The positive regulatory loop of TCF4N/p65 promotes glioblastoma tumourigenesis and chemosensitivity

Clin Transl Med. 2022 Sep;12(9):e1042. doi: 10.1002/ctm2.1042.

Abstract

Background: NF-κB signaling is widely linked to the pathogenesis and treatment resistance in cancers. Increasing attention has been paid to its anti-oncogenic roles, due to its key functions in cellular senescence and the senescence-associated secretory phenotype (SASP). Therefore, thoroughly understanding the function and regulation of NF-κB in cancers is necessary prior to the application of NF-κB inhibitors.

Methods: We established glioblastoma (GBM) cell lines expressing ectopic TCF4N, an isoform of the β-catenin interacting transcription factor TCF7L2, and evaluated its functions in GBM tumorigenesis and chemotherapy in vitro and in vivo. In p65 knock-out or phosphorylation mimic (S536D) cell lines, the dual role and correlation of TCF4N and NF-κB signaling in promoting tumorigenesis and chemosensitivity was investigated by in vitro and in vivo functional experiments. RNA-seq and computational analysis, immunoprecipitation and ubiquitination assay, minigene splicing assay and luciferase reporter assay were performed to identify the underlying mechanism of positive feedback regulation loop between TCF4N and the p65 subunit of NF-κB. A eukaryotic cell-penetrating peptide targeting TCF4N, 4N, was used to confirm the therapeutic significance.

Results: Our results indicated that p65 subunit phosphorylation at Ser 536 (S536) and nuclear accumulation was a promising prognostic marker for GBM, and endowed the dual functions of NF-κB in promoting tumorigenesis and chemosensitivity. p65 S536 phosphorylation and nuclear stability in GBM was regulated by TCF4N. TCF4N bound p65, induced p65 phosphorylation and nuclear translocation, inhibited its ubiquitination/degradation, and subsequently promoted NF-κB activity. p65 S536 phosphorylation was essential for TCF4N-led senescence-independent SASP, GBM tumorigenesis, tumor stem-like cell differentiation and chemosensitivity. Activation of p65 was closely connected to alterative splicing of TCF4N, a likely positive feedback regulation loop between TCF4N and p65 in GBM. 4N increased chemosensitivity, highlighting a novel anti-cancer strategy.

Conclusion: Our study defined key roles of TCF4N as a novel regulator of NF-κB through mutual regulation with p65 and provided a new avenue for GBM inhibition.

Keywords: TCF4N; chemosensitivity; nuclear translocation; p65; stability; tumourigenesis; ubiquitination.

MeSH terms

  • Carcinogenesis / genetics
  • Cell Line, Tumor
  • Cell Transformation, Neoplastic
  • Cell-Penetrating Peptides
  • Central Nervous System Neoplasms* / drug therapy
  • Central Nervous System Neoplasms* / genetics
  • Central Nervous System Neoplasms* / metabolism
  • Drug Resistance, Neoplasm / genetics
  • Drug Resistance, Neoplasm / physiology
  • Glioblastoma* / drug therapy
  • Glioblastoma* / genetics
  • Glioblastoma* / metabolism
  • Humans
  • Luciferases
  • NF-kappa B / genetics
  • NF-kappa B / metabolism
  • Transcription Factor 7-Like 2 Protein* / genetics
  • Transcription Factor 7-Like 2 Protein* / metabolism
  • Transcription Factor RelA* / genetics
  • Transcription Factor RelA* / metabolism
  • beta Catenin

Substances

  • Cell-Penetrating Peptides
  • NF-kappa B
  • RELA protein, human
  • TCF7L2 protein, human
  • Transcription Factor 7-Like 2 Protein
  • Transcription Factor RelA
  • beta Catenin
  • Luciferases