Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Krüppel-like factor 9 and histone deacetylase inhibitors synergistically induce cell death in glioblastoma stem-like cells

Authors: Brian Tung, Ding Ma, Shuyan Wang, Olutobi Oyinlade, John Laterra, Mingyao Ying, Sheng-Qing Lv, Shuang Wei, Shuli Xia

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

The dismal prognosis of patients with glioblastoma (GBM) is attributed to a rare subset of cancer stem cells that display characteristics of tumor initiation, growth, and resistance to aggressive treatment involving chemotherapy and concomitant radiation. Recent research on the substantial role of epigenetic mechanisms in the pathogenesis of cancers has prompted the investigation of the enzymatic modifications of histone proteins for therapeutic drug targeting. In this work, we have examined the function of Krüppel-like factor 9 (KLF9), a transcription factor, in chemotherapy sensitization to histone deacetylase inhibitors (HDAC inhibitors).

Methods

Since GBM neurosphere cultures from patient-derived gliomas are enriched for GBM stem-like cells (GSCs) and form highly invasive and proliferative xenografts that recapitulate the features demonstrated in human patients diagnosed with GBM, we established inducible KLF9 expression systems in these GBM neurosphere cells and investigated cell death in the presence of epigenetic modulators such as histone deacetylase (HDAC) inhibitors.

Results

We demonstrated that KLF9 expression combined with HDAC inhibitor panobinostat (LBH589) dramatically induced glioma stem cell death via both apoptosis and necroptosis in a synergistic manner. The combination of KLF9 expression and LBH589 treatment affected cell cycle by substantially decreasing the percentage of cells at S-phase. This phenomenon is further corroborated by the upregulation of cell cycle inhibitors p21 and p27. Further, we determined that KLF9 and LBH589 regulated the expression of pro- and anti- apoptotic proteins, suggesting a mechanism that involves the caspase-dependent apoptotic pathway. In addition, we demonstrated that apoptosis and necrosis inhibitors conferred minimal protective effects against cell death, while inhibitors of the necroptosis pathway significantly blocked cell death.

