4.7 Article

Epigenetic regulation of p62/SQSTM1 overcomes the radioresistance of head and neck cancer cells via autophagy-dependent senescence induction

期刊

CELL DEATH & DISEASE
卷 12, 期 3, 页码 -

出版社

SPRINGERNATURE
DOI: 10.1038/s41419-021-03539-5

关键词

-

资金

  1. Basic Science Research Program through the National Research Foundation of Korea (NRF) - Ministry of Science, ICT, and Future planning [NRF-2018R1A2A1A05020064, NRF-2020R1A2C1011879, NRF-2020R1A2C201309, NRF2016R1A2B4015049, NRF-2019R1H1A2080034]
  2. MRC - Korean government (MSIT) [2018R1A5A2020732]
  3. Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea [2018-427, 2020IP0014]

向作者/读者索取更多资源

Tumors contain subpopulations of cancer cells with distinct functionalities, which limit the effectiveness of cancer drugs. Understanding the regulatory mechanisms driving subclonal variation within therapy-resistant tumors can provide novel treatment options to overcome resistance to current therapies.
Tumors are composed of subpopulations of cancer cells with functionally distinct features. Intratumoral heterogeneity limits the therapeutic effectiveness of cancer drugs. To address this issue, it is important to understand the regulatory mechanisms driving a subclonal variety within a therapy-resistant tumor. We identified tumor subclones of HN9 head and neck cancer cells showing distinct responses to radiation with different levels of p62 expression. Genetically identical grounds but epigenetic heterogeneity of the p62 promoter regions revealed that radioresistant HN9-R clones displayed low p62 expression via the creation of repressive chromatin architecture, in which cooperation between DNMT1 (DNA methyltransferases 1) and HDAC1 (histone deacetylases 1) resulted in DNA methylation and repressive H3K9me3 and H3K27me3 marks in the p62 promoter. Combined inhibition of DNMT1 and HDAC1 by genetic depletion or inhibitors enhanced the suppressive effects on proliferative capacity and in vivo tumorigenesis following irradiation. Importantly, ectopically p62-overexpressed HN9-R clones increased the induction of senescence along with p62-dependent autophagy activation. These results demonstrate the heterogeneous expression of p62 as the key component of clonal variation within a tumor against irradiation. Understanding the epigenetic diversity of p62 heterogeneity among subclones allows for improved identification of the functional state of subclones and provides a novel treatment option to resolve resistance to current therapies.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据