首页> 外文期刊>Cell death & disease. >ATM-NFκB axis-driven TIGAR regulates sensitivity of glioma cells to radiomimetics in the presence of TNFα
【24h】

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

机译:ATM-NF κ B轴驱动的TIGAR在存在TNF α的情况下调节神经胶质瘤细胞对放射模拟物的敏感性

获取原文
获取外文期刊封面目录资料

摘要

Gliomas are resistant to radiation therapy, as well as to TNF α induced killing. Radiation-induced TNF α triggers Nuclear factor κ B (NF κ B)-mediated radioresistance. As inhibition of NF κ B activation sensitizes glioma cells to TNF α -induced apoptosis, we investigated whether TNF α modulates the responsiveness of glioma cells to ionizing radiation-mimetic Neocarzinostatin (NCS). TNF α 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 NF κ B, Akt and Erk activation. Abrogation of Akt and NF κ B activation further potentiated the death inducing ability of NCS in TNF α 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-I κ B abrogated NCS-induced TIGAR both in the presence and absence of TNF α , TIGAR had no effect on NF κ B activation. Transfection with TIGAR mutant (i) decreased apoptosis and γ H2AX foci formation (ii) decreased p53 (iii) elevated ROS and (iv) increased Akt/Erk activation in cells cotreated with NCS and TNF α . Heightened TIGAR expression was observed in GBM tumors. While NCS induced ATM phosphorylation in a NF κ B independent manner, ATM inhibition abrogated TIGAR and NF κ B activation. Metabolic gene profiling indicated that TNF α affects NCS-mediated regulation of several genes associated with glycolysis. The existence of ATM-NF κ B axis that regulate metabolic modeler TIGAR to overcome prosurvival response in NCS and TNF α 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激活使神经胶质瘤细胞对TNFα诱导的细胞凋亡敏感,因此我们研究了TNFα是否调节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。尽管在存在和不存在TNFα的情况下,DN-1κB都废除了NCS诱导的TIGAR,但TIGAR对NFκB的活化没有影响。用TIGAR突变体转染(i)在用NCS和TNFα共同处理的细胞中,凋亡减少和γH2AX灶形成减少(ii)p53降低(iii)ROS升高和(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NFκB的方式诱导ATM磷酸化时,ATM抑制作用消除了TIGAR和NFκB的活化。代谢基因谱分析表明,TNFα影响NCS介导的与糖酵解相关的几种基因的调控。 ATM-NFκB轴可调节代谢建模工具TIGAR克服NCS和TNFα共同处理的细胞中的生存应答,这表明尽管死亡同时存在,炎症仍可能影响抗药性和对放射模拟物的适应性机制。

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