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:

A mouse knock-in model exposes sequential proteolytic pathways that regulate p27Kip1 in G1 and S phase

An Erratum to this article was published on 11 October 2001

Abstract

The protein p27Kip1 is an inhibitor of cell division1. An increase in p27 causes proliferating cells to exit from the cell cycle, and a decrease in p27 is necessary for quiescent cells to resume division2,3. Abnormally low amounts of p27 are associated with pathological states of excessive cell proliferation, especially cancers4,5,6,7,8. In normal and tumour cells, p27 is regulated primarily at the level of translation9,10,11 and protein turnover. Phosphorylation of p27 on threonine 187 (T187) by cyclin-dependent kinase 2 (Cdk2) is thought to initiate the major pathway for p27 proteolysis12,13,14,15. To critically test the importance of this pathway in vivo, we replaced the murine p27 gene with one that encoded alanine instead of threonine at position 187 (p27T187A). Here we show that cells expressing p27T187A were unable to downregulate p27 during the S and G2 phases of the cell cycle, but that this had a surprisingly modest effect on cell proliferation both in vitro and in vivo. Our efforts to explain this unexpected result led to the discovery of a second proteolytic pathway for controlling p27, one that is activated by mitogens and degrades p27 exclusively during G1.

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: Replacement of the p27 gene with one encoding p27T187A.
Figure 2: Effect of p27T187A on cell proliferation.
Figure 3: Cell-cycle-dependent proteolysis of p27.
Figure 4: Turnover of p27 in G1- and in S-phase MEFs requires Skp2.

Similar content being viewed by others

References

  1. Sherr, C. J. & Roberts, J. M. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 9, 1149–1163 (1995).

    Article  CAS  Google Scholar 

  2. Fero, M. L. et al. A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis, and female sterility in p27(Kip1)-deficient mice. Cell 85, 733–744 (1996).

    Article  CAS  Google Scholar 

  3. Coats, S. et al. A new pathway for mitogen-dependent cdk2 regulation uncovered in p27(Kip1)-deficient cells. Curr. Biol. 9, 163–173 (1999).

    Article  CAS  Google Scholar 

  4. Ophascharoensuk, V., Fero, M. L., Hughes, J., Roberts, J. M. & Shankland, S. J. The cyclin-dependent kinase inhibitor p27Kip1 safeguards against inflammatory injury. Nature Med. 4, 575–580 (1998).

    Article  CAS  Google Scholar 

  5. Fero, M. L., Randel, E., Gurley, K. E., Roberts, J. M. & Kemp, C. J. The murine gene p27Kip1 is haplo-insufficient for tumour suppression. Nature 396, 177–180 (1998).

    Article  CAS  ADS  Google Scholar 

  6. Porter, P. L. et al. Expression of cell-cycle regulators p27Kip1 and cyclin E, alone and in combination, correlate with survival in young breast cancer patients. Nature Med. 3, 222–225 (1997).

    Article  CAS  Google Scholar 

  7. Tan, P. et al. The cell cycle inhibitor p27 is an independent prognostic marker in small (T1a,b) invasive breast carcinomas. Cancer Res. 57, 1259–1263 (1997).

    CAS  PubMed  Google Scholar 

  8. Catzavelos, C. et al. Decreased levels of the cell-cycle inhibitor p27Kip1 protein: prognostic implications in primary breast cancer. Nature Med. 3, 227–230 (1997).

    Article  CAS  Google Scholar 

  9. Agrawal, D. et al. Repression of p27kip1 synthesis by platelet-derived growth factor in BALB/c 3T3 cells. Mol. Cell. Biol. 16, 4327–4336 (1996).

    Article  CAS  Google Scholar 

  10. Hengst, L. & Reed, S. I. Translational control of p27Kip1 accumulation during the cell cycle. Science 271, 1861–1864 (1996).

    Article  CAS  ADS  Google Scholar 

  11. Millard, S. S., Vidal, A., Markus, M. & Koff, A. A U-rich element in the 5′ untranslated region is necessary for the translation of p27 mRNA. Mol. Cell. Biol. 20, 5947–5959 (2000).

    Article  CAS  Google Scholar 

  12. Sheaff, R. J., Groudine, M., Gordon, M., Roberts, J. M. & Clurman, B. E. Cyclin E-CDK2 is a regulator of p27Kip1. Genes Dev. 11, 1464–1478 (1997).

    Article  CAS  Google Scholar 

  13. Vlach, J., Hennecke, S., Alevizopoulos, K., Conti, D. & Amati, B. Growth arrest by the cyclin-dependent kinase inhibitor p27Kip1 is abrogated by c-Myc. EMBO J. 15, 6595–6604 (1996).

    Article  CAS  Google Scholar 

  14. Muller, D. et al. Cdk2-dependent phosphorylation of p27 facilitates its Myc-induced release from cyclin E/cdk2 complexes. Oncogene 15, 2561–2576 (1997).

    Article  CAS  Google Scholar 

  15. Pagano, M. et al. Role of the ubiquitin–proteasome pathway in regulating abundance of the cyclin-dependent kinase inhibitor p27. Science 269, 682–685 (1995).

    Article  CAS  ADS  Google Scholar 

  16. Feldman, R. M., Correll, C. C., Kaplan, K. B. & Deshaies, R. J. A complex of Cdc4p, Skp1p, and Cdc53p/cullin catalyzes ubiquitination of the phosphorylated CDK inhibitor Sic1p. Cell 91, 221–230 (1997).

    Article  CAS  Google Scholar 

  17. Bai, C. et al. SKP1 connects cell cycle regulators to the ubiquitin proteolysis machinery through a novel motif, the F-box. Cell 86, 263–274 (1996).

    Article  CAS  Google Scholar 

  18. Skowyra, D., Craig, K. L., Tyers, M., Elledge, S. J. & Harper, J. W. F-box proteins are receptors that recruit phosphorylated substrates to the SCF ubiquitin-ligase complex. Cell 91, 209–219 (1997).

    Article  CAS  Google Scholar 

  19. Sutterluty, H. et al. p45SKP2 promotes p27Kip1 degradation and induces S phase in quiescent cells. Nature Cell Biol. 1, 207–214 (1999).

    Article  CAS  Google Scholar 

  20. Rolfe, M., Chiu, M. I. & Pagano, M. The ubiquitin-mediated proteolytic pathway as a therapeutic area. J. Mol. Med. 75, 5–17 (1997).

    Article  CAS  Google Scholar 

  21. Carrano, A. C., Eytan, E., Hershko, A. & Pagano, M. SKP2 is required for ubiquitin-mediated degradation of the CDK inhibitor p27. Nature Cell Biol. 1, 193–199 (1999).

    Article  CAS  Google Scholar 

  22. Tsvetkov, L. M., Yeh, K. H., Lee, S. J., Sun, H. & Zhang, H. p27(Kip1) ubiquitination and degradation is regulated by the SCF(Skp2) complex through phosphorylated Thr187 in p27. Curr. Biol. 9, 661–664 (1999).

    Article  CAS  Google Scholar 

  23. Nakayama, K. et al. Targeted disruption of Skp2 results in accumulation of cyclin E and p27(Kip1), polyploidy and centrosome overduplication. EMBO J. 19, 2069–2081 (2000).

    Article  CAS  Google Scholar 

  24. Leone, G., DeGregori, J., Sears, R., Jakoi, L. & Nevins, J. R. Myc and Ras collaborate in inducing accumulation of active cyclin E/Cdk2 and E2F. Nature 387, 422–426 (1997).

    Article  CAS  ADS  Google Scholar 

  25. O'Hagan, R. C. et al. Myc-enhanced expression of Cul1 promotes ubiquitin-dependent proteolysis and cell cycle progression. Genes Dev. 14, 2185–2191 (2000).

    Article  CAS  Google Scholar 

  26. Sherr, C. J. & Roberts, J. M. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 13, 1501–1512 (1999).

    Article  CAS  Google Scholar 

  27. Hatakeyama, M. et al. The cancer cell and the cell cycle clock. Cold Spring Harb. Symp. Quant. Biol. 59, 1–10 (1994).

    Article  CAS  Google Scholar 

  28. Pardee, A. B. A restriction point for control of normal animal cell proliferation. Proc. Natl Acad. Sci. USA 71, 1286–1290 (1974).

    Article  CAS  ADS  Google Scholar 

  29. Nagy, A. et al. Dissecting the role of N-myc in development using a single targeting vector to generate a series of alleles. Curr. Biol. 8, 661–664 (1998).

    Article  CAS  Google Scholar 

  30. Kyriakides, T. R., Tam, J. W. & Bornstein, P. Accelerated wound healing in mice with a disruption of the thrombospondin 2 gene. J. Invest. Dermatol. 113, 782–787 (1999).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Clurman, R. Sheaff, K.-I. Nakayama, B. Carter, M. Groudine, B. Luscher and members of the Roberts laboratory for advice, suggestions and reagents. M. Black and N. Jiang provided expert technical assistance with flow cytometry and with the generation of mutant mouse strains. We thank P. Porter for help with TUNEL staining. N.P.M. was supported by a grant from the Deutche Forschungsgemeinschaft. T.R.K. was supported by The National Science Foundation through the University of Washington Engineered Biomaterials Engineering Research Center. H.S. was supported by a grant from the National Institutes of Health. This work was supported by a grant from the National Institutes of Health to J.M.R. and by a George M. O'Brien Kidney Research Center award. J.M.R. is an Investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to James M. Roberts.

Supplementary information

Figure 1

(GIF 24.1 KB)

The same samples described in Figure 1B were immuneprecipitated with antibodies against cyclin E, and cyclin E-associated histone H1 kinase activity was measured. For convenience, the p27 immunoblots are reproduced here so that the pattern of p27 protein expression can be directly compared to the pattern of cyclin E activation

Figure 2

(JPG 42.3 KB)

Two examples of punch wounds of the skin 4.5 days after wounding. The leading edges of the new epithelial layer in the p27T187A mouse are indicated by arrows. The wound in this control mouse is completely covered by new epithelial cells. Magnification = 100x.

Figure 3

(GIF 17.9 KB)

Mean sizes of thymocytes from 8 week old mice of the indicated genotypes were determined byflow cytometric measurements of forward angle light scatter.

Figure 4

(GIF 28.7 KB)

Cells transfected with a p27 expression plasmid express predominantly the exogenous p27. 293 cells were transfected with the indicated plasmidsand cell extracts immunoblotted for expression of p27 protein. When compared to the expression level of the exogenous p27 protein (lanes 1,2 5,6), the amount of endogenous p27 is far less (lanes 3,4). Moreover, the amount of endogenous p27 is not detectably increased in cells expressing high amounts of exogenous p27.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Malek, N., Sundberg, H., McGrew, S. et al. A mouse knock-in model exposes sequential proteolytic pathways that regulate p27Kip1 in G1 and S phase. Nature 413, 323–327 (2001). https://doi.org/10.1038/35095083

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/35095083

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