Skip to main content

Advertisement

Log in

Vorinostat modulates cell cycle regulatory proteins in glioma cells and human glioma slice cultures

  • Laboratory Investigation - Human/Animal Tissue
  • Published:
Journal of Neuro-Oncology Aims and scope Submit manuscript

Abstract

Chromatin modification through histone deacetylase inhibition has shown evidence of activity against malignancies. The mechanism of action of such agents are pleiotropic and potentially tumor specific. In this study, we studied the mechanisms of vorinostat-induced cellular effects in gliomas. The effects of vorinostat on proliferation, induction of apoptosis and cell cycle effects were studied in vitro (D54, U87 and U373 glioma cell lines). To gain additional insights into its effects on human gliomas, vorinostat-induced changes were examined ex vivo using a novel organotypic human glioma slice model. Vorinostat treatment resulted in increased p21 levels in all glioma cells tested in a p53 independent manner. In addition, cyclin B1 levels were transcriptionally downregulated and resulted in reduced kinase activity of the cyclin B1/cdk1 complex causing a G2 arrest. These effects were associated with a dose- and time-dependent inhibition of cellular proliferation and anchorage-independent growth in association with hyperacetylation of core histones and induction of apoptosis. Of particular significance, we demonstrate histone hyperacetylation and increased p21 levels in freshly resected human glioma specimens maintained as organotypic slice cultures and exposed to vorinostat similar to cell lines suggesting that human glioma can be targeted by this agent. Our data suggest that the effects of vorinostat are associated with modulation of cell cycle related proteins and activation of a G2 checkpoint along with induction of apoptosis. These effects are mediated by both transcriptional and post-translational mechanisms which provide potential options that can be exploited to develop new therapeutic approaches against gliomas.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Holland EC (2001) Gliomagenesis: genetic alterations and mouse models. Nat Rev 2:120–129

    Article  CAS  Google Scholar 

  2. Penas-Prado M, Gilbert MR (2007) Molecularly targeted therapies for malignant gliomas: advances and challenges. Expert Rev Anticancer Ther 7:641–661

    Article  PubMed  Google Scholar 

  3. Baylin SB (2005) DNA methylation and gene silencing in cancer. Nat Clin Pract 2(Suppl 1):S4–S11

    CAS  Google Scholar 

  4. Jones PA, Baylin SB (2007) The epigenomics of cancer. Cell 128:683–692

    Article  PubMed  CAS  Google Scholar 

  5. Amatya VJ, Naumann U, Weller M, Ohgaki H (2005) TP53 promoter methylation in human gliomas. Acta Neuropathol 110:178–184

    Article  PubMed  CAS  Google Scholar 

  6. Fukushima T, Katayama Y, Watanabe T, Yoshino A, Ogino A, Ohta T, Komine C (2005) Promoter hypermethylation of mismatch repair gene hMLH1 predicts the clinical response of malignant astrocytomas to nitrosourea. Clin Cancer Res 11:1539–1544

    Article  PubMed  CAS  Google Scholar 

  7. Hong C, Maunakea A, Jun P, Bollen AW, Hodgson JG, Goldenberg DD, Weiss WA, Costello JF (2005) Shared epigenetic mechanisms in human and mouse gliomas inactivate expression of the growth suppressor SLC5A8. Cancer Res 65:3617–3623

    Article  PubMed  CAS  Google Scholar 

  8. Jiang Z, Li X, Hu J, Zhou W, Jiang Y, Li G, Lu D (2006) Promoter hypermethylation-mediated down-regulation of LATS1 and LATS2 in human astrocytoma. Neurosci Res 56:450–458

    Article  PubMed  CAS  Google Scholar 

  9. Richon VM, Emiliani S, Verdin E, Webb Y, Breslow R, Rifkind RA, Marks PA (1998) A class of hybrid polar inducers of transformed cell differentiation inhibits histone deacetylases. Proc Natl Acad Sci USA 95:3003–3007

    Article  PubMed  CAS  Google Scholar 

  10. Sawa H, Murakami H, Ohshima Y, Sugino T, Nakajyo T, Kisanuki T, Tamura Y, Satone A, Ide W, Hashimoto I, Kamada H (2001) Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol 18:109–114

    Article  PubMed  CAS  Google Scholar 

  11. Wang ZM, Hu J, Zhou D, Xu ZY, Panasci LC, Chen ZP (2002) Trichostatin A inhibits proliferation and induces expression of p21WAF and p27 in human brain tumor cell lines. Ai Zheng 21:1100–1105

    PubMed  Google Scholar 

  12. Arnold NB, Arkus N, Gunn J, Korc M (2007) The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces growth inhibition and enhances gemcitabine-induced cell death in pancreatic cancer. Clin Cancer Res 13:18–26

    Article  PubMed  CAS  Google Scholar 

  13. Duvic M, Zhang C (2006) Clinical and laboratory experience of vorinostat (suberoylanilide hydroxamic acid) in the treatment of cutaneous T-cell lymphoma. Br J Cancer 95(Suppl 1):S13–S19

