Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Neuro Oncol. 2015 Apr; 17(4): 555–565.
Published online 2014 Oct 10. doi: 10.1093/neuonc/nou282
PMCID: PMC4483074
PMID: 25304134

Activation of the NRF2 pathway and its impact on the prognosis of anaplastic glioma patients

Associated Data

Supplementary Materials

Abstract

Background

Nuclear factor erythroid 2–related factor 2 (NRF2) plays pivotal roles in cytoprotection. We aimed at clarifying the contribution of the NRF2 pathway to malignant glioma pathology.

Methods

NRF2 target gene expression and its association with prognosis were examined in 95 anaplastic gliomas with or without isocitrate dehydrogenase (IDH) 1/2 gene mutations and 52 glioblastomas. To explore mechanisms for the altered activity of the NRF2 pathway, we examined somatic mutations and expressions of the NRF2 gene and those encoding NRF2 regulators, Kelch-like ECH-associated protein 1 (KEAP1) and p62/SQSTSM. To clarify the functional interaction between IDH1 mutations and the NRF2 pathway, we introduced a mutant IDH1 to T98 glioblastoma-derived cells and examined the NRF2 activity in these cells.

Results

NRF2 target genes were elevated in 13.7% and 32.7% of anaplastic gliomas and glioblastomas, respectively. Upregulation of NRF2 target genes correlated with poor prognosis in anaplastic gliomas but not in glioblastomas. Neither somatic mutations of NRF2/KEAP1 nor dysregulated expression of KEAP1/p62 explained the increased expression of NRF2 target genes. In most cases of anaplastic glioma with mutated IDH1/2, NRF2 and its target genes were downregulated. This was reproducible in IDH1 R132H–expressing T98 cells. In minor cases of IDH1/2-mutant anaplastic gliomas with increased expression of NRF2 target genes, the clinical outcomes were significantly poor.

Conclusions

The NRF2 activity is increased in a significant proportion of malignant gliomas in general but decreased in the majority of IDH1/2-mutant anaplastic gliomas. It is plausible that the NRF2 pathway plays an important role in tumor progression of anaplastic gliomas with IDH1/2 mutations.

Keywords: IDH1/2 gene, Keap1-NRF2 system, malignant glioma, prognosis

Anaplastic gliomas and glioblastomas are aggressive brain tumors with median survival times ranging from 72.1 to 82.6 months in anaplastic gliomas and 14.6 months in glioblastomas.1,2 One of the main causes of the poor prognosis is chemo- and radioresistance. Thus, elucidation of mechanisms underlying this resistance is urgent to improve the treatment outcomes of malignant gliomas.

The system involving Kelch-like ECH (erythroid cell-derived protein with cap'n'collar homology)-associated protein 1 (KEAP1) and nuclear factor erythroid 2–related factor 2 (NRF2) plays pivotal roles in protecting normal and neoplastic cells from oxidative and electrophilic insults by activating cytoprotective genes. These gene products are involved in glutathione synthesis (eg, glutamate cysteine ligase of catalytic [GCLC] and of modifier subunit [GCLM] genes), reactive oxygen species (ROS) elimination, xenobiotic metabolism (eg, NAD(P)H:quinone oxidoreductase gene [NQO1]), and crossmembrane transport, often resulting in multidrug resistance.3 Under normal conditions, KEAP1 ubiquitinates NRF2, and NRF2 is degraded by the proteosome. In the presence of electrophiles or ROS, KEAP1 is inactivated, and NRF2 is stabilized, binds to antioxidant response elements (AREs), and induces the cytoprotective genes. Recently, constitutive activation of NRF2 was found in various cancers. Multiple causes of the constitutive activation of NRF2 have been described, including somatic mutations of KEAP1 or NRF2, reduced expression of KEAP1, and accumulation of p62.4 Increased NRF2 activity confers cell growth advantage and resistance to chemo- and radiotherapy on lung, prostate, gallbladder, and ovarian cancers59 and leads to poor prognosis in lung, esophagus, gallbladder, and breast cancers.1014 Although similar impacts of NRF2 activation on glioblastoma cell lines and a small cohort of glioblastoma patients have been reported,15,16 the prognostic significance of NRF2 activation in malignant gliomas remains to be elucidated.

Somatic mutations in the isocitrate dehydrogenase 1 (IDH1) gene or IDH2 gene have been identified in adult gliomas.1719 These mutations occur at a very early stage of gliomatogenesis and confer genetic and prognostic differences on anaplastic gliomas and glioblastomas.1719 Malignant gliomas with IDH1/2 mutations had better prognoses than those with wild-type IDH1/2.1719 Although the hypermethylated phenotype is frequently found in World Health Organization grades II and III gliomas with IDH1/2 mutations, underlying mechanisms for better prognosis remain unclear. Recently, IDH1 mutations have been shown to sensitize glioma cells to bis-chloroethylnitrosourea (BCNU)–induced oxidative stress by decreasing the amount of the reduced form of glutathione.20 Because the synthesis and reduction of glutathione is often heavily dependent on NRF2 activity,4 we hypothesized that the NRF2 pathway was suppressed in the presence of mutated IDH1, leading to the better prognosis of malignant gliomas with IDH1/2 mutations.

