Skip to main content
Top
Published in: Cellular Oncology 3/2018

Open Access 01-06-2018 | Original Paper

A HIF-independent, CD133-mediated mechanism of cisplatin resistance in glioblastoma cells

Authors: Eroje M. Ahmed, Gagori Bandopadhyay, Beth Coyle, Anna Grabowska

Published in: Cellular Oncology | Issue 3/2018

Login to get access

Abstract

Purpose

Glioblastoma (GBM) is the commonest brain tumour in adults. A sub-population of cells within these tumours, known as cancer stem cells (CSCs), is thought to mediate their chemo-/radiotherapy resistance. CD133 is a cell surface marker that is used to identify and isolate GBM CSCs. However, its functional significance, as well as the relevant microenvironment in which to study CD133, have so far remained unknown. Here, we examined the effect of hypoxia on the expression of CD133 and on that of the hypoxia-related factors HIF-1α and HIF-2α, and the potential functional significance of CD133 expression on the acquisition of chemo-resistance by GBM cells.

Methods

CD133, HIF-1α, HIF-2α, VEFG and (control) HPRT mRNA expression analyses were carried out on GBM cells (U251, U87 and SNB19; 2D or 3D cultures) under both normoxic and hypoxic conditions, using qRT-PCR. siRNA was used to downregulate CD133, HIF-1α and HIF-2α expression in the GBM cells, which was confirmed by flow cytometry and qRT-PCR, respectively. Drug sensitivity-related IC50 values were established using an Alamar Blue cell viability assay in conjunction with the Graphpad prism software tool.

Results

We found that the expression of CD133 was upregulated under hypoxic conditions in both the 2D and 3D GBM cell culture models. In addition, an increased resistance to cisplatin, temozolomide and etoposide was observed in the GBM cells cultured under hypoxic conditions compared to normoxic conditions. siRNA-mediated knockdown of either HIF-1α or HIF-2α resulted in a reduced CD133 expression, with HIF-2α having a more long-term effect. We also found that HIF-2α downregulation sensitized the GBM cells to cisplatin to a greater extent than HIF-1α, whereas CD133 knockdown had a more marked effect on cisplatin sensitisation than knockdown of either one of the HIFs, suggesting the existence of a HIF-independent cisplatin resistance mechanism mediated by CD133. This same mechanism does not seem to be involved in temozolomide resistance, since we found that HIF-1α downregulation, but not HIF-2α or CD133 downregulation, sensitized GBM cells to temozolomide.

