cardiomyocyte apoptosis

心肌细胞凋亡
  • 文章类型: Journal Article
    背景:葡萄糖波动可能参与心肌细胞凋亡的病理生理过程,但确切的机制仍然难以捉摸。本研究旨在探讨葡萄糖波动诱导心肌细胞凋亡的相关机制。
    方法:通过注射链脲佐菌素建立的糖尿病大鼠随机分为五组:对照糖尿病(CD)组,不受控制的糖尿病(UD)组,血糖波动糖尿病(GFD)组,GFD组大鼠注射0.9%氯化钠(NaCl)(GFD+NaCl),GFD组大鼠注射N-乙酰-L-半胱氨酸(NAC)(GFD+NAC)。十二周后,检测心功能及细胞凋亡相关蛋白表达。进行蛋白质组学分析以进一步分析CD和GFD的差异蛋白表达模式。
    结果:GFD组左心室射血分数水平和缩短率水平降低,与CD和UD组相比。DAB-TUNEL检测的阳性细胞在GFD组中增加,与CD组相比。Bcl-2的表达降低,但是Bax的表达,裂解的caspase-3和裂解的caspase-9响应于葡萄糖波动而增加。与CD相比,GFD组有527个上调蛋白和152个下调蛋白.Txnip是与氧化应激反应相关的差异表达蛋白之一。Txnip表达在GFD组中增加,而Akt磷酸化水平降低。当血糖波动时,Txnip和Akt之间的相互作用增强。此外,应用NAC部分逆转葡萄糖波动诱导的心肌细胞凋亡。
    结论:葡萄糖波动通过上调Txnip表达和增强Txnip-Akt相互作用导致心肌细胞凋亡。
    BACKGROUND: Glucose fluctuations may be involved in the pathophysiological process of cardiomyocyte apoptosis, but the exact mechanism remains elusive. This study focused on exploring the mechanisms related to glucose fluctuation-induced cardiomyocyte apoptosis.
    METHODS: Diabetic rats established via an injection of streptozotocin were randomized to five groups: the controlled diabetic (CD) group, the uncontrolled diabetic (UD) group, the glucose fluctuated diabetic (GFD) group, the GFD group rats with the injection of 0.9% sodium chloride (NaCl) (GFD + NaCl) and the GFD group rats with the injection of N-acetyl-L-cysteine (NAC) (GFD + NAC). Twelve weeks later, cardiac function and apoptosis related protein expressions were tested. Proteomic analysis was performed to further analyze the differential protein expression pattern of CD and GFD.
    RESULTS: The left ventricular ejection fraction levels and fractional shortening levels were decreased in the GFD group, compared with those in the CD and UD groups. Positive cells tested by DAB-TUNEL were increased in the GFD group, compared with those in the CD group. The expression of Bcl-2 was decreased, but the expressions of Bax, cleaved caspase-3 and cleaved caspase-9 were increased in response to glucose fluctuations. Compared with CD, there were 527 upregulated and 152 downregulated proteins in GFD group. Txnip was one of the differentially expressed proteins related to oxidative stress response. The Txnip expression was increased in the GFD group, while the Akt phosphorylation level was decreased. The interaction between Txnip and Akt was enhanced when blood glucose fluctuated. Moreover, the application of NAC partially reversed glucose fluctuations-induced cardiomyocyte apoptosis.
    CONCLUSIONS: Glucose fluctuations lead to cardiomyocyte apoptosis by up-regulating Txnip expression and enhancing Txnip-Akt interaction.
    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
    射血分数保留的心力衰竭(HFpEF)是一种临床综合征,其特征是由左心室舒张功能障碍和充盈压升高引起的肺和全身充血。目前,然而,没有证据表明HFpEF的有效药物治疗.在这项研究中,我们旨在通过建立高脂饮食(HFD)L-NAME诱导的小鼠模型,研究龙胆总黄吨酮(TXG)对HFpEF的治疗作用。超声心动图用于评估TXG对HFpEF小鼠心功能的影响。苏木精和伊红染色,小麦胚芽凝集素染色,用Masson三色染色观察TXG治疗后的组织病理学变化。结果表明,TXG通过降低心肌肥厚相关基因的表达减轻HFpEF,纤维化和凋亡。此外,TXG通过抑制凋亡相关蛋白的表达改善心肌细胞凋亡。机制研究表明,TXG可以激活需要肌醇的酶1α(IRE1α)/X-box结合蛋白1(Xbp1s)信号通路,但是使用IRE1α抑制剂STF083010或siRNA-IRE1α敲低IRE1α会损害TXG改善HFpEF模型中心脏重塑的能力。总之,TXG减轻心肌肥厚,通过激活IRE1α/Xbp1s信号通路,提示其对HFpEF患者的潜在有益作用。
    Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome characterized by pulmonary and systemic congestion resulting from left ventricular diastolic dysfunction and increased filling pressure. Currently, however, there is no evidence on effective pharmacotherapy for HFpEF. In this study, we aimed to investigate the therapeutic effect of total xanthones extracted from Gentianella acuta (TXG) on HFpEF by establishing an high-fat diet (HFD) + L-NAME-induced mouse model. Echocardiography was employed to assess the impact of TXG on the cardiac function in HFpEF mice. Haematoxylin and eosin staining, wheat germ agglutinin staining, and Masson\'s trichrome staining were utilized to observe the histopathological changes following TXG treatment. The results demonstrated that TXG alleviated HFpEF by reducing the expressions of genes associated with myocardial hypertrophy, fibrosis and apoptosis. Furthermore, TXG improved cardiomyocyte apoptosis by inhibiting the expression of apoptosis-related proteins. Mechanistic investigations revealed that TXG could activate the inositol-requiring enzyme 1α (IRE1α)/X-box-binding protein 1 (Xbp1s) signalling pathway, but the knockdown of IRE1α using the IRE1α inhibitor STF083010 or siRNA-IRE1α impaired the ability of TXG to ameliorate cardiac remodelling in HFpEF models. In conclusion, TXG alleviates myocardial hypertrophy, fibrosis and apoptosis through the activation of the IRE1α/Xbp1s signalling pathway, suggesting its potential beneficial effects on HFpEF patients.
    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
    气血双补方(QSP)治疗慢性心力衰竭(CHF)疗效显著。经中药炮制后的QSP(PQSP)可明显改善CHF的治疗效果。本研究阐明了中药炮制后QSP在体外和体内治疗CHF的潜在功效增强机制。缺氧/复氧诱导大鼠心肌细胞H9c2细胞损伤,以模拟体外CHF状态。60只Sprague-Dawley大鼠腹腔注射阿霉素(累计剂量15mg/kg)建立CHF模型。在血清和细胞上清液中进行生化检查,分别。在CHF模型大鼠中评估心功能和组织病理学变化。采用Westernblot和RT-PCR检测ERK1/2、Bcl-2、Bax和Caspase-3蛋白和mRNA水平,分别。以上结果均为低剂量粗QSP治疗组(L-CQSP),高剂量CQSP治疗组(H-CQSP),低剂量PQSP治疗组(L-PQSP),比较大剂量PQSP治疗组(H-PQSP),以系统地探讨中药处理与PQSP治疗CHF的疗效增强之间的相关性。与模型组相比,L-CQSP组在第8周时显示出心脏功能的显着改善,而心肌细胞凋亡和纤维化无显著改善。H-CQSP,L-PQSP和H-PQSP在损伤的H9c2心肌细胞和CHF模型大鼠中发挥有益的治疗作用。L-PQSP和H-PQSP显著提高细胞活力和SOD活性,降低了LDH的活性,MDA和NO,与相同剂量的CQSP相比,ERK1/2和Bcl-2的表达上调,Bax和Caspase-3的表达下调。中药炮制后PQSP治疗CHF的疗效增强机制与ERK/Bcl-2/Bax/Caspases-3信号通路的调节直接相关。
    Qixue Shuangbu prescription (QSP) has been used for the treatment of chronic heart failure (CHF) with remarkable curative effect. Processed QSP (PQSP) could significantly improve the treatment of CHF after traditional Chinese medicine (TCM) processing. This study elucidated the underlying efficacy enhancement mechanism of QSP after TCM processing for treating CHF in vitro and in vivo. The injury of rat cardiomyoblast H9c2 cells was induced by anoxia/reoxygenation to mimic CHF state in vitro. Sixty Sprague-Dawley rats were used to established CHF model by intraperitoneally injecting doxorubicin (the accumulative dose 15 mg/kg). Biochemical examinations were performed in serum and cellular supernatant, respectively. Cardiac functions and histopathological changes were evaluated in CHF model rats. The protein and mRNA levels of ERK1/2, Bcl-2, Bax and Caspase-3 were evaluated by Western blot and RT-PCR, respectively. All above results of low dose crude QSP-treated group (L-CQSP), high dose CQSP-treated group (H-CQSP), low dose PQSP-treated group (L-PQSP), high dose PQSP-treated group (H-PQSP) were compared to systematically explore correlations between TCM processing and the efficacy enhancement for treating CHF of PQSP. Compared with the model group, the L-CQSP group showed significant improvement in cardiac function at 8th weeks, while no significant improvement in cardiomyocyte apoptosis and fibrosis. Both H-CQSP, L-PQSP and H-PQSP exerted beneficial therapeutic effects in injured H9c2 cardiomyocytes and CHF model rats. L-PQSP and H-PQSP significantly increased cell viability and the activity of SOD, decreased the activities of LDH, MDA and NO, up-regulated the expression of ERK1/2 and Bcl-2, down-regulated the expression of Bax and Caspase-3 compared to the same dosage of CQSP. The efficacy enhancement mechanism of PQSP after TCM processing for treating CHF was directly related to the regulation of ERK/Bcl-2/Bax/Caspases-3 signaling pathway.