To test this hypothesis, we characterized the prevalence, possible causes, and prognostic value of the NRF2 activation in anaplastic gliomas with mutated or wild-type IDH1/2 and glioblastomas. We also evaluated the association between IDH1/2 mutations and NRF2 activity by analyzing the expression of NRF2 and its target genes in surgical specimens, and confirmed the effect of IDH1 mutation on NRF2 activity, using IDH1 mutant-expressing cells.

Materials and Methods

Patients

From January 1995 to October 2010, 117 patients with histologically verified anaplastic gliomas and 216 with glioblastomas were treated at the Department of Neurosurgery, Tohoku University Hospital. This study was conducted with the approval of the ethics committee of Tohoku University School of Medicine, and written informed consent was obtained from all patients.

Reverse Transcription and Quantitative Real-time PCR

Quantitative PCR analysis was performed on an ABI7300 system. Primers and probes used for amplification of cDNAs are described in Supplementary Table S1.

Sequencing of IDH1, IDH2, NRF2, and KEAP1 Genes

Genomic DNAs from paired peripheral blood and primary tumor tissues were extracted from snap-frozen samples. Sequencing primers are shown in Supplementary Table S1.

Immunohistochemical Analysis

Tumor samples were fixed in 10% buffered formalin and embedded in paraffin. A guinea pig polyclonal antibody against p62 (Progen GP62, 1 : 3000) was used.

Microarray Analysis

Total RNA was extracted from 12 randomly selected anaplastic glioma tissues. The data were deposited to the Gene Expression Omnibus (accession no. GSE52942).

Quantitative High-resolution DNA Methylation Analysis

DNA methylation was examined using the MassARRAY technique (Sequenom). Primers used in this study are listed in Supplementary Table S1.

Establishment of T98 Cells Expressing IDH1 or IDH1 R132H

An expression vector for FLAG-tagged IDH1 or IDH1 R132H21 was introduced into T98 cells. Stable transformants were selected with 40 μg/mL blasticidin S.

Immunoblot Analysis

Whole cell extracts were prepared from frozen tumor samples. Nuclear extracts and whole-cell extracts were prepared from T98-derived cell lines. Band intensities were quantified using ImageJ64 (ImageJ 1.45s).

Chromatin Immunoprecipitation Assay

Anti-NRF2 antibody (Santa Cruz Biotechnology, sc-13032) was used for ChIP assay. Primers used in the ChIP assay are described in Supplementary Table S1.

NRF2 Knockdown Experiment

NRF2 small interfering (si)RNAs were purchased from Invitrogen and introduced into T98 cells using MP-100 MicroPorator (Digital Bio Technology).

Cell Viability Assay

Cells were challenged with BCNU (LKT Laboratories), and their viability was examined after 48 h using the Cell Counting Kit 8 (Dojindo Molecular Technologies).

Metabolite Measurements and Metabolomic Profiling of Cultured Cells

To measure the levels of intracellular metabolites, extracts were prepared from 2–6 × 106 cells per sample and analyzed on a capillary electrophoresis–connected electrospray ionization–time-of-flight mass spectrometry system as previously described.22

Statistical Analysis

Student's t-test, chi-square test, and log-rank test were utilized. The Cox model was utilized for multivariate analyses. All statistical methods adopted a significance level of P < .05. Details of the methods can be found in the Supplementary Material online.

Results

NQO1 and GCLM Expression in Anaplastic Gliomas and Glioblastomas

To determine the prevalence of tumors with high NRF2 activity, we examined expressions of NRF2 target genes in anaplastic gliomas and randomly selected glioblastomas. Because the NRF2 activity is regulated at multiple levels, target gene expressions provide the most precise parameter for evaluating the overall activity of NRF2. Among the 117 anaplastic gliomas, 22 were excluded from this analysis because the tumor samples were obtained after radiation therapy and/or chemotherapy, which might have modified the NRF2 activity. Characteristics of the remaining 95 anaplastic gliomas are described in Table 1. Regarding the glioblastomas, 52 were randomly selected from patients who underwent surgical resection without any prior therapies (Supplementary Table S2).

Table 1.

Demographics of anaplastic glioma in this study

Cases for Mutation AnalysisCases for Expression and Prognosis Analysis
Number of cases11795
Age, y (median)10–81 (47)10–81 (47)
Sex
 M:F71:4654:41
Histological diagnosis
 AA5840
 AOA2825
 AO3130
Surgery
 Biopsy102
 Resection10893
Radiation therapy
 Yes11191
 none64
Chemotherapy
 ACNU8466
 TMZ99
 PAV86
 None1614
IDH1/2 gene
 Mutated7468
 Wild type4327

Abbreviations: AA, anaplastic astrocytoma; AOA: anaplastic oligoastrocytoma; AO, anaplastic oligodendroglioma; ACNU, nimustine hydrochloride; TMZ, temozolomide; PAV, combination chemotherapy of procarbazine, nimustine hydrochloride, and vincristine.

Expression levels of NQO1 and GCLM, typical NRF2 target genes, positively correlated with each other in both anaplastic gliomas (R = 0.556, P < .001) and glioblastomas (R = 0.716, P < .001) (Fig. 1A and B). Glioblastomas exhibited higher expression of NQO1 and GCLM than did anaplastic gliomas (Fig. 1C). Tumors that displayed increase in NQO1 or GCLM expression of more than 2-fold over that of the normal brain were regarded as high expressers of NRF2 target genes. The prevalence of high expressers among glioblastomas (17 of 52: 32.7%) was significantly higher than that among anaplastic gliomas (13 of 95: 13.7%: P = .0096). These results suggest that expressions of NRF2 target genes are upregulated in some anaplastic glioma and glioblastoma cases.

An external file that holds a picture, illustration, etc.
Object name is nou28201.jpg

Expression of NRF2 target genes NQO1 and GCLM in anaplastic gliomas and glioblastomas. (A and B) Scattergrams demonstrating the correlations between NQO1 and GCLM expression in (A) anaplastic gliomas and (B) glioblastomas. (C) NQO1 and GCLM expression in anaplastic gliomas and glioblastomas relative to the normal brain. Data represent means ± SD. Each sample was analyzed in triplicate. *P < .05. (D and E) Scattergrams demonstrating the correlations between NQO1 and KEAP1 expression in (D) anaplastic gliomas and (E) glioblastomas. (F–I) Kaplan–Meier analysis demonstrating (F and G) PFS and (H and I) OS rates (according to the NQO1 and GCLM expression levels in [F and H] anaplastic gliomas and [G and I] glioblastomas). P-values were calculated with the log-rank test.

Examination of Possible Causes for NRF2 Activation in Anaplastic Gliomas and Glioblastomas

To explore underlying mechanisms for the elevated NRF2 activity, we first investigated whether anaplastic gliomas and glioblastomas harbored mutations in the NRF2 and KEAP1 genes. Among the 117 anaplastic gliomas, somatic missense and heterozygous mutations of KEAP1 were identified in 2 cases (1.7%). The clinical features of these cases are shown in Supplementary Table S3. One case was anaplastic astrocytoma (AA) (1 out of 58 cases; 1.7%) diagnosed as Turcot syndrome, given the familial history and coexistence of colon cancer, and the other was diagnosed as sporadic anaplastic oligodendroglioma (AO) (1 out of 31 cases; 3.2%). NRF2 mutations were not found in the 117 anaplastic gliomas. Mutations in the NRF2 and KEAP1 genes were not found in the 79 randomly selected glioblastomas or 5 glioblastoma cell lines (A172, U87, U251, U373, and T98).

We next examined other possible causes for the NRF2 activation. One is the reduced expression of KEAP1, and the other is the accumulation of p62, both of which inhibit NRF2 degradation and increase the expression of NQO1/GCLM. However, there were no correlations between KEAP1 and NQO1/GCLM expression in the tumor tissues from 95 anaplastic glioma or 52 glioblastoma cases (NQO1 vs KEAP1: R = 0.154, P = .136 and R = 0.070, P = .622 in anaplastic glioma and glioblastoma, respectively; GCLM vs KEAP1: R = 0.070, P = .498 and R = 0.148, P = .294 in anaplastic glioma and glioblastoma, respectively) (Fig. 1D and E and data not shown). Accumulation of p62 was not observed in any of the 95 anaplastic glioma or 10 glioblastoma tissues (data not shown). These results indicated that high expression of NRF2 target genes was not caused by the downregulation of KEAP1 or p62 accumulation. Thus, currently well-known causes did not account for the elevation of NRF2 target gene expression that was observed in the present cohort of anaplastic glioma and glioblastoma cases.

Correlations Between NRF2 Target Gene Expression and Prognosis in Anaplastic Glioma and Glioblastoma Cases

We examined the correlations between NRF2 target gene expression and the progression-free survival (PFS) and overall survival (OS) rates in anaplastic glioma and glioblastoma cases (Fig. 1F–I). In a univariate analysis, high expression of NRF2 target genes was associated with a worse PFS and OS rate in anaplastic gliomas (Fig. 1F and H). In contrast, the expression level was not associated with prognosis in glioblastomas (Fig. 1G and I). These results indicated that elevated expression of NRF2 target genes correlates with poor prognosis in anaplastic gliomas but not in glioblastomas.

Association of IDH1/2 Mutation Status With NRF2 Target Gene Expression

Based on the clear association between NRF2 target gene expression and prognosis, we focused on anaplastic gliomas for further study. Somatic mutations in the IDH1 or IDH2 gene have been found in a substantial portion of anaplastic glioma cases, for which the prognosis is significantly better than for those without the mutations.20 We hypothesized that IDH1/2 mutation status correlates with expression levels of NRF2 target genes. To examine the NRF2 target gene expression in anaplastic gliomas with mutated IDH1/2, we conducted a microarray analysis to measure the expression levels of representative NRF2 target genes, including NQO1, HMOX1, GCLM, TXNRD1, and PRDX1, in 12 anaplastic gliomas with or without mutated IDH1/2 (Fig. 2A) as a pilot study. We found that the average relative expression levels of all 5 genes were lower in anaplastic gliomas with mutated IDH1/2 than in those with wild-type IDH1/2, and this difference was statistically significant for GCLM. To verify this observation that NRF2 target gene expression appeared to be reduced in anaplastic gliomas with mutated IDH1/2, we compared the expression levels of NQO1 and GCLM among all anaplastic glioma cases with and without mutated IDH1/2. The NQO1 and GCLM mRNA levels in IDH1/2-mutated tumors were significantly lower than those in IDH1/2 wild-type tumors (Fig. 2B), which was consistent with the protein abundance (Fig. 2C). When anaplastic gliomas were divided into 2 subgroups—AA and AO/anaplastic oligoastrocytoma (AOA)—an almost similar tendency was observed (Supplementary Fig. S1).

