首页> 美国卫生研究院文献>American Journal of Physiology - Gastrointestinal and Liver Physiology >Hepatocytes produce TNF-α following hypoxia-reoxygenation and liver ischemia-reperfusion in a NADPH oxidase- and c-Src-dependent manner
【2h】

Hepatocytes produce TNF-α following hypoxia-reoxygenation and liver ischemia-reperfusion in a NADPH oxidase- and c-Src-dependent manner

机译:缺氧-再充氧和肝脏缺血-再灌注后肝细胞以NADPH氧化酶和c-Src依赖性方式产生TNF-α

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

Cell line studies have previously demonstrated that hypoxia-reoxygenation (H/R) leads to the production of NADPH oxidase 1 and 2 (NOX1 and NOX2)-dependent reactive oxygen species (ROS) required for the activation of c-Src and NF-κB. We now extend these studies into mouse models to evaluate the contribution of hepatocytes to the NOX- and c-Src-dependent TNF-α production that follows H/R in primary hepatocytes and liver ischemia-reperfusion (I/R). In vitro, c-Src-deficient primary hepatocytes produced less ROS and TNF-α following H/R compared with controls. In vivo, c-Src-KO mice also had impaired TNF-α and NF-κB responses following partial lobar liver I/R. Studies in NOX1 and p47phox knockout primary hepatocytes demonstrated that both NOX1 and p47phox are partially required for H/R-mediated TNF-α production. To further investigate the involvement of NADPH oxidases in the production of TNF-α following liver I/R, we performed additional in vivo experiments in knockout mice deficient for NOX1, NOX2, p47phox, Rac1, and/or Rac2. Cumulatively, these results demonstrate that NOX2 and its activator subunits (p47phox and Rac) control the secretion of TNF-α by the liver following I/R. Interestingly, in the absence of Kupffer cells and NOX2, NOX1 played a dominant role in TNF-α production following hepatic I/R. However, NOX1 deletion alone had little effect on I/R-induced TNF-α. Thus Kupffer cell-derived factors and NOX2 act to suppress hepatic NOX1-dependent TNF-α production. We conclude that c-Src and NADPH oxidase components are necessary for redox-mediated production of TNF-α following liver I/R and that hepatocytes play an important role in this process.
机译:先前的细胞系研究表明,缺氧复氧(H / R)导致产生c-Src和NF-κB活化所需的NADPH氧化酶1和2(NOX1和NOX2)依赖性活性氧(ROS)。 。现在,我们将这些研究扩展到小鼠模型中,以评估肝细胞对NOX和c-Src依赖性TNF-α产生的贡献,其在原代肝细胞中的H / R和肝脏缺血再灌注(I / R)之后发生。在体外,与对照组相比,H / R后缺乏c-Src的原代肝细胞产生较少的ROS和TNF-α。在体内,部分大叶肝I / R后,c-Src-KO小鼠的TNF-α和NF-κB反应也受损。对NOX1和p47 phox 敲除原代肝细胞的研究表明,NOX1和p47 phox 都是H / R介导的TNF-α产生的部分必需。为了进一步研究NADPH氧化酶在肝脏I / R后参与TNF-α的产生,我们在敲除NOX1,NOX2,p47 phox ,Rac1和/或Rac2。累积地,这些结果表明,NO / 2后,NOX2及其激活亚基(p47 phox 和Rac)控制肝脏分泌TNF-α。有趣的是,在没有库普弗细胞和NOX2的情况下,NOX1在肝I / R后在TNF-α产生中起主导作用。但是,单独删除NOX1对I / R诱导的TNF-α几乎没有影响。因此,枯否细胞衍生的因子和NOX2起到抑制肝脏NOX1依赖性TNF-α产生的作用。我们得出结论,肝脏I / R后c-Src和NADPH氧化酶成分对于氧化还原介导的TNF-α产生是必需的,并且肝细胞在此过程中起重要作用。

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