Skip to main content
Erschienen in: Cellular Oncology 1/2017

20.10.2016 | Original Paper

Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine

verfasst von: Mikkel Staberg, Signe Regner Michaelsen, Rikke Darling Rasmussen, Mette Villingshøj, Hans Skovgaard Poulsen, Petra Hamerlik

Erschienen in: Cellular Oncology | Ausgabe 1/2017

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Glioblastoma (GBM) ranks among the deadliest solid cancers worldwide and its prognosis has remained dismal, despite the use of aggressive chemo-irradiation treatment regimens. Limited drug delivery into the brain parenchyma and frequent resistance to currently available therapies are problems that call for a prompt development of novel therapeutic strategies. While only displaying modest efficacies as mono-therapy in pre-clinical settings, histone deacetylase inhibitors (HDACi) have shown promising sensitizing effects to a number of cytotoxic agents. Here, we sought to investigate the sensitizing effect of the HDACi trichostatin A (TSA) to the alkylating agent lomustine (CCNU), which is used in the clinic for the treatment of GBM.

Methods

Twelve primary GBM cell cultures grown as neurospheres were used in this study, as well as one established GBM-derived cell line (U87 MG). Histone deacetylase (HDAC) expression levels were determined using quantitative real-time PCR and Western blotting. The efficacy of either CCNU alone or its combination with TSA was assessed using various assays, i.e., cell viability assays (MTT), cell cycle assays (flow cytometry, FACS), double-strand DNA break (DSB) quantification assays (microscopy/immunofluorescence) and expression profiling assays of proteins involved in apoptosis and cell stress (Western blotting and protein array).

Results

We found that the HDAC1, 3 and 6 expression levels were significantly increased in GBM samples compared to non-neoplastic brain control samples. Additionally, we found that pre-treatment of GBM cells with TSA resulted in an enhancement of their sensitivity to CCNU, possibly via the accumulation of DSBs, decreased cell proliferation and viability rates, and an increased apoptotic rate.

