miR-19a-3p as potential mediators of cardioprotective effects of these EVs.
    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
    免疫球蛋白(Ig)已被广泛接受为仅由B细胞表达。尽管如此,这一理论受到越来越多的证据的挑战,这些证据表明Ig也可以由非B细胞(非B-Ig)产生,包括心肌细胞(CM)。非B-Ig表现出独特的物理和化学特性,独特的可变区序列和功能,与B-Ig不同。例如,非B-Ig显示疏水性,可变区的多样性有限,和细胞外基质蛋白活性。同样,心肌细胞可以表达不同类型的Igs,包括IgM,IgG,和游离的Igκ轻链(心肌细胞衍生的Ig,CM-Ig)。特别是,在各种心血管疾病中,CM-Ig可以分泌到细胞外空间,如心肌缺血和心肌纤维化,它们可能参与补体激活和对心肌细胞的直接损伤。然而,CM-Ig的确切病理活性尚不清楚。最近,朱等人。重点研究了CM-Igκ的序列特征和功能;他们发现CM-Igκ表现出独特的VJ重组模式,高疏水性,并且主要位于心肌细胞的插入盘和交叉条纹上。有趣的是,心肌细胞中Igκ的丢失导致插入椎间盘的结构紊乱和心肌收缩和传导功能障碍。机械上,Igκ促进plectin的稳定,一种细胞骨架交联蛋白,连接desmin和desomsome,以维持插入盘的正常结构。这一发现表明CM-Igκ在维持细胞骨架结构中起着不可或缺的作用。因此,揭示与CM-Igs相关的病理损伤的生理功能和机制势在必行。
    Immunoglobulins (Igs) have been widely accepted to be exclusively expressed by B cells. Nonetheless, this theory is challenged by mounting evidence which suggests that Igs can also be generated by non B cells (non B-Ig), including cardiomyocytes (CM). Non B-Ig exhibits unique physical and chemical characteristics, unique variable region sequences and functions, which diverge from those of B-Ig. For instance, non B-Ig demonstrates hydrophobicity, limited diversity in the variable region, and extracellular matrix protein activity. Likewise, cardiomyocytes can express different classes of Igs, including IgM, IgG, and free Igκ light chains (cardiomyocyte derived-Igs, CM-Igs). In particular, CM-Igs can be secreted into the extracellular space in various cardiovascular diseases, such as myocardial ischaemia and myocardial fibrosis where they might be involved in complement activation and direct damage to cardiomyocytes. Nevertheless, the precise pathological activity of CM-Igs remains unclear. Recently, Zhu et al. focused on studying the sequence characteristics and functions of CM-Igκ; they discovered that the CM-Igκ exhibits a unique VJ recombination pattern, high hydrophobicity, and is principally located on the intercalated discs and cross striations of the cardiomyocytes. Interestingly, loss of Igκ in cardiomyocytes results in structural disorders in intercalated discs and dysfunction in myocardial contraction and conduction. Mechanically, Igκ promotes the stabilisation of plectin, a cytoskeleton cross-linker protein that connects desmin to desomsome, to maintain the normal structure of the intercalated disc. This finding indicates that CM-Igκ plays an integral role in maintaining cytoskeleton structure. Consequently, it is imperative to reveal the physiological functions and mechanisms of pathological injury associated with CM-Igs.
    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
    尽管传统上认为V(D)J重组和免疫球蛋白(Ig)产生仅发生在B淋巴细胞和浆细胞中,Ig在非淋巴细胞中的表达,我们称之为非B细胞衍生的Ig(非BIg),已经被越来越多的研究记录了。已经证明,非B-Ig可以在大多数细胞类型中广泛表达,包括,但不限于,上皮细胞,心肌细胞,造血干/祖细胞,骨髓细胞,和免疫特权位点的细胞,如神经元和生精细胞。特别是,恶性肿瘤细胞表达高水平的IgG。此外,与主要定位于B细胞膜和血清中并主要执行免疫防御功能的B-Ig不同,已发现非B-Ig分布更广泛,在免疫防御中起关键作用,维持细胞增殖和存活,促进进步。非B-Ig的发现可能为多种免疫相关疾病的更多治疗策略提供了更富有的突破口。
    Although V(D)J recombination and immunoglobulin (Ig) production are traditionally recognised to occur only in B lymphocytes and plasma cells, the expression of Igs in non-lymphoid cells, which we call non B cell-derived Igs (non B Igs), has been documented by growing studies. It has been demonstrated that non B-Igs can be widely expressed in most cell types, including, but not limited to, epithelial cells, cardiomyocytes, hematopoietic stem/progenitor cells, myeloid cells, and cells from immune-privileged sites, such as neurons and spermatogenic cells. In particular, malignant tumour cells express high level of IgG. Moreover, different from B-Igs that mainly localised on the B cell membrane and in the serum and perform immune defence function mainly, non B-Igs have been found to distribute more widely and play critical roles in immune defence, maintaining cell proliferation and survival, and promoting progression. The findings of non B-Igs may provide a wealthier breakthrough point for more therapeutic strategies for a wide range of immune-related diseases.
    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
    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

    求助全文

  • 文章类型: Journal Article
    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
    心外膜,以前被视为心脏周围的被动外层,现在被认为是发展的重要组成部分,再生,和修复。在这次审查中,我们探索心外膜的细胞和分子组成,强调其在斑马鱼和sal的心脏再生和修复中的作用,以及它在年轻和成年出生后哺乳动物中的激活。我们还研究了用于研究治疗性干预的心外膜细胞功能的最新技术。对高度再生动物模型的分析表明,心外膜在调节心肌细胞增殖中至关重要。一过性纤维化,和新血管形成。然而,尽管心外膜具有解决心脏损伤的独特细胞程序,目前尚不清楚如何在非再生哺乳动物中复制这些过程.在心肌梗塞期间,心外膜细胞分泌调节纤维化的信号因子,血管,和炎症重塑,差异增强或抑制心脏修复。最近的转录组学研究已经验证了不同物种和发育阶段的心外膜的细胞和分子异质性,进一步阐明其在病理条件下的功能。这些研究还提供了对调节心外膜衍生信号分子在各种疾病中的功能的见解,这可能导致新的疗法和修复性心血管医学的进步。此外,从研究心外膜细胞功能获得的见解已经启动了新技术的发展,包括使用人类多能干细胞和心脏类器官来模拟心血管系统内的修复过程。这种对心外膜功能的日益理解为开发旨在解决发育性心脏病的创新治疗策略提供了潜力。加强再生疗法,和减轻心血管疾病的进展。
    The epicardium, previously viewed as a passive outer layer around the heart, is now recognized as an essential component in development, regeneration, and repair. In this review, we explore the cellular and molecular makeup of the epicardium, highlighting its roles in heart regeneration and repair in zebrafish and salamanders, as well as its activation in young and adult postnatal mammals. We also examine the latest technologies used to study the function of epicardial cells for therapeutic interventions. Analysis of highly regenerative animal models shows that the epicardium is essential in regulating cardiomyocyte proliferation, transient fibrosis, and neovascularization. However, despite the epicardium\'s unique cellular programs to resolve cardiac damage, it remains unclear how to replicate these processes in nonregenerative mammalian organisms. During myocardial infarction, epicardial cells secrete signaling factors that modulate fibrotic, vascular, and inflammatory remodeling, which differentially enhance or inhibit cardiac repair. Recent transcriptomic studies have validated the cellular and molecular heterogeneity of the epicardium across various species and developmental stages, shedding further light on its function under pathological conditions. These studies have also provided insights into the function of regulatory epicardial-derived signaling molecules in various diseases, which could lead to new therapies and advances in reparative cardiovascular medicine. Moreover, insights gained from investigating epicardial cell function have initiated the development of novel techniques, including using human pluripotent stem cells and cardiac organoids to model reparative processes within the cardiovascular system. This growing understanding of epicardial function holds the potential for developing innovative therapeutic strategies aimed at addressing developmental heart disorders, enhancing regenerative therapies, and mitigating cardiovascular disease progression.
    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
    尽管临床和科学进步,心力衰竭是世界范围内发病率和死亡率的主要原因。线粒体功能障碍和炎症都有助于心力衰竭的发展和进展。尽管炎症对急性心肌细胞损伤后的修复性愈合至关重要,慢性炎症会损害心脏,损害功能,减少心输出量.线粒体,占心肌细胞体积的三分之一,可能是心力衰竭的潜在治疗靶点。主要以能源生产闻名,线粒体还参与其他过程,包括钙稳态和细胞凋亡的调节。线粒体功能与形态学密切相关,通过线粒体动力学改变,从而确保满足电池的能量需求。然而,在心力衰竭中,底物使用的变化会导致线粒体功能障碍和心肌细胞功能受损。这篇综述讨论了线粒体和cr的动力学,包括线粒体接触位点和cr组织系统复合物在线粒体超微结构变化中的作用。此外,这篇综述涵盖了线粒体-内质网接触位点的作用,通过纳米隧道的线粒体通讯,心力衰竭期间代谢产物的产生也发生了改变.我们强调了这些经常被忽视的因素和有希望的心力衰竭的临床线粒体靶标。
    Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.
    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
    全角下垂,一种复杂形式的促炎程序性细胞死亡,包括细胞凋亡,焦亡和坏死,近年来已经成为一个新兴的概念,在癌症中被广泛报道,传染病和神经系统疾病。心血管疾病(CVD)是一个重要的全球性健康问题,对个人生命构成严重威胁。越来越多的研究表明,炎症在心血管疾病中起着关键作用,为PANoptosis促进CVD的进展提供了重要的理论依据。迄今为止,仅有零星的关于心血管疾病中PANoptosis的研究报道,其在心血管疾病领域的作用尚未得到充分探索。阐明心肌细胞死亡的各种模式,在各种应激刺激下,特定的分子机制和各种死亡方式之间的联系对于更深入地了解CVD的病理生理学具有重要的临床意义。本文综述了细胞凋亡的分子机制,焦亡,坏死和全视及其在心血管疾病领域的前景。
    PANoptosis, a complex form of proinflammatory programmed cell death, including apoptosis, pyroptosis and necroptosis, has been an emerging concept in recent years that has been widely reported in cancer, infectious diseases and neurological disorders. Cardiovascular diseases (CVDs) are an important global health problem, posing a serious threat to individuals\' lives. An increasing body of research shows that inflammation has a pivotal role in CVDs, which provides an important theoretical basis for PANoptosis to promote the progression of CVDs. To date, only sporadic studies on PANoptosis in CVDs have been reported and its role in the field of CVDs has not been fully explored. Elucidating the various modes of cardiomyocyte death, the specific molecular mechanisms and the links among the various modes of death under various stressful stimuli is of notable clinical significance for a deeper understanding of the pathophysiology of CVDs. The present review summarizes the molecular mechanisms of apoptosis, pyroptosis, necroptosis and PANoptosis and their prospects in the field of CVDs.
    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
    肌节复合物如何在长期存活的心肌细胞中连续翻转尚不清楚。根据肌节维持的流行模式,肌节由细胞质可溶性蛋白质池维持,在池和肌节之间自由循环。
    我们对表达和内源性肌节蛋白的周转进行了成像和定量,包括巨大的蛋白质肌动蛋白,在培养和体内的心肌细胞中,在单细胞和单个肌节水平上,使用带共价配体的Halo标记蛋白的脉冲追踪标记。
    我们反驳了流行的蛋白质库模型,而是显示了一种有序的机制,其中只有新翻译的蛋白质进入肌节复合物,而较旧的蛋白质被去除和降解。我们还表明,降解与蛋白质年龄无关,并且蛋白水解提取是周转中的限速步骤。我们表明肌节蛋白的替换在细胞内和整个心脏中以相似的速率发生,而在成体细胞中更慢。
    我们的发现为心脏肉瘤亚基置换建立了单向置换模型,并确定了它们的周转原理。
    UNASSIGNED: How the sarcomeric complex is continuously turned over in long-living cardiomyocytes is unclear. According to the prevailing model of sarcomere maintenance, sarcomeres are maintained by cytoplasmic soluble protein pools with free recycling between pools and sarcomeres.
    UNASSIGNED: We imaged and quantified the turnover of expressed and endogenous sarcomeric proteins, including the giant protein titin, in cardiomyocytes in culture and in vivo, at the single cell and at the single sarcomere level using pulse-chase labeling of Halo-tagged proteins with covalent ligands.
    UNASSIGNED: We disprove the prevailing protein pool model and instead show an ordered mechanism in which only newly translated proteins enter the sarcomeric complex while older ones are removed and degraded. We also show that degradation is independent of protein age and that proteolytic extraction is a rate-limiting step in the turnover. We show that replacement of sarcomeric proteins occurs at a similar rate within cells and across the heart and is slower in adult cells.
    UNASSIGNED: Our findings establish a unidirectional replacement model for cardiac sarcomeres subunit replacement and identify their turnover principles.
    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:氧化应激可能导致糖尿病心肌病的心脏ryanodine受体(RyR2)功能障碍。人参皂苷Rb1(Rb1)是人参治疗心血管疾病的主要药理活性成分。Rb1是否治疗糖尿病心脏损伤仍然未知。本研究旨在探讨Rb1对糖尿病损伤心肌组织的影响,并进一步探讨其可能的分子药理学机制。
    方法:雄性SD大鼠注射链脲佐菌素溶液2周,随后6周Rb1或胰岛素治疗。SOD的活性,CAT,Gpx,并测量MDA水平;组织学和超微结构分析,进行RyR2活性和磷酸化RyR2(Ser2808)蛋白表达分析;和Tunel测定。
    结果:SOD活性降低,CAT,糖尿病组的Gpx和MDA水平高于对照组。Rb1处理增加了SOD的活性,CAT,与糖尿病大鼠相比,Gpx和MDA水平降低。中和RyR2活性在糖尿病中明显低于对照组,Rb1治疗组较糖尿病组增加。与对照组相比,糖尿病大鼠RyR2Ser2808的磷酸化表达增加,并用胰岛素和Rb1治疗减毒。糖尿病细胞凋亡率增加,Rb1处理降低了细胞凋亡率。Rb1和胰岛素改善糖尿病大鼠心肌损伤。
    结论:这些数据表明Rb1可用于减轻氧化损伤,糖尿病心肌病患者RyR2Ser2808磷酸化降低,心肌细胞凋亡率降低。
    BACKGROUND: Oxidative stress may contribute to cardiac ryanodine receptor (RyR2) dysfunction in diabetic cardiomyopathy. Ginsenoside Rb1 (Rb1) is a major pharmacologically active component of ginseng to treat cardiovascular diseases. Whether Rb1 treat diabetes injured heart remains unknown. This study was to investigate the effect of Rb1 on diabetes injured cardiac muscle tissue and to further investigate its possible molecular pharmacology mechanisms.
    METHODS: Male Sprague-Dawley rats were injected streptozotocin solution for 2 weeks, followed 6 weeks Rb1 or insulin treatment. The activity of SOD, CAT, Gpx, and the levels of MDA was measured; histological and ultrastructure analyses, RyR2 activity and phosphorylated RyR2(Ser2808) protein expression analyses; and Tunel assay were performed.
    RESULTS: There was decreased activity of SOD, CAT, Gpx and increased levels of MDA in the diabetic group from control. Rb1 treatment increased activity of SOD, CAT, Gpx and decreased the levels of MDA as compared with diabetic rats. Neutralizing the RyR2 activity significantly decreased in diabetes from control, and increased in Rb1 treatment group from diabetic group. The expression of phosphorylation of RyR2 Ser2808 was increased in diabetic rats from control, and were attenuated with insulin and Rb1 treatment. Diabetes increased the apoptosis rate, and Rb1 treatment decreased the apoptosis rate. Rb1 and insulin ameliorated myocardial injury in diabetic rats.
    CONCLUSIONS: These data indicate that Rb1 could be useful for mitigating oxidative damage, reduced phosphorylation of RyR2 Ser2808 and decreased the apoptosis rate of cardiomyocytes in diabetic cardiomyopathy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号