An external file that holds a picture, illustration, etc.
Object name is nou28202.jpg

Expression of NRF2 target genes in anaplastic gliomas with and without mutated IDH1/2. (A) Heat map showing the expression of the 5 NRF2 target genes in anaplastic gliomas with (cases 1–6) and without (cases 7–12) mutated IDH1/2. A sample with a median value was defined as the control. The gray scale indicates the level of fold change in gene expression relative to the control value. The darkest and the brightest colors indicate a 4-fold increase and decrease, respectively. (B) Expression of NQO1 and GCLM in anaplastic gliomas with and without IDH1/2 mutations and glioblastomas relative to the normal brain. Data represent means ± SD. Each sample was analyzed in triplicate. *P < .05. (C) Protein abundance of NQO1 and GCLM in anaplastic gliomas with and without IDH1/2 mutations. Band intensities of NQO1 and GCLM were quantified and normalized with those of alpha-tubulin. Values of case 1 were set as one. (D–F) Profiles of CpG methylation ratio in the promoter regions of (D) MGMT, (E) NQO1, and (F) GCLM in anaplastic gliomas with mutated (n = 64) and wild-type IDH1/2 (n = 24). Data are expressed as the averages and SDs of the methylation ratios of each CpG unit. *P < .05. (G) Scattergrams demonstrating the correlations between NQO1 and NRF2 expression and between GCLM and NRF2 expression in anaplastic gliomas. (H) Expression of NRF2 in anaplastic gliomas with mutated and wild-type IDH1/2 genes relative to the normal brain. Data represent means ± SD. Each sample was analyzed in triplicate. *P < .05.

In contrast, there was no difference in NQO1 and GCLM expression between anaplastic gliomas with wild-type IDH1/2 and glioblastomas (Fig. 2B). The prevalence of high expressers of NRF2 target genes was significantly lower in anaplastic gliomas with mutated IDH1/2 than in those with wild-type IDH1/2 (5.8% vs 33%, P = .0012) and in glioblastomas (5.8% vs 33%, P = .0018). These results implied that NRF2 activity was lower in anaplastic gliomas with mutated IDH1/2 than in other malignant gliomas.

Examination of Possible Causes for the Reduced Expression of NRF2 Target Genes in Anaplastic Gliomas With Mutated IDH1/2

Given that hypermethylation phenotypes have been reported for malignant gliomas with IDH1 mutations,23 we suspected that promoter methylation might reduce NRF2 target gene expression. To test this hypothesis, we exploited MassARRAY technology and accessed the methylation statuses of the cytosine–phosphate–guanine (CpG) islands residing in the promoter regions of NQO1 and GCLM. We also examined the O6-DNA methylguanine-methyltransferase gene (MGMT) as a positive control, for which the promoter CpGs are heavily methylated in IDH1-mutant gliomas.24 Methylation ratios that spanned 16–18 CpG sites in the promoter region of each gene were quantified. We examined anaplastic gliomas with (64 cases) and without (24 cases) IDH1/2 mutations. The MGMT promoter was highly methylated in both groups, and, in particular, anaplastic gliomas with mutated IDH1/2 exhibited significantly higher methylation ratios in 7 of the 16 CpG sites, compared with those with wild-type IDH1/2 (Fig. 2D). In contrast, the methylation ratios in the promoter regions of NQO1 and GCLM were relatively low and mostly comparable, irrespective of the IDH1/2 status, except for a single CpG in the GCLM promoter (Fig. 2E and F). Thus, promoter methylation does not seem to account for the reduced expression of NRF2 target genes observed in the presence of IDH1/2 mutations.

We next examined the expression levels of KEAP1 and NRF2. There were no differences in KEAP1 expression between anaplastic gliomas with mutated IDH1/2 and those with wild-type IDH1/2 (data not shown). In contrast, the expression level of NRF2 positively correlated with those of NQO1 (R = 0.391, P < .001) and GCLM (R = 0.266, P = .02) in anaplastic gliomas (Fig. 2G). NRF2 expression was significantly lower in anaplastic gliomas with mutated IDH1/2 than in those with wild-type IDH1/2 (Fig. 2H). These results suggest that the reduced expression of NRF2 at the transcription level is one of the causes for the downregulation of NRF2 target genes in anaplastic gliomas with mutated IDH1/2. In subtypes of anaplastic gliomas, correlation between the decreased NRF2 expression and IDH1/2 mutation was clear in AO/AOA but not in AA (Supplementary Fig. S1). Of note, there was no significant association between the NRF2 mRNA level and the prognosis of anaplastic gliomas, probably due to the multilayered regulation of the NRF2 activity at transcription level and posttranscription level (Supplementary Fig. S2A and B).

An Exogenous IDH1 Mutant Molecule Suppresses NRF2 Activity in T98 Glioblastoma Cells

The clinical analysis revealed a correlation between IDH1/2 mutation and downregulation of NRF2 target genes. To examine whether the IDH1/2 mutation suppressed the NRF2 pathway, either exogenous FLAG-tagged IDH1 or IDH1 R132H, which is the most frequently encountered IDH1 mutant, was introduced into T98 glioblastoma cells, and stable transformants were obtained. Introduction of IDH1 R132H into T98 cells significantly slowed down the proliferation rate (data not shown). We first verified the similar expression levels of exogenous IDH1 and IDH1 R132H in an immunoblot analysis, using anti-FLAG antibody (Fig. 3A). Next, the expression levels of NRF2 target genes were quantified. Three major NRF2 target genes, NQO1, GCLC, and GCLM, were downregulated in IDH1 R132H–expressing cells (Fig. 3B), suggesting that the IDH1 mutation limits the NRF2 activity.