    Article  CAS  Google Scholar 

  14. Emanuele S, Lauricella M, Carlisi D, Vassallo B, D’Anneo A, Di Fazio P, Vento R, Tesoriere G (2007) SAHA induces apoptosis in hepatoma cells and synergistically interacts with the proteasome inhibitor Bortezomib. Apoptosis 12:1327–1338

    Article  PubMed  CAS  Google Scholar 

  15. Rosato RR, Almenara JA, Kolla SS, Maggio SC, Coe S, Gimenez MS, Dent P, Grant S (2007) Mechanism and functional role of XIAP and Mcl-1 down-regulation in flavopiridol/vorinostat antileukemic interactions. Mol Cancer Ther 6:692–702

    Article  PubMed  CAS  Google Scholar 

  16. O’Connor OA (2006) Clinical experience with the novel histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid) in patients with relapsed lymphoma. Br J Cancer 95(Suppl 1):S7–S12

    Article  Google Scholar 

  17. Hockly E, Richon VM, Woodman B, Smith DL, Zhou X, Rosa E, Sathasivam K, Ghazi-Noori S, Mahal A, Lowden PA, Steffan JS, Marsh JL, Thompson LM, Lewis CM, Marks PA, Bates GP (2003) Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease. Proc Natl Acad Sci USA 100:2041–2046

    Article  PubMed  CAS  Google Scholar 

  18. Puduvalli VK, Sampath D, Bruner JM, Nangia J, Xu R, Kyritsis AP (2005) TRAIL-induced apoptosis in gliomas is enhanced by Akt-inhibition and is independent of JNK activation. Apoptosis 10:233–243

    Article  PubMed  CAS  Google Scholar 

  19. Richon VM (2006) Cancer biology: mechanism of antitumour action of vorinostat (suberoylanilide hydroxamic acid), a novel histone deacetylase inhibitor. Br J Cancer 95(Suppl 1):S2–S6

    Article  CAS  Google Scholar 

  20. Arooz T, Yam CH, Siu WY, Lau A, Li KK, Poon RY (2000) On the concentrations of cyclins and cyclin-dependent kinases in extracts of cultured human cells. Biochemistry 39:9494–9501

    Article  PubMed  CAS  Google Scholar 

  21. Takizawa CG, Morgan DO (2000) Control of mitosis by changes in the subcellular location of cyclin-B1-Cdk1 and Cdc25C. Curr Opin Cell Biol 12:658–665

    Article  PubMed  CAS  Google Scholar 

  22. Galanis E, Jaeckle KA, Maurer MJ, Reid JM, Ames MM, Hardwick JS, Reilly JF, Loboda A, Nebozhyn M, Fantin VR, Richon VM, Scheithauer B, Giannini C, Flynn PJ, Moore DF Jr, Zwiebel J, Buckner JC (2009) Phase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study. J Clin Oncol 27(12):2052–2058

    Article  PubMed  CAS  Google Scholar 

  23. Ugur HC, Ramakrishna N, Bello L, Menon LG, Kim SK, Black PM, Carroll RS (2007) Continuous intracranial administration of suberoylanilide hydroxamic acid (SAHA) inhibits tumor growth in an orthotopic glioma model. J Neurooncol 83:267–275

    Article  PubMed  CAS  Google Scholar 

  24. Yin D, Ong JM, Hu J, Desmond JC, Kawamata N, Konda BM, Black KL, Koeffler HP (2007) Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor: effects on gene expression and growth of glioma cells in vitro and in vivo. Clin Cancer Res 13:1045–1052

    Article  PubMed  CAS  Google Scholar 

  25. Eyupoglu IY, Hahnen E, Buslei R, Siebzehnrubl FA, Savaskan NE, Luders M, Trankle C, Wick W, Weller M, Fahlbusch R, Blumcke I (2005) Suberoylanilide hydroxamic acid (SAHA) has potent anti-glioma properties in vitro, ex vivo and in vivo. J Neurochem 93:992–999

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

This study was supported in part by funds from Brain Tumor SPORE 5P50CA12700102, The Gregory J. Jungeblut Fund for Brain Cancer Research, The Center for Targeted Therapy Grant and The Pennebaker Research Funds. The authors acknowledge Susan O. Graham, RN, Angele K. Saleeba, Kristin L. Parks, Lamonne Crutcher and Alicia A. Ledoux for their assistance with the study.

Conflict of interest

The authors declare no conflicts of interest related to this study.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Vinay K. Puduvalli.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Xu, J., Sampath, D., Lang, F.F. et al. Vorinostat modulates cell cycle regulatory proteins in glioma cells and human glioma slice cultures. J Neurooncol 105, 241–251 (2011). https://doi.org/10.1007/s11060-011-0604-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11060-011-0604-7

Keywords

Navigation