Conclusion

From our data we conclude that the combined administration of TSA and CCNU eradicates GBM cells with a higher efficacy than either drug alone, thereby opening a novel avenue for the treatment of GBM.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K. Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O. Mirimanoff, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New Engl. J. Med. 352, 987–996 (2005)CrossRefPubMed R. Stupp, W.P. Mason, M.J. van den Bent, M. Weller, B. Fisher, M.J. Taphoorn, K. Belanger, A.A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R.C. Janzer, S.K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J.G. Cairncross, E. Eisenhauer, R.O. Mirimanoff, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New Engl. J. Med. 352, 987–996 (2005)CrossRefPubMed
2.
Zurück zum Zitat P.O. Sakariassen, H. Immervoll, M. Chekenya, Cancer stem cells as mediators of treatment resistance in brain tumors: status and controversies. Neoplasia 9, 882–892 (2007)CrossRefPubMedPubMedCentral P.O. Sakariassen, H. Immervoll, M. Chekenya, Cancer stem cells as mediators of treatment resistance in brain tumors: status and controversies. Neoplasia 9, 882–892 (2007)CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat M.B. Dhiab, S. Ziadi, S. Mestiri, R.B. Gacem, F. Ksiaa, M. Trimeche, DNA methylation patterns in EBV-positive and EBV-negative Hodgkin lymphomas. Cell. Oncol. 38, 453–462 (2015)CrossRef M.B. Dhiab, S. Ziadi, S. Mestiri, R.B. Gacem, F. Ksiaa, M. Trimeche, DNA methylation patterns in EBV-positive and EBV-negative Hodgkin lymphomas. Cell. Oncol. 38, 453–462 (2015)CrossRef
5.
Zurück zum Zitat X. Xu, X. Chang, Z. Li, J. Wang, P. Deng, X. Zhu, J. Liu, C. Zhang, S. Chen, D. Dai, Aberrant SOX11 promoter methylation is associated with poor prognosis in gastric cancer. Cell. Oncol. 38, 183–194 (2015)CrossRef X. Xu, X. Chang, Z. Li, J. Wang, P. Deng, X. Zhu, J. Liu, C. Zhang, S. Chen, D. Dai, Aberrant SOX11 promoter methylation is associated with poor prognosis in gastric cancer. Cell. Oncol. 38, 183–194 (2015)CrossRef
6.
Zurück zum Zitat R.P. Nagarajan, J.F. Costello, Epigenetic mechanisms in glioblastoma multiforme. Semin. Cancer Biol. 19, 188–197 (2009)CrossRefPubMed R.P. Nagarajan, J.F. Costello, Epigenetic mechanisms in glioblastoma multiforme. Semin. Cancer Biol. 19, 188–197 (2009)CrossRefPubMed
7.
Zurück zum Zitat C. Cortes, S.C. Kozma, A. Tauler, S. Ambrosio, MYCN concurrence with SAHA-induced cell death in human neuroblastoma cells. Cell. Oncol. 38, 341–352 (2015)CrossRef C. Cortes, S.C. Kozma, A. Tauler, S. Ambrosio, MYCN concurrence with SAHA-induced cell death in human neuroblastoma cells. Cell. Oncol. 38, 341–352 (2015)CrossRef
9.
Zurück zum Zitat M.L. Di, G. Canettieri, P. Infante, A. Greco, A. Gulino, Protected from the inside: endogenous histone deacetylase inhibitors and the road to cancer. Biochim. Biophys. Acta 1815, 241–252 (2011) M.L. Di, G. Canettieri, P. Infante, A. Greco, A. Gulino, Protected from the inside: endogenous histone deacetylase inhibitors and the road to cancer. Biochim. Biophys. Acta 1815, 241–252 (2011)
10.
Zurück zum Zitat W. Weichert, HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 280, 168–176 (2009)CrossRefPubMed W. Weichert, HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 280, 168–176 (2009)CrossRefPubMed
11.
Zurück zum Zitat B. Barneda-Zahonero, M. Parra, Histone deacetylases and cancer. Mol. Oncol. 6, 579–589 (2012)CrossRefPubMed B. Barneda-Zahonero, M. Parra, Histone deacetylases and cancer. Mol. Oncol. 6, 579–589 (2012)CrossRefPubMed
13.
Zurück zum Zitat K. Bajbouj, C. Mawrin, R. Hartig, J. Schulze-Luehrmann, A. Wilisch-Neumann, A. Roessner, R. Schneider-Stock, P53-dependent antiproliferative and pro-apoptotic effects of trichostatin A (TSA) in glioblastoma cells. J. Neurooncol. 107, 503–516 (2012)CrossRefPubMed K. Bajbouj, C. Mawrin, R. Hartig, J. Schulze-Luehrmann, A. Wilisch-Neumann, A. Roessner, R. Schneider-Stock, P53-dependent antiproliferative and pro-apoptotic effects of trichostatin A (TSA) in glioblastoma cells. J. Neurooncol. 107, 503–516 (2012)CrossRefPubMed
14.