An external file that holds a picture, illustration, etc.
Object name is nou28203.jpg

Effect of IDH1 R132H expression on NRF2 activity in T98 glioblastoma cells. (A) Expression levels of exogenously introduced IDH1 and IDH1 R132H. Exogenous IDH1 and IDH1 R132H were detected with anti-FLAG antibody. Lamin B was used as a loading control. (B and C) Relative mRNA levels of (B) NQO1, GCLC, and GCLM and (C) NRF2. Data represent the means ± SD of 5 independent experiments, and each sample was analyzed in triplicate. The expression levels in IDH1-expressing cells were set as one. **P < .005, ***P < .001. (D) NRF2 protein abundance in nuclear extracts. Lamin B was detected as a loading control. Band intensities of NRF2 were quantified and normalized with those of Lamin B. A value of IDH1-expressing cells was set as one. (E) Location and sequence of AREs in NQO1, GCLC, and GCLM gene loci. (F) ChIP assay with anti-NRF2 antibody in IDH1- and IDH1 R132H–expressing cells. In the control samples, rabbit IgG was added instead of anti-NRF2 antibody. NQO1 promoter, GCLC enhancer, and GCLM promoter, which contain AREs, and NQO1 exon 2, which does not contain AREs, were examined. Data represent the means ± SD of triplicate samples. A representative result of 3 independent experiments is shown. (G) Metabolomic profiling. Data represent the means ± SD of 3 independent samples. The expression levels in IDH1-expressing cells were set as one. P-values were calculated with Student's t-test. *P < .05, **P < .005, ***P < .001. Promoted and inhibited processes in IDH1 R132H–expressing cells are indicated with red and blue arrows, respectively. Abbreviations of the metabolites are described in the Supplementary information.

We next examined NRF2 expression at mRNA and protein levels. NRF2 mRNA level was decreased in the IDH1 R132H–expressing cells (Fig. 3C), which is consistent with the results obtained in the clinical study. Almost parallel to the mRNA reduction, the nuclear accumulation of NRF2 protein was also decreased in the presence of IDH1 R132H (Fig. 3D and Supplementary Fig. S3). To evaluate the DNA binding activity of NRF2, we conducted a ChIP assay with anti-NRF2 antibody. We examined the recruitment of NRF2 to AREs located in the NQO1 promoter, GCLC enhancer, and GCLM promoter (Fig. 3E). The second exon of NQO1 was used as a negative control locus without AREs. NRF2 was clearly recruited to the NQO1 promoter,25 GCLC enhancer,26 and GCLM promoter27 in control cells expressing wild-type IDH1. In the presence of IDH1 R132H, however, the NRF2 recruitment to the NQO1 promoter was reduced, and no significant recruitment was observed to the GCLC enhancer or GCLM promoter (Fig. 3F). Thus, IDH1 mutation reduces the protein abundance and DNA binding of NRF2 in T98 cells. As the latter is more dramatic than the former, we surmise that the mutant IDH1 might affect, for unknown reasons, the DNA binding ability of NRF2.

We further conducted metabolomic profiling of T98 cells expressing IDH1 or IDH1 R132H to examine whether any NRF2-deficient signatures were detected in the latter cells (Fig. 3G). The elevation of 2-hydroxyglutarate (2-HG) (Fig. 3G, red box) verified the expected enzymatic activity of IDH1 R132H.28 The clearest difference was observed in the glutathione synthesis pathway. In the presence of IDH1 R132H, glutathione and γ-glutamylcysteine were decreased (Fig. 3G, blue boxes), whereas glutamine and glutamate were increased. Because γ-glutamylcysteine is the first-step product in the glutathione synthesis catalyzed by γ-glutamylcysteine ligase (GCL), these results indicate that GCL activity was inhibited in IDH1 R132H–expressing cells and are consistent with the reduced expression of GCLC and GCLM, which encode the 2 subunits of GCL. Thus, the metabolomic profile of the IDH1 R132H–expressing cells exhibited the signature of NRF2 deficiency and supported our hypothesis that the IDH1 mutation suppressed glutathione synthesis of the NRF2 activities.

Prognostic Impact of NRF2 Target Gene Expression Under the Influence of IDH1/2 Mutation Status

Based on previous studies using glioma cell lines,15,16 reduced NRF2 activity was strongly expected to improve the clinical outcome. Consistent with these reports, IDH1 R132H–expressing cells, where NRF2 activity is low, were more susceptible to the chemotherapeutic reagent BCNU than control cells (Fig. 4A). Importantly, NRF2 knockdown in T98 cells with wild-type IDH1 (Supplementary Fig. S4) was sufficient to drive the cells toward the vulnerable phenotype (Fig. 4B). Thus, we consider that the reduced NRF2 activity, at least in part, sensitizes IDH1-mutant tumors to anticancer therapy resulting in the better prognosis.

An external file that holds a picture, illustration, etc.
Object name is nou28204.jpg

Impact of NRF2 activity on drug sensitivity and prognosis under the influence of IDH1/2 mutation status. (A and B) BCNU sensitivity of T98 cells expressing (A) IDH1 or IDH1 R132H and (B) T98 cells with NRF2 knockdown. Cells were treated with increasing concentration of BCNU, and their viability was examined at 48 h after the addition of BCNU. All samples were prepared in triplicate. Three independent experiments were conducted, and the means and SDs were calculated. *P < .05, ***P < .001 ([A] and control vs NRF2 siRNA-1 in [B]), ##P < .005, ###P < .001 (control vs NRF2 siRNA-2 in [B]). (C–F) Kaplan–Meier analyses demonstrating (C and D) the PFS and (E and F) OS rates according to NQO1/GCLM expression levels in anaplastic gliomas with (C and E) mutated and (D and F) wild-type IDH1/2 genes. P-values were calculated with the log-rank test.

To verify this notion, we analyzed the impact of NRF2 target gene expression on the prognosis of anaplastic glioma patients with mutated or wild-type IDH1/2. In anaplastic gliomas with mutated IDH1/2, the high expression levels of NQO1/GCLM were significantly associated with worse PFS (P = .0013; Fig. 4C) and OS (P = .0089; Fig. 4E), while NRF2 expression itself was not (Supplementary Fig. S2C and D). In contrast, NQO1/GCLM expression did not have any clear impact on the prognosis of anaplastic glioma patients with wild-type IDH1/2 (Fig. 4D and F), similar to the glioblastoma cases (see Fig. 1G and I). Thus, higher expression of NRF2 target genes correlated with poor prognosis in anaplastic glioma patients with mutated IDH1/2, but not in those with wild-type IDH1/2. These results indicate that the better prognosis seen in IDH1/2-mutant anaplastic gliomas is canceled by the increased expression of the NRF2 target genes, implying that NRF2 pathway inhibition is responsible for the better prognosis of IDH1/2-mutant anaplastic gliomas.

To further characterize the association between NRF2 target gene expression and clinical outcomes, a multivariate analysis was performed. We previously reported the prognostic values of the IDH1/2 and p53 mutation statuses, codeletion of chromosome 1p and 19q, histological diagnosis, MGMT promoter methylation status, and EGFR amplification in anaplastic glioma cases.20 When 95 anaplastic glioma cases from the present cohort were evaluated, the IDH1/2 mutation status and EGFR amplification were found to be independent prognostic factors for PFS, whereas age and EGFR amplification were independent prognostic factors for OS (Table 2). When the analysis was limited to anaplastic gliomas with mutated IDH1/2, high NQO1/GCLM expression alone was an independent prognostic factor for progression. These results implicate that upregulation of NRF2 target genes is a prognostic marker and that they play an important role in the progression of anaplastic glioma with mutated IDH1/2.

Table 2.

Multivariate analysis of independent factors associated with survival in all of anaplastic glioma and anaplastic glioma with IDH1/2 mutation

All of Anaplastic Glioma
Anaplastic Glioma With IDH1/2 Mutation
RecurrencePOSPRecurrencePOSP
Age1.5 (0.7–3.5).3022.5 (1.1–5.9).0302.6 (0.9–7.7).0874.6 (1.6–13.4).005
Wild-type IDH1/2 gene2.6 (1.1–6.0).0312.2 (0.9–5.5).077
Histological diagnosis1.3 (0.8–2.0).2501.2 (0.8–2.2).3651.7 (0.9–2.9).0561.3 (0.7–2.4).486
NQO1/GCLM high expression1.3 (0.5–3.6).5700.7 (0.2–2.5).6245.8 (1.3–24.5).0164.3 (0.6–29.1).138
Unmethylated MGMT promoter1.0 (0.5–2.2).9861.0 (0.4–2.3).9791.1 (0.2–5.6).9180.3 (0.03–3.3).354
Codeletion of chromosome 1p and 19q0.6 (0.2–1.8).3500.5 (0.1–1.5).1951.5 (0.2–10.4).7121.2 (0.2–6.2).869
p53 mutation1.7 (0.7–4.1).2201.4 (0.6–3.2).4614.2 (0.6–29.0).1392.8 (0.6–13.5).203
EGFR amplification2.4 (1.1–5.4).0353.9 (1.7–9.1).0012.4 (0.5–12.5).2977.6 (1.6–35.9).010

Hazard ratios (95% confidence interval) are shown in parentheses.

P-values <.05 were considered statistically significant and are shown in bold.

Discussion

We demonstrated that NRF2 target genes were upregulated in a significant proportion of malignant gliomas but downregulated in anaplastic gliomas with mutated IDH1/2. IDH1 R132H was found to suppress NRF2 activity in T98 cells. Notably, high expression of NRF2 target genes had prognostic value in anaplastic gliomas with mutated IDH1/2. This study has provided a new insight into the contribution of NRF2 to the pathological conditions of malignant gliomas, particularly in relation to IDH1/2 status.

While the KEAP1 and NRF2 genes are frequently mutated in various solid tumors, relevant somatic KEAP1 mutations were detected in only 1.7% of the anaplastic gliomas (2 cases). These KEAP1 mutations, Q82H and R483H, are identical to those found in gastric adenocarcinomas29 and lung squamous carcinomas,30 respectively. Interestingly, these cases did not carry IDH1/2 mutations, comprising 5.1% of the anaplastic gliomas with wild-type IDH1/2. While genetic aberrations underlying tumorigenesis have been well elucidated in IDH1/2-mutated tumors31 and IDH1/2 wild-type low-grade gliomas,32,33 very few genetic aberrations have been described in anaplastic glioma wild-type IDH1/2. Thus, identification of somatic KEAP1 mutations in anaplastic gliomas with wild-type IDH1/2 is expected to be one of the clues to delineate their features.