Conclusions

From our data we conclude that the mechanisms underlying hypoxia-induced CD133-mediated cisplatin resistance may be instrumental for the design of new GBM treatment strategies.
Appendix
Available only for authorised users
Literature
1.
go back to reference H. Zong, R.G. Verhaak, P. Canoll, The cellular origin for malignant glioma and prospects for clinical advancements. Expert. Rev. Mol. Diagn. 12, 383–394 (2012)CrossRefPubMedPubMedCentral H. Zong, R.G. Verhaak, P. Canoll, The cellular origin for malignant glioma and prospects for clinical advancements. Expert. Rev. Mol. Diagn. 12, 383–394 (2012)CrossRefPubMedPubMedCentral
2.
go back to reference M. Staberg, S.R. Michaelsen, R.D. Rasmussen, M. Villingshøj, H.S. Poulsen, P. Hamerlik, Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine. Cell. Oncol. 40, 21–32 (2017)CrossRef M. Staberg, S.R. Michaelsen, R.D. Rasmussen, M. Villingshøj, H.S. Poulsen, P. Hamerlik, Inhibition of histone deacetylases sensitizes glioblastoma cells to lomustine. Cell. Oncol. 40, 21–32 (2017)CrossRef
3.
go back to reference A. Omuro, L.M. DeAngelis, Glioblastoma and other malignant gliomas: a clinical review. JAMA 310, 1842–1850 (2013)CrossRefPubMed A. Omuro, L.M. DeAngelis, Glioblastoma and other malignant gliomas: a clinical review. JAMA 310, 1842–1850 (2013)CrossRefPubMed
4.
go back to reference R. Stupp, M.E. Hegi, M.R. Gilbert, A. Chakravarti, Chemoradiotherapy in malignant glioma: standard of care and future directions. J. Clin. Oncol. 25, 4127–4136 (2007)CrossRefPubMed R. Stupp, M.E. Hegi, M.R. Gilbert, A. Chakravarti, Chemoradiotherapy in malignant glioma: standard of care and future directions. J. Clin. Oncol. 25, 4127–4136 (2007)CrossRefPubMed
5.
go back to reference S.S. Agarwala, J.M. Kirkwood, Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist 5, 144–151 (2000)CrossRefPubMed S.S. Agarwala, J.M. Kirkwood, Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist 5, 144–151 (2000)CrossRefPubMed
6.
go back to reference D. Matias, J. Balça-Silva, L.G. Dubois, B. Pontes, V.P. Ferrer, L. Rosário, A. do Carmo, J. Echevarria-Lima, A.B. Sarmento-Ribeiro, M.C. Lopes, Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition. Cell. Oncol. 40, 247–261 (2017)CrossRef D. Matias, J. Balça-Silva, L.G. Dubois, B. Pontes, V.P. Ferrer, L. Rosário, A. do Carmo, J. Echevarria-Lima, A.B. Sarmento-Ribeiro, M.C. Lopes, Dual treatment with shikonin and temozolomide reduces glioblastoma tumor growth, migration and glial-to-mesenchymal transition. Cell. Oncol. 40, 247–261 (2017)CrossRef
7.
go back to reference S. Yip, J. Miao, D.P. Cahill, A.J. Iafrate, K. Aldape, C.L. Nutt, D.N. Louis, MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin. Cancer Res. 15, 4622–4629 (2009)CrossRefPubMedPubMedCentral S. Yip, J. Miao, D.P. Cahill, A.J. Iafrate, K. Aldape, C.L. Nutt, D.N. Louis, MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clin. Cancer Res. 15, 4622–4629 (2009)CrossRefPubMedPubMedCentral
8.
go back to reference L. Capdevila, S. Cros, J.-L. Ramirez, C. Sanz, C. Carrato, M. Romeo, O. Etxaniz, C. Hostalot, A. Massuet, J.L. Cuadra, Neoadjuvant cisplatin plus temozolomide versus standard treatment in patients with unresectable glioblastoma or anaplastic astrocytoma: a differential effect of MGMT methylation. J. Neuro-Oncol. 117, 77–84 (2014)CrossRef L. Capdevila, S. Cros, J.-L. Ramirez, C. Sanz, C. Carrato, M. Romeo, O. Etxaniz, C. Hostalot, A. Massuet, J.L. Cuadra, Neoadjuvant cisplatin plus temozolomide versus standard treatment in patients with unresectable glioblastoma or anaplastic astrocytoma: a differential effect of MGMT methylation. J. Neuro-Oncol. 117, 77–84 (2014)CrossRef
