cardiomyocyte

心肌细胞
  • 文章类型: Journal Article
    新生啮齿动物的心脏再生取决于预先存在的心肌细胞增殖和分裂的能力。当这些细胞迅速从增生转变为肥大时,这种能力在出生后发育的第一周内丧失。退出细胞周期,变成双核,并增加尺寸。细胞大小和成核的这些动态变化如何影响心肌细胞增殖潜力尚不清楚。在这项研究中,我们创新了商用数字全息成像显微镜的应用,HolomonitorM4,以评估CHIR99021治疗后单核细胞和双核心肌细胞的增殖反应,增殖刺激模型。该系统能够以单细胞分辨率实时对原代心肌细胞动力学进行长期无标记定量跟踪。我们的结果证实,CHIR99021对糖原合酶激酶3的化学抑制促进了单核细胞和双核心肌细胞的高频率完整细胞分裂。在这些增殖事件期间对心肌细胞体积动力学的定量跟踪表明,在尝试细胞分裂之前,单核和双核心肌细胞均达到相似的大小增加阈值。双核心肌细胞试图以比单核心肌细胞更低的频率分裂,这可能与细胞大小的不充分增加有关。通过定义心肌细胞大小之间的相互关系,成核,和细胞周期控制,我们可能会更好地了解导致哺乳动物出生后心脏再生能力丧失的细胞机制。
    Cardiac regeneration in newborn rodents depends on the ability of pre-existing cardiomyocytes to proliferate and divide. This capacity is lost within the first week of postnatal development when these cells rapidly switch from hyperplasia to hypertrophy, withdraw from the cell cycle, become binucleated, and increase in size. How these dynamic changes in cell size and nucleation impact cardiomyocyte proliferative potential is not well understood. In this study, we innovate the application of a commercially available digital holographic imaging microscope, the Holomonitor M4, to evaluate the proliferative responses of mononucleated and binucleated cardiomyocytes after CHIR99021 treatment, a model proliferative stimulus. This system enables long-term label-free quantitative tracking of primary cardiomyocyte dynamics in real-time with single-cell resolution. Our results confirm that chemical inhibition of glycogen synthase kinase 3 with CHIR99021 promotes complete cell division of both mononucleated and binucleated cardiomyocytes with high frequency. Quantitative tracking of cardiomyocyte volume dynamics during these proliferative events revealed that both mononucleated and binucleated cardiomyocytes reach a similar size-increase threshold prior to attempted cell division. Binucleated cardiomyocytes attempt to divide with lower frequency than mononucleated cardiomyocytes, which may be associated with inadequate increases in cell size. By defining the interrelationship between cardiomyocyte size, nucleation, and cell cycle control, we may better understand the cellular mechanisms that drive the loss of mammalian cardiac regenerative capacity after birth.
    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
    胚胎发育过程中的缺氧可以永久重塑脊椎动物的心脏,通常在成年期引起心血管异常。虽然这种现象大多是有害的,最近的证据表明,发育性缺氧会在某些放热脊椎动物中产生耐受应激的表型。经历慢性缺氧的胚胎普通飞龟(Chelydraserpentina)在孵化后显示出改善的心脏缺氧耐受性,这与心脏细胞(心肌细胞)中Ca2+稳态的改变有关。在这里,我们研究了Ca2+循环变化的可能性,通过肌浆网(SR),是飞龟的发育编程心脏表型的基础。我们通过从正常氧(21%O2;“N21”)或慢性缺氧(10%O2;“H10”)的幼龟中分离心肌细胞来研究这一假设,并对细胞进行缺氧/复氧。在存在或不存在SRCa2+循环抑制剂的情况下。我们同时测量细胞缩短,细胞内[Ca2+],和细胞内pH(pHi)。在常氧条件下,N21和H10心肌细胞同样缩短,但H10Ca2+瞬变(Δ[Ca2+]i)比N21细胞小两倍,和SR抑制仅减少N21缩短和Δ[Ca2]i。缺氧随后抑制缩短,Δ[Ca2+]i,和pHi在对照N21和H10心肌细胞中,然而,H10缩短和Δ[Ca2+]i恢复到缺氧前水平,部分原因是肌丝Ca2+敏感性增强。SR阻断消除了缺氧H10心肌细胞的恢复,并增强了缩短的减少,Δ[Ca2+]i,和pHi。我们的新结果为SR功能的发育编程提供了第一个证据,并证明发育缺氧赋予了海龟耐缺氧的心脏表型,通过增强肌丝Ca2+敏感性和修饰SR功能。
