...
首页> 外文期刊>BMC Cancer >Bifidobacterial recombinant thymidine kinase-ganciclovir gene therapy system induces FasL and TNFR2 mediated antitumor apoptosis in solid tumors
【24h】

Bifidobacterial recombinant thymidine kinase-ganciclovir gene therapy system induces FasL and TNFR2 mediated antitumor apoptosis in solid tumors

机译:双歧杆菌重组胸苷激酶-更昔洛韦基因治疗系统诱导FasL和TNFR2介导的实体瘤抗肿瘤细胞凋亡

获取原文
           

摘要

Background Directly targeting therapeutic suicide gene to a solid tumor is a hopeful approach for cancer gene therapy. Treatment of a solid tumor by an effective vector for a suicide gene remains a challenge. Given the lack of effective treatments, we constructed a bifidobacterial recombinant thymidine kinase (BF-rTK) -ganciclovir (GCV) targeting system (BKV) to meet this requirement and to explore antitumor mechanisms. Methods Bifidobacterium (BF) or BF-rTK was injected intratumorally with or without ganciclovir in a human colo320 intestinal xenograft tumor model. The tumor tissues were analyzed using apoptosis antibody arrays, real time PCR and western blot. The colo320 cell was analyzed by the gene silencing method. Autophagy and necroptosis were also detected in colo320 cell. Meanwhile, three human digestive system xenograft tumor models (colorectal cancer colo320, gastric cancer MKN-45 and liver cancer SSMC-7721) and a breast cancer (MDA-MB-231) model were employed to validate the universality of BF-rTK?+?GCV in solid tumor gene therapy. The survival rate was evaluated in three human cancer models after the BF-rTK?+?GCV intratumor treatment. The analysis of inflammatory markers (TNF-α) in tumor indicated that BF-rTK?+?GCV significantly inhibited TNF-α expression. Results The results suggested that BF-rTK?+?GCV induced tumor apoptosis without autophagy and necroptosis occurrence. The apoptosis was transduced by multiple signaling pathways mediated by FasL and TNFR2 and mainly activated the mitochondrial control of apoptosis via Bid and Bim, which was rescued by silencing Bid or/and Bim . However, BF?+?GCV only induced apoptosis via Fas/FasL signal pathway accompanied with increased P53 expression. We further found that BF-rTK?+?GCV inhibited the expression of the inflammatory maker of TNF-α. However, BF-rTK?+?GCV did not result in necroptosis and autophagy. Conclusions BF-rTK?+?GCV induced tumor apoptosis mediated by FasL and TNFR2 through the mitochondrial control of apoptosis via Bid and Bim without inducing necroptosis and autophagy. Furthermore, BF-rTK?+?GCV showed to repress the inflammation of tumor through downregulating TNF-α expression. Survival analysis results of multiple cancer models confirmed that BF-rTK?+?GCV system has a wide field of application in solid tumor gene therapy.
机译:背景技术将治疗性自杀基因直接靶向实体瘤是用于癌症基因治疗的有希望的方法。用有效的自杀基因载体治疗实体瘤仍然是一个挑战。鉴于缺乏有效的治疗方法,我们构建了双歧杆菌重组胸苷激酶(BF-rTK)-更昔洛韦(GCV)靶向系统(BKV),以满足这一要求并探索抗肿瘤机制。方法在人结肠癌colo320肠道肿瘤模型中,将双歧杆菌(BF)或BF-rTK分别注射或不注射更昔洛韦。使用凋亡抗体阵列,实时PCR和Western印迹分析肿瘤组织。通过基因沉默法分析colo320细胞。在colo320细胞中也检测到自噬和坏死。同时,采用三种人类消化系统异种移植肿瘤模型(结肠癌结肠癌colo320,胃癌MKN-45和肝癌SSMC-7721)和乳腺癌模型(MDA-MB-231)来验证BF-rTK?+的普遍性。 ?GCV在实体瘤基因治疗中。在BF-rTKβ+ΔGCV肿瘤内治疗后,在三种人类癌症模型中评估了存活率。肿瘤中炎症标志物(TNF-α)的分析表明,BF-rTKβ+βGCV显着抑制了TNF-α的表达。结果结果表明BF-rTKβ+ΔGCV诱导肿瘤细胞凋亡,而没有发生自噬和坏死性坏死。细胞凋亡通过FasL和TNFR2介导的多种信号传导途径进行转导,并主要通过Bid和Bim激活线粒体对细胞凋亡的控制,并通过沉默Bid或/和Bim得以挽救。但是,BF + + GCV仅通过Fas / FasL信号途径诱导凋亡,并伴有P53表达的增加。我们进一步发现BF-rTKβ+βGCV抑制了TNF-α的炎性产生子的表达。但是,BF-rTKβ+βGCV没有导致坏死和自噬。结论BF-rTKβ+ΔGCV通过Bid和Bim的线粒体控制凋亡而诱导FasL和TNFR2介导的肿瘤凋亡,而不会引起坏死和自噬。此外,BF-rTKβ+βGCV显示出通过下调TNF-α表达来抑制肿瘤的炎症。多种癌症模型的生存分析结果证实,BF-rTKβ+ΔGCV系统在实体瘤基因治疗中具有广泛的应用领域。

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