9.
go back to reference W.A. Yung, T.J. Janus, M. Maor, L.G. Feun, Adjuvant chemotherapy with carmustine and cisplatin for patients with malignant gliomas. J. Neuro-Oncol. 12, 131–135 (1992)CrossRef W.A. Yung, T.J. Janus, M. Maor, L.G. Feun, Adjuvant chemotherapy with carmustine and cisplatin for patients with malignant gliomas. J. Neuro-Oncol. 12, 131–135 (1992)CrossRef
10.
go back to reference F. Zustovich, G. Lombardi, A. Della Puppa, A. Rotilio, R. Scienza, D. Pastorelli, A phase II study of cisplatin and temozolomide in heavily pre-treated patients with temozolomide-refractory high-grade malignant glioma. Anticancer Res. 29, 4275–4279 (2009)PubMed F. Zustovich, G. Lombardi, A. Della Puppa, A. Rotilio, R. Scienza, D. Pastorelli, A phase II study of cisplatin and temozolomide in heavily pre-treated patients with temozolomide-refractory high-grade malignant glioma. Anticancer Res. 29, 4275–4279 (2009)PubMed
11.
go back to reference A.A. Brandes, U. Basso, M. Reni, F. Vastola, A. Tosoni, G. Cavallo, L. Scopece, A.J. Ferreri, M.G. Panucci, S. Monfardini, First-line chemotherapy with cisplatin plus fractionated temozolomide in recurrent glioblastoma multiforme: a phase II study of the Gruppo Italiano Cooperativo di Neuro-Oncologia. J. Clin. Oncol. 22, 1598–1604 (2004)CrossRefPubMed A.A. Brandes, U. Basso, M. Reni, F. Vastola, A. Tosoni, G. Cavallo, L. Scopece, A.J. Ferreri, M.G. Panucci, S. Monfardini, First-line chemotherapy with cisplatin plus fractionated temozolomide in recurrent glioblastoma multiforme: a phase II study of the Gruppo Italiano Cooperativo di Neuro-Oncologia. J. Clin. Oncol. 22, 1598–1604 (2004)CrossRefPubMed
12.
go back to reference S.S. Byun, S.W. Kim, H. Choi, C. Lee, E. Lee, Augmentation of cisplatin sensitivity in cisplatin-resistant human bladder cancer cells by modulating glutathione concentrations and glutathione-related enzyme activities. BJU Int. 95, 1086–1090 (2005)CrossRefPubMed S.S. Byun, S.W. Kim, H. Choi, C. Lee, E. Lee, Augmentation of cisplatin sensitivity in cisplatin-resistant human bladder cancer cells by modulating glutathione concentrations and glutathione-related enzyme activities. BJU Int. 95, 1086–1090 (2005)CrossRefPubMed
13.
go back to reference A. Leonard, J.E. Wolff, Etoposide improves survival in high-grade glioma: a meta-analysis. Anticancer Res. 33, 3307–3315 (2013)PubMed A. Leonard, J.E. Wolff, Etoposide improves survival in high-grade glioma: a meta-analysis. Anticancer Res. 33, 3307–3315 (2013)PubMed
14.
go back to reference F.R. Balkwill, M. Capasso, T. Hagemann, The tumour microenvironment at a glance. J. Cell Sci. 125, 5591–5596 (2012)CrossRefPubMed F.R. Balkwill, M. Capasso, T. Hagemann, The tumour microenvironment at a glance. J. Cell Sci. 125, 5591–5596 (2012)CrossRefPubMed
15.
go back to reference J.M. Brown, W.R. Wilson, Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 4, 437–447 (2004)CrossRefPubMed J.M. Brown, W.R. Wilson, Exploiting tumour hypoxia in cancer treatment. Nat. Rev. Cancer 4, 437–447 (2004)CrossRefPubMed
16.
17.
go back to reference L. Yang, C. Lin, L. Wang, H. Guo, X. Wang, Hypoxia and hypoxia-inducible factors in glioblastoma multiforme progression and therapeutic implications. Exp. Cell Res. 318, 2417–2426 (2012)CrossRefPubMed L. Yang, C. Lin, L. Wang, H. Guo, X. Wang, Hypoxia and hypoxia-inducible factors in glioblastoma multiforme progression and therapeutic implications. Exp. Cell Res. 318, 2417–2426 (2012)CrossRefPubMed
18.
