UBE2S interacting with TRIM28 in the nucleus accelerates cell cycle by ubiquitination of p27 to promote hepatocellular carcinoma development

Genomic sequencing analysis of tumors provides potential molecular therapeutic targets for precision medicine. However, identifying a key driver gene or mutation that can be used for hepatocellular carcinoma (HCC) treatment remains difficult. Here, we performed whole-exome sequencing on genomic DNA...

Full description

Saved in:
Bibliographic Details
Published inSignal transduction and targeted therapy Vol. 6; no. 1; p. 64
Main Authors Zhang, Ren-Yu, Liu, Ze-Kun, Wei, Ding, Yong, Yu-Le, Lin, Peng, Li, Hao, Liu, Man, Zheng, Nai-Shan, Liu, Ke, Hu, Cai-Xia, Yang, Xiao-Zhen, Chen, Zhi-Nan, Bian, Huijie
Format Journal Article
LanguageEnglish
Published London Nature Publishing Group UK 16.02.2021
Nature Publishing Group
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Genomic sequencing analysis of tumors provides potential molecular therapeutic targets for precision medicine. However, identifying a key driver gene or mutation that can be used for hepatocellular carcinoma (HCC) treatment remains difficult. Here, we performed whole-exome sequencing on genomic DNA obtained from six pairs of HCC and adjacent tissues and identified two novel somatic mutations of UBE2S (p. Gly57Ala and p. Lys63Asn). Predictions of the functional effects of the mutations showed that two amino-acid substitutions were potentially deleterious. Further, we observed that wild-type UBE2S, especially in the nucleus, was significantly higher in HCC tissues than that in adjacent tissues and closely related to the clinicopathological features of patients with HCC. Functional assays revealed that overexpression of UBE2S promoted the proliferation, invasion, metastasis, and G1/S phase transition of HCC cells in vitro, and promoted the tumor growth significantly in vivo. Mechanistically, UBE2S interacted with TRIM28 in the nucleus, both together enhanced the ubiquitination of p27 to facilitate its degradation and cell cycle progression. Most importantly, the small-molecule cephalomannine was found by a luciferase-based sensitive high-throughput screen (HTS) to inhibit UBE2S expression and significantly attenuate HCC progression in vitro and in vivo, which may represent a promising strategy for HCC therapy.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2059-3635
2095-9907
2059-3635
DOI:10.1038/s41392-020-00432-z