For the remaining majority of the malignant gliomas without KEAP1/NRF2 somatic mutations, investigations of either KEAP1 expression or p62 accumulation did not provide conclusive answers regarding NRF2 target gene upregulation. Similar to the malignant gliomas, 22 of 29 ovarian carcinomas with NRF2 accumulation exhibited neither low KEAP1 expression nor KEAP1/NRF2 somatic mutations.9 However, Cong et al15 demonstrated that the extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3 kinase (PI3K) signaling cascades induce NRF2 activation and enhance cell viability partly through NRF2 in human glioblastoma cells. We also observed that NRF2 activity is enhanced under the sustained activation of the PI3K pathway.34 Given that the ERK and PI3K pathways are frequently activated in malignant gliomas, these pathways might have contributed to NRF2 activation in the tumor samples examined in this study.

Intriguingly, NRF2 activity was reduced in the presence of IDH1/2 mutations in clinical tumor samples, and this finding was reproducible in T98 cells that expressed IDH1 R132H. Admitting that T98 cells were established from glioblastoma, whose malignancy was not correlated with the expression levels of NRF2 target genes in this study, we confirmed that T98 cells are dependent on NRF2 activity for their proliferation rate (data not shown) and drug resistance (see Fig. 4B). Thus, we consider that the effect of mutant IDH1 on the NRF2 activity in T98 cells mostly reflects that in primary tumor tissues of patients. Considering the multiple levels of regulation for NRF2 activity, including transcription, protein stability, and DNA binding, clarifying which steps are affected by mutant IDH1 is an important issue to understand the molecular mechanism for the inhibition of the NRF2 pathway in anaplastic gliomas with mutated IDH1/2.

Our study demonstrated that expression levels of NRF2 target genes had prognostic significance in limited cases of malignant gliomas (ie, anaplastic gliomas with mutated IDH1/2), whereas NRF2 activation in lung,10,11 gallbladder,13 ovarian,9 and breast14 cancers associate with poor prognosis irrespective of histological and/or biochemical features. This result implicates that malignant gliomas contain heterogeneous populations with distinct pathological bases. Although the current cohort contained a small number of high expressers of NRF2 target genes among IDH1/2-mutant anaplastic gliomas, our result provides important information on the relation between NRF2 activity and sensitivity to anticancer treatments in anaplastic gliomas with mutated IDH1/2. An increased number of cases will be analyzed for further verification of our current conclusion.

Funding

This work was supported by JSPS KAKENHI grant nos. 24592153; (M.K., T.K., and H.M.), 24249015; (M.Y.), 24390075; (H.M.), and 24790307 (K.T.); MEXT KAKENHI grant nos. 23116002; (H.M.) and 25117703 (K.T.); the Gushinkai Foundation (K.T.), Naito Foundation (M.Y.), Takeda Scientific Foundation (H.M. and M.Y.), and the Core Research for Evolutional Science and Technology from the JST (H.M. and M.Y.).

Supplementary Material

Supplementary Data:

Acknowledgments

We would like to thank Drs Tomoyoshi Soga and Masahiro Sugimoto for providing their special skills of metabolome analysis; Nao Ota for technical support regarding metabolome analysis; and the Biomedical Research Core of the Tohoku University Graduate School of Medicine for their technical support.

Conflict of interest statement. None declared.

References

1. Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol. 2009;27(35):5874–5880. [PubMed] [Google Scholar]
2. Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–466. [PubMed] [Google Scholar]
3. Motohashi H, Yamamoto M. Nrf2-Keap1 defines a physiologically important stress response mechanism. Trends Mol Med. 2004;10(11):549–557. [PubMed] [Google Scholar]
4. Taguchi K, Motohashi H, Yamamoto M. Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution. Genes Cells. 2011;16(2):123–140. [PubMed] [Google Scholar]
5. Singh A, Misra V, Thimmulappa RK, et al. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med. 2006;3(10):e420. [PMC free article] [PubMed] [Google Scholar]
6. Shibata T, Ohta T, Tong KI, et al. Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc Natl Acad Sci U S A. 2008;105(36):13568–13573. [PMC free article] [PubMed] [Google Scholar]
7. Shibata T, Kokubu A, Gotoh M, et al. Genetic alteration of Keap1 confers constitutive Nrf2 activation and resistance to chemotherapy in gallbladder cancer. Gastroenterology. 2008;135(4):1358–1368. [PubMed] [Google Scholar]
8. Zhang P, Singh A, Yegnasubramanian S, et al. Loss of Kelch-like ECH-associated protein 1 function in prostate cancer cells causes chemoresistance and radioresistance and promotes tumor growth. Mol Cancer Ther. 2010;9(2):336–346. [PMC free article] [PubMed] [Google Scholar]
9. Konstantinopoulos PA, Spentzos D, Fountzilas E, et al. Keap1 mutations and NRF2 pathway activation in epithelial ovarian cancer. Cancer Res. 2011;71(15):5081–5089. [PubMed] [Google Scholar]
10. Solis LM, Behrens C, Dong W, et al. Nrf2 and Keap1 abnormalities in non-small cell lung carcinoma and association with clinicopathologic features. Clin Cancer Res. 2010;16(14):3743–3753. [PMC free article] [PubMed] [Google Scholar]
11. Inoue D, Suzuki T, Mitsuishi Y, et al. Accumulation of p62/SQSTM1 is associated with poor prognosis in patients with lung adenocarcinoma. Cancer Sci. 2012;103(4):760–766. [PMC free article] [PubMed] [Google Scholar]
12. Shibata T, Kokubu A, Saito S, et al. NRF2 mutation confers malignant potential and resistance to chemoradiation therapy in advanced esophageal squamous cancer. Neoplasia. 2011;13(9):864–873. [PMC free article] [PubMed] [Google Scholar]
13. Wang J, Zhang M, Zhang L, et al. Correlation of Nrf2, HO-1, and MRP3 in gallbladder cancer and their relationships to clinicopathologic features and survival. J Surg Res. 2010;164(1):99–105. [PubMed] [Google Scholar]
14. Onodera Y, Motohashi H, Takagi K, et al. NRF2 immunolocalization in human breast cancer patients as a prognostic factor. Endocr Relat Cancer. 2014;21(2):241–252. [PubMed] [Google Scholar]
15. Cong ZX, Wang HD, Wang JW, et al. ERK and PI3 K signaling cascades induce Nrf2 activation and regulate cell viability partly through Nrf2 in human glioblastoma cells. Oncol Rep. 2013;30(2):715–722. [PubMed] [Google Scholar]
16. Ji XJ, Chen SH, Zhu L, et al. Knockdown of NF-E2-related factor 2 inhibits the proliferation and growth of U251MG human glioma cells in a mouse xenograft model. Oncol Rep. 2013;30(1):157–164. [PubMed] [Google Scholar]
17. Balss J, Meyer J, Mueller W, et al. Analysis of the IDH1 codon 132 mutation in brain tumors. Acta Neuropathol. 2008;116(6):597–602. [PubMed] [Google Scholar]
18. Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321(5897):1807–1812. [PMC free article] [PubMed] [Google Scholar]
19. Sonoda Y, Kumabe T, Nakamura T, et al. Analysis of IDH1 and IDH2 mutations in Japanese glioma patients. Cancer Sci. 2009;100(10):1996–1998. [PMC free article] [PubMed] [Google Scholar]
20. Mohrenz IV, Antonietti P, Pusch S, et al. Isocitrate dehydrogenase 1 mutant R132H sensitizes glioma cells to BCNU-induced oxidative stress and cell death. Apoptosis. 2013;18(11):1416–1425. [PubMed] [Google Scholar]
21. Shibata T, Kokubu A, Miyamoto M, et al. Mutant IDH1 confers an in vivo growth in a melanoma cell line with BRAF mutation. Am J Pathol. 2011;178(3):1395–1402. [PMC free article] [PubMed] [Google Scholar]
22. Ohashi Y, Hirayama A, Ishikawa T, et al. Depiction of metabolome changes in histidine-starved Escherichia coli by CE-TOFMS. Mol Biosyst. 2008;4(2):135–147. [PubMed] [Google Scholar]
23. Turcan S, Rohle D, Goenka A, et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature. 2012;483(7390):479–483. [PMC free article] [PubMed] [Google Scholar]
24. Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol. 2009;27(25):4150–4154. [PubMed] [Google Scholar]
25. Li Y, Jaiswal AK. Regulation of human NAD(P)H:quinone oxidoreductase gene. Role of AP-1 binding site contained within human antioxidant response element. J Biol Chem. 1992;267(21):15097–15104. [PubMed] [Google Scholar]
26. Mulcahy RT, Wartman MA, Bailey HH, et al. Constitutive and beta-naphthoflavone-induced expression of the human gamma-glutamylcysteine synthetase heavy subunit gene is regulated by a distal antioxidant response element/TRE sequence. J Biol Chem. 1997;272(11):7445–7454. [PubMed] [Google Scholar]
27. Solis WA, Dalton TP, Dieter MZ, et al. Glutamate-cysteine ligase modifier subunit: mouse Gclm gene structure and regulation by agents that cause oxidative stress. Biochem Pharmacol. 2002;63(9):1739–1754. [PubMed] [Google Scholar]
28. Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462(7274):739–744. [PMC free article] [PubMed] [Google Scholar]
29. Yoo NJ, Kim HR, Kim YR, et al. Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology. 2012;60(6):943–952. [PubMed] [Google Scholar]
30. Takahashi T, Sonobe M, Menju T, et al. Mutations in Keap1 are a potential prognostic factor in resected non-small cell lung cancer. J Surg Oncol. 2010;101(6):500–506. [PubMed] [Google Scholar]
31. Watanabe T, Nobusawa S, Kleihues P, et al. IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am J Pathol. 2009;174(4):1149–1153. [PMC free article] [PubMed] [Google Scholar]
32. Kim YH, Lachuer J, Mittelbronn M, et al. Alterations in the RB1 pathway in low-grade diffuse gliomas lacking common genetic alterations. Brain Pathol. 2011;21(6):645–651. [PMC free article] [PubMed] [Google Scholar]
33. Kim YH, Pierscianek D, Mittelbronn M, et al. TET2 promoter methylation in low-grade diffuse gliomas lacking IDH1/2 mutations. J Clin Pathol. 2011;64(10):850–852. [PubMed] [Google Scholar]
34. Mitsuishi Y, Taguchi K, Kawatani Y, et al. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell. 2012;22(1):66–79. [PubMed] [Google Scholar]

Articles from Neuro-Oncology are provided here courtesy of Society for Neuro-Oncology and Oxford University Press