Mutant p53 drives the loss of heterozygosity by the upregulation of Nek2 in breast cancer cells

Breast Cancer Res. 2020 Dec 2;22(1):133. doi: 10.1186/s13058-020-01370-y.

Abstract

Background: Mutations in one allele of the TP53 gene in early stages are frequently followed by the loss of the remaining wild-type p53 (wtp53) allele (p53LOH) during tumor progression. Despite the strong notion of p53LOH as a critical step in tumor progression, its oncogenic outcomes that facilitate the selective pressure for p53LOH occurrence were not elucidated.

Methods: Using MMTV;ErbB2 mouse model of breast cancer carrying heterozygous R172H p53 mutation, we identified a novel gain-of-function (GOF) activity of mutant p53 (mutp53): the exacerbated loss of wtp53 allele in response to γ-irradiation.

Results: As consequences of p53LOH in mutp53 heterozygous cells, we observed profound stabilization of mutp53 protein, the loss of p21 expression, the abrogation of G2/M checkpoint, chromosomal instability, centrosome amplification, and transcriptional upregulation of mitotic kinase Nek2 (a member of Never in Mitosis (NIMA) Kinases family) involved in the regulation of centrosome function. To avoid the mitotic catastrophe in the absence of G2/M checkpoint, cells with centrosome amplification adapt Nek2-mediated centrosomes clustering as pro-survival mutp53 GOF mechanism enabling unrestricted proliferation and clonal expansion of cells with p53LOH. Thus, the clonal dominance of mutp53 cells with p53LOH may represent the mechanism of irradiation-induced p53LOH. We show that pharmacological and genetic ablation of Nek2 decreases centrosome clustering and viability of specifically mutp53 cells with p53LOH.

Conclusion: In a heterogeneous tumor population, Nek2 inhibition may alter the selective pressure for p53LOH by contraction of the mutp53 population with p53LOH, thus, preventing the outgrowth of genetically unstable, more aggressive cells.

Keywords: Breast cancer; Centrosome clustering; Mutant p53; Nek2, LOH.

Publication types

  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Breast Neoplasms / genetics*
  • Breast Neoplasms / pathology
  • Cell Line, Tumor
  • Centrosome / metabolism
  • Chromosomal Instability
  • Datasets as Topic
  • Disease Models, Animal
  • Female
  • G2 Phase Cell Cycle Checkpoints / genetics
  • Gain of Function Mutation
  • Gene Expression Regulation, Neoplastic / drug effects
  • Humans
  • Loss of Heterozygosity / drug effects
  • Loss of Heterozygosity / genetics*
  • Mice
  • Mice, Transgenic
  • NIMA-Related Kinases / antagonists & inhibitors
  • NIMA-Related Kinases / genetics*
  • Receptor, ErbB-2 / genetics
  • Transcriptional Activation
  • Tumor Suppressor Protein p53 / genetics*
  • Tumor Suppressor Protein p53 / metabolism
  • Up-Regulation

Substances

  • TP53 protein, human
  • Trp53 protein, mouse
  • Tumor Suppressor Protein p53
  • ERBB2 protein, human
  • Erbb2 protein, rat
  • Receptor, ErbB-2
  • NEK2 protein, human
  • NIMA-Related Kinases
  • Nek2 protein, mouse