Zurück zum Zitat F.D. Sassi, L. Caesar, M. Jaeger, C. Nor, A.L. Abujamra, G. Schwartsmann, C.B. de Farias, A.L. Brunetto, P.L. Lopez, R. Roesler, Inhibitory activities of trichostatin A in U87 glioblastoma cells and tumorsphere-derived cells. J. Mol. Neurosci. 54, 27–40 (2014)CrossRef F.D. Sassi, L. Caesar, M. Jaeger, C. Nor, A.L. Abujamra, G. Schwartsmann, C.B. de Farias, A.L. Brunetto, P.L. Lopez, R. Roesler, Inhibitory activities of trichostatin A in U87 glioblastoma cells and tumorsphere-derived cells. J. Mol. Neurosci. 54, 27–40 (2014)CrossRef
15.
Zurück zum Zitat J.H. Kim, J.H. Shin, I.H. Kim, Susceptibility and radiosensitization of human glioblastoma cells to trichostatin A, a histone deacetylase inhibitor. Int. J. Radiat. Oncol. Biol. Phys. 59, 1174–1180 (2004)CrossRefPubMed J.H. Kim, J.H. Shin, I.H. Kim, Susceptibility and radiosensitization of human glioblastoma cells to trichostatin A, a histone deacetylase inhibitor. Int. J. Radiat. Oncol. Biol. Phys. 59, 1174–1180 (2004)CrossRefPubMed
16.
Zurück zum Zitat J. Bartkova, P. Hamerlik, M.T. Stockhausen, J. Ehrmann, A. Hlobilkova, H. Laursen, O. Kalita, Z. Kolar, H.S. Poulsen, H. Broholm, J. Lukas, J. Bartek, Replication stress and oxidative damage contribute to aberrant constitutive activation of DNA damage signalling in human gliomas. Oncogene 29, 5095–5102 (2010)CrossRefPubMed J. Bartkova, P. Hamerlik, M.T. Stockhausen, J. Ehrmann, A. Hlobilkova, H. Laursen, O. Kalita, Z. Kolar, H.S. Poulsen, H. Broholm, J. Lukas, J. Bartek, Replication stress and oxidative damage contribute to aberrant constitutive activation of DNA damage signalling in human gliomas. Oncogene 29, 5095–5102 (2010)CrossRefPubMed
17.
Zurück zum Zitat K. A. Harvey, Z. Xu, M. R. Saaddatzadeh, H. Wang, K. Pollok, A. A. Cohen-Gadol, R. A. Siddiqui, Enhanced anticancer properties of lomustine in conjunction with docosahexaenoic acid in glioblastoma cell lines. J. Neurosurg. 122, 547-556 (2015) K. A. Harvey, Z. Xu, M. R. Saaddatzadeh, H. Wang, K. Pollok, A. A. Cohen-Gadol, R. A. Siddiqui, Enhanced anticancer properties of lomustine in conjunction with docosahexaenoic acid in glioblastoma cell lines. J. Neurosurg. 122, 547-556 (2015)
18.
Zurück zum Zitat S.H. Yang, Y.K. Hong, S.S. Jeun, I.S. Kim, J.T. Hong, J.H. Sung, B.C. Son, S.W. Lee, M.C. Kim, K.S. Lee, Assessment of cetuximab efficacy by bioluminescence monitoring of intracranial glioblastoma xenograft in mouse. J. Neurooncol. 95, 23–28 (2009)CrossRefPubMed S.H. Yang, Y.K. Hong, S.S. Jeun, I.S. Kim, J.T. Hong, J.H. Sung, B.C. Son, S.W. Lee, M.C. Kim, K.S. Lee, Assessment of cetuximab efficacy by bioluminescence monitoring of intracranial glioblastoma xenograft in mouse. J. Neurooncol. 95, 23–28 (2009)CrossRefPubMed
19.
Zurück zum Zitat W. Taal, H.M. Oosterkamp, A.M. Walenkamp, H.J. Dubbink, L.V. Beerepoot, M.C. Hanse, J. Buter, A.H. Honkoop, D. Boerman, F.Y. de Vos, W.N. Dinjens, R.H. Enting, M.J. Taphoorn, F.W. van den Berkmortel, R.L. Jansen, D. Brandsma, J.E. Bromberg, I. van Heuvel, R.M. Vernhout, B. van der Holt, M.J. van den Bent, Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 15, 943–953 (2014)CrossRefPubMed W. Taal, H.M. Oosterkamp, A.M. Walenkamp, H.J. Dubbink, L.V. Beerepoot, M.C. Hanse, J. Buter, A.H. Honkoop, D. Boerman, F.Y. de Vos, W.N. Dinjens, R.H. Enting, M.J. Taphoorn, F.W. van den Berkmortel, R.L. Jansen, D. Brandsma, J.E. Bromberg, I. van Heuvel, R.M. Vernhout, B. van der Holt, M.J. van den Bent, Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 15, 943–953 (2014)CrossRefPubMed
20.
Zurück zum Zitat A.A. Brandes, M. Bartolotti, A. Tosoni, E. Franceschi, Nitrosoureas in the management of malignant gliomas. Curr. Neurol. Neurosci. Rep. 16, 13 (2016)CrossRefPubMed A.A. Brandes, M. Bartolotti, A. Tosoni, E. Franceschi, Nitrosoureas in the management of malignant gliomas. Curr. Neurol. Neurosci. Rep. 16, 13 (2016)CrossRefPubMed
21.