    Oxygen deprivation during embryonic development can permanently remodel the vertebrate heart, often causing cardiovascular abnormalities in adulthood. While this phenomenon is mostly damaging, recent evidence suggests developmental hypoxia produces stress-tolerant phenotypes in some ectothermic vertebrates. Embryonic common snapping turtles (Chelydra serpentina) subjected to chronic hypoxia display improved cardiac anoxia tolerance after hatching, which is associated with altered Ca2+ homeostasis in heart cells (cardiomyocytes). Here we examined the possibility that changes in Ca2+ cycling, through the sarcoplasmic reticulum (SR), underlie the developmentally programmed cardiac phenotype of snapping turtles. We investigated this hypothesis by isolating cardiomyocytes from juvenile turtles that developed in either normoxia (21% O2; \"N21\") or chronic hypoxia (10% O2; \"H10\") and subjected the cells to anoxia/reoxygenation, either in the presence or absence of SR Ca2+-cycling inhibitors. We simultaneously measured cellular shortening, intracellular [Ca2+], and intracellular pH (pHi). Under normoxic conditions, N21 and H10 cardiomyocytes shortened equally, but H10 Ca2+ transients (Δ[Ca2+]i) were twofold smaller than N21 cells, and SR inhibition only decreased N21 shortening and Δ[Ca2+]i. Anoxia subsequently depressed shortening, Δ[Ca2+]i, and pHi in control N21 and H10 cardiomyocytes, yet H10 shortening and Δ[Ca2+]i recovered to pre-anoxic levels, partly due to enhanced myofilament Ca2+ sensitivity. SR blockade abolished the recovery of anoxic H10 cardiomyocytes and potentiated decreases in shortening, Δ[Ca2+]i, and pHi. Our novel results provide the first evidence of developmental programming of SR function and demonstrate that developmental hypoxia confers a long-lasting, superior anoxia-tolerant cardiac phenotype in snapping turtles, by enhancing myofilament Ca2+ sensitivity and modifying SR function.
    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
    流行病学表明细颗粒物(PM)暴露与心血管疾病之间存在密切的关系。然而,PM是否加重心肌缺血再灌注(I/R)损伤,相关机制尚不清楚。我们之前的研究表明,脂肪干细胞来源的外泌体(ADSC-Exos)含有高水平的Mir221和Mir222。本研究探讨了PM暴露通过线粒体自噬和凋亡对I/R诱导的心脏损伤的影响。以及Mir221和Mir222在ADSC-Exos中的潜在作用。野生型,mir221-和mir222-敲除(KO),将过表达Mir221和Mir222的转基因(TG)小鼠气管内注射PM(10mg/kg)。24小时后,小鼠接受左冠状动脉结扎30分钟,然后再灌注3小时(I/R)。H9c2心肌细胞在1%O2下培养6h,然后复氧12小时(缺氧-复氧[H/R])。PM通过增加ROS水平并引起线粒体功能障碍而加重I/R(或H/R)心脏损伤,在体内和体外增加了线粒体裂变相关蛋白(DNM1L/Drp1和MFF)和线粒体自噬相关蛋白(BNIP3和MAP1LC3B/LC3B)的表达。用ADSC-Exos或Mir221-和Mir222-模拟物治疗显著减少PM+I/R-诱导的心脏损伤。重要的是,ADSC-Exos包含Mir221和Mir222,它们直接针对BNIP3,MAP1LC3B/LC3B,和BBC3/PUMA,降低其表达并最终减少心肌细胞的线粒体自噬和凋亡。目前的数据表明,ADSC-Exos治疗通过Mir221和Mir222-BNIP3-MAP1LC3B-BBC3/PUMA通路调节线粒体自噬和凋亡,并显着减少PMI/R引起的心脏损伤。本研究揭示了ADSC-Exos在减轻PM引起的心肌I/R损伤加重方面的新治疗潜力。缩写:ADSC-Exos:脂肪干细胞外泌体;AL:自溶酶体;ATP:三磷酸腺苷;BBC3/PUMA:BCL2结合成分3;BNIP3:BCL2/腺病毒E1B相互作用蛋白3;CASP3:caspase3;CASP9:caspase9;CDKN1B/p27:细胞周期蛋白依赖性激酶抑制剂DCH疾病:心血管7;二氯二氢荧光素二乙酸盐;DHE:二氢乙啶;DNM1L/Drp1:动力蛋白1样;EF:射血分数;FS:分数缩短;H/R:缺氧复氧;I/R:缺血再灌注;LDH:乳酸脱氢酶;MAP1LC3B/LC3B:微管相关蛋白PR3AMPT蛋白;NTEM-dpA-N-N-
