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.

  • Letter
  • Published:

Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation

An Author Correction to this article was published on 30 April 2020

Abstract

Telomere shortening limits the proliferative lifespan of human cells by activation of DNA damage pathways, including upregulation of the cell cycle inhibitor p21 (encoded by Cdkn1a, also known as Cip1 and Waf1)) (refs. 15). Telomere shortening in response to mutation of the gene encoding telomerase is associated with impaired organ maintenance and shortened lifespan in humans6 and in mice7,8,9. The in vivo function of p21 in the context of telomere dysfunction is unknown. Here we show that deletion of p21 prolongs the lifespan of telomerase-deficient mice with dysfunctional telomeres. p21 deletion improved hematolymphopoiesis and the maintenance of intestinal epithelia without rescuing telomere function. Moreover, deletion of p21 rescued proliferation of intestinal progenitor cells and improved the repopulation capacity and self-renewal of hematopoietic stem cells from mice with dysfunctional telomeres. In these mice, apoptotic responses remained intact, and p21 deletion did not accelerate chromosomal instability or cancer formation. This study provides experimental evidence that telomere dysfunction induces p21-dependent checkpoints in vivo that can limit longevity at the organismal level.

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: Cdkn1a deletion prolongs lifespan and improves maintenance of high-turnover organs in aging mice with dysfunctional telomeres.
Figure 2: Impaired maintenance and function of stem and progenitor cells in aged mice with dysfunctional telomeres.
Figure 3: Cdkn1a deletion does not rescue telomere function or DNA damage signal induction in intestinal crypts of aging iG4 mice.
Figure 4: Deletion of Cdkn1a rescues cell cycle arrest in intestinal progenitor cells and improves repopulation capacity and self-renewal of HSCs from mice with dysfunctional telomeres.
Figure 5: Cdkn1a deletion leaves apoptotic responses intact and does not increase chromosomal instability and carcinogenesis in mice with dysfunctional telomeres.

Similar content being viewed by others

References

  1. Wright, W.E. & Shay, J.W. The two-stage mechanism controlling cellular senescence and immortalization. Exp. Gerontol. 27, 383–389 (1992).

    Article  CAS  Google Scholar 

  2. Allsopp, R.C. et al. Telomere length predicts replicative capacity of human fibroblasts. Proc. Natl. Acad. Sci. USA 89, 10114–10118 (1992).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Brown, J.P., Wei, W. & Sedivy, J.M. Bypass of senescence after disruption of p21CIP1/WAF1 gene in normal diploid human fibroblasts. Science 277, 831–834 (1997).

    Article  CAS  Google Scholar 

  5. 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 

  6. Vulliamy, T. et al. Disease anticipation is associated with progressive telomere shortening in families with dyskeratosis congenita due to mutations in TERC. Nat. Genet. 36, 447–449 (2004).

    Article  CAS  Google Scholar 

  7. Rudolph, K.L. et al. Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 96, 701–712 (1999).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Hao, L.Y. et al. Short telomeres, even in the presence of telomerase, limit tissue renewal capacity. Cell 123, 1121–1131 (2005).

    Article  CAS  Google Scholar 

  10. Lombard, D.B. et al. DNA repair, genome stability, and aging. Cell 120, 497–512 (2005).

    Article  CAS  Google Scholar 

  11. Lieber, M.R. & Karanjawala, Z.E. Aging, repetitive genomes and DNA damage. Nat. Rev. Mol. Cell Biol. 5, 69–75 (2004).

    Article  CAS  Google Scholar 

  12. Min, H., Montecino-Rodriguez, E. & Dorshkind, K. Effects of aging on early B- and T-cell development. Immunol. Rev. 205, 7–17 (2005).

    Article  CAS  Google Scholar 

  13. Lipschitz, D.A., Udupa, K.B., Milton, K.Y. & Thompson, C.O. Effect of age on hematopoiesis in man. Blood 63, 502–509 (1984).

    Article  CAS  Google Scholar 

  14. Nishimura, E.K., Granter, S.R. & Fisher, D.E. Mechanisms of hair graying: incomplete melanocyte stem cell maintenance in the niche. Science 307, 720–724 (2005).

    Article  CAS  Google Scholar 

  15. Wagers, A.J. & Conboy, I.M. Cellular and molecular signatures of muscle regeneration: current concepts and controversies in adult myogenesis. Cell 122, 659–667 (2005).

    Article  CAS  Google Scholar 

  16. Lechel, A. et al. The cellular level of telomere dysfunction determines induction of senescence or apoptosis in vivo. EMBO Rep. 6, 275–281 (2005).

    Article  CAS  Google Scholar 

  17. Lee, H.W. et al. Essential role of mouse telomerase in highly proliferative organs. Nature 392, 569–574 (1998).

    Article  CAS  Google Scholar 

  18. Chin, L. et al. p53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell 97, 527–538 (1999).

    Article  CAS  Google Scholar 

  19. Artandi, S.E. et al. Telomere dysfunction promotes non-reciprocal translocations and epithelial cancers in mice. Nature 406, 641–645 (2000).

    Article  CAS  Google Scholar 

  20. 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 

  21. Deng, C., Zhang, P., Harper, J.W., Elledge, S.J. & Leder, P. Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82, 675–684 (1995).

    Article  CAS  Google Scholar 

  22. Hemann, M.T., Strong, M.A., Hao, L.Y. & Greider, C.W. The shortest telomere, not average telomere length, is critical for cell viability and chromosomal stability. Cell 107, 67–77 (2001).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  24. Cheng, T. et al. Haematopoeitic stem cell quiescence maintained by p21Waf1/Cip1. Science 287, 1804–1808 (2000).

    Article  CAS  Google Scholar 

  25. Rudolph, K.L. et al. Telomere dysfunction and evolution of intestinal carcinoma in mice and humans. Nat. Genet. 28, 155–159 (2001).

    Article  CAS  Google Scholar 

  26. Greenberg, R.A. et al. Short dysfunctional telomeres impair tumorigenesis in the INK4a(delta2/3) cancer-prone mouse. Cell 97, 515–525 (1999).

    Article  CAS  Google Scholar 

  27. Carnero, A. & Beach, D.H. Absence of p21WAF1 cooperates with c-myc in bypassing Ras-induced senescence and enhances oncogenic cooperation. Oncogene 23, 6006–6011 (2004).

    Article  CAS  Google Scholar 

  28. Prowse, K.R. & Greider, C.W. Developmental and tissue-specific regulation of mouse telomerase and telomere length. Proc. Natl. Acad. Sci. USA 92, 4818–4822 (1995).

    Article  CAS  Google Scholar 

  29. Randall, T.D. & Weissman, I.L. Characterization of a population of cells in the bone marrow that phenotypically mimics hematopoietic stem cells: resting stem cells or mystery population? Stem Cells 16, 38–48 (1998).

    Article  CAS  Google Scholar 

  30. Satyanarayana, A. et al. Telomere shortening impairs organ regeneration by inhibiting cell cycle re-entry of a subpopulation of cells. EMBO J. 22, 4003–4013 (2003).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Leder (Department of Genetics, Harvard Medical School) for providing Cdkn1a knockout mice. We thank R. Greenberg and N. Bardeesy for critical discussion and M. Ballmaier (Cell Sorting Core Facility, Hannover Medical School) for cell sorting. K.L.R. is supported by the Deutsche Forschungsgemeinschaft (Heisenberg Professorship: Ru 745/8-1, Ru745 4-1 and KFO119) and the Deutsche Krebshilfe e.V. (10-2236-Ru 2) as well as the Roggenbuck Foundation, the Wilhelm Sander Foundation and the Fritz Thyssen Foundation. H.W.L. was supported by grants from the 21C Frontier Functional Human Genome Project (FG05-22-02) and the BRC Frontier (M103KV010018-05K2201-01830).

Author information

Authors and Affiliations

Authors

Contributions

A.R.C., Z.J., A.L., S.S., A. Stepczynska, H.J., C.W., J.B., Tv.Z., A.G., P.S. and H.N. performed phenotype assessment of aging mice; M.W.D., A. Schienke and H.W.L. generated mouse crosses; A.R.C., Z.J., P.S. and H.N. contributed to writing the paper and K.L.R. designed the studies and wrote the paper.

Corresponding author

Correspondence to K Lenhard Rudolph.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Fig. 1

p21 deletion does not rescue critical telomere shortening in iG4mice. (PDF 17 kb)

Supplementary Fig. 2

The reduced lifespan of telomere dysfunctional mice is not associated with impaired organ maintenance at a young age. (PDF 56 kb)

Supplementary Fig. 3

The reduction in function and maintenance of hematopoietic and intestinal stem and progenitor cells in telomere dysfunctional mice is age dependent. (PDF 47 kb)

Supplementary Fig. 4

Absence of widespread accumulation of SA-bGal-positive cells in different organs of aged iG4 mice. (PDF 108 kb)

Supplementary Fig. 5

p21-independent apoptosis in aging telomere-dysfunctional mice. (PDF 86 kb)

Supplementary Table 1

The prevalence and severity of colonic crypt dysplasia for individual mice analyzed at a histological level in 12- to 15-month-old mice. (PDF 22 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Choudhury, A., Ju, Z., Djojosubroto, M. et al. Cdkn1a deletion improves stem cell function and lifespan of mice with dysfunctional telomeres without accelerating cancer formation. Nat Genet 39, 99–105 (2007). https://doi.org/10.1038/ng1937

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng1937

This article is cited by

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