Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

BRCA2 acts as a RAD51 loader to facilitate telomere replication and capping

Abstract

The tumor suppressor protein BRCA2 is a key component of the homologous recombination pathway of DNA repair, acting as the loader of RAD51 recombinase at sites of double-strand breaks. Here we show that BRCA2 associates with telomeres during the S and G2 phases of the cell cycle and facilitates the loading of RAD51 onto telomeres. Conditional deletion of Brca2 and inhibition of Rad51 in mouse embryonic fibroblasts (MEFs), but not inactivation of Brca1, led to shortening of telomeres and accumulation of fragmented telomeric signals—a hallmark of telomere fragility that is associated with replication defects. These findings suggest that BRCA2-mediated homologous recombination reactions contribute to the maintenance of telomere length by facilitating telomere replication and imply that BRCA2 has an essential role in maintaining telomere integrity during unchallenged cell proliferation. Mouse mammary tumors that lacked Brca2 accumulated telomere dysfunction–induced foci. Human breast tumors in which BRCA2 was mutated had shorter telomeres than those in which BRCA1 was mutated, suggesting that the genomic instability in BRCA2-deficient tumors was due in part to telomere dysfunction.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: BRCA2 is required to recruit RAD51 to the telomeres during the S and G2 phases.
Figure 2: Conditional deletion of Brca2 causes telomere shortening.
Figure 3: BRCA2 and RAD51 are required for maintenance of telomere length in MEFs.
Figure 4: Increased telomere fragility in BRCA2- and RAD51-deficient MEFs.
Figure 5: Homologous recombination activities and the telomeric factor TRF1 act independently in facilitating telomere replication.
Figure 6: Telomere dysfunction–induced foci (TIFs) accumulate in BRCA2- and RAD51-deficient MEFs and in Brca2-deficient mammary tumors.
Figure 7: The function of BRCA2, but not that of BRCA1, is essential for telomere maintenance in human breast tumors.

Similar content being viewed by others

References

  1. Narod, S.A. & Foulkes, W.D. BRCA1 and BRCA2: 1994 and beyond. Nat. Rev. Cancer 4, 665–676 (2004).

    Article  CAS  Google Scholar 

  2. Evers, B. & Jonkers, J. Mouse models of BRCA1 and BRCA2 deficiency: past lessons, current understanding and future prospects. Oncogene 25, 5885–5897 (2006).

    Article  CAS  Google Scholar 

  3. Thorslund, T. & West, S.C. BRCA2: a universal recombinase regulator. Oncogene 26, 7720–7730 (2007).

    Article  CAS  Google Scholar 

  4. Venkitaraman, A.R. Linking the cellular functions of BRCA genes to cancer pathogenesis and treatment. Annu. Rev. Pathol. 4, 461–487 (2009).

    Article  CAS  Google Scholar 

  5. Nagaraju, G. & Scully, R. Minding the gap: the underground functions of BRCA1 and BRCA2 at stalled replication forks. DNA Repair (Amst.) 6, 1018–1031 (2007).

    Article  CAS  Google Scholar 

  6. Tsang, E. & Carr, A.M. Replication fork arrest, recombination and the maintenance of ribosomal DNA stability. DNA Repair (Amst.) 7, 1613–1623 (2008).

    Article  CAS  Google Scholar 

  7. Henson, J.D., Neumann, A.A., Yeager, T.R. & Reddel, R.R. Alternative lengthening of telomeres in mammalian cells. Oncogene 21, 598–610 (2002).

    Article  CAS  Google Scholar 

  8. Cesare, A.J. & Reddel, R.R. Telomere uncapping and alternative lengthening of telomere. Mech. Ageing Dev. 129, 99–108 (2008).

    Article  CAS  Google Scholar 

  9. Verdun, R.E. & Karlseder, J. The DNA damage machinery and homologous recombination pathway act consecutively to protect human telomeres. Cell 127, 709–720 (2006).

    Article  CAS  Google Scholar 

  10. West, S.C. Molecular views of recombination proteins and their control. Nat. Rev. Mol. Cell Biol. 4, 1–11 (2003).

    Article  Google Scholar 

  11. Thacker, J. & Zdzienicka, M.Z. The mammalian XRCC genes: their roles in DNA repair and genetic stability. DNA Repair (Amst.) 2, 655–672 (2003).

    Article  CAS  Google Scholar 

  12. Badie, S. et al. RAD51C facilitates checkpoint signalling by promoting CHK2 phosphorylation. J. Cell Biol. 185, 587–600 (2009).

    Article  CAS  Google Scholar 

  13. Palm, W. & de Lange, T. How shelterin protects mammalian telomeres. Annu. Rev. Genet. 42, 16.1–16.34 (2008).

    Article  Google Scholar 

  14. Tarsounas, M. et al. Telomere maintenance requires the RAD51D recombination/repair protein. Cell 117, 337–347 (2004).

    Article  CAS  Google Scholar 

  15. Jaco, I. et al. Role of mammalian Rad54 in telomere length maintenance. Mol. Cell. Biol. 23, 5572–5580 (2003).

    Article  CAS  Google Scholar 

  16. Tarsounas, M. & West, S.C. Recombination at mammalian telomeres: an alternative mechanism for telomere protection and elongation. Cell Cycle 4, 672–674 (2005).

    Article  CAS  Google Scholar 

  17. Maser, R.S. & DePinho, R.A. Telomeres and the DNA damage response: why the fox is guarding the henhouse. DNA Repair (Amst.) 8–9, 979–988 (2004).

    Article  Google Scholar 

  18. Maser, R.S. & DePinho, R.A. Connecting chromosomes, crisis, and cancer. Science 297, 565–569 (2002).

    Article  CAS  Google Scholar 

  19. Jacobs, J.J.L. et al. Senescence bypass screen identifies TBX2, which represses Cdkn2a(p19ARF) and is amplified in a subset of human breast cancers. Nat. Genet. 26, 291–299 (2000).

    Article  CAS  Google Scholar 

  20. Silver, D.P. & Livingston, D.M. Self-excising retroviral vectors encoding the Cre recombinase overcome Cre-mediated cellular toxicity. Mol. Cell 8, 233–243 (2001).

    Article  CAS  Google Scholar 

  21. Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat. Genet. 29, 418–425 (2001).

    Article  CAS  Google Scholar 

  22. Herrera, E. et al. Disease states associated to telomere deficiency appear earlier in mice with short telomeres. EMBO J. 18, 2950–2960 (1999).

    Article  CAS  Google Scholar 

  23. Blasco, M.A. et al. Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 91, 25–34 (1997).

    Article  CAS  Google Scholar 

  24. Muñoz, P., Blanco, R., Flores, J.M. & Blasco, M.A. XPF nuclease-dependent telomere loss and increased DNA damage in mice overexpressing TRF2 result in premature aging and cancer. Nat. Genet. 37, 1063–1071 (2005).

    Article  Google Scholar 

  25. Blanco, R., Munoz, P., Flores, J.M., Klatt, P. & Blasco, M.A. Telomerase abrogation dramatically accelerates TRF2-induced epithelial carcinogenesis. Genes Dev. 21, 206–220 (2007).

    Article  CAS  Google Scholar 

  26. Okamoto, K., Iwano, T., Tachibana, M. & Shinkai, Y. Distinct roles of TRF1 in the regulation of telomere structure and lengthening. J. Biol. Chem. 283, 23981–23988 (2008).

    Article  CAS  Google Scholar 

  27. Sfeir, A. et al. Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell 138, 90–103 (2009).

    Article  CAS  Google Scholar 

  28. Martínez, P. et al. Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice. Genes Dev. 23, 2060–2075 (2009).

    Article  Google Scholar 

  29. Durkin, S.G. & Glover, T.W. Chromosome fragile sites. Annu. Rev. Genet. 41, 169–192 (2007).

    Article  CAS  Google Scholar 

  30. d'Adda di Fagagna, F. et al. A DNA damage checkpoint response in telomere-initiated senescence. Nature 426, 194–198 (2003).

    Article  CAS  Google Scholar 

  31. Takai, H., Smogorzewska, A. & de Lange, T. DNA damage foci at dysfunctional telomeres. Curr. Biol. 13, 1549–1556 (2003).

    Article  CAS  Google Scholar 

  32. Bouwman, P. et al. 53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers. Nat. Struct. Mol. Biol. 17, 688–695 (2010).

    Article  CAS  Google Scholar 

  33. Palacios, J. et al. Immunohistochemical characteristics defined by tissue microarray of hereditary breast cancer not attributable to BRCA1 or BRCA2 mutations: differences from breast carcinomas arising in BRCA1 and BRCA2 mutation carriers. Clin. Cancer Res. 9, 3606–3614 (2003).

    CAS  PubMed  Google Scholar 

  34. Connor, F. et al. Tumorigenesis and a DNA-repair defect in mice with a truncating BRCA2 mutation. Nat. Genet. 17, 423–430 (1997).

    Article  CAS  Google Scholar 

  35. Patel, K.J. et al. Involvement of BRCA2 in DNA repair. Mol. Cell 1, 347–357 (1998).

    Article  CAS  Google Scholar 

  36. Bugreev, D.V., Mazina, O.M. & Mazin, A.V. Rad54 protein promotes branch migration of Holliday junctions. Nature 442, 590–593 (2006).

    Article  CAS  Google Scholar 

  37. Miller, K.M., Rog, O. & Cooper, J.P. Semi-conservative DNA replication through telomeres requires Taz1. Nature 440, 824–828 (2006).

    Article  CAS  Google Scholar 

  38. Karlseder, J. Telomeric proteins: clearing the way for the replication fork. Nat. Struct. Mol. Biol. 13, 386–387 (2006).

    Article  CAS  Google Scholar 

  39. Ye, J. et al. TRF2 and apollo cooperate with topoisomerase 2α to protect human telomeres from replicative damage. Cell 142, 230–242 (2010).

    Article  CAS  Google Scholar 

  40. Kinzler, K.W. & Vogelstein, B. Cancer-susceptibility genes: gatekeepers and caretakers. Nature 386, 761 (1997).

    Article  CAS  Google Scholar 

  41. Meeker, A.K. & Argani, P. Telomere shortening occurs early during breast tumorigenesis: a cause of chromosome destabilization underlying malignant transformation? J. Mammary Gland Biol. Neoplasia 9, 285–296 (2004).

    Article  Google Scholar 

  42. Jacks, T. et al. Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4, 1–7 (1994).

    Article  CAS  Google Scholar 

  43. Dirac, A.M. & Bernards, R. Reversal of senescence in mouse fibroblasts through lentiviral suppression of p53. J. Biol. Chem. 278, 11731–11734 (2003).

    Article  CAS  Google Scholar 

  44. Thanasoula, M. et al. p53 prevents entry into mitosis with uncapped telomeres. Curr. Biol. 20, 521–526 (2010).

    Article  CAS  Google Scholar 

  45. Tarsounas, M., Davies, D. & West, S.C. BRCA2-dependent and independent formation of RAD51 nuclear foci. Oncogene 22, 1115–1123 (2003).

    Article  CAS  Google Scholar 

  46. Herbig, U., Jobling, W.A., Chen, B.P., Chen, D.J. & Sedivy, J.M. Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol. Cell 14, 501–513 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Biggs for generating the targeting construct for the Brca2sko allele, I. Roswell (Cancer Research UK, Clare Hall Laboratories) for help with establishing Trp53−/− MEFs and F. Uhlmann for reading and comments on the manuscript. Work in M. Tarsounas's laboratory is supported by Cancer Research UK and Breast Cancer Campaign. Travel related to this project was funded by a Joint International Award to M. Tarsounas and M.A.B. from The Royal Society. Work in J.J.'s laboratory is supported by the Dutch Cancer Society (KWF) and the Netherlands Organisation for Scientific Research (NWO). M.A.B.'s laboratory was funded by the Spanish Ministry of Innovation and Science. U.H. was supported by the New Jersey Commission on Cancer Research (NJCCR) grant no. 09-1124-CCR-EO.

Author information

Authors and Affiliations

Authors

Contributions

M. Tarsounas, S.B. and J.M.E. designed and planned the experiments. S.B. and J.M.E. performed most of the experiments. P.B. and J.J. generated the Brca2sko conditional mouse model, established immortalized MEFs and contributed to the results in Figure 2a. A.R.C. contributed the results in Figure 1a,b and Supplementary Figure 6a. M. Thanasoula performed the IF-FISH experiments in Figure 6a–c, Supplementary Figure 1d and Supplementary Figure 6b,c. M.M.G., J.B. and M.A.B. performed the experiments in Figure 7b,c. A.S. and U.H. contributed to the results in Figure 6d–f. I.J. designed and validated the shRNA against mouse RAD51. M. Tarsounas made the figures and wrote the paper.

Corresponding author

Correspondence to Madalena Tarsounas.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Methods (PDF 775 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Badie, S., Escandell, J., Bouwman, P. et al. BRCA2 acts as a RAD51 loader to facilitate telomere replication and capping. Nat Struct Mol Biol 17, 1461–1469 (2010). https://doi.org/10.1038/nsmb.1943

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.1943

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer