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:

Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells

Abstract

Rare cells with the properties of stem cells are integral to the development and perpetuation of leukaemias. A defining characteristic of stem cells is their capacity to self-renew, which is markedly extended in leukaemia stem cells. The underlying molecular mechanisms, however, are largely unknown. Here we demonstrate that expression of the cell-cycle inhibitor p21 is indispensable for maintaining self-renewal of leukaemia stem cells. Expression of leukaemia-associated oncogenes in mouse haematopoietic stem cells (HSCs) induces DNA damage and activates a p21-dependent cellular response, which leads to reversible cell-cycle arrest and DNA repair. Activated p21 is critical in preventing excess DNA-damage accumulation and functional exhaustion of leukaemic stem cells. These data unravel the oncogenic potential of p21 and suggest that inhibition of DNA repair mechanisms might function as potent strategy for the eradication of the slowly proliferating leukaemia stem cells.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: p21 maintains self-renewal of PML–RAR-expressing HSCs.
Figure 2: Fusion proteins induce DNA damage.
Figure 3: p21 upregulation and cell-cycle restriction in HSCs.
Figure 4: p21 limits DNA damage in LSCs and is indispensable for leukaemogenesis.
Figure 5: Effects of fusion proteins on progenitors and fibroblasts.

Similar content being viewed by others

References

  1. Harrison, D. E. Long-term erythropoietic repopulating ability of old, young, and fetal stem cells. J. Exp. Med. 157, 1496–1504 (1983)

    Article  CAS  PubMed  Google Scholar 

  2. Harrison, D. E. & Astle, C. M. Loss of stem cell repopulating ability upon transplantation. Effects of donor age, cell number, and transplantation procedure. J. Exp. Med. 156, 1767–1779 (1982)

    Article  CAS  PubMed  Google Scholar 

  3. Shepherd, B. E. et al. Hematopoietic stem cell behavior in non-human primates. Blood 110, 1806–1813 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cheng, T. Cell cycle inhibitors in normal and tumor stem cells. Oncogene 23, 7256–7266 (2004)

    Article  CAS  PubMed  Google Scholar 

  5. Cheng, T. et al. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287, 1804–1808 (2000)

    Article  ADS  CAS  PubMed  Google Scholar 

  6. Bonnet, D. & Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nature Med. 3, 730–737 (1997)

    Article  CAS  PubMed  Google Scholar 

  7. Hope, K. J., Jin, L. & Dick, J. E. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nature Immunol. 5, 738–743 (2004)

    Article  CAS  Google Scholar 

  8. Warner, J. K., Wang, J. C., Hope, K. J., Jin, L. & Dick, J. E. Concepts of human leukemic development. Oncogene 23, 7164–7177 (2004)

    Article  CAS  PubMed  Google Scholar 

  9. Minucci, S. et al. PML–RAR induces promyelocytic leukemias with high efficiency following retroviral gene transfer into purified murine hematopoietic progenitors. Blood 100, 2989–2995 (2002)

    Article  CAS  PubMed  Google Scholar 

  10. Westervelt, P. et al. High-penetrance mouse model of acute promyelocytic leukemia with very low levels of PML–RARα expression. Blood 102, 1857–1865 (2003)

    Article  CAS  PubMed  Google Scholar 

  11. Christensen, J. L. & Weissman, I. L. Flk-2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-term stem cells. Proc. Natl Acad. Sci. USA 98, 14541–14546 (2001)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–245 (1996)

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Yang, L. et al. Identification of Lin(-)Sca1(+)kit(+)CD34(+)Flt3- short-term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients. Blood 105, 2717–2723 (2005)

    Article  CAS  PubMed  Google Scholar 

  14. Szilvassy, S. J., Humphries, R. K., Lansdorp, P. M., Eaves, A. C. & Eaves, C. J. Quantitative assay for totipotent reconstituting hematopoietic stem cells by a competitive repopulation strategy. Proc. Natl Acad. Sci. USA 87, 8736–8740 (1990)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Szilvassy, S. J., Lansdorp, P. M., Humphries, R. K., Eaves, A. C. & Eaves, C. J. Isolation in a single step of a highly enriched murine hematopoietic stem cell population with competitive long-term repopulating ability. Blood 74, 930–939 (1989)

    CAS  PubMed  Google Scholar 

  16. Janzen, V. et al. Stem-cell ageing modified by the cyclin-dependent kinase inhibitor p16INK4a. Nature 443, 421–426 (2006)

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Passegue, E., Wagers, A. J., Giuriato, S., Anderson, W. C. & Weissman, I. L. Global analysis of proliferation and cell cycle gene expression in the regulation of hematopoietic stem and progenitor cell fates. J. Exp. Med. 202, 1599–1611 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rossi, D. J. et al. Cell intrinsic alterations underlie hematopoietic stem cell aging. Proc. Natl Acad. Sci. USA 102, 9194–9199 (2005)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age-associated characteristics of murine hematopoietic stem cells. J. Exp. Med. 192, 1273–1280 (2000)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Rossi, D. J. et al. Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age. Nature 447, 725–729 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Nijnik, A. et al. DNA repair is limiting for haematopoietic stem cells during ageing. Nature 447, 686–690 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  22. Rogakou, E. P., Pilch, D. R., Orr, A. H., Ivanova, V. S. & Bonner, W. M. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J. Biol. Chem. 273, 5858–5868 (1998)

    Article  CAS  PubMed  Google Scholar 

  23. Branzei, D. & Foiani, M. The DNA damage response during DNA replication. Curr. Opin. Cell Biol. 17, 568–575 (2005)

    Article  CAS  PubMed  Google Scholar 

  24. Furuta, T. et al. p21CDKN1A allows the repair of replication-mediated DNA double-strand breaks induced by topoisomerase I and is inactivated by the checkpoint kinase inhibitor 7-hydroxystaurosporine. Oncogene 25, 2839–2849 (2006)

    Article  CAS  PubMed  Google Scholar 

  25. Ishikawa, K., Ishii, H. & Saito, T. DNA damage-dependent cell cycle checkpoints and genomic stability. DNA Cell Biol. 25, 406–411 (2006)

    Article  CAS  PubMed  Google Scholar 

  26. Wang, Y. A., Elson, A. & Leder, P. Loss of p21 increases sensitivity to ionizing radiation and delays the onset of lymphoma in atm-deficient mice. Proc. Natl Acad. Sci. USA 94, 14590–14595 (1997)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Insinga, A. et al. Impairment of p53 acetylation, stability and function by an oncogenic transcription factor. EMBO J. 23, 1144–1154 (2004)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bartkova, J. et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature 444, 633–637 (2006)

    ADS  CAS  PubMed  Google Scholar 

  29. Di Micco, R. et al. Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature 444, 638–642 (2006)

    Article  ADS  CAS  PubMed  Google Scholar 

  30. Paulson, Q. X. et al. Transgenic expression of E2F3a causes DNA damage leading to ATM-dependent apoptosis. Oncogene 27, 4954–4961 (2008)

    Article  CAS  PubMed  Google Scholar 

  31. Bartek, J., Bartkova, J. & Lukas, J. DNA damage signalling guards against activated oncogenes and tumour progression. Oncogene 26, 7773–7779 (2007)

    Article  CAS  PubMed  Google Scholar 

  32. Campisi, J. & d’Adda di Fagagna, F. Cellular senescence: when bad things happen to good cells. Nature Rev. Mol. Cell Biol. 8, 729–740 (2007)

    Article  CAS  Google Scholar 

  33. Dimri, G. P. et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo . Proc. Natl Acad. Sci. USA 92, 9363–9367 (1995)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Speck, N. A. & Gilliland, D. G. Core-binding factors in haematopoiesis and leukaemia. Nature Rev. Cancer 2, 502–513 (2002)

    Article  CAS  Google Scholar 

  35. Heyting, C., Huigen, A. & Den Engelse, L. Repair of ethylnitrosourea-induced DNA damage in the newborn rat. I. Alkali-labile lesions and in situ breaks. Carcinogenesis 1, 769–778 (1980)

    Article  CAS  PubMed  Google Scholar 

  36. Zhou, C. et al. DNA damage evaluated by gammaH2AX foci formation by a selective group of chemical/physical stressors. Mutat. Res. 604, 8–18 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Friedberg, E. C. Suffering in silence: the tolerance of DNA damage. Nature Rev. Mol. Cell Biol. 6, 943–953 (2005)

    Article  CAS  Google Scholar 

  38. Livneh, Z. DNA damage control by novel DNA polymerases: translesion replication and mutagenesis. J. Biol. Chem. 276, 25639–25642 (2001)

    Article  CAS  PubMed  Google Scholar 

  39. Guan, Y., Gerhard, B. & Hogge, D. E. Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML). Blood 101, 3142–3149 (2003)

    Article  CAS  PubMed  Google Scholar 

  40. Ishikawa, F. et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nature Biotechnol. 25, 1315–1321 (2007)

    Article  CAS  Google Scholar 

  41. Grignani, F. et al. High-efficiency gene transfer and selection of human hematopoietic progenitor cells with a hybrid EBV/retroviral vector expressing the green fluorescence protein. Cancer Res. 58, 14–19 (1998)

    CAS  PubMed  Google Scholar 

  42. Minucci, S. et al. Oligomerization of RAR and AML1 transcription factors as a novel mechanism of oncogenic activation. Mol. Cell 5, 811–820 (2000)

    Article  CAS  PubMed  Google Scholar 

  43. Alcalay, M. et al. Acute myeloid leukemia fusion proteins deregulate genes involved in stem cell maintenance and DNA repair. J. Clin. Invest. 112, 1751–1761 (2003)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Insinga, A. et al. Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nature Med. 11, 71–76 (2005)

    Article  CAS  PubMed  Google Scholar 

  45. van Os, R. et al. A Limited role for p21Cip1/Waf1 in maintaining normal hematopoietic stem cell functioning. Stem Cells 25, 836–843 (2007)

    Article  CAS  PubMed  Google Scholar 

  46. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I. L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000)

    Article  ADS  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank L. Luzi, D. Shing and M. Faretta for discussions and suggestions; M. Capillo, M. Stendardo and D. Sardella for assistance in maintenance of mouse colonies and for mouse genotyping; B. Amati, G. McVie, P. P. Di Fiore and C. Basilico for reviewing the manuscript; and P. Dalton for editing the manuscript. This study was supported by grants from the Italian Association on Cancer Research (AIRC), Ministero Italiano della Salute, Cariplo and the European Community (FP6: EPITRON and GENICA) to P.G.P. A.V. is a fellow of the Vollaro Foundation.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Andrea Viale or Pier Giuseppe Pelicci.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-11 with Legends (PDF 540 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Viale, A., De Franco, F., Orleth, A. et al. Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells. Nature 457, 51–56 (2009). https://doi.org/10.1038/nature07618

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature07618

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing