Elsevier

Biochemical Pharmacology

Volume 81, Issue 3, 1 February 2011, Pages 412-424
Biochemical Pharmacology

NFκB inhibitors induce cell death in glioblastomas

https://doi.org/10.1016/j.bcp.2010.10.014Get rights and content

Abstract

Identification of novel target pathways in glioblastoma (GBM) remains critical due to poor prognosis, inefficient therapies and recurrence associated with these tumors. In this work, we evaluated the role of nuclear-factor-kappa-B (NFκB) in the growth of GBM cells, and the potential of NFκB inhibitors as antiglioma agents. NFκB pathway was found overstimulated in GBM cell lines and in tumor specimens compared to normal astrocytes and healthy brain tissues, respectively. Treatment of a panel of established GBM cell lines (U138MG, U87, U373 and C6) with pharmacological NFκB inhibitors (BAY117082, parthenolide, MG132, curcumin and arsenic trioxide) and NFκB-p65 siRNA markedly decreased the viability of GBMs as compared to inhibitors of other signaling pathways such as MAPKs (ERK, JNK and p38), PKC, EGFR and PI3K/Akt. In addition, NFκB inhibitors presented a low toxicity to normal astrocytes, indicating selectivity to cancerous cells. In GBMs, mitochondrial dysfunction (membrane depolarization, bcl-xL downregulation and cytochrome c release) and arrest in the G2/M phase were observed at the early steps of NFκB inhibitors treatment. These events preceded sub-G1 detection, apoptotic body formation and caspase-3 activation. Also, NFκB was found overstimulated in cisplatin-resistant C6 cells, and treatment of GBMs with NFκB inhibitors overcame cisplatin resistance besides potentiating the effects of the chemotherapeutics, cisplatin and doxorubicin. These findings support NFκB as a potential target to cell death induction in GBMs, and that the NFκB inhibitors may be considered for in vivo testing on animal models and possibly on GBM therapy.

Introduction

Glioblastoma (GBM) is an aggressive, invasive, and difficult to treat primary brain tumor. Standard therapy includes surgical resection, external beam radiation and chemotherapy, with no known curative therapy [1]. A number of dysregulated signaling cascades have been described in GBMs, including the MEK/ERK pathway, PLC/PKC pathway, and the PI3K/Akt pathway. Dysregulation of these pathways is driven by mutation, amplification, or overexpression of multiple genes such as PTEN, EGFR, PDGFR-a, p53, and mTOR [2], [3], [4]. Understanding these dysregulated pathways has provideded the basis for designing molecular targeted therapies as monoclonal antibodies against EGF, VEGF and PDGF receptors, as well as new combination therapies and drug delivery systems [4], [5], [6]. Despite these new treatment strategies, median survival has remained approximately 1 year for decades [1]. Thus, it is urgent to determine novel molecular targets in GBMs in order to develop more effective therapies and new therapeutic opportunities to patients.

Studies have compelling evidence that the transcription factor NFκB (Nuclear factor κB) plays a role in the control of oncogenesis, tumor progression and chemotherapy resistance of diverse types of malignances as lymphoma, leukemia, breast and ovarian cancers [7], [8], [9], [10], [11]. NFκB is formed by homo or heterodimers comprising members of the Rel family of proteins (p50/p105, p52/p100, p65, c-Rel and RelB) which form, upon non-stimulated conditions, a ternary and inactive cytoplasmic complex by interacting with inhibitory proteins of the IκB family. Upon stimulation by cytokines (TNF-alpha and IL1-beta), growth factors (EGF and PDGF), anticancer drugs (cisplatin, doxorubicin and vincristine) and other stressor stimuli, IκBs are phosphorylated by IKK (IκB kinase) proteins thus releasing the active NFκB, which translocates into nucleus and binds to DNA sequences in gene promoters. NFκB binding to DNA modulates the expression of a wide range of genes as that involved in inflammation (IL1-beta, IL-6, COX2, and TNF), apoptosis resistance (bcl-xL, cIAP1/2, XIAP, FLICE and survivin), cell invasion (ICAM-1, VCAM-1, MMP2, MMP9), angiogenesis (VEGF), proliferation (cyclin D1, MYC) and metastasis (CXCR4 and TWIST) [7], [8], [9], [10], [11], [12]. These groups of genes are directly related to tumor-associated phenomena suggesting NFκB as a potential target to cell death induction and chemosensitization in cancer [9], [13], [14], [15].

Aberrant NFκB activity has been described in some types of cancer cells upon basal and following anticancer drugs treatments. Besides, constitutive NFκB up-regulation has been found in chemotherapy-resistant cell lines, which correlates with therapy failure [9], [13], [15], [16], [17]. In this context, studies have suggested that molecules with NFκB inhibitory properties are potential anticancer agents [9], [15], [17], [18]. In this intent, researchers have blocking NFκB activity via IKK/NFκB inhibitors (as BAY117082, BAY117085, parthenolide, curcumin, arsenic trioxide and PDTC) or proteasome/NFκB inhibitors (PS-341 and MG132) in order to inhibit the growth of myeloma [10], leukemia [17], [19], esophageal [14] and breast cancer cells [20]. Recently, analysis of brain tumor biopsies identified that NFκB and its target genes are overexpressed in GBM and astrocytoma tumors compared to normal brain tissues [21], [22]. In addition, a positive correlation between NFκB activation and poor GBM prognosis was reported, suggesting that the known role of NFκB as a survival factor in other cancers could also be considered in GBMs [23]. Taken into account the aforementioned, this study was undertaken in order to determine the role of NFκB in GBM growth, and the selectivity, apoptotic potential and chemosensitizing activity of the NFκB inhibitors BAY117082, parthenolide, curcumin, arsenic trioxide, MG132, and p65 small-interfering RNA in GBM cell lines.

Section snippets

Reagents and antibodies

BAY117082 ((E)3-[(4-methylphenyl)-sulfonyl]-2-propenenitrile); MG132 (Z-Leu-Leu-Leu-CHO), curcumin, arsenic trioxide, propidium iodide, Hepes, CHAPS, dithiothreitol, EDTA, trypsin, MTT (3-(4,5-dimethyl)-2,5-diphenyl tetrazolium bromide), Nonidet-P40, spermin tetrahydrochloride, RNAse A and culture analytical grade reagents were purchased from Sigma Chemical Co. (St. Louis, MO, USA). Anti-NFκB p65 rabbit polyclonal antibody and anti-Lamin B were from Santa Cruz Biotechnologies; anti-β-actin was

NFκB as potential target to cell death induction in GBMs

Initially, we treated U138MG and C6 cells with inhibitors of some signaling pathways which are described as dysregulated in GBMs and other cancers [2], [3], [4]. Inhibitors of PI3K/Akt (LY294002, wortmannin), EGFR (PD158780), MEK/ERK1/2 (UO126), JNK1/2 (SP600129), p38 MAPK (SB203580), PKC (Gö6983), proteasome/NFκB (MG132) and IKK/NFκB (BAY117082) were tested at different concentrations (1–60 μM) based on the literature range and manufacturer instructions, which describe IC50 values < 20 μM for all

Discussion

GBMs are associated with a poor prognosis due to intrinsic malignance and drug/radiation resistance, besides limited therapeutic opportunities such as neurosurgery and temozolomide/radiotherapy regimens [1], [15], [25]. Studies reported that NFκB activation correlates with therapeutic implications and worse prognosis in human gliomas [21], [22], [23], [37], [38], [39]. Aberrant NFκB activity was found critical for focal necrosis formation, invasive phenotype establishment [22] and resistance to

Disclosure statement

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

Acknowledgements

We acknowledge the Brazilian funds CAPES, FINEP/IBNNet (01060842-00) and CNPq for financial support, and Dr Rafael Roesler (Hospital de Clinicas de Porto Alegre, HCPA) for kindly providing U87 and U373 cell lines.

References (70)

  • K.N. Anderson et al.

    Parthenolide induces apoptosis in glioblastomas without affecting NF-kappaB

    J Pharmacol Sci

    (2008)
  • L.A. Stewart

    Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials

    Lancet

    (2002)
  • C. Yu et al.