Conclusions

Our findings suggest a detailed understanding of how KLF9 expression in cancer cells with epigenetic modulators like HDAC inhibitors may promote synergistic cell death through a mechanism involving both apoptosis and necroptosis that will benefit novel combinatory antitumor strategies to treat malignant brain tumors.
Literature
1.
go back to reference Huse JT, Holland E, DeAngelis LM. Glioblastoma: molecular analysis and clinical implications. Annu Rev Med. 2013;64:59–70.CrossRef Huse JT, Holland E, DeAngelis LM. Glioblastoma: molecular analysis and clinical implications. Annu Rev Med. 2013;64:59–70.CrossRef
2.
go back to reference Enderling H, Hlatky L, Hahnfeldt P. Cancer stem cells: a minor Cancer subpopulation that redefines global Cancer features. Front Oncol. 2013;3:76.PubMedPubMedCentral Enderling H, Hlatky L, Hahnfeldt P. Cancer stem cells: a minor Cancer subpopulation that redefines global Cancer features. Front Oncol. 2013;3:76.PubMedPubMedCentral
3.
go back to reference Hale JS, Sinyuk M, Rich JN, Lathia JD. Decoding the cancer stem cell hypothesis in glioblastoma. CNS Oncol. 2013;2:319–30.CrossRef Hale JS, Sinyuk M, Rich JN, Lathia JD. Decoding the cancer stem cell hypothesis in glioblastoma. CNS Oncol. 2013;2:319–30.CrossRef
4.
go back to reference Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol, 2013; Chapter 14: Unit 14 25. Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. Curr Protoc Pharmacol, 2013; Chapter 14: Unit 14 25.
5.
go back to reference Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–21.CrossRef Galli R, Binda E, Orfanelli U, et al. Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–21.CrossRef
6.
go back to reference Piccirillo SG, Reynolds BA, Zanetti N, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 2006;444:761–5.CrossRef Piccirillo SG, Reynolds BA, Zanetti N, et al. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 2006;444:761–5.CrossRef
7.
go back to reference Sun P, Xia S, Lal B, et al. DNER, an epigenetically modulated gene, regulates glioblastoma-derived neurosphere cell differentiation and tumor propagation. Stem Cells. 2009;27:1473–86.CrossRef Sun P, Xia S, Lal B, et al. DNER, an epigenetically modulated gene, regulates glioblastoma-derived neurosphere cell differentiation and tumor propagation. Stem Cells. 2009;27:1473–86.CrossRef
8.
go back to reference Ying M, Wang S, Sang Y, et al. Regulation of glioblastoma stem cells by retinoic acid: role for notch pathway inhibition. Oncogene. 2011;30:3454–67.CrossRef Ying M, Wang S, Sang Y, et al. Regulation of glioblastoma stem cells by retinoic acid: role for notch pathway inhibition. Oncogene. 2011;30:3454–67.CrossRef
9.
go back to reference Ying M, Sang Y, Li Y, et al. Kruppel-like family of transcription factor 9, a differentiation-associated transcription factor, suppresses Notch1 signaling and inhibits glioblastoma-initiating stem cells. Stem Cells. 2011;29:20–31.CrossRef Ying M, Sang Y, Li Y, et al. Kruppel-like family of transcription factor 9, a differentiation-associated transcription factor, suppresses Notch1 signaling and inhibits glioblastoma-initiating stem cells. Stem Cells. 2011;29:20–31.CrossRef
10.
go back to reference Kaczynski J, Cook T, Urrutia R. Sp1- and Kruppel-like transcription factors. Genome Biol. 2003;4:206.CrossRef Kaczynski J, Cook T, Urrutia R. Sp1- and Kruppel-like transcription factors. Genome Biol. 2003;4:206.CrossRef
11.
go back to reference Pearson R, Fleetwood J, Eaton S, Crossley M, Bao S. Kruppel-like transcription factors: a functional family. Int J Biochem Cell Biol. 2008;40:1996–2001.CrossRef Pearson R, Fleetwood J, Eaton S, Crossley M, Bao S. Kruppel-like transcription factors: a functional family. Int J Biochem Cell Biol. 2008;40:1996–2001.CrossRef
12.
go back to reference Wang S, Shi X, Wei S, et al. Kruppel-like factor 4 (KLF4) induces mitochondrial fusion and increases spare respiratory capacity of human glioblastoma cells. J Biol Chem. 2018;293:6544–55.CrossRef Wang S, Shi X, Wei S, et al. Kruppel-like factor 4 (KLF4) induces mitochondrial fusion and increases spare respiratory capacity of human glioblastoma cells. J Biol Chem. 2018;293:6544–55.CrossRef
13.