go back to reference S.M. Evans, K.D. Judy, I. Dunphy, W.T. Jenkins, W.-T. Hwang, P.T. Nelson, R.A. Lustig, K. Jenkins, D.P. Magarelli, S.M. Hahn, Hypoxia is important in the biology and aggression of human glial brain tumors. Clin. Cancer Res. 10, 8177–8184 (2004)CrossRefPubMed S.M. Evans, K.D. Judy, I. Dunphy, W.T. Jenkins, W.-T. Hwang, P.T. Nelson, R.A. Lustig, K. Jenkins, D.P. Magarelli, S.M. Hahn, Hypoxia is important in the biology and aggression of human glial brain tumors. Clin. Cancer Res. 10, 8177–8184 (2004)CrossRefPubMed
19.
go back to reference C.W. Chou, C.C. Wang, C.P. Wu, Y.J. Lin, Y.C. Lee, Y.W. Cheng, C.H. Hsieh, Tumor cycling hypoxia induces chemoresistance in glioblastoma multiforme by upregulating the expression and function of ABCB1. Neuro-Oncology 14, 1227–1238 (2012)CrossRefPubMedPubMedCentral C.W. Chou, C.C. Wang, C.P. Wu, Y.J. Lin, Y.C. Lee, Y.W. Cheng, C.H. Hsieh, Tumor cycling hypoxia induces chemoresistance in glioblastoma multiforme by upregulating the expression and function of ABCB1. Neuro-Oncology 14, 1227–1238 (2012)CrossRefPubMedPubMedCentral
20.
go back to reference S. Seidel, B.K. Garvalov, V. Wirta, L. von Stechow, A. Schänzer, K. Meletis, M. Wolter, D. Sommerlad, A.-T. Henze, M. Nistér, A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2α. Brain 133, 983–995 (2010)CrossRefPubMed S. Seidel, B.K. Garvalov, V. Wirta, L. von Stechow, A. Schänzer, K. Meletis, M. Wolter, D. Sommerlad, A.-T. Henze, M. Nistér, A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2α. Brain 133, 983–995 (2010)CrossRefPubMed
21.
22.
go back to reference A. Loboda, A. Jozkowicz, J. Dulak, HIF-1 and HIF-2 transcription factors—similar but not identical. Mol. Cell 29, 435–442 (2010)CrossRef A. Loboda, A. Jozkowicz, J. Dulak, HIF-1 and HIF-2 transcription factors—similar but not identical. Mol. Cell 29, 435–442 (2010)CrossRef
23.
go back to reference L. Holmquist-mengelbier, E. Fredlund, T. Löfstedt, R. Noguera, S. Navarro, H. Nilsson, A. Pietras, J. Vallon-christersson, Å. Borg, K. Gradin, Recruitment of HIF-1α and HIF-2α to common target genes is differentially regulated in neuroblastoma: HIF-2α promotes an aggressive phenotype. Cancer Cell 10, 413–423 (2006)CrossRefPubMed L. Holmquist-mengelbier, E. Fredlund, T. Löfstedt, R. Noguera, S. Navarro, H. Nilsson, A. Pietras, J. Vallon-christersson, Å. Borg, K. Gradin, Recruitment of HIF-1α and HIF-2α to common target genes is differentially regulated in neuroblastoma: HIF-2α promotes an aggressive phenotype. Cancer Cell 10, 413–423 (2006)CrossRefPubMed
24.
go back to reference A. Soeda, M. Park, D. Lee, A. Mintz, A. Androutsellis-Theotokis, R. McKay, J. Engh, T. Iwama, T. Kunisada, A. Kassam, Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1α. Oncogene 28, 3949–3959 (2009)CrossRefPubMed A. Soeda, M. Park, D. Lee, A. Mintz, A. Androutsellis-Theotokis, R. McKay, J. Engh, T. Iwama, T. Kunisada, A. Kassam, Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1α. Oncogene 28, 3949–3959 (2009)CrossRefPubMed
25.
go back to reference P. Brescia, B. Ortensi, L. Fornasari, D. Levi, G. Broggi, G. Pelicci, CD133 is essential for glioblastoma stem cell maintenance. Stem Cells 31, 857–869 (2013)CrossRefPubMed P. Brescia, B. Ortensi, L. Fornasari, D. Levi, G. Broggi, G. Pelicci, CD133 is essential for glioblastoma stem cell maintenance. Stem Cells 31, 857–869 (2013)CrossRefPubMed
27.
go back to reference U. Karsten, S. Goletz, What makes cancer stem cell markers different? Spring 2, 301 (2013)CrossRef U. Karsten, S. Goletz, What makes cancer stem cell markers different? Spring 2, 301 (2013)CrossRef
28.