Zurück zum Zitat W. Wick, V.K. Puduvalli, M.C. Chamberlain, M.J. van den Bent, A.F. Carpentier, L.M. Cher, W. Mason, M. Weller, S. Hong, L. Musib, A.M. Liepa, D.E. Thornton, H.A. Fine, Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J. Clin. Oncol. 28, 1168–1174 (2010)CrossRefPubMedPubMedCentral W. Wick, V.K. Puduvalli, M.C. Chamberlain, M.J. van den Bent, A.F. Carpentier, L.M. Cher, W. Mason, M. Weller, S. Hong, L. Musib, A.M. Liepa, D.E. Thornton, H.A. Fine, Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J. Clin. Oncol. 28, 1168–1174 (2010)CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat M.T. Stockhausen, H. Broholm, M. Villingshoj, M. Kirchhoff, T. Gerdes, K. Kristoffersen, M. Kosteljanetz, M. Spang-Thomsen, H.S. Poulsen, Maintenance of EGFR and EGFRvIII expressions in an in vivo and in vitro model of human glioblastoma multiforme. Exp. Cell Res. 317, 1513–1526 (2011)CrossRefPubMed M.T. Stockhausen, H. Broholm, M. Villingshoj, M. Kirchhoff, T. Gerdes, K. Kristoffersen, M. Kosteljanetz, M. Spang-Thomsen, H.S. Poulsen, Maintenance of EGFR and EGFRvIII expressions in an in vivo and in vitro model of human glioblastoma multiforme. Exp. Cell Res. 317, 1513–1526 (2011)CrossRefPubMed
23.
Zurück zum Zitat R.D. Rasmussen, M.K. Gajjar, K.E. Jensen, P. Hamerlik, Enhanced efficacy of combined HDAC and PARP targeting in glioblastoma. Mol. Oncol. 10, 751–763 (2016)CrossRefPubMed R.D. Rasmussen, M.K. Gajjar, K.E. Jensen, P. Hamerlik, Enhanced efficacy of combined HDAC and PARP targeting in glioblastoma. Mol. Oncol. 10, 751–763 (2016)CrossRefPubMed
24.
Zurück zum Zitat P. Hamerlik, J.D. Lathia, R. Rasmussen, Q. Wu, J. Bartkova, M. Lee, P. Moudry, J. Bartek Jr., W. Fischer, J. Lukas, J.N. Rich, J. Bartek, Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J. Exp. Med. 209, 507–520 (2012)CrossRefPubMedPubMedCentral P. Hamerlik, J.D. Lathia, R. Rasmussen, Q. Wu, J. Bartkova, M. Lee, P. Moudry, J. Bartek Jr., W. Fischer, J. Lukas, J.N. Rich, J. Bartek, Autocrine VEGF-VEGFR2-Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J. Exp. Med. 209, 507–520 (2012)CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat M. Wetzel, D.R. Premkumar, B. Arnold, I.F. Pollack, Effect of trichostatin A, a histone deacetylase inhibitor, on glioma proliferation in vitro by inducing cell cycle arrest and apoptosis. J. Neurosurg. 103, 549–556 (2005)PubMed M. Wetzel, D.R. Premkumar, B. Arnold, I.F. Pollack, Effect of trichostatin A, a histone deacetylase inhibitor, on glioma proliferation in vitro by inducing cell cycle arrest and apoptosis. J. Neurosurg. 103, 549–556 (2005)PubMed
26.
Zurück zum Zitat M. Koprinarova, P. Botev, G. Russev, Histone deacetylase inhibitor sodium butyrate enhances cellular radiosensitivity by inhibiting both DNA nonhomologous end joining and homologous recombination. DNA Repair 10, 970–977 (2011)CrossRefPubMed M. Koprinarova, P. Botev, G. Russev, Histone deacetylase inhibitor sodium butyrate enhances cellular radiosensitivity by inhibiting both DNA nonhomologous end joining and homologous recombination. DNA Repair 10, 970–977 (2011)CrossRefPubMed
27.
Zurück zum Zitat A.S. Balajee, C.R. Geard, Replication protein A and gamma-H2AX foci assembly is triggered by cellular response to DNA double-strand breaks. Exp. Cell Res. 300, 320–334 (2004)CrossRefPubMed A.S. Balajee, C.R. Geard, Replication protein A and gamma-H2AX foci assembly is triggered by cellular response to DNA double-strand breaks. Exp. Cell Res. 300, 320–334 (2004)CrossRefPubMed
28.
Zurück zum Zitat C. Adamson, O.O. Kanu, A.I. Mehta, C. Di, N. Lin, A.K. Mattox, D.D. Bigner, Glioblastoma multiforme: a review of where we have been and where we are going. Expert Opin. Investig. Drugs 18, 1061–1083 (2009)CrossRefPubMed C. Adamson, O.O. Kanu, A.I. Mehta, C. Di, N. Lin, A.K. Mattox, D.D. Bigner, Glioblastoma multiforme: a review of where we have been and where we are going. Expert Opin. Investig. Drugs 18, 1061–1083 (2009)CrossRefPubMed
29.
Zurück zum Zitat W. Weichert, A. Roske, V. Gekeler, T. Beckers, C. Stephan, K. Jung, F.R. Fritzsche, S. Niesporek, C. Denkert, M. Dietel, G. Kristiansen, Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br. J. Cancer 98, 604–610 (2008)CrossRefPubMedPubMedCentral W. Weichert, A. Roske, V. Gekeler, T. Beckers, C. Stephan, K. Jung, F.R. Fritzsche, S. Niesporek, C. Denkert, M. Dietel, G. Kristiansen, Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br. J. Cancer 98, 604–610 (2008)CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat W. Weichert, A. Roske, S. Niesporek, A. Noske, A.C. Buckendahl, M. Dietel, V. Gekeler, M. Boehm, T. Beckers, C. Denkert, Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo. Clin. Cancer Res. 14, 1669–1677 (2008)CrossRefPubMed W. Weichert, A. Roske, S. Niesporek, A. Noske, A.C. Buckendahl, M. Dietel, V. Gekeler, M. Boehm, T. Beckers, C. Denkert, Class I histone deacetylase expression has independent prognostic impact in human colorectal cancer: specific role of class I histone deacetylases in vitro and in vivo. Clin. Cancer Res. 14, 1669–1677 (2008)CrossRefPubMed