    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
    多柔比星(DOX)的临床应用受到DOX心脏毒性的严重阻碍。关于DOX心脏毒性,包括DNA损伤和DNA损伤反应(DDR),尽管所涉及的机制仍有待阐明。这项研究评估了TBC结构域家族成员15(TBC1D15)在DOX心脏毒性中的潜在作用。在DOX攻击后1周或4周进行心脏评估之前,将他莫昔芬诱导的心脏特异性Tbc1d15敲除(Tbc1d15CKO)或Tbc1d15敲入(Tbc1d15CKI)雄性小鼠用单剂量DOX攻击。将编码TBC1D15或含有靶向Tbc1d15的shRNA的腺病毒用于分离的原代小鼠心肌细胞中的Tbc1d15过表达或敲低。我们的结果表明,DOX引起TBC1D15的上调,心肌功能受损和明显的死亡率,其效应通过Tbc1d15缺失和Tbc1d15过表达得到改善和增强,分别。DOX明显诱发凋亡性细胞死亡,Tbc1d15基因敲除和过表达减轻和加剧了这种效应,分别。同时,DOX激起线粒体膜电位塌陷,氧化应激和DNA损伤,Tbc1d15敲低和过表达减轻和加剧了其影响,分别。进一步的审查显示,TBC1D15促进了主要DDR元件DNA依赖性蛋白激酶催化亚基(DNA-PKcs)的胞浆积累。液相色谱-串联质谱和共免疫沉淀表明TBC1D15与TBC1D15的片段594-624处的DNA-PKcs之间的相互作用。此外,TBC1D15突变体的过表达(Δ594-624,片段594-624的缺失)未能引起DOX诱导的DNA-PKcs的细胞溶质保留的加重,TBC1D15野生型的DNA毁伤和心肌细胞凋亡。然而,Tbc1d15缺失改善DOX诱导的心肌细胞收缩异常,凋亡,线粒体异常,DNA损伤和胞质DNA-PKcs积累,它们被DNA-PKcs抑制或ATM激活所抵消。一起来看,我们的发现表明TBC1D15在DOX诱导的DNA损伤中的关键作用,线粒体损伤,和凋亡可能通过与DNA-PKcs结合,从而保持其细胞溶质保留,在DOX诱导的心脏毒性中加重心脏收缩功能障碍的途径。
    Clinical application of doxorubicin (DOX) is heavily hindered by DOX cardiotoxicity. Several theories were postulated for DOX cardiotoxicity including DNA damage and DNA damage response (DDR), although the mechanism(s) involved remains to be elucidated. This study evaluated the potential role of TBC domain family member 15 (TBC1D15) in DOX cardiotoxicity. Tamoxifen-induced cardiac-specific Tbc1d15 knockout (Tbc1d15CKO) or Tbc1d15 knockin (Tbc1d15CKI) male mice were challenged with a single dose of DOX prior to cardiac assessment 1 week or 4 weeks following DOX challenge. Adenoviruses encoding TBC1D15 or containing shRNA targeting Tbc1d15 were used for Tbc1d15 overexpression or knockdown in isolated primary mouse cardiomyocytes. Our results revealed that DOX evoked upregulation of TBC1D15 with compromised myocardial function and overt mortality, the effects of which were ameliorated and accentuated by Tbc1d15 deletion and Tbc1d15 overexpression, respectively. DOX overtly evoked apoptotic cell death, the effect of which was alleviated and exacerbated by Tbc1d15 knockout and overexpression, respectively. Meanwhile, DOX provoked mitochondrial membrane potential collapse, oxidative stress and DNA damage, the effects of which were mitigated and exacerbated by Tbc1d15 knockdown and overexpression, respectively. Further scrutiny revealed that TBC1D15 fostered cytosolic accumulation of the cardinal DDR element DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Liquid chromatography-tandem mass spectrometry and co-immunoprecipitation denoted an interaction between TBC1D15 and DNA-PKcs at the segment 594-624 of TBC1D15. Moreover, overexpression of TBC1D15 mutant (∆594-624, deletion of segment 594-624) failed to elicit accentuation of DOX-induced cytosolic retention of DNA-PKcs, DNA damage and cardiomyocyte apoptosis by TBC1D15 wild type. However, Tbc1d15 deletion ameliorated DOX-induced cardiomyocyte contractile anomalies, apoptosis, mitochondrial anomalies, DNA damage and cytosolic DNA-PKcs accumulation, which were canceled off by DNA-PKcs inhibition or ATM activation. Taken together, our findings denoted a pivotal role for TBC1D15 in DOX-induced DNA damage, mitochondrial injury, and apoptosis possibly through binding with DNA-PKcs and thus gate-keeping its cytosolic retention, a route to accentuation of cardiac contractile dysfunction in DOX-induced cardiotoxicity.
    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:心力衰竭(HF)是以心脏功能受损为特征的全球健康问题。心脏重塑和细胞死亡有助于HF的发展。尽管已经使用了地高辛和血管紧张素受体阻滞剂药物等治疗方法,它们在降低死亡率方面的有效性尚不确定。研究人员正在探索使用脂肪间充质干细胞(ADMSC)外泌体(Exos)作为HF的潜在疗法。这些囊泡,由细胞分泌,可能有助于组织修复和调节炎症和免疫反应。然而,需要进一步研究以了解这些囊泡在HF治疗中的具体作用.
    目的:探讨AMSC细胞外囊泡治疗HF的作用机制。
    方法:发现ADMSCs的外源性表面标记,并培养ADMSCs。
    结果:表面标志物的鉴定表明,脂肪干细胞(ADSCs)的表面标志物CD44和CD29表达良好,而ADSCs的表面标志物CD45和CD34为阴性,所以培养的细胞被认为是ADSCs。蛋白质印迹检测到Exo表面标记蛋白,表达CD63蛋白,但不表达calnexin蛋白,表明ADSC衍生的Exos已成功提取。
    结论:从脂肪组织中分泌MSCs可以增加ATP水平,阻断心肌细胞凋亡,并增强易患HF的动物的心脏功能。Bax的抑制作用,caspase-3和p53蛋白的表达可能与这一过程有关。
    BACKGROUND: Heart failure (HF) is a global health problem characterized by impaired heart function. Cardiac remodeling and cell death contribute to the development of HF. Although treatments such as digoxin and angiotensin receptor blocker drugs have been used, their effectiveness in reducing mortality is uncertain. Researchers are exploring the use of adipose-derived mesenchymal stem cell (ADMSC) exosomes (Exos) as a potential therapy for HF. These vesicles, secreted by cells, may aid in tissue repair and regulation of inflammation and immune responses. However, further investigation is needed to understand the specific role of these vesicles in HF treatment.
    OBJECTIVE: To investigate the mechanism of extracellular vesicles produced by ADMSC s in the treatment of HF.
    METHODS: Exogenous surface markers of ADMSCs were found, and ADMSCs were cultured.
    RESULTS: The identification of surface markers showed that the surface markers CD44 and CD29 of adipose-derived stem cells (ADSCs) were well expressed, while the surface markers CD45 and CD34 of ADSCs were negative, so the cultured cells were considered ADSCs. Western blotting detected the Exo surface marker protein, which expressed CD63 protein but did not express calnexin protein, indicating that ADSC-derived Exos were successfully extracted.
    CONCLUSIONS: The secretion of MSCs from adipose tissue can increase ATP levels, block cardiomyocyte apoptosis, and enhance the heart function of animals susceptible to HF. The inhibition of Bax, caspase-3 and p53 protein expression may be related to this process.
    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
    缺血性心脏病的患病率已达到全球流行水平。早期血运重建是目前缺血性心脏病最有效的治疗方法,但矛盾的是会引起心肌缺血/再灌注(MI/R)损伤。心肌炎症反应和氧化应激主要参与MI/R损伤的病理过程。低强度脉冲超声(LIPUS)已被证明可以通过保护许多疾病的炎症反应和氧化应激来减少细胞损伤。包括心血管疾病,但很少发生MI/R损伤。
    本研究旨在阐明LIPUS是否通过减轻炎症反应和氧化应激来减轻MI/R损伤。同时,我们还试图确认哪种强度的LIPUS可能更适合改善MI/R损伤,以及阐明信令机制。分别在SpragueDawley大鼠和人多能干细胞来源的心肌细胞(hPSC-CMs)中诱导MI/R和模拟缺血/再灌注(SI/R)。LIPUS治疗,生化测量,细胞死亡测定,心脏氧化应激和炎症反应的估计,根据方案进行蛋白质印迹和蛋白质检测.
    在我们的研究中,在体内和体外,在常氧条件下,LIPUS为0.1W/cm2(LIPUS0.1)和0.5W/cm2(LIPUS0.5)在心肌细胞中没有显着差异。在缺氧条件下,MI/R损伤,炎症反应,LIPUS0.5部分改善了氧化应激,但体内和体外2.5W/cm2的LIPUS(LIPUS2.5)显着加重。在MI/R损伤的心肌细胞中,凋亡信号调节激酶1(ASK1)/c-JunN末端激酶(JNK)途径的激活在体内和体外均部分纠正了LIPUS0.5。
    我们的研究首次证明不同强度的LIPUS通过调节心脏炎症反应和氧化应激对MI/R损伤有不同的影响。对ASK1/JNK途径的调节是LIPUS0.5发挥心脏保护作用的信号机制。LIPUS0.5有望用于预防MI/R损伤的临床翻译。这对于患有MI/R损伤的患者将是极大的福利。
    The prevalence of ischemic heart disease has reached pandemic levels worldwide. Early revascularization is currently the most effective therapy for ischemic heart diseases but paradoxically induces myocardial ischemia/reperfusion (MI/R) injury. Cardiac inflammatory reaction and oxidative stress are primarily involved in the pathology of MI/R injury. Low-intensity pulsed ultrasound (LIPUS) has been demonstrated to reduce cell injury by protecting against inflammatory reaction and oxidative stress in many diseases, including cardiovascular diseases, but rarely on MI/R injury.
    This study was designed to clarify whether LIPUS alleviates MI/R injury by alleviating inflammatory reaction and oxidative stress. Simultaneously, we have also tried to confirm which intensity of the LIPUS might be more suitable to ameliorate the MI/R injury, as well as to clarify the signaling mechanisms. MI/R and simulated ischemia/reperfusion (SI/R) were respectively induced in Sprague Dawley rats and human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). LIPUS treatment, biochemical measurements, cell death assay, estimation of cardiac oxidative stress and inflammatory reaction, and protein detections by western blotting were performed according to the protocol.
    In our study, both in vivo and in vitro, LIPUS of 0.1 W/cm2 (LIPUS0.1) and 0.5 W/cm2 (LIPUS0.5) make no significant difference in the cardiomyocytes under normoxic condition. Under the hypoxic condition, MI/R injury, inflammatory reaction, and oxidative stress were partially ameliorated by LIPUS0.5 but were significantly aggravated by LIPUS of 2.5 W/cm2 (LIPUS2.5) both in vivo and in vitro. The activation of the apoptosis signal-regulating kinase 1 (ASK1)/c-Jun N-terminal kinase (JNK) pathway in cardiomyocytes with MI/R injury was partly rectified LIPUS0.5 both in vivo and in vitro.
