1,090
Views
27
CrossRef citations to date
0
Altmetric
Report

The p27–Skp2 axis mediates glucocorticoid-induced cell cycle arrest in T-lymphoma cells

, , , , , , , , , & show all
Pages 2625-2635 | Received 08 Feb 2013, Accepted 03 Jul 2013, Published online: 09 Jul 2013
 

Abstract

Glucocorticoid therapy is an important treatment modality of hematological malignancies, especially T-cell acute lymphoblastic leukemia (T-ALL). Glucocorticoids are known to induce a cell cycle arrest and apoptosis in T-lymphoma cells. We could demonstrate that the cell cycle arrest induced by the synthetic glucocorticoid dexamethasone (Dex) clearly precedes apoptosis in human CEM T-ALL and murine S49.1 T-lymphoma cells. Cyclin D3 is strongly downregulated, whereas the CDK inhibitor p27Kip1 (p27) is strongly upregulated in response to dexamethasone in these cells. RNAi-mediated knockdown of p27 as well as overexpression of its negative regulator Skp2 revealed the critical function of p27 in the Dex-induced G1 arrest of CEM cells. Our studies indicate that several mechanisms contribute to the increase of p27 protein in our T-lymphoma cell lines. We found a significant upregulation of p27 mRNA in S49.1 and CEM cells. In addition, Dex treatment activated the mouse p27 promotor in reporter gene experiments, indicating a transcriptional regulation. However, the relatively moderate induction of p27 mRNA levels by Dex did not explain the strong increase of p27 protein in CEM and S49.1 cells. We found clear evidence for a posttranslational mechanism responsible for the robust increase in p27 protein. Dex treatment of S49.1 and CEM cells increases the half-life of p27 protein, which indicates that decreased protein degradation is the primary mechanism of p27 induction by glucocorticoids. Interestingly, we found that Dex treatment decreased the protein and mRNA levels of the negative regulator of p27 protein and E3 ubiquitin ligase subunit Skp2. We conclude that the cell cycle inhibitor p27 and its negative regulator Skp2 are key players in the glucocorticoid-induced growth suppression of T-lymphoma cells and should be considered as potential drug targets to improve therapies of T-cell malignancies.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank Peter Herrlich and Martin Göttlicher for critical and helpful comments during the preparation of the manuscript, Jonathan Vosper and all members of the Hengst lab for support, stimulating discussions and critical reading of the manuscript, Johanna Gostner for help with the RT-qPCR and statistical analysis of the data, and Barbara Gschirr for TaqMan real-time RT-PCR. We appreciate Albert Nordin providing us the p27 reporter constructs. Part of the work was funded by the FWF (Grant P24031-B20 and SFB F21-B12). AT was supported by ONCOTYROL, a COMET Center funded by the Austrian Research Promotion Agency (FFG), the Tiroler Zukunftsstiftung and the Styrian Business Promotion Agency (SFG). The Tyrolean Cancer Research Institute is supported by the Tiroler Krankenanstalten Ges.m.b.H (TILAK), the Tyrolean Cancer Aid Society, various businesses, financial institutions, and the people of Tyrol.

Supplemental Materials

Supplemental materials may be found here:
http://www.landesbioscience.com/journals/cc/article/25622