首页> 美国卫生研究院文献>Cell Death Disease >ATM-NFκB axis-driven TIGAR regulates sensitivity of glioma cells to radiomimetics in the presence of TNFα
【2h】

ATM-NFκB axis-driven TIGAR regulates sensitivity of glioma cells to radiomimetics in the presence of TNFα

机译:ATM-NFκB轴驱动的TIGAR在TNFα存在下调节神经胶质瘤细胞对放射模拟物的敏感性

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

Gliomas are resistant to radiation therapy, as well as to TNFα induced killing. Radiation-induced TNFα triggers Nuclear factor κB (NFκB)-mediated radioresistance. As inhibition of NFκB activation sensitizes glioma cells to TNFα-induced apoptosis, we investigated whether TNFα modulates the responsiveness of glioma cells to ionizing radiation-mimetic Neocarzinostatin (NCS). TNFα enhanced the ability of NCS to induce glioma cell apoptosis. NCS-mediated death involved caspase-9 activation, reduction of mitochondrial copy number and lactate production. Death was concurrent with NFκB, Akt and Erk activation. Abrogation of Akt and NFκB activation further potentiated the death inducing ability of NCS in TNFα cotreated cells. NCS-induced p53 expression was accompanied by increase in TP53-induced glycolysis and apoptosis regulator (TIGAR) levels and ATM phosphorylation. siRNA-mediated knockdown of TIGAR abrogated NCS-induced apoptosis. While DN-IκB abrogated NCS-induced TIGAR both in the presence and absence of TNFα, TIGAR had no effect on NFκB activation. Transfection with TIGAR mutant (i) decreased apoptosis and γH2AX foci formation (ii) decreased p53 (iii) elevated ROS and (iv) increased Akt/Erk activation in cells cotreated with NCS and TNFα. Heightened TIGAR expression was observed in GBM tumors. While NCS induced ATM phosphorylation in a NFκB independent manner, ATM inhibition abrogated TIGAR and NFκB activation. Metabolic gene profiling indicated that TNFα affects NCS-mediated regulation of several genes associated with glycolysis. The existence of ATM-NFκB axis that regulate metabolic modeler TIGAR to overcome prosurvival response in NCS and TNFα cotreated cells, suggests mechanisms through which inflammation could affect resistance and adaptation to radiomimetics despite concurrent induction of death.
机译:神经胶质瘤对放射疗法以及TNFα诱导的杀伤具有抵抗力。辐射诱导的TNFα触发核因子κB(NFκB)介导的放射抗性。由于抑制NFκB激活可使神经胶质瘤细胞对TNFα诱导的凋亡敏感,因此我们研究了TNFα是否调节了神经胶质瘤细胞对电离辐射模拟新拟抑制素(NCS)的反应性。 TNFα增强了NCS诱导神经胶质瘤细胞凋亡的能力。 NCS介导的死亡涉及caspase-9激活,线粒体拷贝数减少和乳酸产生。死亡与NFκB,Akt和Erk激活同时发生。 Akt和NFκB激活的废止进一步增强了TNFα共处理细胞中NCS的死亡诱导能力。 NCS诱导的p53表达伴随着TP53诱导的糖酵解和凋亡调节剂(TIGAR)水平以及ATM磷酸化的增加。 siRNA介导的TIGAR敲除消除了NCS诱导的细胞凋亡。在存在和不存在TNFα的情况下,DN-IκB都消除了NCS诱导的TIGAR,而TIGAR对NFκB的激活没有影响。用TIGAR突变体转染(i)在用NCS和TNFα处理的细胞中,凋亡减少,γH2AX灶形成减少(ii)p53降低(iii)ROS升高,以及(iv)Akt / Erk激活增加。在GBM肿瘤中观察到TIGAR表达升高。 NCS以独立于NF κ B的方式诱导ATM磷酸化,而ATM抑制则消除了TIGAR和NF κ B的活化。代谢基因分析表明,TNF α影响NCS介导的糖酵解相关基因的调控。 ATM-NF κ B轴可调节代谢建模工具TIGAR克服NCS和TNF α共同处理的细胞中的生存应答,提示炎症可能通过这种机制影响抗药性和适应性放射模拟物,尽管同时诱发死亡。

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