Cardioprotection

心脏保护
  • 文章类型: Journal Article
    运动在一定程度上是减轻乳腺癌心脏损伤的有效途径。然而,自愿性运动(VE)是否激活心脏信号转导和转录激活因子3(STAT3)及其潜在机制尚不清楚.这项研究调查了STAT3-microRNA(miRNA)靶向蛋白轴在VE抗乳腺癌诱导的心脏损伤中的作用。与没有癌症的同窝小鼠(MMTV-PyMT-)相比,VE4周不仅改善了转基因乳腺癌雌性小鼠[小鼠乳腺肿瘤病毒-多瘤病毒中间T抗原(MMTV-PyMT)]的心脏功能,而且增加心肌STAT3酪氨酸705磷酸化。明显更明显的心脏纤维化,较小的心肌细胞大小,较低的细胞活力,与MMTV-PyMT小鼠相比,MMTV-PyMT小鼠的血清肿瘤坏死因子(TNF)-α更高,这些都是由VE改善的。然而,VE不影响肿瘤的生长。miRNA测序鉴定miR-181a-5p在VE诱导的心脏保护中上调,miR-130b-3p下调。驱动STAT3酪氨酸705突变的腺相关病毒血清型9的心肌注射消除了上述心脏保护作用。心肌STAT3被鉴定为结合pri-miR-181a(miR-181a-5p的前体)和HOX转录反义RNA(HOTAIR,海绵化miR-130b-3p)在分离的心肌细胞中。此外,在AC-16细胞中证明了miR-181a-5p靶向PTEN和miR-130b-3p靶向锌指和含BTB结构域的蛋白20(Zbtb20)。这些发现表明,VE通过激活STAT3来促进靶向PTEN的miR-181a-5p并促进HOTAIR海绵靶向Zbtb20的miR-130b-3p,从而预防乳腺癌引起的心脏损伤,从而有助于开发运动疗法的新靶标。
    Exercise is an effective way to alleviate breast cancer-induced cardiac injury to a certain extent. However, whether voluntary exercise (VE) activates cardiac signal transducer and activator of transcription 3 (STAT3) and the underlying mechanisms remain unclear. This study investigated the role of STAT3-microRNA(miRNA)-targeted protein axis in VE against breast cancer-induced cardiac injury.VE for 4 weeks not only improved cardiac function of transgenic breast cancer female mice [mouse mammary tumor virus-polyomavirus middle T antigen (MMTV-PyMT +)] compared with littermate mice with no cancer (MMTV-PyMT -), but also increased myocardial STAT3 tyrosine 705 phosphorylation. Significantly more obvious cardiac fibrosis, smaller cardiomyocyte size, lower cell viability, and higher serum tumor necrosis factor (TNF)-α were shown in MMTV-PyMT + mice compared with MMTV-PyMT - mice, which were ameliorated by VE. However, VE did not influence the tumor growth. MiRNA sequencing identified that miR-181a-5p was upregulated and miR-130b-3p was downregulated in VE induced-cardioprotection. Myocardial injection of Adeno-associated virus serotype 9 driving STAT3 tyrosine 705 mutations abolished cardioprotective effects above. Myocardial STAT3 was identified as the transcription factor binding the promoters of pri-miR-181a (the precursor of miR-181a-5p) and HOX transcript antisense RNA (HOTAIR, sponged miR-130b-3p) in isolated cardiomyocytes. Furthermore, miR-181a-5p targeting PTEN and miR-130b-3p targeting Zinc finger and BTB domain containing protein 20 (Zbtb20) were proved in AC-16 cells. These findings indicated that VE protects against breast cancer-induced cardiac injury via activating STAT3 to promote miR-181a-5p targeting PTEN and to promote HOTAIR to sponge miR-130b-3p targeting Zbtb20, helping to develop new targets in exercise therapy for breast cancer-induced cardiac injury.
    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
    新出现的证据表明,肠道微生物组通过产生多种代谢物对心血管疾病产生深远影响。使用心肌缺血再灌注(I/R)损伤的动物模型,我们发现预防性施用一种众所周知的益生菌,婴儿双歧杆菌(B.婴儿),在I/R后保留心脏收缩功能和预防不良心脏重塑方面表现出心脏保护作用,并且这些心脏保护作用由其代谢物肌苷概括。转录组学分析进一步揭示肌苷减轻I/R诱导的心脏炎症和细胞死亡。机制研究表明,肌苷抑制了促炎细胞因子的产生,并减少了树突状细胞和自然杀伤细胞的数量,通过激活腺苷A2A受体(A2AR)来实现,该受体在抑制时消除了肌苷的心脏保护作用。此外,使用C2C12成肌细胞的体外研究表明,当经历模拟心肌I/R损伤的氧-葡萄糖剥夺/复氧时,肌苷通过嘌呤补救途径作为三磷酸腺苷(ATP)生成的替代碳源来减轻细胞死亡。同样,肌苷逆转了I/R诱导的小鼠心脏ATP水平的降低。一起来看,我们的发现表明婴儿双歧杆菌或其代谢产物肌苷通过抑制心脏炎症和减轻心脏细胞死亡而对I/R发挥心脏保护作用。提示急性缺血性心脏损伤的预防性治疗选择。
    Emerging evidence has demonstrated the profound impact of the gut microbiome on cardiovascular diseases through the production of diverse metabolites. Using an animal model of myocardial ischemia-reperfusion (I/R) injury, we found that the prophylactic administration of a well-known probiotic, Bifidobacterium infantis (B. infantis), exhibited cardioprotective effects in terms of preserving cardiac contractile function and preventing adverse cardiac remodeling following I/R and that these cardioprotective effects were recapitulated by its metabolite inosine. Transcriptomic analysis further revealed that inosine mitigated I/R-induced cardiac inflammation and cell death. Mechanistic investigations elucidated that inosine suppressed the production of pro-inflammatory cytokines and reduced the numbers of dendritic cells and natural killer cells, achieved through the activation of the adenosine A2A receptor (A2AR) that when inhibited abrogated the cardioprotective effects of inosine. Additionally, in vitro studies using C2C12 myoblasts revealed that inosine attenuated cell death by serving as an alternative carbon source for adenosine triphosphate (ATP) generation through the purine salvage pathway when subjected to oxygen-glucose deprivation/reoxygenation that simulated myocardial I/R injury. Likewise, inosine reversed the I/R-induced decrease in ATP levels in mouse hearts. Taken together, our findings indicate that B. infantis or its metabolite inosine exerts cardioprotective effects against I/R by suppressing cardiac inflammation and attenuating cardiac cell death, suggesting prophylactic therapeutic options for acute ischemic cardiac injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    围手术期心肌损伤是大手术常见的并发症。许多药理学和非药理学研究已经研究了围手术期心脏保护。然而,这些方法不足以满足日益增长的心脏保护临床需求。间充质干细胞来源的外泌体(MSC-Exos)的应用是一种新型的无细胞治疗策略,并显着使患有各种疾病的患者受益。在这次审查中,我们综合分析了MSC-Exos通过调节炎症反应预防心肌梗死/损伤的应用,抑制心肌细胞凋亡和自噬,促进血管生成,介导心脏重塑。最后,我们从临床角度评估了MSC-Exos应用的治疗效果和挑战.
    Perioperative myocardial injury is a common complication caused by major surgery. Many pharmacological and nonpharmacological studies have investigated perioperative cardioprotection. However, the methods are insufficient to meet the increasing clinical needs for cardioprotection. The application of Mesenchymal Stem Cell-Derived Exosomes (MSC-Exos) is a novel cell-free therapeutic strategy and has significantly benefitted patients suffering from various diseases. In this review, we comprehensively analyzed the application of MSC-Exos to prevent myocardial infarction/injury by regulating inflammatory reactions, inhibiting cardiomyocyte apoptosis and autophagy, promoting angiogenesis, and mediating cardiac remodeling. Finally, we assessed the therapeutic effects and the challenges associated with the application of MSC-Exos from a clinical perspective.
    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
    本研究旨在探讨过氧化物酶体增殖物激活受体α(PPAR-α),一种已知的铁凋亡抑制剂,心肌缺血/再灌注损伤(MIRI)及其相关机制。建立体内和体外MIRI模型。我们的结果表明,PPAR-α的激活减少了心肌梗死的大小,维持心脏功能,降低血清肌酸激酶同工酶(CK-MB)含量,乳酸脱氢酶(LDH),和Fe2+在缺血/再灌注(I/R)处理的小鼠中。此外,H&E染色结果,DHE染色,TUNEL染色,透射电镜显示PPAR-α的激活抑制MIRI诱导的心脏组织和线粒体损伤。还发现PPAR-α的激活减弱了MIRI诱导的铁凋亡,如丙二醛的减少所示。总铁,和活性氧(ROS)。体外实验表明,细胞内丙二醛含量,总铁,LDH,活性氧(ROS),脂质ROS,氧化型谷胱甘肽二硫化物(GSSG),缺氧/复氧(A/R)处理的H9c2细胞中PPAR-α的激活使Fe2减少,而PPAR-α激活后细胞活力和GSH增加。此外,铁凋亡标志物蛋白质水平的变化进一步证实了PPAR-α活化对MIRI诱导的铁凋亡的有益作用。此外,免疫荧光和双荧光素酶报告基因分析的结果表明,PPAR-α通过与14-3-3η启动子结合实现其活性,提高其表达水平。此外,PPAR-α的心脏保护作用可被pAd/14-3-3η-shRNA或化合物C11(14-3-3η抑制剂)消除。总之,我们的结果表明,铁中毒在加重MIRI中起关键作用,PPAR-α/14-3-3η途径介导的铁凋亡和线粒体损伤可能是针对MIRI的有效治疗靶标。
    This study aimed to explore the effects of peroxisome proliferator-activated receptor α (PPAR-α), a known inhibitor of ferroptosis, in Myocardial ischemia/reperfusion injury (MIRI) and its related mechanisms. In vivo and in vitro MIRI models were established. Our results showed that activation of PPAR-α decreased the size of the myocardial infarct, maintained cardiac function, and decreased the serum contents of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and Fe2+ in ischemia/reperfusion (I/R)-treated mice. Additionally, the results of H&E staining, DHE staining, TUNEL staining, and transmission electron microscopy demonstrated that activation of PPAR-α inhibited MIRI-induced heart tissue and mitochondrial damage. It was also found that activation of PPAR-α attenuated MIRI-induced ferroptosis as shown by a reduction in malondialdehyde, total iron, and reactive oxygen species (ROS). In vitro experiments showed that intracellular contents of malondialdehyde, total iron, LDH, reactive oxygen species (ROS), lipid ROS, oxidized glutathione disulphide (GSSG), and Fe2+ were reduced by the activation of PPAR-α in H9c2 cells treated with anoxia/reoxygenation (A/R), while the cell viability and GSH were increased after PPAR-α activation. Additionally, changes in protein levels of the ferroptosis marker further confirmed the beneficial effects of PPAR-α activation on MIRI-induced ferroptosis. Moreover, the results of immunofluorescence and dual-luciferase reporter assay revealed that PPAR-α achieved its activity via binding to the 14-3-3η promoter, promoting its expression level. Moreover, the cardioprotective effects of PPAR-α could be canceled by pAd/14-3-3η-shRNA or Compound C11 (14-3-3η inhibitor). In conclusion, our results indicated that ferroptosis plays a key role in aggravating MIRI, and PPAR-α/14-3-3η pathway-mediated ferroptosis and mitochondrial injury might be an effective therapeutic target against MIRI.
    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)是一种严重的公共卫生风险,迫切需要预防和治疗工作。目前仍缺乏有效的心血管疾病预防和治疗方案,因为CVD的原因是不同的,并且可能是多因素组合的结果。线粒体自噬是细胞选择性自噬的一种形式,越来越多的证据表明线粒体自噬参与心脏保护过程。最近,许多研究表明,含FUN14结构域的蛋白1(FUNDC1)水平和磷酸化状态与许多疾病,包括心脏病.这里,我们回顾了FUNDC1的结构和功能及其介导的线粒体自噬的途径,并表明线粒体自噬可以通过Ser13和Tyr18位点的去磷酸化被有效激活,FUNDC1中Ser17位点的磷酸化和Lys119位点的泛素化。通过有效激活或抑制过度的线粒体自噬,线粒体质量可以得到有效控制。主要原因是,一方面,避免了线粒体的不当清除和受损线粒体的积累,另一方面,避免过度的线粒体自噬导致细胞凋亡,都是为了保护心脏.此外,我们探讨了FUNDC1介导的线粒体自噬参与运动预处理(EP)保护心脏的可能机制.最后,我们还指出了FUNDC1及其介导的线粒体自噬中尚未解决的问题,并给出了可能需要进一步研究的方向.
    Cardiovascular disease (CVD) is a serious public health risk, and prevention and treatment efforts are urgently needed. Effective preventive and therapeutic programs for cardiovascular disease are still lacking, as the causes of CVD are varied and may be the result of a multifactorial combination. Mitophagy is a form of cell-selective autophagy, and there is increasing evidence that mitophagy is involved in cardioprotective processes. Recently, many studies have shown that FUN14 domain-containing protein 1 (FUNDC1) levels and phosphorylation status are highly associated with many diseases, including heart disease. Here, we review the structure and functions of FUNDC1 and the path-ways of its mediated mitophagy, and show that mitophagy can be effectively activated by dephosphorylation of Ser13 and Tyr18 sites, phosphorylation of Ser17 site and ubiquitination of Lys119 site in FUNDC1. By effectively activating or inhibiting excessive mitophagy, the quality of mitochondria can be effectively controlled. The main reason is that, on the one hand, improper clearance of mitochondria and accumulation of damaged mitochondria are avoided, and on the other hand, excessive mitophagy causing apoptosis is avoided, both serving to protect the heart. In addition, we explore the possible mechanisms by which FUNDC1-mediated mitophagy is involved in exercise preconditioning (EP) for cardioprotection. Finally, we also point out unresolved issues in FUNDC1 and its mediated mitophagy and give directions where further research may be needed.
    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
    人参被认为能够补气益气和增强体力,三七可以缓解血瘀和疼痛。人参和三七经常用于治疗由心脏血管阻塞引起的缺血性心脏病。线粒体功能障碍常与线粒体形态异常并存,线粒体可塑性和动力学在心血管疾病中起关键作用。在这项研究中,将原代新生大鼠心肌细胞暴露于4小时的缺氧(H),然后进行2小时的复氧(R)。MitoTracker深红和Hoechst33342用于标记线粒体和细胞核,分别。然后使用ImageXpress微共焦获取荧光图像。使用ImageProPlus进行自动图像处理和参数提取/计算。随后,选择代表性参数作为评估线粒体形态和功能改变的指标.根据UPLC-Triple-TOF-MS结果和线粒体形态参数筛选并鉴定了人参和三七的活性化合物。研究结果表明,RS-2,RS-4,SQ-1和SQ-4显着增加了三个关键形态参数的值。包括线粒体长度,分支,和面积,这可能有助于挽救因H/R损伤而受损的心肌细胞的形态特征。在这两种草药的活性成分中,20(R)-人参皂苷Rg_3,人参皂苷Re,和绞股蓝皂甙对心肌细胞线粒体的保护作用最强。具体来说,20(R)-人参皂苷Rg_3可能上调视神经萎缩1(OPA1)和肌萎缩素2(MFN2)的表达,人参皂苷Re和绞股蓝皂苷XVII可以选择性上调OPA1的表达。总的来说,它们促进线粒体膜融合,减轻线粒体损伤,从而对心肌细胞发挥保护作用。本研究为从人参和三七中发现心肌缺血再灌注损伤的新型治疗药物提供了实验支持,并为大规模筛选具有心脏保护作用的中药生物活性化合物提供了新的方法。
    Panax ginseng is reputed to be capable of replenishing healthy Qi and bolstering physical strength, and P. notoginseng can resolve blood stasis and alleviate pain. P. ginseng and P. notoginseng are frequently employed to treat ischemic heart diseases caused by blockages in the heart vessels. Mitochondrial dysfunction often coexists with abnormal mitochondrial morphology, and mitochondrial plasticity and dynamics play key roles in cardiovascular diseases. In this study, primary neonatal rat cardiomyocytes were exposed to 4 hours of hypoxia(H) followed by 2 hours of reoxygenation(R). MitoTracker Deep Red and Hoechst 33342 were used to label mitochondria and nuclei, respectively. Fluorescence images were then acquired using ImageXpress Micro Confocal. Automated image processing and parameter extraction/calculation were carried out using ImagePro Plus. Subsequently, representative parameters were selected as indicators to assess alterations in mitochondrial morphology and function. The active compounds of P. ginseng and P. notoginseng were screened out and identified based on the UPLC-Triple-TOF-MS results and mitochondrial morphometric parameters. The findings demonstrated that RS-2, RS-4, SQ-1, and SQ-4 significantly increased the values of three key morphometric parameters, including mitochondrial length, branching, and area, which might contribute to rescuing morphological features of myocardial cells damaged by H/R injury. Among the active components of the two medicinal herbs, 20(R)-ginsenoside Rg_3, ginsenoside Re, and gypenoside ⅩⅦ exhibited the strongest protective effects on mitochondria in cardiomyocytes. Specifically, 20(R)-ginsenoside Rg_3 might upregulate expression of optic atrophy 1(OPA1) and mitofusin 2(MFN2), and ginsenoside Re and gypenoside ⅩⅦ might selectively upregulate OPA1 expression. Collectively, they promoted mitochondrial membrane fusion and mitigated mitochondrial damage, thereby exerting protective effects on cardiomyocytes. This study provides experimental support for the discovery of novel therapeutic agents for myocardial ischemia-reperfusion injury from P. ginseng and P. notoginseng and offers a novel approach for large-scale screening of bioactive compounds with cardioprotective effects from traditional Chinese medicines.
    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:糖尿病心肌病(DCM)是长期慢性糖尿病的重要并发症,可导致心肌肥厚,心肌纤维化,和心力衰竭。越来越多的证据表明DCM与焦亡有关,一种与炎症相关的程序性细胞死亡。生长分化因子11(GDF11)是转化生长因子β超家族的一员,调节氧化应激,炎症,和细胞存活以减轻心肌肥大,心肌梗塞,和血管损伤。然而,GDF11在调节DCM细胞焦凋亡中的作用仍有待阐明。本研究旨在探讨GDF11在调节DCM细胞焦凋亡中的作用及相关机制。
    结果:给小鼠注射链脲佐菌素(STZ)以诱导糖尿病模型。H9c2心肌细胞在高糖(50mM)中培养,建立糖尿病的体外模型。C57BL/6J小鼠经尾静脉内预先注射腺相关病毒9(AAV9)以特异性过表达心肌GDF11。GDF11减弱了高糖治疗后H9c2心肌细胞的焦亡。在糖尿病小鼠中,GDF11减轻心肌细胞焦亡,减少心肌纤维化,和改善心脏功能。机械上,GDF11通过防止炎性体激活来抑制焦亡。GDF11通过与含有CARD(ASC)的凋亡相关斑点样蛋白特异性结合并阻止炎性小体的组装和激活来实现这一目标。此外,GDF11的表达受过氧化物酶体增殖物激活受体α(PPARα)调控。
    结论:这些发现表明GDF11可以通过减轻焦凋亡来治疗糖尿病性心肌病,并揭示了PPARα-GDF11-ASC通路在DCM中的作用,为心脏保护新策略提供思路。
    BACKGROUND: Diabetic cardiomyopathy (DCM) is a crucial complication of long-term chronic diabetes that can lead to myocardial hypertrophy, myocardial fibrosis, and heart failure. There is increasing evidence that DCM is associated with pyroptosis, a form of inflammation-related programmed cell death. Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor β superfamily, which regulates oxidative stress, inflammation, and cell survival to mitigate myocardial hypertrophy, myocardial infarction, and vascular injury. However, the role of GDF11 in regulating pyroptosis in DCM remains to be elucidated. This research aims to investigate the role of GDF11 in regulating pyroptosis in DCM and the related mechanism.
    RESULTS: Mice were injected with streptozotocin (STZ) to induce a diabetes model. H9c2 cardiomyocytes were cultured in high glucose (50 mM) to establish an in vitro model of diabetes. C57BL/6J mice were preinjected with adeno-associated virus 9 (AAV9) intravenously via the tail vein to specifically overexpress myocardial GDF11. GDF11 attenuated pyroptosis in H9c2 cardiomyocytes after high-glucose treatment. In diabetic mice, GDF11 alleviated cardiomyocyte pyroptosis, reduced myocardial fibrosis, and improved cardiac function. Mechanistically, GDF11 inhibited pyroptosis by preventing inflammasome activation. GDF11 achieved this by specifically binding to apoptosis-associated speck-like protein containing a CARD (ASC) and preventing the assembly and activation of the inflammasome. Additionally, the expression of GDF11 during pyroptosis was regulated by peroxisome proliferator-activated receptor α (PPARα).
    CONCLUSIONS: These findings demonstrate that GDF11 can treat diabetic cardiomyopathy by alleviating pyroptosis and reveal the role of the PPARα-GDF11-ASC pathway in DCM, providing ideas for new strategies for cardioprotection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:本研究旨在探讨电针对脂多糖(LPS)诱导的心肌病的心肌保护作用及其分子机制。
    结果:在腹腔注射LPS前30分钟进行EA预处理。心电图观察EA+LPS组小鼠心功能变化,超声心动图,酶联免疫吸附试验(ELISA)并与LPS组比拟。结果表明,电针预处理显著提高了脓毒症小鼠的存活率,减轻了内毒素血症的严重程度,并表现出显著的心脏保护作用。这些影响的特征是心电图上ST段抬高的减少,超声心动图的射血分数(EF)和缩短分数(FS)增加,血清肌钙蛋白I(cTn-I)水平的表达降低。提取EA预处理后获得的血清外泌体,并给予脓毒症小鼠,揭示了EA衍生的外泌体的显着心脏保护作用。此外,小鼠循环外泌体的拮抗作用显著抑制了EA预处理赋予的心脏保护作用.使用定量逆转录-聚合酶链反应(qRT-PCR)对血清外来体的分析揭示了在EA预处理后miR-381表达的显著上调。通过血清型9腺相关病毒(AAV9)介导的基因传递抑制或过表达miR-381表明miR-381的过表达具有心脏保护作用。而miR-381的抑制显著减弱了EA预处理赋予的心脏保护作用。
    结论:我们的研究发现揭示了一种新的内源性心脏保护机制,其中源自EA预处理的循环外泌体通过miR-381减轻LPS诱导的心脏功能障碍。
    OBJECTIVE: This study aims to investigate the cardiac protective effects and molecular mechanisms of electroacupuncture (EA) pre-treatment in lipopolysaccharide (LPS)-Induced Cardiomyopathy.
    RESULTS: Pre-treatment with EA was performed 30 min before intraperitoneal injection of LPS. Cardiac function changes in mice of the EA + LPS group were observed using electrocardiography, echocardiography, and enzyme linked immunosorbent assay (ELISA) and compared with the LPS group. The results demonstrated that EA pre-treatment significantly improved the survival rate of septic mice, alleviated the severity of endotoxemia, and exhibited notable cardiac protective effects. These effects were characterized by a reduction in ST-segment elevation on electrocardiography, an increase in ejection fraction (EF) and fraction shortening (FS) on echocardiography and a decrease in the expression of serum cardiac troponin I (cTn-I) levels. Serum exosomes obtained after EA pre-treatment were extracted and administered to septic mice, revealing significant cardiac protective effects of EA-derived exosomes. Furthermore, the antagonism of circulating exosomes in mice markedly suppressed the cardiac protective effects conferred by EA pre-treatment. Analysis of serum exosomes using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) revealed a significant upregulation of miR-381 expression after EA pre-treatment. Inhibition or overexpression of miR-381 through serotype 9 adeno-associated virus (AAV9)-mediated gene delivery demonstrated that overexpression of miR-381 exerted a cardiac protective effect, while inhibition of miR-381 significantly attenuated the cardiac protective effects conferred by EA pre-treatment.
    CONCLUSIONS: Our research findings have revealed a novel endogenous cardiac protection mechanism, wherein circulating exosomes derived from EA pre-treatment mitigate LPS-induced cardiac dysfunction via miR-381.
    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
    目的:该研究旨在评估右美托咪定(Dex)预处理对体外循环下心脏瓣膜置换术患者的影响。
    方法:对于Dex组的患者(n=52),麻醉诱导前给予Dex0.5μg/kg,然后在主动脉闭塞前泵入0.5μg/kg/h。对于对照组的患者(n=52),给予0.125ml/kg生理盐水代替Dex。
    结果:Dex组患者首次使用丙泊酚的时间比对照组长(P=0.003)。Dex组需要的丙泊酚总用量少于对照组(P=0.0001)。心肌肌钙蛋白I(cTnI)水平,肌酸激酶同工酶MB(CK-MB),丙二醛(MDA),肿瘤坏死因子-α(TNF-α)在术后T4、8h(T5)时低于对照组,术后24h(T6)(P<0.01)。Dex组机械通气所需时间少于对照组(P=0.003)。
    结论:研究表明,0.50µg/kgDex预处理可以减少异丙酚的使用和机械通气的持续时间,并在心脏瓣膜置换术期间赋予心肌保护而不增加不良事件。
    OBJECTIVE: The study aims to assess the effects of dexmedetomidine (Dex) pretreatment on patients during cardiac valve replacement under cardiopulmonary bypass.
    METHODS: For patients in the Dex group (n = 52), 0.5 μg/kg Dex was given before anesthesia induction, followed by 0.5 μg/kg/h pumping injection before aortic occlusion. For patients in the control group (n = 52), 0.125 ml/kg normal saline was given instead of Dex.
    RESULTS: The patients in the Dex group had longer time to first dose of rescue propofol than the control group (P = 0.003). The Dex group required less total dosage of propofol than the control group (P = 0.0001). The levels of cardiac troponin I (cTnI), creatine kinase isoenzyme MB (CK-MB), malondialdehyde (MDA), and tumor necrosis factor-α (TNF-α) were lower in the Dex group than the control group at T4, 8 h after the operation (T5), and 24 h after the operation (T6) (P <0.01). The Dex group required less time for mechanical ventilation than the control group (P = 0.003).
    CONCLUSIONS: The study suggests that 0.50 µg/kg Dex pretreatment could reduce propofol use and the duration of mechanical ventilation, and confer myocardial protection without increased adverse events during cardiac valve replacement.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号