31.
Zurück zum Zitat C.A. Krusche, P. Wulfing, C. Kersting, A. Vloet, W. Bocker, L. Kiesel, H.M. Beier, J. Alfer, Histone deacetylase-1 and −3 protein expression in human breast cancer: a tissue microarray analysis. Breast Cancer Res. Treat. 90, 15–23 (2005)CrossRefPubMed C.A. Krusche, P. Wulfing, C. Kersting, A. Vloet, W. Bocker, L. Kiesel, H.M. Beier, J. Alfer, Histone deacetylase-1 and −3 protein expression in human breast cancer: a tissue microarray analysis. Breast Cancer Res. Treat. 90, 15–23 (2005)CrossRefPubMed
32.
Zurück zum Zitat Y. Minamiya, T. Ono, H. Saito, N. Takahashi, M. Ito, S. Motoyama, J. Ogawa, Strong expression of HDAC3 correlates with a poor prognosis in patients with adenocarcinoma of the lung. Tumour Biol. 31, 533–539 (2010)CrossRefPubMed Y. Minamiya, T. Ono, H. Saito, N. Takahashi, M. Ito, S. Motoyama, J. Ogawa, Strong expression of HDAC3 correlates with a poor prognosis in patients with adenocarcinoma of the lung. Tumour Biol. 31, 533–539 (2010)CrossRefPubMed
33.
Zurück zum Zitat T. Rikimaru, A. Taketomi, Y. Yamashita, K. Shirabe, T. Hamatsu, M. Shimada, Y. Maehara, Clinical significance of histone deacetylase 1 expression in patients with hepatocellular carcinoma. Oncology 72, 69–74 (2007)CrossRefPubMed T. Rikimaru, A. Taketomi, Y. Yamashita, K. Shirabe, T. Hamatsu, M. Shimada, Y. Maehara, Clinical significance of histone deacetylase 1 expression in patients with hepatocellular carcinoma. Oncology 72, 69–74 (2007)CrossRefPubMed
34.
Zurück zum Zitat W. Weichert, A. Roske, V. Gekeler, T. Beckers, M.P. Ebert, M. Pross, M. Dietel, C. Denkert, C. Rocken, Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol. 9, 139–148 (2008)CrossRefPubMed W. Weichert, A. Roske, V. Gekeler, T. Beckers, M.P. Ebert, M. Pross, M. Dietel, C. Denkert, C. Rocken, Association of patterns of class I histone deacetylase expression with patient prognosis in gastric cancer: a retrospective analysis. Lancet Oncol. 9, 139–148 (2008)CrossRefPubMed
35.
Zurück zum Zitat M. Venere, P. Hamerlik, Q. Wu, R.D. Rasmussen, L.A. Song, A. Vasanji, N. Tenley, W.A. Flavahan, A.B. Hjelmeland, J. Bartek, J.N. Rich, Therapeutic targeting of constitutive PARP activation compromises stem cell phenotype and survival of glioblastoma-initiating cells. Cell Death Differ. 21, 258–269 (2014)CrossRefPubMed M. Venere, P. Hamerlik, Q. Wu, R.D. Rasmussen, L.A. Song, A. Vasanji, N. Tenley, W.A. Flavahan, A.B. Hjelmeland, J. Bartek, J.N. Rich, Therapeutic targeting of constitutive PARP activation compromises stem cell phenotype and survival of glioblastoma-initiating cells. Cell Death Differ. 21, 258–269 (2014)CrossRefPubMed
36.
Zurück zum Zitat T.C. Karagiannis, K.N. Harikrishnan, A. El-Osta, The histone deacetylase inhibitor, Trichostatin A, enhances radiation sensitivity and accumulation of gammaH2A.X. Cancer Biol. Ther. 4, 787–793 (2005)CrossRefPubMed T.C. Karagiannis, K.N. Harikrishnan, A. El-Osta, The histone deacetylase inhibitor, Trichostatin A, enhances radiation sensitivity and accumulation of gammaH2A.X. Cancer Biol. Ther. 4, 787–793 (2005)CrossRefPubMed
37.
Zurück zum Zitat B. Sarcar, S. Kahali, P. Chinnaiyan, Vorinostat enhances the cytotoxic effects of the topoisomerase I inhibitor SN38 in glioblastoma cell lines. J. Neurooncol. 99, 201–207 (2010)CrossRefPubMed B. Sarcar, S. Kahali, P. Chinnaiyan, Vorinostat enhances the cytotoxic effects of the topoisomerase I inhibitor SN38 in glioblastoma cell lines. J. Neurooncol. 99, 201–207 (2010)CrossRefPubMed
38.
Zurück zum Zitat C.H. Ryu, W.S. Yoon, K.Y. Park, S.M. Kim, J.Y. Lim, J.S. Woo, C.H. Jeong, Y. Hou, S.S. Jeun, Valproic acid downregulates the expression of MGMT and sensitizes temozolomide-resistant glioma cells. J. Biomed. Biotechnol. 2012(987495) (2012) C.H. Ryu, W.S. Yoon, K.Y. Park, S.M. Kim, J.Y. Lim, J.S. Woo, C.H. Jeong, Y. Hou, S.S. Jeun, Valproic acid downregulates the expression of MGMT and sensitizes temozolomide-resistant glioma cells. J. Biomed. Biotechnol. 2012(987495) (2012)
39.
Zurück zum Zitat A. Ferraro, Altered primary chromatin structures and their implications in cancer development. Cell. Oncol. 39, 195–210 (2016)CrossRef A. Ferraro, Altered primary chromatin structures and their implications in cancer development. Cell. Oncol. 39, 195–210 (2016)CrossRef
40.
Zurück zum Zitat A.J. de Ruijter, A.H. van Gennip, H.N. Caron, S. Kemp, A.B. van Kuilenburg, Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem. J. 370, 737–749 (2003)CrossRefPubMedPubMedCentral A.J. de Ruijter, A.H. van Gennip, H.N. Caron, S. Kemp, A.B. van Kuilenburg, Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem. J. 370, 737–749 (2003)CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat A.K. Lucio-Eterovic, M.A. Cortez, E.T. Valera, F.J. Motta, R.G. Queiroz, H.R. Machado, C.G. Carlotti Jr., L. Neder, C.A. Scrideli, L.G. Tone, Differential expression of 12 histone deacetylase (HDAC) genes in astrocytomas and normal brain tissue: class II and IV are hypoexpressed in glioblastomas. BMC Cancer 8, 243 (2008)CrossRefPubMedPubMedCentral A.K. Lucio-Eterovic, M.A. Cortez, E.T. Valera, F.J. Motta, R.G. Queiroz, H.R. Machado, C.G. Carlotti Jr., L. Neder, C.A. Scrideli, L.G. Tone, Differential expression of 12 histone deacetylase (HDAC) genes in astrocytomas and normal brain tissue: class II and IV are hypoexpressed in glioblastomas. BMC Cancer 8, 243 (2008)CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat V. Medina, B. Edmonds, G.P. Young, R. James, S. Appleton, P.D. Zalewski, Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway. Cancer Res. 57, 3697–3707 (1997)PubMed V. Medina, B. Edmonds, G.P. Young, R. James, S. Appleton, P.D. Zalewski, Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway. Cancer Res. 57, 3697–3707 (1997)PubMed
43.
Zurück zum Zitat H. Sawa, H. Murakami, Y. Ohshima, T. Sugino, T. Nakajyo, T. Kisanuki, Y. Tamura, A. Satone, W. Ide, I. Hashimoto, H. Kamada, 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 (2001)CrossRefPubMed H. Sawa, H. Murakami, Y. Ohshima, T. Sugino, T. Nakajyo, T. Kisanuki, Y. Tamura, A. Satone, W. Ide, I. Hashimoto, H. Kamada, 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 (2001)CrossRefPubMed
44.
Zurück zum Zitat G. Aquilina, M. Crescenzi, M. Bignami, Mismatch repair, G(2)/M cell cycle arrest and lethality after DNA damage. Carcinogenesis 20, 2317–2326 (1999)CrossRefPubMed G. Aquilina, M. Crescenzi, M. Bignami, Mismatch repair, G(2)/M cell cycle arrest and lethality after DNA damage. Carcinogenesis 20, 2317–2326 (1999)CrossRefPubMed
45.
Zurück zum Zitat Z. Shinwari, H. Al-Hindi, E. Al-Shail, Y. Khafaga, A. Al-Kofide, N. El-Kum, A. Aboussekhra, Response of medulloblastoma cells to vincristine and lomustine: role of TRKC, CTNNB1 and STK15. Anticancer Res. 31, 1721–1733 (2011)PubMed Z. Shinwari, H. Al-Hindi, E. Al-Shail, Y. Khafaga, A. Al-Kofide, N. El-Kum, A. Aboussekhra, Response of medulloblastoma cells to vincristine and lomustine: role of TRKC, CTNNB1 and STK15. Anticancer Res. 31, 1721–1733 (2011)PubMed
46.
Zurück zum Zitat M. Cornago, C. Garcia-Alberich, N. Blasco-Angulo, N. Vall-Llaura, M. Nager, J. Herreros, J.X. Comella, D. Sanchis, M. Llovera, Histone deacetylase inhibitors promote glioma cell death by G2 checkpoint abrogation leading to mitotic catastrophe. Cell Death Dis. 5, e1435 (2014)CrossRefPubMedPubMedCentral M. Cornago, C. Garcia-Alberich, N. Blasco-Angulo, N. Vall-Llaura, M. Nager, J. Herreros, J.X. Comella, D. Sanchis, M. Llovera, Histone deacetylase inhibitors promote glioma cell death by G2 checkpoint abrogation leading to mitotic catastrophe. Cell Death Dis. 5, e1435 (2014)CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat K.M. Miller, J.V. Tjeertes, J. Coates, G. Legube, S.E. Polo, S. Britton, S.P. Jackson, Human HDAC1 and HDAC2 function in the DNA-damage response to promote DNA nonhomologous end-joining. Nat. Struct. Mol. Biol. 17, 1144–1151 (2010)CrossRefPubMedPubMedCentral K.M. Miller, J.V. Tjeertes, J. Coates, G. Legube, S.E. Polo, S. Britton, S.P. Jackson, Human HDAC1 and HDAC2 function in the DNA-damage response to promote DNA nonhomologous end-joining. Nat. Struct. Mol. Biol. 17, 1144–1151 (2010)CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat S. Bhaskara, B.J. Chyla, J.M. Amann, S.K. Knutson, D. Cortez, Z.W. Sun, S.W. Hiebert, Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control. Mol. Cell 30, 61–72 (2008)CrossRefPubMedPubMedCentral S. Bhaskara, B.J. Chyla, J.M. Amann, S.K. Knutson, D. Cortez, Z.W. Sun, S.W. Hiebert, Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control. Mol. Cell 30, 61–72 (2008)CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat E.P. Rogakou, W. Nieves-Neira, C. Boon, Y. Pommier, W.M. Bonner, Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139. J. Biol. Chem. 275, 9390–9395 (2000)CrossRefPubMed E.P. Rogakou, W. Nieves-Neira, C. Boon, Y. Pommier, W.M. Bonner, Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139. J. Biol. Chem. 275, 9390–9395 (2000)CrossRefPubMed
50.
Zurück zum Zitat A. Duensing, X. Teng, Y. Liu, M. Tseng, N. Spardy, S. Duensing, A role of the mitotic spindle checkpoint in the cellular response to DNA replication stress. J. Cell. Biochem. 99, 759–769 (2006)CrossRefPubMedPubMedCentral A. Duensing, X. Teng, Y. Liu, M. Tseng, N. Spardy, S. Duensing, A role of the mitotic spindle checkpoint in the cellular response to DNA replication stress. J. Cell. Biochem. 99, 759–769 (2006)CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat E. Diss, N. Nalabothula, D. Nguyen, E. Chang, Y. Kwok, F. Carrier, Vorinostat promotes hyper-radiosensitivity in wild type p53 human glioblastoma cells. J. Clin. Oncol. Res. 2, 1–9 (2014) E. Diss, N. Nalabothula, D. Nguyen, E. Chang, Y. Kwok, F. Carrier, Vorinostat promotes hyper-radiosensitivity in wild type p53 human glioblastoma cells. J. Clin. Oncol. Res. 2, 1–9 (2014)
52.
Zurück zum Zitat A. Min, S.A. Im, D.K. Kim, S.H. Song, H.J. Kim, K.H. Lee, T.Y. Kim, S.W. Han, D.Y. Oh, T.Y. Kim, M.J. O’Connor, Y.J. Bang, Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances anti-tumor effects of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib in triple-negative breast cancer cells. Breast Cancer Res. 17, 33 (2015)CrossRefPubMedPubMedCentral A. Min, S.A. Im, D.K. Kim, S.H. Song, H.J. Kim, K.H. Lee, T.Y. Kim, S.W. Han, D.Y. Oh, T.Y. Kim, M.J. O’Connor, Y.J. Bang, Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances anti-tumor effects of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib in triple-negative breast cancer cells. Breast Cancer Res. 17, 33 (2015)CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat O.S. Chao, O.B. Goodman Jr., Synergistic loss of prostate cancer cell viability by coinhibition of HDAC and PARP. Mol. Cancer Res. 12, 1755–1766 (2014)CrossRefPubMed O.S. Chao, O.B. Goodman Jr., Synergistic loss of prostate cancer cell viability by coinhibition of HDAC and PARP. Mol. Cancer Res. 12, 1755–1766 (2014)CrossRefPubMed
54.
Zurück zum Zitat P.A. Konstantinopoulos, A.J. Wilson, J. Saskowski, E. Wass, D. Khabele, Suberoylanilide hydroxamic acid (SAHA) enhances olaparib activity by targeting homologous recombination DNA repair in ovarian cancer. Gynecol. Oncol. 133, 599–606 (2014)CrossRefPubMedPubMedCentral P.A. Konstantinopoulos, A.J. Wilson, J. Saskowski, E. Wass, D. Khabele, Suberoylanilide hydroxamic acid (SAHA) enhances olaparib activity by targeting homologous recombination DNA repair in ovarian cancer. Gynecol. Oncol. 133, 599–606 (2014)CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat A. Kretsovali, C. Hadjimichael, N. Charmpilas, Histone deacetylase inhibitors in cell pluripotency, differentiation, and reprogramming. Stem Cells Int. 2012, 1–10 184154 (2012) A. Kretsovali, C. Hadjimichael, N. Charmpilas, Histone deacetylase inhibitors in cell pluripotency, differentiation, and reprogramming. Stem Cells Int. 2012, 1–10 184154 (2012)
Metadaten
Titel
Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine
verfasst von
Mikkel Staberg
Signe Regner Michaelsen
Rikke Darling Rasmussen
Mette Villingshøj
Hans Skovgaard Poulsen
Petra Hamerlik
Publikationsdatum
20.10.2016
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 1/2017
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-016-0301-9