    Epidemiology has shown a strong relationship between fine particulate matter (PM) exposure and cardiovascular disease. However, it remains unknown whether PM aggravates myocardial ischemia-reperfusion (I/R) injury, and the related mechanisms are unclear. Our previous study has shown that adipose stem cell-derived exosomes (ADSC-Exos) contain high levels of Mir221 and Mir222. The present study investigated the effects of PM exposure on I/R-induced cardiac injury through mitophagy and apoptosis, as well as the potential role of Mir221 and Mir222 in ADSC-Exos. Wild-type, mir221- and mir222-knockout (KO), and Mir221- and Mir222-overexpressing transgenic (TG) mice were intratracheally injected with PM (10 mg/kg). After 24 h, mice underwent left coronary artery ligation for 30 min, followed by 3 h of reperfusion (I/R). H9c2 cardiomyocytes were cultured under 1% O2 for 6 h, then reoxygenated for 12 h (hypoxia-reoxygenation [H/R]). PM aggravated I/R (or H/R) cardiac injury by increasing ROS levels and causing mitochondrial dysfunction, which increased the expression of mitochondrial fission-related proteins (DNM1L/Drp1 and MFF) and mitophagy-related proteins (BNIP3 and MAP1LC3B/LC3B) in vivo and in vitro. Treatment with ADSC-Exos or Mir221- and Mir222-mimics significantly reduced PM+I/R-induced cardiac injury. Importantly, ADSC-Exos contain Mir221 and Mir222, which directly targets BNIP3, MAP1LC3B/LC3B, and BBC3/PUMA, decreasing their expression and ultimately reducing cardiomyocyte mitophagy and apoptosis. The present data showed that ADSC-Exos treatment regulated mitophagy and apoptosis through the Mir221 and Mir222-BNIP3-MAP1LC3B-BBC3/PUMA pathway and significantly reduced the cardiac damage caused by PM+I/R. The present study revealed the novel therapeutic potential of ADSC-Exos in alleviating PM-induced exacerbation of myocardial I/R injury.Abbreviation: ADSC-Exos: adipose-derived stem cell exosomes; AL: autolysosome; ATP: adenosine triphosphate; BBC3/PUMA: BCL2 binding component 3; BNIP3: BCL2/adenovirus E1B interacting protein 3; CASP3: caspase 3; CASP9: caspase 9; CDKN1B/p27: cyclin dependent kinase inhibitor 1B; CVD: cardiovascular disease; DCFH-DA: 2\',7\'-dichlorodihydrofluorescein diacetate; DHE: dihydroethidium; DNM1L/Drp1: dynamin 1-like; EF: ejection fraction; FS: fractional shortening; H/R: hypoxia-reoxygenation; I/R: ischemia-reperfusion; LDH: lactate dehydrogenase; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MFF: mitochondrial fission factor; miRNA: microRNA; NAC: N-acetylcysteine; OCR: oxygen consumption rate; PIK3C3/Vps34: phosphatidylinositol 3-kinase catalytic subunit type 3; PM: particulate matter; PRKAA1/AMPK: protein kinase AMP-activated catalytic subunit alpha 1; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TRP53/p53: transformation related protein 53; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling.