go back to reference Leng Z, Tao K, Xia Q, et al. Kruppel-like factor 4 acts as an oncogene in colon cancer stem cell-enriched spheroid cells. PLoS One. 2013;8:e56082.CrossRef Leng Z, Tao K, Xia Q, et al. Kruppel-like factor 4 acts as an oncogene in colon cancer stem cell-enriched spheroid cells. PLoS One. 2013;8:e56082.CrossRef
14.
go back to reference Lin ZS, Chu HC, Yen YC, Lewis BC, Chen YW. Kruppel-like factor 4, a tumor suppressor in hepatocellular carcinoma cells reverts epithelial mesenchymal transition by suppressing slug expression. PLoS One. 2012;7:e43593.CrossRef Lin ZS, Chu HC, Yen YC, Lewis BC, Chen YW. Kruppel-like factor 4, a tumor suppressor in hepatocellular carcinoma cells reverts epithelial mesenchymal transition by suppressing slug expression. PLoS One. 2012;7:e43593.CrossRef
15.
go back to reference Kang L, Lu B, Xu J, Hu H, Lai M. Downregulation of Kruppel-like factor 9 in human colorectal cancer. Pathol Int. 2008;58:334–8.CrossRef Kang L, Lu B, Xu J, Hu H, Lai M. Downregulation of Kruppel-like factor 9 in human colorectal cancer. Pathol Int. 2008;58:334–8.CrossRef
16.
go back to reference Simmen FA, Su Y, Xiao R, Zeng Z, Simmen RC. The Kruppel-like factor 9 (KLF9) network in HEC-1-a endometrial carcinoma cells suggests the carcinogenic potential of dys-regulated KLF9 expression. Reprod Biol Endocrinol. 2008;6:41.CrossRef Simmen FA, Su Y, Xiao R, Zeng Z, Simmen RC. The Kruppel-like factor 9 (KLF9) network in HEC-1-a endometrial carcinoma cells suggests the carcinogenic potential of dys-regulated KLF9 expression. Reprod Biol Endocrinol. 2008;6:41.CrossRef
17.
go back to reference Ying M, Tilghman J, Wei Y, et al. Kruppel-like factor-9 (KLF9) inhibits glioblastoma stemness through global transcription repression and integrin alpha6 inhibition. J Biol Chem. 2014;289:32742–56.CrossRef Ying M, Tilghman J, Wei Y, et al. Kruppel-like factor-9 (KLF9) inhibits glioblastoma stemness through global transcription repression and integrin alpha6 inhibition. J Biol Chem. 2014;289:32742–56.CrossRef
18.
go back to reference Kanwal R, Gupta S. Epigenetic modifications in cancer. Clin Genet. 2012;81:303–11.CrossRef Kanwal R, Gupta S. Epigenetic modifications in cancer. Clin Genet. 2012;81:303–11.CrossRef
19.
go back to reference Tambaro FP, Dell’aversana C, Carafa V, et al. Histone deacetylase inhibitors: clinical implications for hematological malignancies. Clin Epigenetics. 2010;1:25–44.CrossRef Tambaro FP, Dell’aversana C, Carafa V, et al. Histone deacetylase inhibitors: clinical implications for hematological malignancies. Clin Epigenetics. 2010;1:25–44.CrossRef
20.
go back to reference Bellarosa D, Bressan A, Bigioni M, et al. SAHA/Vorinostat induces the expression of the CD137 receptor/ligand system and enhances apoptosis mediated by soluble CD137 receptor in a human breast cancer cell line. Int J Oncol. 2012;41:1486–94.CrossRef Bellarosa D, Bressan A, Bigioni M, et al. SAHA/Vorinostat induces the expression of the CD137 receptor/ligand system and enhances apoptosis mediated by soluble CD137 receptor in a human breast cancer cell line. Int J Oncol. 2012;41:1486–94.CrossRef
21.
go back to reference Gaur V, Connor T, Sanigorski A, et al. Disruption of the class IIa HDAC corepressor complex increases energy expenditure and lipid oxidation. Cell Rep. 2016;16:2802–10.CrossRef Gaur V, Connor T, Sanigorski A, et al. Disruption of the class IIa HDAC corepressor complex increases energy expenditure and lipid oxidation. Cell Rep. 2016;16:2802–10.CrossRef
22.
go back to reference Rosato RR, Maggio SC, Almenara JA, et al. The histone deacetylase inhibitor LAQ824 induces human leukemia cell death through a process involving XIAP down-regulation, oxidative injury, and the acid sphingomyelinase-dependent generation of ceramide. Mol Pharmacol. 2006;69:216–25.PubMed Rosato RR, Maggio SC, Almenara JA, et al. The histone deacetylase inhibitor LAQ824 induces human leukemia cell death through a process involving XIAP down-regulation, oxidative injury, and the acid sphingomyelinase-dependent generation of ceramide. Mol Pharmacol. 2006;69:216–25.PubMed
23.
go back to reference Laubach JP, Moreau P, San-Miguel JF, Richardson PG. Panobinostat for the treatment of multiple myeloma. Clin Cancer Res. 2015;21:4767–73.CrossRef Laubach JP, Moreau P, San-Miguel JF, Richardson PG. Panobinostat for the treatment of multiple myeloma. Clin Cancer Res. 2015;21:4767–73.CrossRef
24.
go back to reference Sun P, Xia S, Lal B, et al. Lipid metabolism enzyme ACSVL3 supports glioblastoma stem cell maintenance and tumorigenicity. BMC Cancer. 2014;14:401.CrossRef Sun P, Xia S, Lal B, et al. Lipid metabolism enzyme ACSVL3 supports glioblastoma stem cell maintenance and tumorigenicity. BMC Cancer. 2014;14:401.CrossRef
25.
go back to reference Goodwin CR, Lal B, Ho S, et al. PTEN reconstitution alters glioma responses to c-met pathway inhibition. Anti-Cancer Drugs. 2011;22:905–12.CrossRef Goodwin CR, Lal B, Ho S, et al. PTEN reconstitution alters glioma responses to c-met pathway inhibition. Anti-Cancer Drugs. 2011;22:905–12.CrossRef
26.
go back to reference Woodard CL, Goodwin CR, Wan J, et al. Profiling the dynamics of a human phosphorylome reveals new components in HGF/c-met signaling. PLoS One. 2013;8:e72671.CrossRef Woodard CL, Goodwin CR, Wan J, et al. Profiling the dynamics of a human phosphorylome reveals new components in HGF/c-met signaling. PLoS One. 2013;8:e72671.CrossRef
27.
go back to reference Wan J, Su Y, Song Q, et al. Methylated cis-regulatory elements mediate KLF4-dependent gene transactivation and cell migration. Elife. 2017:6. Wan J, Su Y, Song Q, et al. Methylated cis-regulatory elements mediate KLF4-dependent gene transactivation and cell migration. Elife. 2017:6.
28.
go back to reference Menendez JA, Vellon L, Colomer R, Lupu R. Pharmacological and small interference RNA-mediated inhibition of breast cancer-associated fatty acid synthase (oncogenic antigen-519) synergistically enhances Taxol (paclitaxel)-induced cytotoxicity. Int J Cancer. 2005;115:19–35.CrossRef Menendez JA, Vellon L, Colomer R, Lupu R. Pharmacological and small interference RNA-mediated inhibition of breast cancer-associated fatty acid synthase (oncogenic antigen-519) synergistically enhances Taxol (paclitaxel)-induced cytotoxicity. Int J Cancer. 2005;115:19–35.CrossRef
29.
go back to reference Oyinlade O, Wei S, Lal B, et al. Targeting UDP-alpha-D-glucose 6-dehydrogenase inhibits glioblastoma growth and migration. Oncogene. 2018;37:2615–29.CrossRef Oyinlade O, Wei S, Lal B, et al. Targeting UDP-alpha-D-glucose 6-dehydrogenase inhibits glioblastoma growth and migration. Oncogene. 2018;37:2615–29.CrossRef
30.
go back to reference Wei S, Wang J, Oyinlade O, et al. Heterozygous IDH1 (R132H/WT) created by “single base editing” inhibits human astroglial cell growth by downregulating YAP. Oncogene. 2018. Wei S, Wang J, Oyinlade O, et al. Heterozygous IDH1 (R132H/WT) created by “single base editing” inhibits human astroglial cell growth by downregulating YAP. Oncogene. 2018.
31.
go back to reference Li Y, Fan X, Goodwin CR, Laterra J, Xia S. Hepatocyte growth factor enhances death receptor-induced apoptosis by up-regulating DR5. BMC Cancer. 2008;8:325.CrossRef Li Y, Fan X, Goodwin CR, Laterra J, Xia S. Hepatocyte growth factor enhances death receptor-induced apoptosis by up-regulating DR5. BMC Cancer. 2008;8:325.CrossRef
32.
go back to reference Xia S, Laterra J. Hepatocyte growth factor increases mitochondrial mass in glioblastoma cells. Biochem Biophys Res Commun. 2006;345:1358–64.CrossRef Xia S, Laterra J. Hepatocyte growth factor increases mitochondrial mass in glioblastoma cells. Biochem Biophys Res Commun. 2006;345:1358–64.CrossRef
33.
go back to reference Tilghman J, Schiapparelli P, Lal B, et al. Regulation of glioblastoma tumor-propagating cells by the integrin partner Tetraspanin CD151. Neoplasia. 2016;18:185–98.CrossRef Tilghman J, Schiapparelli P, Lal B, et al. Regulation of glioblastoma tumor-propagating cells by the integrin partner Tetraspanin CD151. Neoplasia. 2016;18:185–98.CrossRef
34.
go back to reference Wu Y, Richard JP, Wang SD, et al. Regulation of glioblastoma multiforme stem-like cells by inhibitor of DNA binding proteins and oligodendroglial lineage-associated transcription factors. Cancer Sci. 2012;103:1028–37.CrossRef Wu Y, Richard JP, Wang SD, et al. Regulation of glioblastoma multiforme stem-like cells by inhibitor of DNA binding proteins and oligodendroglial lineage-associated transcription factors. Cancer Sci. 2012;103:1028–37.CrossRef
35.
go back to reference Li Y, Rory Goodwin C, Sang Y, et al. Camptothecin and Fas receptor agonists synergistically induce medulloblastoma cell death: ROS-dependent mechanisms. Anti-Cancer Drugs. 2009;20:770–8.CrossRef Li Y, Rory Goodwin C, Sang Y, et al. Camptothecin and Fas receptor agonists synergistically induce medulloblastoma cell death: ROS-dependent mechanisms. Anti-Cancer Drugs. 2009;20:770–8.CrossRef
36.
go back to reference Pollard SM, Yoshikawa K, Clarke ID, et al. Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell. 2009;4:568–80.CrossRef Pollard SM, Yoshikawa K, Clarke ID, et al. Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell. 2009;4:568–80.CrossRef
37.
go back to reference Chatzispyrou IA, Held NM, Mouchiroud L, Auwerx J, Houtkooper RH. Tetracycline antibiotics impair mitochondrial function and its experimental use confounds research. Cancer Res. 2015;75:4446–9.CrossRef Chatzispyrou IA, Held NM, Mouchiroud L, Auwerx J, Houtkooper RH. Tetracycline antibiotics impair mitochondrial function and its experimental use confounds research. Cancer Res. 2015;75:4446–9.CrossRef
38.
go back to reference Xia S, Li Y, Rosen EM, Laterra J. Ribotoxic stress sensitizes glioblastoma cells to death receptor induced apoptosis: requirements for c-Jun NH2-terminal kinase and Bim. Mol Cancer Res. 2007;5:783–92.CrossRef Xia S, Li Y, Rosen EM, Laterra J. Ribotoxic stress sensitizes glioblastoma cells to death receptor induced apoptosis: requirements for c-Jun NH2-terminal kinase and Bim. Mol Cancer Res. 2007;5:783–92.CrossRef
39.
go back to reference Jin H, Liang L, Liu L, Deng W, Liu J. HDAC inhibitor DWP0016 activates p53 transcription and acetylation to inhibit cell growth in U251 glioblastoma cells. J Cell Biochem. 2013;114:1498–509.CrossRef Jin H, Liang L, Liu L, Deng W, Liu J. HDAC inhibitor DWP0016 activates p53 transcription and acetylation to inhibit cell growth in U251 glioblastoma cells. J Cell Biochem. 2013;114:1498–509.CrossRef
40.
go back to reference Castedo M, Perfettini JL, Roumier T, et al. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37.CrossRef Castedo M, Perfettini JL, Roumier T, et al. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37.CrossRef
41.
go back to reference Cornago M, Garcia-Alberich C, Blasco-Angulo N, et al. Histone deacetylase inhibitors promote glioma cell death by G2 checkpoint abrogation leading to mitotic catastrophe. Cell Death Dis. 2014;5:e1435.CrossRef Cornago M, Garcia-Alberich C, Blasco-Angulo N, et al. Histone deacetylase inhibitors promote glioma cell death by G2 checkpoint abrogation leading to mitotic catastrophe. Cell Death Dis. 2014;5:e1435.CrossRef
42.
go back to reference Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517:311–20.CrossRef Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517:311–20.CrossRef
43.
go back to reference Wu YT, Tan HL, Huang Q, et al. zVAD-induced necroptosis in L929 cells depends on autocrine production of TNFalpha mediated by the PKC-MAPKs-AP-1 pathway. Cell Death Differ. 2011;18:26–37.CrossRef Wu YT, Tan HL, Huang Q, et al. zVAD-induced necroptosis in L929 cells depends on autocrine production of TNFalpha mediated by the PKC-MAPKs-AP-1 pathway. Cell Death Differ. 2011;18:26–37.CrossRef
44.
go back to reference Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1:112–9.CrossRef Degterev A, Huang Z, Boyce M, et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 2005;1:112–9.CrossRef
45.
go back to reference Dong W, Zhang M, Zhu Y, et al. Protective effect of NSA on intestinal epithelial cells in a necroptosis model. Oncotarget. 2017;8:86726–35.PubMedPubMedCentral Dong W, Zhang M, Zhu Y, et al. Protective effect of NSA on intestinal epithelial cells in a necroptosis model. Oncotarget. 2017;8:86726–35.PubMedPubMedCentral
46.
go back to reference Silke J, Rickard JA, Gerlic M. The diverse role of RIP kinases in necroptosis and inflammation. Nat Immunol. 2015;16:689–97.CrossRef Silke J, Rickard JA, Gerlic M. The diverse role of RIP kinases in necroptosis and inflammation. Nat Immunol. 2015;16:689–97.CrossRef
Metadata
Title
Krüppel-like factor 9 and histone deacetylase inhibitors synergistically induce cell death in glioblastoma stem-like cells
Authors
Brian Tung
Ding Ma
Shuyan Wang
Olutobi Oyinlade
John Laterra
Mingyao Ying
Sheng-Qing Lv
Shuang Wei
Shuli Xia
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4874-8

Other articles of this Issue 1/2018

BMC Cancer 1/2018 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