Weitere Artikel der Ausgabe 1/2017

Cellular Oncology 1/2017 Zur Ausgabe

Neu im Fachgebiet Pathologie

Molekularpathologische Untersuchungen im Wandel der Zeit

Open Access Biomarker Leitthema

Um auch an kleinen Gewebeproben zuverlässige und reproduzierbare Ergebnisse zu gewährleisten ist eine strenge Qualitätskontrolle in jedem Schritt des Arbeitsablaufs erforderlich. Eine nicht ordnungsgemäße Prüfung oder Behandlung des …

Vergleichende Pathologie in der onkologischen Forschung

Pathologie Leitthema

Die vergleichende experimentelle Pathologie („comparative experimental pathology“) ist ein Fachbereich an der Schnittstelle von Human- und Veterinärmedizin. Sie widmet sich der vergleichenden Erforschung von Gemeinsamkeiten und Unterschieden von …

Gastrointestinale Stromatumoren

Open Access GIST CME-Artikel

Gastrointestinale Stromatumoren (GIST) stellen seit über 20 Jahren ein Paradigma für die zielgerichtete Therapie mit Tyrosinkinaseinhibitoren dar. Eine elementare Voraussetzung für eine mögliche neoadjuvante oder adjuvante Behandlung bei …

Personalisierte Medizin in der Onkologie

Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender …