Journal of Experimental & Clinical Cancer Research (Dec 2022)

The A-to-I editing of KPC1 promotes intrahepatic cholangiocarcinoma by attenuating proteasomal processing of NF-κB1 p105 to p50

  • Chengming Gao,
  • Guangming Zhou,
  • Jie Shi,
  • Peipei Shi,
  • Liang Jin,
  • Yuanfeng Li,
  • Xiaowen Wang,
  • Song Liao,
  • Han Yan,
  • Junjie Wu,
  • Yiming Lu,
  • Yun Zhai,
  • Jinxu Zhang,
  • Haitao Zhang,
  • Hongxing Zhang,
  • Chenning Yang,
  • Pengbo Cao,
  • Shuqun Cheng,
  • Gangqiao Zhou

DOI
https://doi.org/10.1186/s13046-022-02549-1
Journal volume & issue
Vol. 41, no. 1
pp. 1 – 20

Abstract

Read online

Abstract Background Aberrant RNA editing of adenosine-to-inosine (A-to-I) has been linked to multiple human cancers, but its role in intrahepatic cholangiocarcinoma (iCCA) remains unknown. We conducted an exome-wide investigation to search for dysregulated RNA editing that drive iCCA pathogenesis. Methods An integrative whole-exome and transcriptome sequencing analysis was performed to elucidate the RNA editing landscape in iCCAs. Putative RNA editing sites were validated by Sanger sequencing. In vitro and in vivo experiments were used to assess the effects of an exemplary target gene Kip1 ubiquitination-promoting complex 1 (KPC1) and its editing on iCCA cells growth and metastasis. Crosstalk between KPC1 RNA editing and NF-κB signaling was analyzed by molecular methods. Results Through integrative omics analyses, we revealed an adenosine deaminases acting on RNA 1A (ADAR1)-mediated over-editing pattern in iCCAs. ADAR1 is frequently amplified and overexpressed in iCCAs and plays oncogenic roles. Notably, we identified a novel ADAR1-mediated A-to-I editing of KPC1 transcript, which results in substitution of methionine with valine at residue 8 (p.M8V). KPC1 p.M8V editing confers loss-of-function phenotypes through blunting the tumor-suppressive role of wild-type KPC1. Mechanistically, KPC1 p.M8V weakens the affinity of KPC1 to its substrate NF-κB1 p105, thereby reducing the ubiquitinating and proteasomal processing of p105 to p50, which in turn enhances the activity of oncogenic NF-κB signaling. Conclusions Our findings established that amplification-driven ADAR1 overexpression results in overediting of KPC1 p.M8V in iCCAs, leading to progression via activation of the NF-κB signaling pathway, and suggested ADAR1-KPC1-NF-κB axis as a potential therapeutic target for iCCA.

Keywords