CircNFIB inhibits tumor growth and metastasis through suppressing MEK1/ERK signaling in intrahepatic cholangiocarcinoma

Mol Cancer. 2022 Jan 17;21(1):18. doi: 10.1186/s12943-021-01482-9.

Abstract

Background: Considerable evidence shows that circular RNAs (circRNAs) play an important role in tumor development. However, their function in intrahepatic cholangiocarcinoma (ICC) metastasis and the underlying mechanisms are incompletely understood.

Methods: circNFIB (hsa_circ_0086376, termed as cNFIB hereafter) was identified in human ICC tissues through circRNAs sequencing. The biological role of cNFIB was determined in vitro and in vivo by gain or loss of functional experiments. Fluorescence in situ hybridization (FISH), RNA immunoprecipitation (RIP) and RNA pull-down assays were conducted to analyze the interaction of cNFIB with dual specificity mitogen-activated protein kinase kinase1 (MEK1). Duolink in situ proximity ligation assay (PLA) and coimmunoprecipitation (co-IP) assay were used to investigate the effects of cNFIB on the interaction between MEK1 and mitogen-activated protein kinase 2 (ERK2). Finally, a series of in vitro and in vivo experiments were performed to explore the influences of cNFIB on the anti-tumor activity of trametinib (a MEK inhibitor).

Results: cNFIB was significantly down-regulated in human ICC tissues with postoperative metastases. The loss of cNFIB was highly associated with aggressive characteristics and predicted unfavorable prognosis in ICC patients. Functional studies revealed that cNFIB inhibited the proliferation and metastasis of ICC cells in vitro and in vivo. Mechanistically, cNFIB competitively interacted with MEK1, which induced the dissociation between MEK1 and ERK2, thereby resulting in the suppression of ERK signaling and tumor metastasis. Moreover, we found that ICC cells with high levels of cNFIB held the potential to delay the trametinib resistance. Consistently, in vivo and in vitro studies demonstrated that cotreatment with trametinib and lentivirus vector encoding cNFIB showed greater inhibitory effect than isolated trametinib treatment.

Conclusions: Our findings identified that cNFIB played a key role in ICC growth and metastasis by regulating MEK1/ERK signaling. Given the efficacy of cNFIB modulation on ICC suppression and trametinib sensitivity, cNFIB appears to be a potential therapeutic molecule for ICC treatment.

Keywords: Circular RNA; Dual specificity mitogen-activated protein kinase kinase 1 (MEK1); Intrahepatic cholangiocarcinoma (ICC); Metastasis; Trametinib.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adult
  • Aged
  • Animals
  • Bile Duct Neoplasms / diagnosis
  • Bile Duct Neoplasms / etiology*
  • Bile Duct Neoplasms / metabolism*
  • Bile Duct Neoplasms / mortality
  • Biomarkers, Tumor
  • Cell Line, Tumor
  • Cholangiocarcinoma / diagnosis
  • Cholangiocarcinoma / etiology*
  • Cholangiocarcinoma / metabolism*
  • Cholangiocarcinoma / mortality
  • Disease Models, Animal
  • Disease Susceptibility
  • Female
  • Gene Expression Regulation, Neoplastic*
  • Humans
  • MAP Kinase Signaling System*
  • Male
  • Mice
  • Middle Aged
  • NFI Transcription Factors / genetics*
  • Neoplasm Metastasis
  • Neoplasm Staging
  • Prognosis
  • RNA, Circular*
  • Xenograft Model Antitumor Assays

Substances

  • Biomarkers, Tumor
  • NFI Transcription Factors
  • NFIB protein, human
  • RNA, Circular