首页> 美国卫生研究院文献>Wiley-Blackwell Online Open >Human Liver Stem Cell-Derived Microvesicles Inhibit Hepatoma Growth in SCID Mice by Delivering Antitumor MicroRNAs
【2h】

Human Liver Stem Cell-Derived Microvesicles Inhibit Hepatoma Growth in SCID Mice by Delivering Antitumor MicroRNAs

机译:人类肝干细胞衍生的微泡通过提供抗肿瘤MicroRNA抑制SCID小鼠肝癌的生长。

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

Microvesicles (MVs) play a pivotal role in cell-to-cell communication. Recent studies demonstrated that MVs may transfer genetic information between cells. Here, we show that MVs derived from human adult liver stem cells (HLSC) may reprogram in vitro HepG2 hepatoma and primary hepatocellular carcinoma cells by inhibiting their growth and survival. In vivo intratumor administration of MVs induced regression of ectopic tumors developed in SCID mice. We suggest that the mechanism of action is related to the delivery of microRNAs (miRNAs) from HLSC-derived MVs (MV-HLSC) to tumor cells on the basis of the following evidence: (a) the rapid, CD29-mediated internalization of MV-HLSC in HepG2 and the inhibition of tumor cell growth after MV uptake; (b) the transfer by MV-HLSC of miRNAs with potential antitumor activity that was downregulated in HepG2 cells with respect to normal hepatocytes; (c) the abrogation of the MV-HLSC antitumor effect after MV pretreatment with RNase or generation of MVs depleted of miRNAs; (d) the relevance of selected miRNAs was proven by transfecting HepG2 with miRNA mimics. The antitumor effect of MV-HLSC was also observed in tumors other than liver such as lymphoblastoma and glioblastoma. These results suggest that the delivery of selected miRNAs by MVs derived from stem cells may inhibit tumor growth and stimulate apoptosis. Stem Cells2012;30:1985–1998
机译:微泡(MVs)在细胞间通讯中起着关键作用。最近的研究表明,MVs可以在细胞之间转移遗传信息。在这里,我们显示源自成人肝干细胞(HLSC)的MVs可能会通过抑制其生长和存活而在体外重编程HepG2肝癌和原发性肝癌细胞。 MV的体内肿瘤内施用诱导了SCID小鼠中发展的异位肿瘤的消退。我们建议基于以下证据,其作用机制与从HLSC衍生的MV(MV-HLSC)向肿瘤细胞的microRNA(miRNA)传递有关:(a)快速,CD29介导的MV内在化HepG2中的-HLSC和MV摄取后对肿瘤细胞生长的抑制; (b)通过MV-HLSC转移具有潜在抗肿瘤活性的miRNA在HepG2细胞中相对于正常肝细胞被下调; (c)用RNase进行MV预处理或产生缺失miRNA的MV后,取消MV-HLSC的抗肿瘤作用; (d)通过用miRNA模拟物转染HepG2证明了所选miRNA的相关性。 MV-HLSC的抗肿瘤作用在除肝脏以外的其他肿瘤如淋巴母细胞瘤和胶质母细胞瘤中也观察到。这些结果表明,由干细胞衍生的MV传递选定的miRNA可能抑制肿瘤生长并刺激细胞凋亡。干细胞2012; 30:1985–1998

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

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