首页> 外文期刊>Cell death & disease. >Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis
【24h】

Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis

机译:来自人胚胎干细胞的细胞外囊衍生心血管祖细胞通过减少心肌细胞死亡和促进血管生成来促进心脏梗塞愈合

获取原文
           

摘要

Human pluripotent stem cells (hPSCs)-derived cardiovascular progenitor cells (CVPCs) are a promising source for myocardial repair, while the mechanisms remain largely unknown. Extracellular vesicles (EVs) are known to mediate cell-cell communication, however, the efficacy and mechanisms of hPSC-CVPC-secreted EVs (hCVPC-EVs) in the infarct healing when given at the acute phase of myocardial infarction (MI) are unknown. Here, we report the cardioprotective effects of the EVs secreted from hESC-CVPCs under normoxic (EV-N) and hypoxic (EV-H) conditions in the infarcted heart and the long noncoding RNA (lncRNA)-related mechanisms. The hCVPC-EVs were confirmed by electron microscopy, nanoparticle tracking, and immunoblotting analysis. Injection of hCVPC-EVs into acutely infracted murine myocardium significantly improved cardiac function and reduced fibrosis at day 28 post MI, accompanied with the improved vascularization and cardiomyocyte survival at border zones. Consistently, hCVPC-EVs enhanced the tube formation and migration of human umbilical vein endothelial cells (HUVECs), improved the cell viability, and attenuated the lactate dehydrogenase release of neonatal rat cardiomyocytes (NRCMs) with oxygen glucose deprivation (OGD) injury. Moreover, the improvement of the EV-H in cardiomyocyte survival and tube formation of HUVECs was significantly better than these in the EV-N. RNA-seq analysis revealed a high abundance of the lncRNA MALAT1 in the EV-H. Its abundance was upregulated in the infarcted myocardium and cardiomyocytes treated with hCVPC-EVs. Overexpression of human MALAT1 improved the cell viability of NRCM with OGD injury, while knockdown of MALAT1 inhibited the hCVPC-EV-promoted tube formation of HUVECs. Furthermore, luciferase activity assay, RNA pull-down, and manipulation of miR-497 levels showed that MALAT1 improved NRCMs survival and HUVEC tube formation through targeting miR-497. These results reveal that hCVPC-EVs promote the infarct healing through improvement of cardiomyocyte survival and angiogenesis. The cardioprotective effects of hCVPC-EVs can be enhanced by hypoxia-conditioning of hCVPCs and are partially contributed by MALAT1 via targeting the miRNA.
机译:人多能干细胞(HPSC)的心血管祖细胞(CVPC)是心肌修复的有前途的源,而机制仍然很大程度上是未知的。已知细胞外囊泡(EVS)介导细胞 - 细胞通信,然而,当在心肌梗死(MI)的急性阶段给出时,HPSC-CVPC分泌的EVS(HCVPC-EV)中HPSC-CVPC分泌的EVS(HCVPC-EV)的疗效和机制是未知的。在这里,我们报告了在梗死的心脏和鼻窦炎(EV-N)和缺氧(EV-H)条件下HESC-CVPCs分泌的EVS的心脏保护作用以及长的非分量RNA(LNCRNA)的机制。通过电子显微镜,纳米粒子跟踪和免疫印迹分析证实HCVPC-EV。将HCVPC-eV注入急性碎断的小鼠心肌中的心脏功能显着提高,第28天的心脏功能和纤维化降低,伴随着边境区域的改善血管化和心肌细胞存活。始终如一地,HCVPC-EVS增强了人脐静脉内皮细胞(HUVEC)的管形成和迁移,改善了细胞活力,并通过氧葡萄糖剥夺(OGD)损伤来减毒新生大鼠心肌细胞(NRCMS)的乳酸脱氢酶释放。此外,在EV-N中,Huvecs的心肌细胞存活和管形成的EV-H的改善明显优于这些。 RNA-SEQ分析显示EV-H中的LNCRNA MALAT1的高丰度。它的丰富在用HCVPC-EVS处理的梗死的心肌和心肌细胞中上调。人类malat1的过度表达改善了NRCM与OGD损伤的细胞活力,而Malat1的敲低抑制Huvecs的HCVPC-EV促进管形成。此外,荧光素酶活性测定,RNA下拉和MiR-497水平的操纵表明,MALAT1通过靶向MIR-497改善了NRCMS存活率和HUVEC管形成。这些结果表明,通过改善心肌细胞存活和血管生成,HCVPC-EVS促进梗塞愈合。 HCVPC-EV可以通过HCVPC的缺氧调节增强HCVPC-EV的心脏保护作用,并且通过靶向miRNA部分通过MALAT1贡献。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号