    Our study firstly demonstrated that LIPUS of different intensities differently affects MI/R injury by regulating cardiac inflammatory reaction and oxidative stress. Modulations on the ASK1/JNK pathway are the signaling mechanism by which LIPUS0.5 exerts cardioprotective effects. LIPUS0.5 is promising for clinical translation in protecting against MI/R injury. This will be great welfare for patients suffering from MI/R 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: Journal Article
    目的:分子氢在心脏疾病中具有保护作用。我们先前的研究表明,富氢盐水(HRS)可以选择性清除自由基并减轻心肌缺血/再灌注(I/R)损伤中的炎症反应。但是潜在的机制尚未完全阐明。
    结果:用成年(10周)C57BL/6雄性小鼠和新生大鼠心肌细胞建立I/R和缺氧/复氧(H/R)损伤模型。用HRS处理I/R和H/R模型以对心脏控制功能的机制进行分类。在这项研究中,我们发现miR-124-3p在I/R和H/R模型中均显著降低,而HRS预处理部分改善。HRS治疗还减轻了缺血诱导的凋亡性细胞死亡,并增加了I/R过程中的细胞活力。而miR-124-3p的沉默表达消除了这种保护作用。此外,我们确定calpain1是miR-124-3p的直接靶标,miR-124-3的上调产生了calpain1的活性和表达。还发现,与HRS组相比,钙蛋白酶1的过表达增加了caspase-3的活性,促进cleaved-caspase3和Bax蛋白表达,和相应地降低Bcl-2,进一步降低细胞活力。这些结果表明,在H/R模型中,calpain1的过表达减弱了HRS对心肌细胞的保护作用。
    结论:本研究表明HRS对I/R损伤具有保护作用,这可能与miR-124-3p-calpain1信号通路有关。
    Molecular hydrogen has been exhibited a protective function in heart diseases. Our previous study demonstrated that hydrogen-rich saline (HRS) could scavenge free radicals selectively and alleviate the inflammatory response in the myocardial ischaemia/reperfusion (I/R) injury, but the underlying mechanism has not been fully clarified.
    Adult (10 weeks) C57BL/6 male mice and neonatal rat cardiomyocytes were used to establish I/R and hypoxia/reoxygenation (H/R) injury models. I/R and H/R models were treated with HRS to classify the mechanisms of cardioproctective function. In this study, we found that miR-124-3p was significantly decreased in both I/R and H/R models, while it was partially ameliorated by HRS pretreatment. HRS treatment also alleviated ischaemia-induced apoptotic cell death and increased cell viability during I/R process, whereas silencing expression of miR-124-3p abolished this protective effect. In addition, we identified calpain1 as a direct target of miR-124-3p, and up-regulation of miR-124-3 produced both activity and expression of calpain1. It was also found that compared with the HRS group, overexpression of calpain1 increased caspase-3 activities, promoted cleaved-caspase3 and Bax protein expressions, and correspondingly decreased Bcl-2, further reducing cell viability. These results illustrated that calpain1 overexpression attenuated protective effect of HRS on cardiomyocytes in H/R model.
    The present study showed a protective effect of HRS on I/R injury, which may be associated with miR-124-3p-calpain1 signalling pathway.
    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
    缺血性心力衰竭(HF)已成为严重危害人们生命和健康的疾病。作为一种广泛应用于临床的中药配方,新优化的生麦散(NO-SMS)已被证明对改善心脏功能显着有效,增加运动耐受力,在中国各地区的多中心临床研究中,可以减缓心力衰竭患者心肌纤维化的进展。在我们之前的药效学和毒理学研究中,我们发现,中等剂量的制剂(8.1克原药/公斤)是最有效的治疗心力衰竭,但其作用机制仍在研究中。本研究正在探索其与心肌细胞凋亡的关系。
    我们通过两部分实验进行了研究和验证,体内和体外。首先,通过结扎冠状动脉左前降支(EF≤50%)制备雄性SD大鼠心力衰竭模型,用NO-SMS配方治疗(8.1g原药/kg/d),依芬地尔(5.4mg/kg/d)或依那普利(0.9mg/kg/d)通过管饲法制备悬浮液4周。通过超声心动图评估心脏和结构的变化,H&E,和MASSON染色。Westernblot检测各组心肌细胞凋亡,qRT-PCR,和ELISA。体外细胞实验包括H2O2和NMDA分别诱导H9c2心肌细胞损伤,各组用含NO-SMS和Ifenprodil的血清孵育24h。AnnexinV-FITC/PI双染色法检测细胞凋亡,和其余的测定与体内实验一致。
    与模型组相比,NO-SMS组和依芬地尔组能够显著改善心功能,延缓心肌纤维化,减少促凋亡蛋白的表达,mRNA以及H2O2和NMDA诱导的心力衰竭大鼠和H9c2心肌细胞中Ca2+和ROS的浓度水平,能显著降低受损心肌细胞的凋亡率,有效抑制心肌细胞凋亡。
    NO-SMS配方改善心功能,抑制HF大鼠心室重构和心肌细胞凋亡,其机制可能与NMDAR信号通路的调控有关,抑制大的细胞内Ca2+向内流动,和心肌细胞中的ROS产生。
    UNASSIGNED: Ischemic heart failure (HF) has become a disease that seriously endangers people\'s life and health. As a herbal formula widely used in clinical practice, new optimized Sheng-Mai-San (NO-SMS) has been shown to be significantly effective in improving cardiac function, increasing exercise tolerance, and slowing the progression of myocardial fibrosis in heart failure patients in multi-center clinical studies in various regions of China. In our previous pharmacodynamic and toxicological studies, we found that a medium-dose formulation (8.1 g of raw drug/kg) was the most effective in the treatment of heart failure, but its mechanism of action is still being investigated. The present study is exploring its relationship with cardiomyocyte apoptosis.
