Epigenetic modifications promote the expression of the orphan nuclear receptor NR0B1 in human lung adenocarcinoma cells

2016 
// Yongjie Lu 1, * , Yunqiang Liu 1, * , Shunyao Liao 2, * , Wenling Tu 1 , Ying Shen 1 , Yuanlong Yan 1 , Dachang Tao 1 , Yilu Lu 1 , Yongxin Ma 1 , Yuan Yang 1 , Sizhong Zhang 1 1 Department of Medical Genetics and Division of Human Morbid Genomics, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China 2 Diabetic Center and Institute of Transplantation, Sichuan Academy of Medical Science & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China * These authors have contributed equally to this work Correspondence to: Yunqiang Liu, email: yq_liu@scu.edu.cn Keywords: NR0B1, lung adenocarcinoma, DNA methylation, histone modifications, cell self-renewal Received: March 03, 2016      Accepted: April 15, 2016      Published: April 26, 2016 ABSTRACT The ectopic activation of NR0B1 is involved in the development of some cancers. However, the regulatory mechanisms controlling NR0B1 expression are not well understood. Therefore, the epigenetic modifications promoting NR0B1 activation were examined in this study. NR0B1 protein was detected in cancerous tissues of more than 50% of human lung adenocarcinoma (ADCA) cases and tended to be expressed in low-differentiated cancerous tissues obtained from males. Nevertheless, NR0B1 activation in ADCA has not previously been correlated with DNA demethylation. NR0B1 expression was not detected in 293T cells, although it contains a hypomethylated NR0B1 promoter. Treating 293T cells with a histone deacetylase inhibitor increased acetylated histone H4 binding to the NR0B1 promoter and activated NR0B1 expression. In contrast, treatment with histone methylase inhibitors decreased the methylation of histones H3K9 and H3K27 and slightly induced NR0B1 transcription. Furthermore, the level of acetyl-histone H4 binding to the NR0B1 promoter increased, whereas the occupancy of H3K27me3 was lower in cancerous tissues than in non-cancerous tissues. Similar histone occupancies were confirmed in a comparison of cancerous tissues with strong, moderate and negative NR0B1 expression. In conclusion, this study shows that CpG methylation within the NR0B1 promoter is not involved in the in vivo regulation of NR0B1 expression, whereas the hyperacetylation of histone H4 and the unmethylation of histones H3K9 and H3K27, and their binding to the NR0B1 promoter results in decondensed euchromatin for NR0B1 activation.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    44
    References
    5
    Citations
    NaN
    KQI
    []