go back to reference S.M. Pollard, K. Yoshikawa, I.D. Clarke, D. Danovi, S. Stricker, R. Russell, J. Bayani, R. Head, M. Lee, M. Bernstein, Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell 4, 568–580 (2009)CrossRefPubMed S.M. Pollard, K. Yoshikawa, I.D. Clarke, D. Danovi, S. Stricker, R. Russell, J. Bayani, R. Head, M. Lee, M. Bernstein, Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell 4, 568–580 (2009)CrossRefPubMed
29.
go back to reference L. Hutchinson, R. Kirk, High drug attrition rates-where are we going wrong? Nat. Rev. Clin. Oncol. 8, 189–190 (2011)CrossRefPubMed L. Hutchinson, R. Kirk, High drug attrition rates-where are we going wrong? Nat. Rev. Clin. Oncol. 8, 189–190 (2011)CrossRefPubMed
30.
go back to reference A.M. Grabowska, J. Hughes, S.A. Watson, Use of interfering RNA to investigate the role of endogenous gastrin in the survival of gastrointestinal cancer cells. Br. J. Cancer 96, 464–473 (2007)CrossRefPubMedPubMedCentral A.M. Grabowska, J. Hughes, S.A. Watson, Use of interfering RNA to investigate the role of endogenous gastrin in the survival of gastrointestinal cancer cells. Br. J. Cancer 96, 464–473 (2007)CrossRefPubMedPubMedCentral
32.
go back to reference P. Vaupel, L. Harrison, Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 9, 4–9 (2004)CrossRefPubMed P. Vaupel, L. Harrison, Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 9, 4–9 (2004)CrossRefPubMed
33.
go back to reference C. Petit, F. Gouel, I. Dubus, C. Heuclin, K. Roget, J. Vannier, Hypoxia promotes chemoresistance in acute lymphoblastic leukemia cell lines by modulating death signaling pathways. BMC Cancer 16, 746 (2016)CrossRefPubMedPubMedCentral C. Petit, F. Gouel, I. Dubus, C. Heuclin, K. Roget, J. Vannier, Hypoxia promotes chemoresistance in acute lymphoblastic leukemia cell lines by modulating death signaling pathways. BMC Cancer 16, 746 (2016)CrossRefPubMedPubMedCentral
34.
go back to reference S.D. Fouse, J.L. Nakamura, C.D. James, S. Chang, J.F. Costello, Response of primary glioblastoma cells to therapy is patient specific and independent of cancer stem cell phenotype. Neuro-Oncology 16, 361–371 (2013)CrossRefPubMedPubMedCentral S.D. Fouse, J.L. Nakamura, C.D. James, S. Chang, J.F. Costello, Response of primary glioblastoma cells to therapy is patient specific and independent of cancer stem cell phenotype. Neuro-Oncology 16, 361–371 (2013)CrossRefPubMedPubMedCentral
35.
go back to reference E.R. Blazek, J.L. Foutch, G. Maki, Daoy medulloblastoma cells that express CD133 are radioresistant relative to CD133− cells, and the CD133+ sector is enlarged by hypoxia. Int. J. Radiat. Oncol. Biol. Phys. 67, 1–5 (2007)CrossRefPubMed E.R. Blazek, J.L. Foutch, G. Maki, Daoy medulloblastoma cells that express CD133 are radioresistant relative to CD133− cells, and the CD133+ sector is enlarged by hypoxia. Int. J. Radiat. Oncol. Biol. Phys. 67, 1–5 (2007)CrossRefPubMed
36.
go back to reference K. Wang, F.M. Kievit, S.J. Florczyk, Z.R. Stephen, M. Zhang, 3D porous chitosan–alginate scaffolds as an in vitro model for evaluating nanoparticle-mediated tumor targeting and gene delivery to prostate cancer. Biomacromolecules 16, 3362–3372 (2015)CrossRefPubMedPubMedCentral K. Wang, F.M. Kievit, S.J. Florczyk, Z.R. Stephen, M. Zhang, 3D porous chitosan–alginate scaffolds as an in vitro model for evaluating nanoparticle-mediated tumor targeting and gene delivery to prostate cancer. Biomacromolecules 16, 3362–3372 (2015)CrossRefPubMedPubMedCentral
37.