    UNASSIGNED: We investigated and verified this through two parts of experiments, in vivo and in vitro. Firstly, we prepared male SD rats with heart failure models by ligating the left anterior descending branch of the coronary artery (EF ≤ 50%), which were treated with NO-SMS Formula (8.1 g of raw drug/kg/d), Ifenprodil (5.4 mg/kg/d) or Enalapril (0.9 mg/kg/d) prepared suspensions by gavage for 4 weeks. The cardiac and structural changes were evaluated by echocardiography, H&E, and MASSON staining. The apoptosis of cardiomyocytes in each group was detected by Western blot, qRT-PCR, and ELISA. In vitro cell experiments include H9c2 cardiomyocyte injury induced by H2O2 and NMDA respectively, and the groups were incubated with NO-SMS and Ifenprodil-containing serum for 24 h. Apoptosis was detected by Annexin V-FITC/PI double-staining method, and the rest of the assays were consistent with the in vivo experiments.
    UNASSIGNED: Compared with the model group, the NO-SMS formula group and the Ifenprodil group could significantly improve cardiac function, delay myocardial fibrosis, reduce the expression of pro-apoptotic proteins, mRNA, and the concentration levels of Ca2+ and ROS in heart failure rats and H9c2 cardiomyocytes with H2O2 and NMDA-induced injury, which could significantly reduce the apoptosis rate of damaged cardiomyocytes and effectively inhibit the apoptosis of cardiomyocytes.
    UNASSIGNED: NO-SMS Formula improved cardiac function, inhibited ventricular remodeling and cardiomyocyte apoptosis in HF rats, and its mechanism may be related to the regulation of the NMDAR signaling pathway, inhibition of large intracellular Ca2+ inward flow, and ROS production in cardiomyocytes.
    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
    探讨lncRNAMALAT1在糖尿病心肌病(DCM)发病中的作用机制。在db/db小鼠中确认DCM模型。通过miRNA测序检测心肌中的miRNA。miR-185-5p与MALAT1和RhoA的相互作用通过双荧光素酶报告基因测定进行验证。在存在或不存在MALAT1-shRNA和法舒地尔的情况下,用5.5或30mmol/L的D-葡萄糖(HG)培养原代新生心肌细胞,岩石抑制剂。通过实时定量PCR检测MALAT1和miR-185-5p的表达。使用流式细胞术和TUNEL染色评估凋亡心肌细胞。测定SOD活性和MDA含量。ROCK活动,Drp1S616的磷酸化,通过Western印迹分析了mitofusin2和凋亡相关蛋白。通过JC-1检查线粒体膜电位。在db/db小鼠和HG诱导的心肌细胞中,MALAT1显著上调,而miR-185-5p下调。MALAT1通过在HG心肌细胞中生成miR-185-5p来调节RhoA/ROCK途径。MALAT1和法舒地尔的敲除均能抑制HG诱导的氧化应激,并减轻线粒体动力学和线粒体功能障碍的失衡,伴有心肌细胞凋亡减少。MALAT1通过激活miR-185-5p激活RhoA/ROCK通路,并介导HG诱导的氧化应激,小鼠心肌细胞线粒体损伤和凋亡。
    To explore the underlying mechanism of lncRNA MALAT1 in the pathogenesis of diabetic cardiomyopathy (DCM). DCM models were confirmed in db/db mice. MiRNAs in myocardium were detected by miRNA sequencing. The interactions of miR-185-5p with MALAT1 and RhoA were validated by dual-luciferase reporter assays. Primary neonatal cardiomyocytes were cultured with 5.5 or 30 mmol/L D-glucose (HG) in the presence or absence of MALAT1-shRNA and fasudil, a ROCK inhibitor. MALAT1 and miR-185-5p expression were determined by real-time quantitative PCR. The apoptotic cardiomyocytes were evaluated using flow cytometry and TUNEL staining. SOD activity and MDA contents were measured. The ROCK activity, phosphorylation of Drp1S616 , mitofusin 2 and apoptosis-related proteins were analysed by Western blotting. Mitochondrial membrane potential was examined by JC-1. MALAT1 was significantly up-regulated while miR-185-5p was down-regulated in myocardium of db/db mice and HG-induced cardiomyocytes. MALAT1 regulated RhoA/ROCK pathway via sponging miR-185-5p in cardiomyocytes in HG. Knockdown of MALAT1 and fasudil all inhibited HG-induced oxidative stress, and alleviated imbalance of mitochondrial dynamics and mitochondrial dysfunction, accompanied by reduced cardiomyocyte apoptosis. MALAT1 activated the RhoA/ROCK pathway via sponging miR-185-5p and mediated HG-induced oxidative stress, mitochondrial damage and apoptosis of cardiomyocytes in mice.
    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
    背景心肌梗死(MI)是一种高发病率和死亡率的心血管疾病。PI16(肽酶抑制剂16),作为一种分泌的蛋白质,在心脏疾病如心力衰竭中高度表达。然而,PI16在MI中的功能作用尚不清楚。本研究旨在探讨PI16在MI后的作用及其机制。方法和结果采用酶联免疫吸附法和免疫荧光法检测MI后PI16水平,这表明PI16在急性MI患者的血浆和小鼠心脏梗死区的血浆中上调。使用PI16的功能增益和功能丧失实验来研究MI后PI16的潜在作用。体外,PI16过表达抑制氧糖剥夺诱导的乳鼠心肌细胞凋亡,而PI16敲低会加剧新生大鼠心肌细胞凋亡。在体内,对PI16转基因小鼠进行左冠状动脉前降支结扎,PI16基因敲除小鼠,和他们的同窝伙伴。PI16转基因小鼠在MI后24小时显示心肌细胞凋亡减少,在MI后28天改善左心室重构。相反,PI16敲除小鼠表现出加重的尺寸和重塑。机械上,PI16下调Wnt3a(无翼MMTV集成站点系列,成员3a)/β-连环蛋白途径,重组Wnt3a逆转了PI16在氧糖剥夺诱导的新生大鼠心肌细胞中的抗凋亡作用。PI16也抑制HDAC1(I类组蛋白脱乙酰酶)的表达,HDAC1的过表达消除了PI16对细胞凋亡和Wnt信号的抑制作用。结论总之,PI16通过HDAC1-Wnt3a-β-catenin轴保护MI后心肌细胞凋亡和左心室重构。
    Background Myocardial infarction (MI) is a cardiovascular disease with high morbidity and mortality. PI16 (peptidase inhibitor 16), as a secreted protein, is highly expressed in heart diseases such as heart failure. However, the functional role of PI16 in MI is unknown. This study aimed to investigate the role of PI16 after MI and its underlying mechanisms. Methods and Results PI16 levels after MI were measured by enzyme-linked immunosorbent assay and immunofluorescence staining, which showed that PI16 was upregulated in the plasma of patients with acute MI and in the infarct zone of murine hearts. PI16 gain- and loss-of-function experiments were used to investigate the potential role of PI16 after MI. In vitro, PI16 overexpression inhibited oxygen-glucose deprivation-induced apoptosis in neonatal rat cardiomyocytes, whereas knockdown of PI16 exacerbated neonatal rat cardiomyocyte apoptosis. In vivo, left anterior descending coronary artery ligation was performed on PI16 transgenic mice, PI16 knockout mice, and their littermates. PI16 transgenic mice showed decreased cardiomyocyte apoptosis at 24 hours after MI and improved left ventricular remodeling at 28 days after MI. Conversely, PI16 knockout mice showed aggravated infract size and remodeling. Mechanistically, PI16 downregulated Wnt3a (wingless-type MMTV integration site family, member 3a)/β-catenin pathways, and the antiapoptotic role of PI16 was reversed by recombinant Wnt3a in oxygen-glucose deprivation-induced neonatal rat cardiomyocytes. PI16 also inhibited HDAC1 (class I histone deacetylase) expression, and overexpression HDAC1 abolished the inhibition of apoptosis and Wnt signaling of PI16. Conclusions In summary, PI16 protects against cardiomyocyte apoptosis and left ventricular remodeling after MI through the HDAC1-Wnt3a-β-catenin axis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号