...
首页> 外文期刊>International journal of oncology >Synergistic antineoplastic effect of DLC1 tumor suppressor protein and histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), on prostate and liver cancer cells: Perspectives for therapeutics
【24h】

Synergistic antineoplastic effect of DLC1 tumor suppressor protein and histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), on prostate and liver cancer cells: Perspectives for therapeutics

机译:DLC1肿瘤抑制蛋白和组蛋白脱乙酰基酶抑制剂,亚磺酰苯胺异羟肟酸(SAHA)对前列腺癌和肝癌细胞的协同抗肿瘤作用:治疗的前景

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Inactivation of tumor suppressor genes is a major contributing alteration in the initiation or progression of cancer. The human tumor suppressor gene DLC1 (deleted in liver cancer 1) is frequently downregulated or silenced in multiple cancers, predominantly by epigenetic mechanisms. With the current considerable interest and progress in epigenetic therapy, a number of promising antineoplastic agents, particularly histone deacetylase (HDAC) inhibitors, have been developed and used successfully in clinical trials. Both DLC1 and HDAC inhibitors exert antineoplastic functions, and their combined action could be exploited for a more effective cancer therapy. To evaluate the potential benefits of this approach, we examined the antineoplastic effects of adenoviral (Ad)-DLC1-mediated transduction and exposure to suberoylanilide hydroxamic acid (SAHA), a powerful HDAC inhibitor, in two human cancer cell lines that lack intrinsic DLC1 expression, 22Rv1 prostate cancer cells and 7703K human hepatocellular carcinoma cells. Consistent with the oncosuppressive function of DLC1 in several cancers, including prostate and liver cancer, transduction of 22Rv1 and 7703K cells with an Ad-DLC1 expression vector resulted in alterations of cell morphology, induction of apoptosis, and inhibition of cell proliferation, migration, and anchorage-independent growth. A low concentration of SAHA (5 μM) efficiently restored the expression of DLC1 in 22Rv1 cells that lack DLC1 expression due to histone deacetylation but had a minimal effect in 7703K cells in which silencing of the DLC1 gene is due mainly to promoter hypermethylation. Regardless of the epigenetic mechanism of DLC1 inactivation, SAHA treatment of DLC1-transduced cells had a synergistic inhibitory effect on tumor cell proliferation and tumorigenesis in both cell lines. In 22Rv1 cells, this combination regimen nearly abolished the formation of colonies in semisolid media as a measure of tumorigenicity in vitro. Current in vitro results validate this protocol as a potentially new therapeutic option in certain cancers.
机译:肿瘤抑制基因的失活是癌症的起始或进展的主要贡献改变。在多种癌症中,人类肿瘤抑制基因DLC1(在肝癌1中缺失)经常被下调或沉默,主要是通过表观遗传机制。随着表观遗传学治疗方面的当前巨大兴趣和进展,已经开发了许多有希望的抗肿瘤药,尤其是组蛋白脱乙酰基酶(HDAC)抑制剂,并已成功用于临床试验。 DLC1和HDAC抑制剂均具有抗肿瘤功能,它们的联合作用可用于更有效的癌症治疗。为了评估这种方法的潜在益处,我们在两种缺乏内在DLC1表达的人类癌细胞系中,研究了腺病毒(Ad)-DLC1介导的转导作用以及暴露于强力HDAC抑制剂异戊酰苯胺异羟肟酸(SAHA)的抗肿瘤作用。 ,22Rv1前列腺癌细胞和7703K人肝细胞癌细胞。与DLC1在包括前列腺癌和肝癌在内的多种癌症中的抑癌功能一致,用Ad-DLC1表达载体转导22Rv1和7703K细胞可导致细胞形态改变,凋亡诱导以及对细胞增殖,迁移和抑制的抑制。独立于锚地的生长。低浓度的SAHA(5μM)有效地恢复了22Rv1细胞中DLC1的表达,这些细胞由于组蛋白去乙酰化而缺乏DLC1表达,但是在7703K细胞中影响最小,其中DLC1基因的沉默主要是由于启动子超甲基化。无论DLC1失活的表观遗传机制如何,SAHA处理DLC1转导的细胞对两种细胞系中的肿瘤细胞增殖和肿瘤发生均具有协同抑制作用。在22Rv1细胞中,这种组合方案几乎消除了半固体培养基中集落的形成,以此来衡量体外致瘤性。当前的体外结果证实该方案在某些癌症中是潜在的新治疗选择。

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