...
首页> 外文期刊>Molecular Carcinogenesis >Bortezomib-induced sensitization of malignant human glioma cells to vorinostat-induced apoptosis depends on reactive oxygen species production, mitochondrial dysfunction, Noxa upregulation, Mcl-1 cleavage, and DNA damage
【24h】

Bortezomib-induced sensitization of malignant human glioma cells to vorinostat-induced apoptosis depends on reactive oxygen species production, mitochondrial dysfunction, Noxa upregulation, Mcl-1 cleavage, and DNA damage

机译:硼替佐米诱导的恶性人类神经胶质瘤细胞对伏立诺他诱导的凋亡的敏感性取决于活性氧的产生,线粒体功能障碍,Noxa上调,Mcl-1裂解和DNA损伤

获取原文
获取原文并翻译 | 示例
   

获取外文期刊封面封底 >>

       

摘要

Glioblastomas are invasive tumors with poor prognosis despite current therapies. Histone deacetylase inhibitors (HDACIs) represent a class of agents that can modulate gene expression to reduce tumor growth, and we and others have noted some antiglioma activity from HDACIs, such as vorinostat, although insufficient to warrant use as monotherapy. We have recently demonstrated that proteasome inhibitors, such as bortezomib, dramatically sensitized highly resistant glioma cells to apoptosis induction, suggesting that proteasomal inhibition may be a promising combination strategy for glioma therapeutics. In this study, we examined whether bortezomib could enhance response to HDAC inhibition in glioma cells. Although primary cells from glioblastoma multiforme (GBM) patients and established glioma cell lines did not show significant induction of apoptosis with vorinostat treatment alone, the combination of vorinostat plus bortezomib significantly enhanced apoptosis. The enhanced efficacy was due to proapoptotic mitochondrial injury and increased generation of reactive oxygen species. Our results also revealed that combination of bortezomib with vorinostat enhanced apoptosis by increasing Mcl-1 cleavage, Noxa upregulation, Bak and Bax activation, and cytochrome c release. Further downregulation of Mcl-1 using shRNA enhanced cell killing by the bortezomib/vorinostat combination. Vorinostat induced a rapid and sustained phosphorylation of histone H2AX in primary GBM and T98G cells, and this effect was significantly enhanced by co-administration of bortezomib. Vorinostat/bortezomib combination also induced Rad51 downregulation, which plays an important role in the synergistic enhancement of DNA damage and apoptosis. The significantly enhanced antitumor activity that results from the combination of bortezomib and HDACIs offers promise as a novel treatment for glioma patients.
机译:胶质母细胞瘤是尽管有当前疗法但预后较差的浸润性肿瘤。组蛋白脱乙酰基酶抑制剂(HDACIs)代表一类可以调节基因表达以减少肿瘤生长的药物,我们和其他人已经注意到HDACIs产生的一些抗神经胶质瘤活性,例如伏立诺他,尽管不足以保证用作单一疗法。最近,我们证明了蛋白酶体抑制剂(如硼替佐米)可​​使高度耐药的神经胶质瘤细胞对凋亡诱导产生显着的敏感性,这表明蛋白酶体抑制可能是神经胶质瘤治疗的有希望的联合策略。在这项研究中,我们检查了硼替佐米是否可以增强神经胶质瘤细胞对HDAC抑制的反应。尽管单独使用伏立诺他治疗后,来自多形性胶质母细胞瘤(GBM)患者的原代细胞和已建立的神经胶质瘤细胞系均未显示出明显的凋亡诱导作用,但伏立诺他与硼替佐米的组合显着增强了凋亡。增强的功效归因于促凋亡的线粒体损伤和活性氧种类的产生增加。我们的研究结果还表明,硼替佐米与伏立诺他合用可通过增加Mcl-1裂解,Noxa上调,Bak和Bax活化以及细胞色素c释放来增强细胞凋亡。使用shRNA进一步下调Mcl-1可以增强硼替佐米/伏立诺他联合治疗对细胞的杀伤作用。伏立诺他诱导了原代GBM和T98G细胞中组蛋白H2AX的快速持续磷酸化,并且与硼替佐米合用显着增强了这种作用。伏立诺他/硼替佐米的组合还诱导了Rad51的下调,这在DNA损伤和细胞凋亡的协同增强中起着重要作用。硼替佐米和HDACIs联合使用可显着增强抗肿瘤活性,有望成为神经胶质瘤患者的新型治疗方法。

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