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:

Oncogene-induced senescence as an initial barrier in lymphoma development

Abstract

Acute induction of oncogenic Ras provokes cellular senescence involving the retinoblastoma (Rb) pathway, but the tumour suppressive potential of senescence in vivo remains elusive. Recently, Rb-mediated silencing of growth-promoting genes by heterochromatin formation associated with methylation of histone H3 lysine 9 (H3K9me) was identified as a critical feature of cellular senescence, which may depend on the histone methyltransferase Suv39h1. Here we show that Eµ-N-Ras transgenic mice harbouring targeted heterozygous lesions at the Suv39h1, or the p53 locus for comparison, succumb to invasive T-cell lymphomas that lack expression of Suv39h1 or p53, respectively. By contrast, most N-Ras-transgenic wild-type (‘control’) animals develop a non-lymphoid neoplasia significantly later. Proliferation of primary lymphocytes is directly stalled by a Suv39h1-dependent, H3K9me-related senescent growth arrest in response to oncogenic Ras, thereby cancelling lymphomagenesis at an initial step. Suv39h1-deficient lymphoma cells grow rapidly but, unlike p53-deficient cells, remain highly susceptible to adriamycin-induced apoptosis. In contrast, only control, but not Suv39h1-deficient or p53-deficient, lymphomas senesce after drug therapy when apoptosis is blocked. These results identify H3K9me-mediated senescence as a novel Suv39h1-dependent tumour suppressor mechanism whose inactivation permits the formation of aggressive but apoptosis-competent lymphomas in response to oncogenic Ras.

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: Suv39h1 - and p53 -defects license aggressive lymphomas in response to oncogenic Ras.
Figure 2: Oncogenic Ras selects against Suv39h1, ARF or p53 in primary lymphomas.
Figure 3: Suv39h1 loss produces chromosomally stable senescence-defective, but apoptosis-competent, Ras lymphomas.
Figure 4: Suv39h1/H3K9me-controlled senescence limits Ras-mediated oncogenicity.
Figure 5: Methylation-targeting therapy mimics Suv39h1 loss in a Ras transgenic context in vivo.

Similar content being viewed by others

References

  1. Campisi, J. Cellular senescence as a tumour-suppressor mechanism. Trends Cell Biol. 11, S27–S31 (2001)

    Article  CAS  Google Scholar 

  2. Schmitt, C. A. Senescence, apoptosis and therapy—cutting the lifelines of cancer. Nature Rev. Cancer 3, 286–295 (2003)

    Article  CAS  Google Scholar 

  3. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997)

    Article  CAS  Google Scholar 

  4. Trimarchi, J. M. & Lees, J. A. Sibling rivalry in the E2F family. Nature Rev. Mol. Cell Biol. 3, 11–20 (2002)

    Article  CAS  Google Scholar 

  5. Narita, M. et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113, 703–716 (2003)

    Article  CAS  Google Scholar 

  6. Rea, S. et al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature 406, 593–599 (2000)

    Article  ADS  CAS  Google Scholar 

  7. Nielsen, S. J. et al. Rb targets histone H3 methylation and HP1 to promoters. Nature 412, 561–565 (2001)

    Article  ADS  CAS  Google Scholar 

  8. Bannister, A. J. et al. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. Nature 410, 120–124 (2001)

    Article  ADS  CAS  Google Scholar 

  9. Lachner, M., O'Carroll, D., Rea, S., Mechtler, K. & Jenuwein, T. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 410, 116–120 (2001)

    Article  ADS  CAS  Google Scholar 

  10. Ait-Si-Ali, S. et al. A Suv39h-dependent mechanism for silencing S-phase genes in differentiating but not in cycling cells. EMBO J. 23, 605–615 (2004)

    Article  CAS  Google Scholar 

  11. Haupt, Y., Harris, A. W. & Adams, J. M. Retroviral infection accelerates T lymphomagenesis in E mu-N-ras transgenic mice by activating c-myc or N-myc. Oncogene 7, 981–986 (1992)

    CAS  PubMed  Google Scholar 

  12. Peters, A. H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107, 323–337 (2001)

    Article  CAS  Google Scholar 

  13. Kemp, C. J., Donehower, L. A., Bradley, A. & Balmain, A. Reduction of p53 gene dosage does not increase initiation or promotion but enhances malignant progression of chemically induced skin tumors. Cell 74, 813–822 (1993)

    Article  CAS  Google Scholar 

  14. Lin, A. W. et al. Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signalling. Genes Dev. 12, 3008–3019 (1998)

    Article  CAS  Google Scholar 

  15. Lyon, M. F. Gene action in the X-chromosome of the mouse (Mus musculus L.). Nature 190, 372–373 (1961)

    Article  ADS  CAS  Google Scholar 

  16. Ohtani, N. et al. Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence. Nature 409, 1067–1070 (2001)

    Article  ADS  CAS  Google Scholar 

  17. Chang, B. D. et al. A senescence-like phenotype distinguishes tumour cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 59, 3761–3767 (1999)

    CAS  PubMed  Google Scholar 

  18. Schmitt, C. A. et al. A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell 109, 335–346 (2002)

    Article  CAS  Google Scholar 

  19. 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  Google Scholar 

  20. Nguyen, C. T. et al. Histone H3-lysine 9 methylation is associated with aberrant gene silencing in cancer cells and is rapidly reversed by 5-aza-2′-deoxycytidine. Cancer Res. 62, 6456–6461 (2002)

    CAS  Google Scholar 

  21. Fuks, F., Hurd, P. J., Deplus, R. & Kouzarides, T. The DNA methyltransferases associate with HP1 and the SUV39H1 histone methyltransferase. Nucleic Acids Res. 31, 2305–2312 (2003)

    Article  CAS  Google Scholar 

  22. Leone, G., Voso, M. T., Teofili, L. & Lubbert, M. Inhibitors of DNA methylation in the treatment of hematological malignancies and MDS. Clin. Immunol. 109, 89–102 (2003)

    Article  CAS  Google Scholar 

  23. Rane, S. G., Cosenza, S. C., Mettus, R. V. & Reddy, E. P. Germ line transmission of the Cdk4(R24C) mutation facilitates tumorigenesis and escape from cellular senescence. Mol. Cell. Biol. 22, 644–656 (2002)

    Article  CAS  Google Scholar 

  24. Guerra, C. et al. Tumor induction by an endogenous K-ras oncogene is highly dependent on cellular context. Cancer Cell 4, 111–120 (2003)

    Article  MathSciNet  CAS  Google Scholar 

  25. Tuveson, D. A. et al. Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell 5, 375–387 (2004)

    Article  CAS  Google Scholar 

  26. Kim, K. C., Geng, L. & Huang, S. Inactivation of a histone methyltransferase by mutations in human cancers. Cancer Res. 63, 7619–7623 (2003)

    CAS  PubMed  Google Scholar 

  27. Steele-Perkins, G. et al. Tumor formation and inactivation of RIZ1, an Rb-binding member of a nuclear protein-methyltransferase superfamily. Genes Dev. 15, 2250–2262 (2001)

    Article  CAS  Google Scholar 

  28. Schmitt, C. A., McCurrach, M. E., de Stanchina, E., Wallace-Brodeur, R. R. & Lowe, S. W. INK4a/ARF mutations accelerate lymphomagenesis and promote chemoresistance by disabling p53. Genes Dev. 13, 2670–2677 (1999)

    Article  CAS  Google Scholar 

  29. Schmitt, C. A. et al. Dissecting p53 tumour suppressor functions in vivo. Cancer Cell 1, 289–298 (2002)

    Article  CAS  Google Scholar 

  30. Frank, O. et al. Tumor cells escape suicide gene therapy by genetic and epigenetic instability. Blood 104, 3543–3549 (2004)

    Article  CAS  Google Scholar 

  31. Hamrouni, A., Aublin, A., Guillaume, P. & Maryanski, J. L. T cell receptor gene rearrangement lineage analysis reveals clues for the origin of highly restricted antigen-specific repertoires. J. Exp. Med. 197, 601–614 (2003)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Harris, T. Jacks and M. Serrano for mice; S. W. Lowe for retroviral constructs; B. Teichmann and S. Spieckermann for technical assistance; and A. Lee, M. Reimann and P. Kahlem for discussions and editorial advice. This work was supported by grants from the European Union (to B.S.) and from the Deutsche Krebshilfe (to C.A.S.). S.L. is a postdoctoral fellow of the José Carreras Leukemia Foundation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Clemens A. Schmitt.

Ethics declarations

Competing interests

Reprints and permissions information is available at npg.nature.com/reprintsandpermissions. The authors declare no competing financial interests.

Supplementary information

Supplementary Figure S1

Characterization of Ras-induced neoplasms. (PDF 21776 kb)

Supplementary Figure S2

Representative dual-colour flow cytometric analyses of the indicated antigens reflecting the haematopoietic differentiation status in different haematopoietic compartments of Suv39h1+/+ and Suv39h1-/- mice of matched age. (PDF 33 kb)

Supplementary Figure S3

Invasiveness, growth characteristics and expression of related genes in mouse embryo fibroblasts and Ras-driven lymphomas. (PDF 38073 kb)

Supplementary Figure S4

Characterization of TSA/DAC-promoted lymphoma and leukaemia in Eµ-N-Ras transgenic mice. (PDF 16142 kb)

Supplementary Table S1

Clinical and pathological characteristics of terminal disease conditions in Eµ-N-Ras mice of the indicated genotypes grouped according to their time-to-death. (PDF 156 kb)

Supplementary Table S2

Comprehensive summary of haematological differentiation measured in nucleated cell populations isolated from the indicated compartments of Suv39h1+/+ and Suv39h1-/- mice. (PDF 109 kb)

Supplementary Table S3

Chromosomal aberrations of individual primary control (n = 2), Suv39h1-null (n = 4), and p53-null Ras-lymphomas (n = 3) assessed by spectral karyotyping. (PDF 110 kb)

Supplementary Notes

Legends for Figures S1-S4 and Tables S1-S3 (DOC 25 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Braig, M., Lee, S., Loddenkemper, C. et al. Oncogene-induced senescence as an initial barrier in lymphoma development. Nature 436, 660–665 (2005). https://doi.org/10.1038/nature03841

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature03841

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