...
首页> 外文期刊>Cell death & disease. >Transmissible gastroenteritis virus targets Paneth cells to inhibit the self-renewal and differentiation of Lgr5 intestinal stem cells via Notch signaling
【24h】

Transmissible gastroenteritis virus targets Paneth cells to inhibit the self-renewal and differentiation of Lgr5 intestinal stem cells via Notch signaling

机译:传染性胃肠炎病毒靶向PANETH细胞,抑制LGR5肠道干细胞通过缺口信号传导的自我更新和分化

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Infection with transmissible gastroenteritis virus (TGEV) has been associated with villous atrophy within 48 h, which seriously disrupts intestinal homeostasis. However, the underlying mechanisms remain elusive. In this study, we found that TGEV infection severely disrupted intestinal homeostasis via inhibition of self-renewal and differentiation in Lgr5 intestinal stem cells (ISCs). Profoundly, TGEV-encoded NSP10/NSP16 protein complex-mediated the inactivation of Notch signaling provided a mechanistic explanation for this phenomenon. Initial invasions by TGEV-targeted Paneth cells through aminopeptidase N (APN) receptor, then inducing mitochondrial damage and ROS generation in them, ultimately causing Paneth cell decrease and loss of Notch factors (DII4 and Hes5), which are essential for Lgr5 ISCs selfrenewal and differentiation. Interestingly, loss of Notch signaling induced goblet cells differentiation at the cost of absorptive enterocytes and promoted mucins secretion, which accelerated TGEV replication. Therefore, the more differentiation of goblet cells, the greater TGEV infection in jejunum. These results provide a detailed mechanistic pathway by which villous atrophy sharply occurs in TGEV-infected jejunum within 48 h. Thus, the pathogenesis of TGEV can be described as a “bottom up scenario”, which is contrary to the traditional “top down” hypothesis. Together, our findings provide a potential link between diarrheal virus infection and crypt cells response that regulates Paneth cells function and Lgr5 ISCs fate and could be exploited for therapeutic application.
机译:传染性胃肠炎病毒(TGEV)感染已与绒毛萎缩有关,48小时内,这严重破坏了肠道稳态。但是,潜在机制仍然难以捉摸。在这项研究中,我们发现TGEV感染通过抑制LGR5肠干细胞(ISC)中的自我更新和分化来严重破坏肠道稳态。深刻地,TGEV编码的NSP10 / NSP16蛋白复合介导的凹口信号传导的失活提供了这种现象的机械解释。通过氨基肽酶N(APN)受体的TGEv靶向碱基细胞的初始入侵,然后诱导它们中的线粒体损伤和ROS生成,最终导致群体细胞减少和损失的缺口因子(DII4和HES5),这对于LGR5 ISCs自我和recryewal至关重要差异化。有趣的是,在吸收肠细胞的成本和促进粘蛋白分泌的成本下分化的缺口信号传导诱导的狗丝细胞分化,其加速了TGEV复制。因此,脚牙姆越血珠细胞的分化越多,济纳姆的TGEV感染越大。这些结果提供了一种详细的机制途径,在48小时内在TGEV感染的Jejunum中急剧发生绒毛萎缩途径。因此,TGEv的发病机制可以被描述为“底部上的场景”,这与传统的“自上而下”假设相反。我们的研究结果在一起提供了腹泻病毒感染和隐窝细胞之间的潜在联系,该潜在的联系在调节Paneth细胞功能和LGR5 ISCS命运中,并且可以用于治疗应用。

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