    Mitochondrial Bax translocation partially mediates synergistic cytotoxicity between histone deacetylase inhibitors and proteasome inhibitors in glioma cells

    Neuro Oncol

    (2008)
  • C. Brennan et al.

    Glioblastoma subclasses can be defined by activity among signal transduction pathways and associated genomic alterations

    PLoS ONE

    (2009)
  • E.R. Gerstner et al.

    Anti-vascular endothelial growth factor therapy for malignant glioma

    Curr Neurol Neurosci Rep

    (2009)
  • R.W. Mercer et al.

    Targeted therapies for malignant glioma: progress and potential

    BioDrugs

    (2009)
  • M. Barkett et al.

    Control of apoptosis by Rel/NFkB transcription factors

    Oncogene

    (1999)
  • B. Rayet et al.

    Aberrant rel/NFκB genes and activity in human cancer

    Oncogene

    (1999)
  • C. Nakanishi et al.

    Nuclear factor-kappaB inhibitors as sensitizers to anticancer drugs

    Nat Rev Cancer

    (2005)
  • V. Baud et al.

    Is NF-kB a good target for cancer therapy? Hopes and pitfalls

    Nat Rev Drug Discov

    (2009)
  • T.X. Xie et al.

    Constitutive NF-kappaB activity regulates the expression of VEGF and IL-8 and tumor angiogenesis of human glioblastoma

    Oncol Rep

    (2010)
  • B. Li et al.

    Targeting NF-kappaB signaling pathway suppresses tumor growth, angiogenesis, and metastasis of human esophageal cancer

    Mol Cancer Ther

    (2009)
  • K.M. Dhandapani et al.

    Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFkappaB transcription factors

    J. Neurochem

    (2007)
  • T.X. Xie et al.

    NF-kappaB activity is critical in focal necrosis formation of human glioblastoma by regulation of the expression of tissue factor

    Int J Oncol

    (2008)
  • A. Zanotto-Filho et al.

    The pharmacological NFkappaB inhibitors BAY117082 and MG132 induce cell arrest and apoptosis in leukemia cells through ROS–mitochondria pathway activation

    Cancer Lett

    (2010)
  • C. Kasuga et al.

    Sensitization of human glioblastomas to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) by NF-kappaB inhibitors

    Cancer Sci

    (2004)
  • B.M. Pickering et al.

    Pharmacological inhibitors of NF-kB accelerate apoptosis in chronic lymphocytic leukemia cells

    Oncogene

    (2007)
  • J. Domingo-Domènech et al.

    Inactivation of NF-kappaB by proteasome inhibition contributes to increased apoptosis induced by histone deacetylase inhibitors in human breast cancer cells

    Breast Cancer Res Treat

    (2008)
  • P.A. Robe et al.

    In vitro and in vivo activity of the nuclear factor-kappaB inhibitor sulfasalazine in human glioblastomas

    Clin Cancer Res

    (2004)
  • B. Raychaudhuri et al.

    Aberrant constitutive activation of nuclear factor kappaB in glioblastoma multiforme drives invasive phenotype

    J Neurooncol

    (2007)
  • R.E. Brown et al.

    Morphoproteomic demonstration of constitutive nuclear factor-kappaB activation in glioblastoma multiforme with genomic correlates and therapeutic implications

    Ann Clin Lab Sci

    (2006)
  • M.L. da Frota et al.

    Brazilian marine sponge Polymastia janeirensis induces apoptotic cell death in human U138MG glioma cell line, but not in a normal cell culture

    Invest New Drugs

    (2009)
  • E. Braganhol et al.

    Antiproliferative effect of quercetin in the human U138MG glioma cell line

    Anticancer Drugs

    (2006)
  • Y. Hirose et al.

    p53 affects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells

    Cancer Res

    (2001)
  • J. Kucharczak et al.

    To be, or not to be: NF-kappaB is the answer: role of Rel/NF-kappaB in the regulation of apoptosis

    Oncogene

    (2003)
  • Cited by (0)

    View full text