Multi-Level Control of the ATM/ATR-CHK1 Axis by the Transcription Factor E4F1 in Triple-Negative Breast Cancer

Int J Mol Sci. 2022 Aug 16;23(16):9217. doi: 10.3390/ijms23169217.

Abstract

E4F1 is essential for early embryonic mouse development and for controlling the balance between proliferation and survival of actively dividing cells. We previously reported that E4F1 is essential for the survival of murine p53-deficient cancer cells by controlling the expression of genes involved in mitochondria functions and metabolism, and in cell-cycle checkpoints, including CHEK1, a major component of the DNA damage and replication stress responses. Here, combining ChIP-Seq and RNA-Seq approaches, we identified the transcriptional program directly controlled by E4F1 in Human Triple-Negative Breast Cancer cells (TNBC). E4F1 binds and regulates a limited list of direct target genes (57 genes) in these cells, including the human CHEK1 gene and, surprisingly, also two other genes encoding post-transcriptional regulators of the ATM/ATR-CHK1 axis, namely, the TTT complex component TTI2 and the phosphatase PPP5C, that are essential for the folding and stability, and the signaling of ATM/ATR kinases, respectively. Importantly, E4F1 also binds the promoter of these genes in vivo in Primary Derived Xenograft (PDX) of human TNBC. Consequently, the protein levels and signaling of CHK1 but also of ATM/ATR kinases are strongly downregulated in E4F1-depleted TNBC cells resulting in a deficiency of the DNA damage and replicative stress response in these cells. The E4F1-depleted cells fail to arrest into S-phase upon treatment with the replication-stalling agent Gemcitabine, and are highly sensitized to this drug, as well as to other DNA-damaging agents, such as Cisplatin. Altogether, our data indicate that in breast cancer cells the ATM/ATR-CHK1 signaling pathway and DNA damage-stress response are tightly controlled at the transcriptional and post-transcriptional level by E4F1.

Keywords: ATM/ATR-CHK checkpoint pathway; DNA Damage Response (DDR); E4F1 transcription factor; chemotherapy; triple-negative breast cancer.

MeSH terms

  • Animals
  • Ataxia Telangiectasia Mutated Proteins / metabolism
  • Cell Cycle Proteins / metabolism
  • Checkpoint Kinase 1 / metabolism
  • DNA Damage
  • DNA-Binding Proteins / metabolism
  • Humans
  • Mice
  • Phosphorylation
  • Protein Kinases / metabolism
  • Repressor Proteins* / metabolism
  • Transcription Factors* / metabolism
  • Triple Negative Breast Neoplasms* / metabolism
  • Ubiquitin-Protein Ligases* / metabolism

Substances

  • Cell Cycle Proteins
  • DNA-Binding Proteins
  • Repressor Proteins
  • Transcription Factors
  • E4F1 protein, human
  • E4f1 protein, mouse
  • Ubiquitin-Protein Ligases
  • Protein Kinases
  • ATM protein, human
  • ATR protein, human
  • Ataxia Telangiectasia Mutated Proteins
  • CHEK1 protein, human
  • Checkpoint Kinase 1

Grants and funding

This work was supported by grants to Claude Sardet and Laurent Le Cam by the Agence Nationale pour la Recherche (ANR PyrE4F 2018 N°RPV18008FFA), the Fondation pour la Recherche contre le Cancer ARC (Claude Sardet). The Canceropole GSO, Synergie program (Claude Sardet). Institutional support was provided by the Institut National de la Santé et de la Recherche Médicale INSERM, the Montpellier University, the Institut du Cancer de Montpellier ICM and the Centre National de la Recherche Scientifique CNRS (to Claude Sardet and Geneviève Rodier) and the SIRIC Montpellier Cancer (Grant InCa-DGOS-INSERM6045). Kalil Batnini, Thibault Houles and Mehdi Zaroual were supported by fellowships from the Labex EpiGenMed (Labex EpiGenMed (ANR-10-LABX-12-01), Ligue Contre le Cancer and FHU Evocan, respectively.