4.8 Article

Histone demethylase PHF8 promotes epithelial to mesenchymal transition and breast tumorigenesis

期刊

NUCLEIC ACIDS RESEARCH
卷 45, 期 4, 页码 1687-1702

出版社

OXFORD UNIV PRESS
DOI: 10.1093/nar/gkw1093

关键词

-

资金

  1. Department of Anatomy and Cell Biology
  2. Carver College of Medicine
  3. University of Iowa
  4. Carver Trust Young Investigator Award from Roy J. Carver Charitable Trust [01-224]
  5. The National Institutes of Health
  6. CTSA through ICTS (Institute for Clinical and Translational Science) at University of Iowa [UL1RR024979]
  7. ACSIRG seed grant [IRG-77-004-34]
  8. American Cancer Society
  9. administrated through Holden Comprehensive Cancer Center at University of Iowa
  10. Breast Cancer Research Award
  11. Holden Comprehensive Cancer Center at the University of Iowa
  12. National Institutes of Health grant [P30 CA86862]
  13. Libraries and Provost's Open Access Fund at University of Iowa

向作者/读者索取更多资源

Histone demethylase PHF8 is upregulated and plays oncogenic roles in various cancers; however, the mechanisms underlying its dysregulation and functions in carcinogenesis remain obscure. Here, we report the novel functions of PHF8 in EMT ( epithelial to mesenchymal transition) and breast cancer development. Genome-wide gene expression analysis revealed that PHF8 overexpression induces an EMTlike process, including the upregulation of SNAI1 and ZEB1. PHF8 demethylates H3K9me1, H3K9me2 and sustains H3K4me3 to prime the transcriptional activation of SNAI1 by TGF- beta signaling. We show that PHF8 is upregulated and positively correlated with MYC at protein levels in breast cancer. MYC posttranscriptionally regulates the expression of PHF8 via the repression of microRNAs. Specifically, miR22 directly targets and inhibits PHF8 expression, and mediates the regulation of PHF8 by MYC and TGF- beta signaling. This novel MYC/microRNAs/PHF8 regulatory axis thus places PHF8 as an important downstream effector of MYC. Indeed, PHF8 contributes to MYC-induced cell proliferation and the expression of EMT-related genes. We also report that PHF8 plays important roles in breast cancer cell migration and tumor growth. These oncogenic functions of PHF8 in breast cancer confer its candidacy as a promising therapeutic target for this disease.

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

4.8
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