go back to reference M.Y. Koh, R. Lemos, X. Liu, G. Powis, The hypoxia-associated factor switches cells from HIF-1α-to HIF-2α-dependent signaling promoting stem cell characteristics, aggressive tumor growth and invasion. Cancer Res. 71, 4015–4027 (2011)CrossRefPubMedPubMedCentral M.Y. Koh, R. Lemos, X. Liu, G. Powis, The hypoxia-associated factor switches cells from HIF-1α-to HIF-2α-dependent signaling promoting stem cell characteristics, aggressive tumor growth and invasion. Cancer Res. 71, 4015–4027 (2011)CrossRefPubMedPubMedCentral
38.
go back to reference R. Chen, E.M. Dioum, R.T. Hogg, R.D. Gerard, J.A. Garcia, Hypoxia increases sirtuin 1 expression in a hypoxia-inducible factor-dependent manner. J. Biol. Chem. 286, 13869–13878 (2011)CrossRefPubMedPubMedCentral R. Chen, E.M. Dioum, R.T. Hogg, R.D. Gerard, J.A. Garcia, Hypoxia increases sirtuin 1 expression in a hypoxia-inducible factor-dependent manner. J. Biol. Chem. 286, 13869–13878 (2011)CrossRefPubMedPubMedCentral
40.
go back to reference D. Wang, A. Berglund, R.S. Kenchappa, P.A. Forsyth, J.J. Mulé, A.B. Etame, BIRC3 is a novel driver of therapeutic resistance in glioblastoma. Sci. Rep. 6, 21710 (2016)CrossRefPubMedPubMedCentral D. Wang, A. Berglund, R.S. Kenchappa, P.A. Forsyth, J.J. Mulé, A.B. Etame, BIRC3 is a novel driver of therapeutic resistance in glioblastoma. Sci. Rep. 6, 21710 (2016)CrossRefPubMedPubMedCentral
42.
go back to reference R. Sullivan, G.C. Paré, L.J. Frederiksen, G.L. Semenza, C.H. Graham, Hypoxia-induced resistance to anticancer drugs is associated with decreased senescence and requires hypoxia-inducible factor-1 activity. Mol. Cancer Ther. 7, 1961–1973 (2008)CrossRefPubMed R. Sullivan, G.C. Paré, L.J. Frederiksen, G.L. Semenza, C.H. Graham, Hypoxia-induced resistance to anticancer drugs is associated with decreased senescence and requires hypoxia-inducible factor-1 activity. Mol. Cancer Ther. 7, 1961–1973 (2008)CrossRefPubMed
43.
go back to reference J. Adamski, A. Price, C. Dive, G. Makin, Hypoxia–induced cytotoxic drug resistance in osteosarcoma is independent of HIF-1Alpha. PLoS One 8, e65304 (2013)CrossRefPubMedPubMedCentral J. Adamski, A. Price, C. Dive, G. Makin, Hypoxia–induced cytotoxic drug resistance in osteosarcoma is independent of HIF-1Alpha. PLoS One 8, e65304 (2013)CrossRefPubMedPubMedCentral
44.
go back to reference L. Persano, F. Pistollato, E. Rampazzo, A. Della Puppa, S. Abbadi, C. Frasson, F. Volpin, S. Indraccolo, R. Scienza, G. Basso, BMP2 sensitizes glioblastoma stem-like cells to Temozolomide by affecting HIF-1α stability and MGMT expression. Cell Death Dis. 3, e412 (2012)CrossRefPubMedPubMedCentral L. Persano, F. Pistollato, E. Rampazzo, A. Della Puppa, S. Abbadi, C. Frasson, F. Volpin, S. Indraccolo, R. Scienza, G. Basso, BMP2 sensitizes glioblastoma stem-like cells to Temozolomide by affecting HIF-1α stability and MGMT expression. Cell Death Dis. 3, e412 (2012)CrossRefPubMedPubMedCentral
45.
go back to reference L. Li, X. Lin, A.R. Shoemaker, D.H. Albert, S.W. Fesik, Y. Shen, Hypoxia-inducible factor-1 inhibition in combination with temozolomide treatment exhibits robust antitumor efficacy in vivo. Clin. Cancer Res. 12, 4747–4754 (2006)CrossRefPubMed L. Li, X. Lin, A.R. Shoemaker, D.H. Albert, S.W. Fesik, Y. Shen, Hypoxia-inducible factor-1 inhibition in combination with temozolomide treatment exhibits robust antitumor efficacy in vivo. Clin. Cancer Res. 12, 4747–4754 (2006)CrossRefPubMed
46.