    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
    S-柠檬烯(s-Lim)是在多种植物中发现的单环单萜,已在心肌梗塞的实验模型中显示出抗氧化和心脏保护活性。这项研究的目的是评估s-Lim发挥其抗心律失常作用的潜在机制。关注β-肾上腺素受体(β-AR)的阻断及其对各种体内和体外参数的影响,包括心电图(ECG)测量,左心室发展压(LVDP),β-肾上腺素能途径,肌节缩短剂和L型钙电流(ICa,L).在孤立的心中,10μM的s-Lim不会改变ECG曲线或LVPD。s-Lim在50μM时增加了心率校正的QT间期(QTc)(10.8%),在30(12.4%)和50μM(16.6%)的浓度时降低了心率。s-Lim(10μM)还抑制了异丙肾上腺素(ISO)(1μM)引起的肾上腺素能反应,降低了心率的增加,LVDP和ECG改变。在心室心肌细胞中,s-Lim通过防止肌节缩短的增加来拮抗多巴酚丁胺的作用,与阿替洛尔(阻滞剂β1-AR)相似。在体内,s-Lim拮抗ISO(激动剂β1-AR)的作用,与普萘洛尔(一种非选择性阻断剂β-AR)相似。在心室心肌细胞中,s-Lim没有改变ICa的电压依赖性,L激活或ICa,L密度。此外,s-Lim不影响5μM毛喉素(腺苷酸环化酶的激活剂)介导的ECG效应的变化。在体内咖啡因/ISO诱导的心律失常模型中,s-Lim(1mg/kg)表现出抗心律失常作用,通过降低的心律失常评分验证,心率,室性早搏和不适当的窦性心动过速的发生。这些发现表明s-Lim的抗心律失常活性与心脏中β-AR的阻断有关。
    S-Limonene (s-Lim) is a monocyclic monoterpene found in a variety of plants and has been shown to present antioxidant and cardioprotective activity in experimental models of myocardial infarction. The aim of this study was to evaluate the potential mechanism by which s-Lim exerts its antiarrhythmic effect, focusing on the blockade of β-adrenoceptor (β-AR) and its effects on various in vivo and in vitro parameters, including electrocardiogram (ECG) measurements, left ventricular developed pressure (LVDP), the β-adrenergic pathway, sarcomeric shortening and L-type calcium current (ICa,L). In isolated hearts, 10 μM of s-Lim did not alter the ECG profile or LVPD. s-Lim increased the heart rate corrected QT interval (QTc) (10.8%) at 50 μM and reduced heart rate at the concentrations of 30 (12.4%) and 50 μM (16.6%). s-Lim (10 μM) also inhibited the adrenergic response evoked by isoproterenol (ISO) (1 μM) reducing the increased of heart rate, LVDP and ECG changes. In ventricular cardiomyocyte, s-Lim antagonized the effect of dobutamine by preventing the increase of sarcomeric shortening, demonstrating a similar effect to atenolol (blocker β1-AR). In vivo, s-Lim antagonized the effect of ISO (agonists β1-AR), presenting a similar effect to propranolol (a non-selective blocker β-AR). In ventricular cardiomyocyte, s-Lim did not alter the voltage dependence for ICa,L activation or the ICa,L density. In addition, s-Lim did not affect changes in the ECG effect mediated by 5 μM forskolin (an activator of adenylate cyclase). In an in vivo caffeine/ISO-induced arrhythmia model, s-Lim (1 mg/kg) presented antiarrhythmic action verified by a reduced arrhythmia score, heart rate, and occurrence of ventricular premature beats and inappropriate sinus tachycardia. These findings indicate that the antiarrhythmic activity of s-Lim is related to blockade of β-AR in the heart.
    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
    核和胞质蛋白的翻译后SUMO化维持真核细胞的稳态,并协调对代谢需求或细胞外刺激变化的程序性反应。在兴奋的细胞中,SUMO化调节离子通道的生物物理特性和运输。离子通道SUMO化状态由SUMO连接酶和解连接酶的相反酶活性决定。磷酸化在SUMO化中也起允许作用。已经确定了几种离子通道的SUMO解混合气体,但其相应的E3连接酶仍然未知。这项研究显示PIAS3,又名KChAP,是HEK细胞中Kv4.2和HCN2通道的真正的SUMOE3连接酶,和心肌细胞中的内源性Kv4.2和Kv4.3通道。PIAS3介导的在Kv4.2-K579的SUMO化通过依赖rab11a的再循环机制增加通道表面表达。Kv4.2-S552的PKA磷酸化降低了HEK293细胞中Kv4通道介导的电流,心肌细胞,和神经元。这项研究显示PKA介导的磷酸化阻断了HEK细胞和心肌细胞中的Kv4.2-K579SUMO化。一起,这些数据表明PIAS3是控制离子通道表面表达的信号级联中的关键下游介质.
    Post-translational SUMOylation of nuclear and cytosolic proteins maintains homeostasis in eukaryotic cells and orchestrates programmed responses to changes in metabolic demand or extracellular stimuli. In excitable cells, SUMOylation tunes the biophysical properties and trafficking of ion channels. Ion channel SUMOylation status is determined by the opposing enzyme activities of SUMO ligases and deconjugases. Phosphorylation also plays a permissive role in SUMOylation. SUMO deconjugases have been identified for several ion channels, but their corresponding E3 ligases remain unknown. This study shows PIAS3, a.k.a. KChAP, is a bona fide SUMO E3 ligase for Kv4.2 and HCN2 channels in HEK cells, and endogenous Kv4.2 and Kv4.3 channels in cardiomyocytes. PIAS3-mediated SUMOylation at Kv4.2-K579 increases channel surface expression through a rab11a-dependent recycling mechanism. PKA phosphorylation at Kv4.2-S552 reduces the current mediated by Kv4 channels in HEK293 cells, cardiomyocytes, and neurons. This study shows PKA mediated phosphorylation blocks Kv4.2-K579 SUMOylation in HEK cells and cardiomyocytes. Together, these data identify PIAS3 as a key downstream mediator in signaling cascades that control ion channel surface expression.
    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
    成年哺乳动物心脏具有极其有限的心脏再生能力。大多数心肌细胞生活在永久细胞周期停滞的状态下,无法重新进入周期。在新生儿发育过程中,心肌细胞从细胞增殖转变为成熟状态。尽管几种信号通路参与了这一转变,这些输入协调调节心肌细胞成熟的分子机制尚不完全清楚。维甲酸(RA)在发育中起着举足轻重的作用,形态发生,和再生。尽管RA信号在胚胎心脏发育中的重要性,对其在出生后早期的功能知之甚少。我们发现醛脱氢酶1家族成员A2(Aldh1a2)的mRNA表达,它编码合成全反式维甲酸(ATRA)的关键酶,是RA信号传导的重要调节剂,在新生小鼠心室中短暂上调。单细胞转录组分析和免疫组织化学显示,在出生后早期,Aldh1a2表达在心脏成纤维细胞中富集。施用ATRA抑制培养的新生大鼠心肌细胞和人心肌细胞中的心肌细胞增殖。RNA-seq分析表明,ATRA治疗的产前大鼠心室心肌细胞中细胞增殖相关基因下调,而心肌细胞成熟相关基因上调。这些发现表明,源自心脏成纤维细胞的RA信号是新生儿心脏发育过程中心肌细胞增殖和成熟的关键调节因子之一。
    The adult mammalian heart has extremely limited cardiac regenerative capacity. Most cardiomyocytes live in a state of permanent cell-cycle arrest and are unable to re-enter the cycle. Cardiomyocytes switch from cell proliferation to a maturation state during neonatal development. Although several signaling pathways are involved in this transition, the molecular mechanisms by which these inputs coordinately regulate cardiomyocyte maturation are not fully understood. Retinoic acid (RA) plays a pivotal role in development, morphogenesis, and regeneration. Despite the importance of RA signaling in embryo heart development, little is known about its function in the early postnatal period. We found that mRNA expression of aldehyde dehydrogenase 1 family member A2 (Aldh1a2), which encodes the key enzyme for synthesizing all-trans retinoic acid (ATRA) and is an important regulator for RA signaling, was transiently upregulated in neonatal mouse ventricles. Single-cell transcriptome analysis and immunohistochemistry revealed that Aldh1a2 expression was enriched in cardiac fibroblasts during the early postnatal period. Administration of ATRA inhibited cardiomyocyte proliferation in cultured neonatal rat cardiomyocytes and human cardiomyocytes. RNA-seq analysis indicated that cell proliferation-related genes were downregulated in prenatal rat ventricular cardiomyocytes treated with ATRA, while cardiomyocyte maturation-related genes were upregulated. These findings suggest that RA signaling derived from cardiac fibroblasts is one of the key regulators of cardiomyocyte proliferation and maturation during neonatal heart development.
    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
    心血管疾病(CVD)是全球范围内的主要死亡原因,高龄是CVD患病率的主要原因。细胞衰老是细胞周期停滞的不可逆状态,发生在老年或细胞遭受各种应激后。衰老细胞不仅导致细胞功能的降低,但也产生衰老相关的分泌表型(SASP)影响周围细胞和组织微环境。越来越多的证据表明,衰老心肌细胞的逐渐积累与心血管系统功能的下降有关。为了强调衰老心肌细胞在年龄相关性CVD病理生理学中的作用,我们首先介绍了衰老的心肌细胞可以通过结构变化和几种衰老相关的生物标志物来鉴定。我们随后提供了现有知识的全面总结,概述了关于衰老心肌细胞与年龄相关的CVD表型之间关系的令人信服的证据。此外,我们讨论了显著的治疗潜力代表的加速衰老心肌细胞的预防,以及一些现有的老年保护因子在预防和治疗与年龄相关的CVD方面的现状。一起,综述了心肌细胞衰老在心血管疾病中的作用,并探讨了衰老心肌细胞的分子知识及其在开发基于衰老的疗法中的潜在临床意义,从而为他们的生物学和潜在的治疗探索提供重要的见解。
    Cardiovascular diseases (CVD) are the leading cause of death worldwide, and advanced age is a main contributor to the prevalence of CVD. Cellular senescence is an irreversible state of cell cycle arrest that occurs in old age or after cells encounter various stresses. Senescent cells not only result in the reduction of cellular function, but also produce senescence-associated secretory phenotype (SASP) to affect surrounding cells and tissue microenvironment. There is increasing evidence that the gradual accumulation of senescent cardiomyocytes is causally involved in the decline of cardiovascular system function. To highlight the role of senescent cardiomyocytes in the pathophysiology of age-related CVD, we first introduced that senescent cardiomyoyctes can be identified by structural changes and several senescence-associated biomarkers. We subsequently provided a comprehensive summary of existing knowledge, outlining the compelling evidence on the relationship between senescent cardiomyocytes and age-related CVD phenotypes. In addition, we discussed that the significant therapeutic potential represented by the prevention of accelerated senescent cardiomyocytes, and the current status of some existing geroprotectors in the prevention and treatment of age-related CVD. Together, the review summarized the role of cardiomyocyte senescence in CVD, and explored the molecular knowledge of senescent cardiomyocytes and their potential clinical significance in developing senescent-based therapies, thereby providing important insights into their biology and potential therapeutic exploration.
    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
    2019年冠状病毒病(COVID-19)的心血管表现包括心肌损伤,心力衰竭,和心肌炎,并与长期残疾和死亡率有关。在COVID-19患者的心肌中发现了严重急性呼吸综合征冠状病毒2(SARS-CoV-2)RNA和抗原,和人心肌细胞在细胞或类器官培养物中容易感染。虽然这些观察结果提出了心肌细胞感染可能导致COVID-19心脏后遗症的可能性,但心肌细胞感染与心肌功能障碍和病理之间的因果关系尚未确定。这里,我们通过选择性表达人血管紧张素转换酶2(hACE2),SARS-CoV-2受体,在心肌细胞中。用祖先接种Myh6-CreRosa26loxP-STOP-loxP-hACE2小鼠,非小鼠适应的SARS-CoV-2株导致病毒在心脏内复制,巨噬细胞的积累,和中度左心室(LV)收缩功能障碍。该模型中的心脏病理学是短暂的,并且通过病毒清除得以解决。单核细胞运输的阻断减少了巨噬细胞的积累,抑制左心室收缩功能障碍的发展,并促进心脏的病毒清除。这些发现建立了SARS-CoV-2心肌细胞感染的小鼠模型,该模型概括了COVID-19心脏功能障碍的特征,并表明病毒复制和由此产生的先天免疫反应都有助于心脏病理学。严重急性呼吸综合征冠状病毒2(SARS-CoV-2)感染后的心脏受累有多种方式发生,并与2019年冠状病毒病(COVID-19)患者的不良预后相关。目前尚不清楚心脏病是否是由心脏的原发性感染或对病毒的免疫反应引起的。SARS-CoV-2能够在培养皿中进入心脏的收缩细胞。然而,目前尚不清楚这种感染如何影响心脏在体内的功能。这里,我们设计了一种小鼠,其中只有心肌细胞可以感染SARS-CoV-2菌株,以研究与其他器官系统隔离的心脏感染。在我们的模型中,受感染的小鼠表现出病毒感染,功能更差,和心脏中免疫细胞的积累。免疫细胞的子集促进这种恶化的心脏功能。由于该模型显示出与患者相似的特征,这可能有助于了解作为COVID-19一部分发生的心脏病。
    Cardiovascular manifestations of coronavirus disease 2019 (COVID-19) include myocardial injury, heart failure, and myocarditis and are associated with long-term disability and mortality. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA and antigens are found in the myocardium of COVID-19 patients, and human cardiomyocytes are susceptible to infection in cell or organoid cultures. While these observations raise the possibility that cardiomyocyte infection may contribute to the cardiac sequelae of COVID-19, a causal relationship between cardiomyocyte infection and myocardial dysfunction and pathology has not been established. Here, we generated a mouse model of cardiomyocyte-restricted infection by selectively expressing human angiotensin-converting enzyme 2 (hACE2), the SARS-CoV-2 receptor, in cardiomyocytes. Inoculation of Myh6-Cre Rosa26loxP-STOP-loxP-hACE2 mice with an ancestral, non-mouse-adapted strain of SARS-CoV-2 resulted in viral replication within the heart, accumulation of macrophages, and moderate left ventricular (LV) systolic dysfunction. Cardiac pathology in this model was transient and resolved with viral clearance. Blockade of monocyte trafficking reduced macrophage accumulation, suppressed the development of LV systolic dysfunction, and promoted viral clearance in the heart. These findings establish a mouse model of SARS-CoV-2 cardiomyocyte infection that recapitulates features of cardiac dysfunctions of COVID-19 and suggests that both viral replication and resultant innate immune responses contribute to cardiac pathology.IMPORTANCEHeart involvement after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection occurs in multiple ways and is associated with worse outcomes in coronavirus disease 2019 (COVID-19) patients. It remains unclear if cardiac disease is driven by primary infection of the heart or immune response to the virus. SARS-CoV-2 is capable of entering contractile cells of the heart in a culture dish. However, it remains unclear how such infection affects the function of the heart in the body. Here, we designed a mouse in which only heart muscle cells can be infected with a SARS-CoV-2 strain to study cardiac infection in isolation from other organ systems. In our model, infected mice show viral infection, worse function, and accumulation of immune cells in the heart. A subset of immune cells facilitates such worsening heart function. As this model shows features similar to those observed in patients, it may be useful for understanding the heart disease that occurs as a part of COVID-19.
    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)是由编码肌节相关蛋白的基因变异引起的心血管病理学。然而,HCM相关基因中多种变异的临床意义尚不清楚.CRISPR/Cas9是一种核苷酸序列编辑工具,可以解开不同的生物学任务。在这项研究中,将CRISPR/Cas9突变引入健康供体的诱导多能干细胞(iPSCs),并将等基因iPSC系定向分化为心肌细胞,以评估未知意义的变异的致病性。MYH7中的p.M659I(c.1977G>A),其先前在HCM患者中发现。使用具有和不具有p.M659I(c.1977G>A)突变的两个单链供体寡核苷酸,与CRISPR/Cas9一起,产生在MYH7中的p.M659I(c.1977G>A)变体处杂合的iPSC系。没有观察到CRISPR/Cas9脱靶活性。在MYH7中具有引入的p.M659I(c.1977G>A)突变的iPSC系保留其多能状态和正常核型。与等基因对照相比,MYH7中引入p.M659I(c.1977G>A)突变的iPSC衍生的心肌细胞概括了已知的HCM特征:增大的大小,舒张钙水平升高,HCM相关基因表达的变化,破坏了能量代谢.这些发现表明该变体的致病性。
    Hypertrophic cardiomyopathy (HCM) is a cardiovascular pathology that is caused by variants in genes encoding sarcomere-associated proteins. However, the clinical significance of numerous variants in HCM-associated genes is still unknown. CRISPR/Cas9 is a tool of nucleotide sequence editing that allows for the unraveling of different biological tasks. In this study, introducing a mutation with CRISPR/Cas9 into induced pluripotent stem cells (iPSCs) of a healthy donor and the directed differentiation of the isogenic iPSC lines into cardiomyocytes were used to assess the pathogenicity of a variant of unknown significance, p.M659I (c.1977G > A) in MYH7, which was found previously in an HCM patient. Using two single-stranded donor oligonucleotides with and without the p.M659I (c.1977G > A) mutation, together with CRISPR/Cas9, an iPSC line heterozygous at the p.M659I (c.1977G > A) variant in MYH7 was generated. No CRISPR/Cas9 off-target activity was observed. The iPSC line with the introduced p.M659I (c.1977G > A) mutation in MYH7 retained its pluripotent state and normal karyotype. Compared to the isogenic control, cardiomyocytes derived from the iPSCs with the introduced p.M659I (c.1977G > A) mutation in MYH7 recapitulated known HCM features: enlarged size, elevated diastolic calcium level, changes in the expression of HCM-related genes, and disrupted energy metabolism. These findings indicate the pathogenicity of the variant.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号