Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice

  1. Paula Martínez1,
  2. Maria Thanasoula2,
  3. Purificación Muñoz1,3,
  4. Chunyan Liao2,
  5. Agueda Tejera1,
  6. Carolyn McNees1,
  7. Juana M. Flores4,
  8. Oscar Fernández-Capetillo5,
  9. Madalena Tarsounas2 and
  10. Maria A. Blasco1,6
  1. 1Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid E-28029, Spain;
  2. 2Telomere and Genome Stability Group, The CR-UK/MRC Gray Institute for Radiation Oncology and Biology, Oxford OX3 7DQ, United Kingdom;
  3. 3Epigenetics and Cancer Biology Program (PEBC), Catalan Institute of Oncology (ICO), Barcelona 08907, Spain;
  4. 4Animal Surgery and Medicine Department, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid 28029, Spain;
  5. 5Genetic Instability Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid E-28029, Spain

    Abstract

    The telomere repeat-binding factor 1 (TERF1, referred to hereafter as TRF1) is a component of mammalian telomeres whose role in telomere biology and disease has remained elusive. Here, we report on cells and mice conditionally deleted for TRF1. TRF1-deleted mouse embryonic fibroblasts (MEFs) show rapid induction of senescence, which is concomitant with abundant telomeric γ-H2AX foci and activation of the ATM/ATR downstream checkpoint kinases CHK1 and CHK2. DNA damage foci are rescued by both ATM and ATM/ATR inhibitors, further indicating that both signaling pathways are activated upon TRF1 deletion. Abrogation of the p53 and RB pathways bypasses senescence but leads to chromosomal instability including sister chromatid fusions, chromosome concatenation, and occurrence of multitelomeric signals (MTS). MTS are also elevated in ATR-deficient MEFs or upon treatment with aphidicolin, two conditions known to induce breakage at fragile sites, suggesting that TRF1-depleted telomeres are prone to breakage. To address the impact of these molecular defects in the organism, we deleted TRF1 in stratified epithelia of TRF1Δ/ΔK5-Cre mice. These mice die perinatally and show skin hyperpigmentation and epithelial dysplasia, which are associated with induction of telomere-instigated DNA damage, activation of the p53/p21 and p16 pathways, and cell cycle arrest in vivo. p53 deficiency rescues mouse survival but leads to development of squamous cell carcinomas, demonstrating that TRF1 suppresses tumorigenesis. Together, these results demonstrate that dysfunction of a telomere-binding protein is sufficient to produce severe telomeric damage in the absence of telomere shortening, resulting in premature tissue degeneration and development of neoplastic lesions.

    Keywords

    Footnotes

    • 6 Corresponding author.

      E-MAIL mblasco{at}cnio.es: FAX 34-91-917328028.

    • Article published online ahead of print. Article and publication date are online at http://www.genesdev.org/cgi/doi/10.1101/gad.543509.

    • Supplemental material is available at http://www.genesdev.org.

      • Received June 15, 2009.
      • Accepted July 23, 2009.
    | Table of Contents

    Life Science Alliance