go back to reference A. Hamidian, K. von Stedingk, M.M. Thorén, S. Mohlin, S. Påhlman, Differential regulation of HIF-1α and HIF-2α in neuroblastoma: Estrogen-related receptor alpha (ERRα) regulates HIF2A transcription and correlates to poor outcome. Biochem. Biophys. Res. Commun. 461, 560–567 (2015)CrossRefPubMed A. Hamidian, K. von Stedingk, M.M. Thorén, S. Mohlin, S. Påhlman, Differential regulation of HIF-1α and HIF-2α in neuroblastoma: Estrogen-related receptor alpha (ERRα) regulates HIF2A transcription and correlates to poor outcome. Biochem. Biophys. Res. Commun. 461, 560–567 (2015)CrossRefPubMed
47.
go back to reference G. Perazzoli, J. Prados, R. Ortiz, O. Caba, L. Cabeza, M. Berdasco, B. Gónzalez, C. Melguizo, Temozolomide resistance in glioblastoma cell lines: implication of MGMT, MMR, P-glycoprotein and CD133 expression. PLoS One 10, e0140131 (2015)CrossRefPubMedPubMedCentral G. Perazzoli, J. Prados, R. Ortiz, O. Caba, L. Cabeza, M. Berdasco, B. Gónzalez, C. Melguizo, Temozolomide resistance in glioblastoma cell lines: implication of MGMT, MMR, P-glycoprotein and CD133 expression. PLoS One 10, e0140131 (2015)CrossRefPubMedPubMedCentral
48.
go back to reference G. Liu, X. Yuan, Z. Zeng, P. Tunici, H. Ng, I.R. Abdulkadir, L. Lu, D. Irvin, K.L. Black, S.Y. John, Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol. Cancer 5, 67 (2006)CrossRefPubMedPubMedCentral G. Liu, X. Yuan, Z. Zeng, P. Tunici, H. Ng, I.R. Abdulkadir, L. Lu, D. Irvin, K.L. Black, S.Y. John, Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol. Cancer 5, 67 (2006)CrossRefPubMedPubMedCentral
49.
go back to reference L. Dong, N. Qi, R.-M. Ge, C.-L. Cao, F. Lan, L. Shen, Overexpression of CD133 promotes the phosphorylation of Erk in U87MG human glioblastoma cells. Neurosci. Lett. 484, 210–214 (2010)CrossRefPubMed L. Dong, N. Qi, R.-M. Ge, C.-L. Cao, F. Lan, L. Shen, Overexpression of CD133 promotes the phosphorylation of Erk in U87MG human glioblastoma cells. Neurosci. Lett. 484, 210–214 (2010)CrossRefPubMed
50.
go back to reference Y. Wei, Y. Jiang, F. Zou, Y. Liu, S. Wang, N. Xu, W. Xu, C. Cui, Y. Xing, Y. Liu, B. Cao, C. Liu, G. Wu, H. Ao, X. Zhang, J. Jiang, Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells. Proc. Natl. Acad. Sci. 110, 6829–6834 (2013)CrossRefPubMedPubMedCentral Y. Wei, Y. Jiang, F. Zou, Y. Liu, S. Wang, N. Xu, W. Xu, C. Cui, Y. Xing, Y. Liu, B. Cao, C. Liu, G. Wu, H. Ao, X. Zhang, J. Jiang, Activation of PI3K/Akt pathway by CD133-p85 interaction promotes tumorigenic capacity of glioma stem cells. Proc. Natl. Acad. Sci. 110, 6829–6834 (2013)CrossRefPubMedPubMedCentral
51.
go back to reference K.A. West, S. Sianna Castillo, P.A. Dennis, Activation of the PI3K/Akt pathway and chemotherapeutic resistance. Drug Resist. Updat. 5, 234–248 (2002) CrossRefPubMed K.A. West, S. Sianna Castillo, P.A. Dennis, Activation of the PI3K/Akt pathway and chemotherapeutic resistance. Drug Resist. Updat. 5, 234–248 (2002) CrossRefPubMed
52.
go back to reference M. Kase, A. Minajeva, K. Niinepuu, S. Kase, M. Vardja, T. Asser, J. Jaal, Impact of CD133 positive stem cell proportion on survival in patients with glioblastoma multiforme. Radiol. Oncol. 47, 405–410 (2013)CrossRefPubMedPubMedCentral M. Kase, A. Minajeva, K. Niinepuu, S. Kase, M. Vardja, T. Asser, J. Jaal, Impact of CD133 positive stem cell proportion on survival in patients with glioblastoma multiforme. Radiol. Oncol. 47, 405–410 (2013)CrossRefPubMedPubMedCentral
Metadata
Title
A HIF-independent, CD133-mediated mechanism of cisplatin resistance in glioblastoma cells
Authors
Eroje M. Ahmed
Gagori Bandopadhyay
Beth Coyle
Anna Grabowska
Publication date
01-06-2018
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 3/2018
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-018-0374-8

Other articles of this Issue 3/2018

Cellular Oncology 3/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine