Home The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
Article Open Access

The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma

  • Maher Kurdi ORCID logo EMAIL logo , Motaz M. Fadul , Bassam J. Addas , Eyad Faizo , Shadi Alkhayyat , Fahad Okal , Alaa Alkhotani , Abdulrahman J. Sabbagh , Mohammed Abutalib , Ahmed K. Bamaga , Amany A. Fathaddin and Saleh Baeesa
Published/Copyright: September 21, 2023

Abstract

Objectives

Nuclear factor erythroid-2-related factor 2 (NRF2), a transcriptional gene factor related to nuclear factor erythroid 2, plays a role in the development of gliomas with isocitrate dehydrogenase (IDH) mutation. Its impact on tumour recurrence has seldom been investigated.

Methods

A group of 34 patients diagnosed with Grade 4 astrocytoma was included in a retrospective cohort. NRF2 protein and gene expressions were assessed using different profiling assays. The association between IDH mutation, NRF2 expression, and tumor recurrence was investigated.

Results

The mean patients’ age in this study was 50 years. Out of the total number of tumors analyzed, 21 of them had IDH mutation. NRF2 protein expression was found to be overexpressed in 27 tumors and reduced in 7 tumors. Additionally, NRF2 gene was upregulated in 24 tumors and downregulated in 10 tumors. Insignificant statistical difference was observed in recurrence-free interval (RFI) between patients with upregulated or downregulated NRF2 gene or protein expressions (p-value>0.05). However, this relationship is distinctive when NRF2 expression was compared to IDH mutation. Tumors with different levels of NRF2 expressions and IDH mutations showed significant statistical variation in RFI (p-value=0.001). There was insignificant impact on RFI among patients receiving different chemotherapies (TMZ or TMZ plus) who had abnormal NRF2 gene activities (p-value=0.97).

Conclusions

WHO-Grade 4 astrocytoma with IDH mutation and altered NRF2 expression showed a delayed tumor recurrence compared to IDH-wildtype glioblastoma.

Introduction

The most severe form of primary malignant brain tumor in adults is Grade-4 astrocytoma. Despite decades of treatment options available after surgical resection, the overall survival (OS) remains unchanged [1]. Thus, many biomarkers have been investigated to prevent disease progression [2]. Although various genetic and epigenetic changes have been linked to Grade-4 astrocytoma, exploring a key mechanism for treatment resistance continues to be a difficult therapeutic hurdle. As other methods have yielded inconclusive outcomes, the analysis of gene expression has become crucial in formulating effective treatment plans for malignant brain tumors [3].

Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) is a transcriptional gene that has an anti-apoptotic, anti-oxidative and anti-cancer effects due to its interaction with different signaling regulatory pathways [4]. NRF2 is inactive in normally oxygenated environment; however, it migrates to the nucleus during oxidative stress to promote hemostasis [5]. NRF2 has the ability to counteract oxidative stress by facilitating the release of enzymes that are associated with nicotinamide adenine dinucleotide phosphate (NADPH), including isocitrate dehydrogenase (IDH) and other metabolic regulators. This pathway leads to the production of NADPH as a way to address oxidative stress (Figure 1) [6].

Figure 1: 
A schematic diagram illustrating NRF2 pathway during oxidative stress condition, and how it influences WHO-Grade 4 astrocytoma. It also shows the potential therapeutic targets. NRF2 is a protein that is regulated by KEAP1 to maintain its degradation. When there is oxidative stress, KEAP1 is uncoupled from NRF2, allowing NRF2 to move to the nucleus and bind to other proteins to form a complex. This complex regulates the expression of genes involved in antioxidation and metabolism. Mutations in the NRF2 gene can affect this process. KEAP1, Kelch-like ECH-associated protein 1; ROS, reactive oxygen species.
Figure 1:

A schematic diagram illustrating NRF2 pathway during oxidative stress condition, and how it influences WHO-Grade 4 astrocytoma. It also shows the potential therapeutic targets. NRF2 is a protein that is regulated by KEAP1 to maintain its degradation. When there is oxidative stress, KEAP1 is uncoupled from NRF2, allowing NRF2 to move to the nucleus and bind to other proteins to form a complex. This complex regulates the expression of genes involved in antioxidation and metabolism. Mutations in the NRF2 gene can affect this process. KEAP1, Kelch-like ECH-associated protein 1; ROS, reactive oxygen species.

Based on the old reports, tumour degree was found to be associated with NRF2 activity. Nonetheless, patients with benign tumor do not exhibit elevated NRF2 levels, whereas the levels of NRF2 are variably regulated in patients with high-grade tumours such as glioblastoma [5], [6], [7], [8], [9]. Although this finding has not been broadly and specifically correlated with IDH mutation, it has been observed that some gliomas with IDH mutations demonstrate atypical NRF2 expression [7], [8], [9].

Because NRF2 plays a critical role in cell cycle regulation, its pathways have been identified as potential targets for treating high-grade gliomas. Multiple studies are currently targeting NRF2 molecular pathways to overcome treatment resistance, inhibit the proliferation of glioma cells, and promote apoptosis [10]. NRF2 has a dual characteristic that has been identified, which brings up some concerns when examining it as a potential treatment option [11]. Upon detection of harmful substances, the cell initiates the activation of NRF2, which subsequently transcribes defensive genes and regulates cytoprotective mechanisms. The cell will keep producing NRF2 until it reaches a certain threshold. Once the threshold is crossed, the role of NRF2 shifts from being cytoprotective to promoting apoptosis. For this reason, NRF2 overexpression has been observed to safeguard against antitumor therapies in glioblastoma, while suppressing NRF2 could impede the progression of the disease [9].

NRF2 expression aids glioma cell growth by up-regulating Hypoxia Inducible Factor-1α (HIF-1α) and Vascular Endothelial Growth Factor (VEGF) [12]. X Ji et al., discovered that NRF2 signaling is linked to HIF-1α in glioma angiogenesis [13]. Additionally, it was found that inhibiting NRF2 in gliomas can decrease the activation of HIF-1α and VEGF signaling, possibly by reducing oxygen consumption [13, 14]. Similarly, NRF2 gene downregulation was shown to increase glioblastoma sensitivity to chemotherapeutic agents [4]. It also regulates the expression of antioxidant enzymes for tumor cells against oxidative stressors such as chemotherapeutic drugs and radiation [15]. Zhang et al., has demonstrated that the combination of Temozolomide (TMZ) and radiotherapy has been shown to induce resistance via the NRF2 pathway. Consequently, the inhibition of NRF2 activity can potentially restore the effectiveness of therapy [16]. Although there is no concrete evidence to support the efficacy of the compounds currently being tested in clinical trials, there is a potential for combination therapies targeting several NRF2-mediated pathways. The molecular interactions associated with NRF2 in malignant gliomas are still being actively investigated and multiple new compounds have been identified.

The aims of our study are to examine the correlation between NRF2 gene expression and IDH mutation and the impact of this association on tumour recurrence in Grade 4 astrocytoma.

Materials and methods

Selection of patients

This study has been granted approval by a combined ethical agreement between King Faisal Specialist Hospital and King Abdulaziz University (agreement # CA-2020-06), to utilize patient tissue for our study. The study involved a cohort of 34 patients who were diagnosed with Grade 4 astrocytoma following gross resection (Table 1). The pathological diagnosis followed the 5th edition of WHO classification of Central Nervous System (CNS) tumours [1, 17].

Table 1:

The biological data of 34 tumours enrolled in our study.

Age Sex IDH status IHC grading NRF2 Ct NRF2 RE NRF2 expression CTX RFI
80 Male IDH-mutant II 36.428 0.612 Upregulated TMZ 600
67 Female IDH-wildtype II 30.663 0.097 Upregulated TMZ 198
40 Male IDH-mutant II 32.450 0.513 Upregulated TMZ+ 534
44 Male IDH-mutant II 29.963 0.300 Upregulated TMZ 76
56 Male IDH-wildtype II 29.877 0.066 Upregulated TMZ 191
50 Female IDH-wildtype II 29.952 0.156 Upregulated None 217
48 Male IDH-mutant II 32.317 0.195 Upregulated TMZ 747
51 Male IDH-wildtype II 28.743 0.233 Upregulated TMZ 180
31 Female IDH-mutant II 30.147 0.360 Upregulated TMZ 359
25 Male IDH-wildtype II 30.681 0.116 Upregulated TMZ 430
66 Female IDH-mutant II 30.885 0.127 Upregulated TMZ 350
59 Female IDH-mutant II 29.885 0.147 Upregulated TMZ 293
56 Female IDH-mutant II 32.675 0.105 Upregulated TMZ 566
45 Male IDH-wildtype II 33.029 0.225 Upregulated TMZ 190
85 Male IDH-wildtype II 31.457 0.228 Upregulated TMZ 211
51 Male IDH-mutant II 33.029 0.225 Upregulated None 1128
54 Male IDH-mutant II 29.585 0.258 Upregulated TMZ 339
31 Male IDH-mutant II 32.005 0.140 Upregulated TMZ 623
21 Male IDH-wildtype II 30.688 0.377 Upregulated TMZ 485
29 Female IDH-mutant II 33.958 0.328 Upregulated TMZ 550
29 Male IDH-wildtype II 33.645 0.175 Upregulated None 80
64 Female IDH-wildtype II 33.148 0.189 Upregulated None 130
83 Male IDH-mutant II 33.180 0.290 Upregulated TMZ + 684
47 Male IDH-mutant II 30.843 0.139 Upregulated TMZ 762
31 Male IDH-mutant I 31.226 0.460 Downregulated TMZ 548
14 Female IDH-mutant I 31.911 0.107 Downregulated TMZ 229
15 Female IDH-mutant I 32.476 0.412 Downregulated TMZ 461
59 Male IDH-mutant II 28.212 0.301 Downregulated TMZ + 549
78 Male IDH-mutant I 30.302 0.363 Downregulated TMZ 723
43 Female IDH-wildtype II 30.302 0.136 Downregulated TMZ 208
59 Male IDH-wildtype I 32.769 0.099 Downregulated None 455
51 Male IDH-mutant II 34.462 0.273 Downregulated TMZ 210
64 Male IDH-wildtype I 30.655 0.611 Downregulated TMZ 433
60 Male IDH-mutant I 29.045 0.171 Downregulated TMZ 1100
  1. IDH, isocitrate dehydrogenase; IHC, immunohistochemistry; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; NRF2 nuclear factor erythroid 2-related factor 2; RE, relative expression, CTX, chemotherapy; RFI: recurrence free-interval.

Radiation was given to all patients and TMZ was given only to 29 patients at a rate of 150–200 mg/m2 up to 12 cycles (Table 1) [18]. All patients enrolled in this project are unfortunately deceased. The data collected from the patients encompassed their age, gender, IDH mutation status, treatment history, and Recurrence-Free Interval (RFI). RFI was determined by estimating the period from the date of gross tumor resection to the first day of tumor recurrence.

Tissue processing

A 4-μm section from each 34 formalin-fixed paraffin-embedding (FFPE) tissue block was obtained for expression assays. The thick section slides were employed for the protein expression, utilizing Immunohistochemistry (IHC) with an anti-NRF2 antibody. On the other hand, the tissue rolls were used in RNA extraction to assess NRF2 gene expression using Real Time-Polymerase Chain Reaction (RT-qPCR).

Anti-NRF2 protein expression assay

Anti-NRF2 antibody (Rabbit polyclonal, Elabscience, Cat# E-AB-64023, Houston, Texas, USA) was targeted on 34 tissue sections using IHC assay. The Ultra-View detection Kit from Ventana (Tucson, AZ, USA) was employed on automated auto-stainer for IHC processing. The protocol involved deparaffinization using EZ Prep and followed by heat pre-treatment. Subsequently, the antibody was adjusted to a dilution of 1:100 and incubated for 20 min at 75 °C. The tumor microenvironment was assessed for the presence of tumor cells (TC) using anti-NRF2 antibody. Tissue sections were initially examined at (×10) magnification to identify a focal living area exhibiting anti-NRF2 expression. Subsequently, this area was further examined at (×25) magnification. Cells displaying anti-NRF2 expression were classified as NRF2 positive, while the total cell population demonstrated both NRF2-expressing cells and non-stained cells. The labelling index (LI) of NRF2 expression in each selected area was assessed using the following equation:

Labelling index  ( % ) = NRF 2 + TC Total cells × 100

Three expression arrays were estimated: Grade [0]: <10 % expression; Grade [1]: 10–50 % expression; Grade [2]: >50 % expression (Figure 2). Expression with Grade [0] and [I] was categorized as “low expression, while expression with Grade [II] was categorized as “high expression”.

Figure 2: 
Anti-NRF2 stains tumour cells in microenvironment using IHC. (a) low expression (b) high expression. Magnification [×25].
Figure 2:

Anti-NRF2 stains tumour cells in microenvironment using IHC. (a) low expression (b) high expression. Magnification [×25].

RNA extraction and cDNA synthesis

RNA was isolated from a total of 34 tumor samples and two control samples using the RNeasy FFPE Kit (QIAGEN). Each FFPE sample was transferred to RNase-free Eppendorf tube after being mixed with m-Xylene for deparaffinization. The samples were then centrifuged to collect the capsule, followed by a wash with ethanol and another round of centrifugation. The ethanol was subsequently evaporated in a 37 °C dry bath and all samples were incubated for 15 min followed by a short cooling. A mixture of Buffer RBC and the sample was vortexed, and then 100 % ethanol was added and mixed was then centrifuged. The eluates-containing RNA was used for spectrophotometric analysis. The High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems™, Waltham, MA, USA) was utilized for cDNA synthesis. A master mix was prepared by a mixture of Reverse Transcriptase (RT) Buffer, dNTP Mix, Random Primers, and MultiScribe™ RT. This mixture was then combined with 80 ng of RNA, and the final volume was adjusted to 10 µL.

Gene expression profiling using RT-qPCR

The primers for NRF2 gene and one reference gene, Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH), were designed. The following primer sequences for NRF2 have been used in the processing: forward: 5′-TCCATTCCTGAGTTACAGTGTCT-3′, Reverse 5′-CCATGGTAGTCTCAACCAGC-3′. The RT-qPCR analysis was conducted using the EverGreen Master Mix (Haven Scientific, KAUST, KSA), following the manufacturer’s instructions in triplicate reactions. In brief, the previously synthesized cDNAs were combined with the master mix and enough amount of oligo in order to achieve optimum PCR reactions. The reactions were performed in semi-skirted plates which were preserved with the RT-qPCR Adhesive Seal (Haven Scientific, KAUST, KSA). An initial denaturation step at 95 °C for 3 min was performed and followed by 40 cycles of denaturation for 15 seconds and 60 s annealing process.

Two replicates of threshold cycle (CT) values were used for both the targeting gene and the reference gene. The mean CT and standard deviation for the reference gene (GAPDH) and the target gene (NRF2) were calculated from the RT-PCR data. Relative quantification (Rq), fold change (FC) and ΔCT values were also assessed for each sample. The results are summarized in Table 1.

Statistical analysis

To assess the correlation between NRF2 gene and protein expression, a sensitivity and specificity analysis was conducted using the McNemar test. The distribution of RFI in relation to other variables was compared using Kaplan-Meier curves (KMC). Fisher’s Exact test was also employed to investigate the association between NRF2 gene expression and IDH mutation. Statistical significance was determined at a p-value of less than 0.05.

Results

The average age of the patients was 50 years (SD: 17.3). IDH was mutant in 21 tumors. NRF2 protein was overexpressed in 27 (79.4 %) tumors and reduced in 7 (20.6 %) tumors. NRF2 gene expression was upregulated in 24 (70.6 %) tumors and downregulated in 10 (29.4 %) tumors. Tumors with low levels of NRF2 protein expression as detected through IHC showed NRF2 gene downregulation in RT-qPCR. Conversely, 24 tumors with high levels of NRF2 expression as detected by IHC demonstrated NRF2 gene upregulation in RT-qPCR. The specificity and sensitivity of protein expression to gene expression were 100 and 70 %, respectively. The overall diagnostic accuracy between the two testing methods was 91 % (Figure 3).

Figure 3: 
Matching consistency of NRF2 expression between IHC and RT-PCR.
Figure 3:

Matching consistency of NRF2 expression between IHC and RT-PCR.

There was no significant statistical distinction observed in RFI between tumors exhibiting upregulated or downregulated NRF2 gene or protein expressions (p-value >0.05) (Figure 4a and b). This relationship is distinctive when NRF2 expressions were compared to IDH mutation. There was a significant correlation found between abnormal NRF2 gene expression and IDH mutation. Tumors with different levels of NRF2 gene expression and IDH mutations showed significant statistical variation in RFI (p-value=0.001) (Figure 5a). Out of the 14 IDH-mutant tumors with upregulated NRF2, 10 were wildtype for IDH. Nonetheless, IDH-mutant tumors with either upregulated or downregulated NRF2 expressions exhibited a delayed tumor recurrence compared to tumors with IDH wildtype, regardless of NRF2 abnormality, highlighting the greater impact of IDH mutation on RFI. Furthermore, there was no significant statistical impact observed on patients receiving TMZ or TMZ and other chemotherapeutic agents with abnormal NRF2 gene activities (p-value=0.97) (Figure 5b). However, an interesting finding was noted among patients with NRF2 downregulation who received TMZ treatment, as they experienced late tumor recurrence.

Figure 4: 
(a and b) The impact of NRF2 gene and protein expressions on RFI. RFI, Recurrence-free interval.
Figure 4:

(a and b) The impact of NRF2 gene and protein expressions on RFI. RFI, Recurrence-free interval.

Figure 5: 
(a and b) The impact of NRF2 gene and protein expressions, IDH mutation and chemotherapies on RFI. RFI, Recurrence-free interval.
Figure 5:

(a and b) The impact of NRF2 gene and protein expressions, IDH mutation and chemotherapies on RFI. RFI, Recurrence-free interval.

Discussion

The activation of NRF2 gene signaling pathways provides a novel therapeutic strategy in many cancers including brain tumours. NRF2 has been found to be upregulated in malignant astrocytoma, mainly Grade 4 type, which can contribute to uncontrolled tumour growth and resistance to chemotherapy. By promoting antioxidant defense, reducing inflammation, and enhancing DNA repair, NRF2 inhibition could potentially help to overcome the resistance of malignant astrocytoma to conventional treatments. Currently, the exact process that causes NRF2 upregulation in malignant astrocytoma remains unclear. However, one of the commonly linked factors is IDH mutation. Studies indicate that IDH mutation significantly impacts the regulation of the NRF2 gene, suppressing its expression [4]. This can cause less cellular oxidative stress [9, 15].

While NRF2 is frequently present in IDH-mutant WHO-Grade 4 astrocytoma, the association between these two factors has not been scientifically considered as a cause for delayed tumor recurrence. Our study did not reveal a substantial correlation between IDH mutation and NRF2 upregulation. Out of the 14 IDH-mutant tumors with upregulated NRF2, 10 cases with upregulated NRF2 expression were IDH wildtype. Nonetheless, IDH mutation with upregulated or downregulated NRF2 exhibited a delayed tumor recurrence compared to IDH-wildtype tumors. This indicates that the IDH drivers could potentially halt NRF2 activity through unknown mechanisms.

Two studies have shown that IDH1 and IDH2 mutations lead to increased levels of reactive oxygen species (ROS), which in turn activate NRF2 [19, 20]. IDH-mutant gliomas were shown to increase levels of NRF2 and their downstream targets. These findings suggest that there is a link between IDH mutation and NRF2 upregulation, which could have important implications for cancer treatment [19]. Tonelli et al. found that IDH-mutant gliomas cause excessive release of Nicotinamide Adenine Dinucleotide Phosphate (NADP) oxidative coenzyme which in turn uncouples NRF2 inside the nucleus [20]. Keap1, a component of the E3-ubiquitin ligase complex, binds to NRF2 and keeps it continually ubiquitinated and degraded. However, when the body is under stress, Keap1’s cysteine residues get oxidized by ROS, causing it to separate from NRF2. NRF2 can then travel to the nucleus and forms a complex with musculoaponeurotic fibrosarcoma (Maf) oncogene homolog known as the NRF2-Maf complex. This complex binds to antioxidant response elements and helps in regulating cellular processes during stress (Figure 1) [20].

Because NRF2 and IDH both share similar chromosomal location, their functions are concurrently related. The tricarboxylic acid cycle (TCA cycle) is a center for cellular energy production and the precursor to metabolic pathways. Hence, NRF2 regulates TCA cycle genes including IDH (Reactome pathway ID: R-HSA-9755511.4) which cause excessive production of D2-hydroxyglutarate (D2-HG). Elevated levels of D2-HG are found in IDH mutant tumors as shown in (Figure 6). The mechanism by which the NRF2 protein is involved with other related mutant genes to develop glioma is described in the Supplementary file.

Figure 6: 
Mechanism of NRF2 mediated IDH regulation leading to WHO Grade IV astrocytoma.
Figure 6:

Mechanism of NRF2 mediated IDH regulation leading to WHO Grade IV astrocytoma.

Several drugs have been found to affect NRF2 function either directly or indirectly. PDI inhibitor, corilagin, valproic acid, melatonin, and trans-retinoic acids are examples of drugs that indirectly impact NRF2 signaling in TMZ-resistant glioblastoma by inducing apoptosis and inhibiting its activity [10, 21, 22]. In our study, patients with NRF2 downregulation who had TMZ treatment experienced delayed tumor recurrence. Nevertheless, there was an insignificant impact on tumor recurrence among patients receiving TMZ and TMZ plus, regardless NRF2 gene alteration. Studies have shown that the use of a compound similar to sphingosine-1-phosphate can decrease the presence of NRF2 in glioblastoma cells, thus increasing their sensitivity to TMZ. Another method of impeding tumor growth is by targeting vascular and growth factor receptors. By controlling NRF2 expression, angiogenesis can be reduced, and tumor growth becomes inhibited [23].

Although the sample size in our study was limited, it is crucial to recognize that our research is the initial endeavor to establish a connection between NRF2, IDH, and tumor recurrence, and explore the underlying mechanism.

Conclusions

Our study concluded that WHO-Grade 4 astrocytoma with IDH mutation and altered NRF2 expression exhibits a delayed tumor recurrence compared to IDH-wildtype glioblastoma. This explains that the IDH driver could potentially halt NRF2 activity through unknown mechanisms. It also indicates that NRF2 is not a direct prognostic indicator.


Corresponding author: Maher Kurdi, Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Kingdom of Saudi Arabia; and Neuromuscular Unit, King Fahad Medical Research Center, Jeddah, Saudi Arabia, E-mail:

Acknowledgments

Special thanks to Deanship of Scientific Research at King Abdulaziz University for their support.

  1. Research ethics: The local Institutional Review Board deemed the study exempt from review. This study was performed in line with the principles of the Declaration of Helsinki. The approval was granted by Biomedical Ethics Committee at King Faisal Specialist Hospital and Research Center [CA-2020-06] and King Abdulaziz University [HA-02-J-008] to authorize using Patient Samples in Research.

  2. Informed consent: Informed consent was obtained from all individual participants included in the study.

  3. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  4. Competing interests: Authors state no conflict of interest.

  5. Research funding: No funding.

  6. Data availability: The datasets generated during and/or analysed during the current study are available from the corresponding author on reasonable request.

References

1. Louis, DN, Perry, A, Wesseling, P, Brat, DJ, Cree, IA, Figarella-Branger, D, et al.. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol 2021;23:1231–51.10.1093/neuonc/noab106Search in Google Scholar PubMed PubMed Central

2. Weller, M, van den Bent, M, Preusser, M, Le Rhun, E, Tonn, JC, Minniti, G, et al.. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol 2021;18:170–86.10.1038/s41571-020-00447-zSearch in Google Scholar PubMed PubMed Central

3. D’Alessio, A, Proietti, G, Sica, G, Scicchitano, BM. Pathological and molecular features of glioblastoma and its peritumoral tissue. Cancers 2019;11:469.10.3390/cancers11040469Search in Google Scholar PubMed PubMed Central

4. Awuah, WA, Toufik, AR, Yarlagadda, R, Mikhailova, T, Mehta, A, Huang, H, et al.. Exploring the role of NRF2 signaling in glioblastoma multiforme. Discov Oncol 2022;13:94.10.1007/s12672-022-00556-4Search in Google Scholar PubMed PubMed Central

5. Fan, Z, Wirth, AK, Chen, D, Wruck, CJ, Rauh, M, Buchfelder, M, et al.. NRF2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis 2017;8:e371.10.1038/oncsis.2017.65Search in Google Scholar PubMed PubMed Central

6. Lin, TY, Cantley, LC, DeNicola, GM. NRF2 rewires cellular metabolism to support the antioxidant response In: A master regulator of oxidative stress - the transcription factor Nrf2, InTech; 2017, Available from: https://doi.org/10.5772/6514110.5772/65141Search in Google Scholar

7. Haapasalo, J, Nordfors, K, Granberg, KJ, Kivioja, T, Nykter, M, Haapasalo, H, et al.. NRF2, DJ1 and SNRX1 and their prognostic impact in astrocytic gliomas. Histol Histopathol 2018;33:791–801.Search in Google Scholar

8. Kanamori, M, Higa, T, Sonoda, Y, Murakami, S, Dodo, M, Kitamura, H, et al.. Activation of the NRF2 pathway and its impact on the prognosis of anaplastic glioma patients. Neuro Oncol 2015;17:555–65.10.1093/neuonc/nou282Search in Google Scholar PubMed PubMed Central

9. Zhu, J, Wang, H, Ji, X, Zhu, L, Sun, Q, Cong, Z, et al.. Differential Nrf2 expression between glioma stem cells and non-stem-like cells in glioblastoma. Oncol Lett 2014;7:693–8.10.3892/ol.2013.1760Search in Google Scholar PubMed PubMed Central

10. Pan, H, Wang, H, Jia, Y, Wang, Q, Li, L, Wu, Q, et al.. VPA and MEL induce apoptosis by inhibiting the Nrf2-ARE signaling pathway in TMZ-resistant U251 cells. Mol Med Rep 2017;16:908–14.10.3892/mmr.2017.6621Search in Google Scholar PubMed

11. Catanzaro, E, Calcabrini, C, Turrini, E, Sestili, P, Fimognari, C Nrf2: a potential therapeutic target for naturally occurring anticancer drugs? Expert Opin Ther Targets 2017;21:781–93.10.1080/14728222.2017.1351549Search in Google Scholar PubMed

12. Ji, XJ, Chen, SH, Zhu, L, Pan, H, Zhou, Y, Li, W, 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;30:157–64.10.3892/or.2013.2476Search in Google Scholar PubMed

13. Ji, X, Wang, H, Zhu, J, Zhu, L, Pan, H, Li, W, et al.. Knockdown of Nrf2 suppresses glioblastoma angiogenesis by inhibiting hypoxia-induced activation of HIF-1α. Int J Cancer 2014;135:574–84.10.1002/ijc.28699Search in Google Scholar PubMed

14. Kim, TH, Hur, EG, Kang, SJ, Kim, JA, Thapa, D, Lee, YM, et al.. NRF2 blockade suppresses colon tumor angiogenesis by inhibiting hypoxia-induced activation of HIF-1α. Cancer Res 2011;71:2260–75.10.1158/0008-5472.CAN-10-3007Search in Google Scholar PubMed

15. Meyer, UA. Overview of enzymes of drug metabolism. J Pharmacokinetic Biopharm 1996;24:449–59.10.1007/BF02353473Search in Google Scholar PubMed

16. Zhang, L, Wang, H. FTY720 inhibits the Nrf2/ARE pathway in human glioblastoma cell lines and sensitizes glioblastoma cells to temozolomide. Pharmacol Rep 2017;69:1186–93.10.1016/j.pharep.2017.07.003Search in Google Scholar PubMed

17. Kurdi, M, Moshref, RH, Katib, Y, Faizo, E, Najjar, AA, Bahakeem, B, et al.. Simple approach for the histomolecular diagnosis of central nervous system gliomas based on 2021 World Health Organization Classification. World J Clin Oncol 2022;13:567–76.10.5306/wjco.v13.i7.567Search in Google Scholar PubMed PubMed Central

18. Stupp, R, Mason, WP, van den Bent, MJ, Weller, M, Fisher, B, Taphoorn, MJ, et al.. Radiotherapy plus concomitant and adjuvant TMZ for glioblastoma. N Engl J Med 2005;352:987–96.10.1056/NEJMoa043330Search in Google Scholar PubMed

19. Poursheikhani, A, Abbaszadegan, MR, Nokhandani, N, Kerachian, MA, Mollazadeh, S. Targeting NRF2 for cancer therapy: a promising strategy for cancer therapy. Mol Biol Rep 2018;45:2011–34.Search in Google Scholar

20. Tonelli, C, Chio, IIC, Tuveson, DA. Transcriptional regulation by Nrf2. Antioxid Redox Signal 2018;29:1727–45.10.1089/ars.2017.7342Search in Google Scholar PubMed PubMed Central

21. Liu, J, Qin, X, Ma, W, Jia, S, Zhang, X, Yang, X, et al.. Corilagin induces apoptosis and autophagy in NRF2-addicted U251 glioma cell line. Mol Med Rep 2021;23:320.10.3892/mmr.2021.11959Search in Google Scholar PubMed PubMed Central

22. Pokkunuri, ID, Lokhandwala, MF, Banday, AA. Protein disulfide isomerase inhibition impairs Keap1/Nrf2 signaling and mitochondrial function and induces apoptosis in renal proximal tubular cells. Am J Physiol Ren Physiol 2022;319:F686–96.10.1152/ajprenal.00049.2020Search in Google Scholar PubMed PubMed Central

23. Cong, ZX, Wang, HD, Wang, JW, Zhou, Y, Pan, H, Zhang, DD, et al.. ERK and PI3K signaling cascades induce Nrf2 activation and regulate cell viability partly through Nrf2 in human glioblastoma cells. Oncol Rep 2013;30:715–22.10.3892/or.2013.2485Search in Google Scholar PubMed


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/oncologie-2023-0262).


Received: 2023-07-01
Accepted: 2023-09-06
Published Online: 2023-09-21

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Frontmatter
  2. Review Articles
  3. Mitochondrial thermogenesis in cancer cells
  4. Application of indocyanine green in the management of oral cancer: a literature review
  5. Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
  6. The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
  7. Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
  8. Research Articles
  9. Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
  10. The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
  11. More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
  12. DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
  13. IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
  14. MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
  15. Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
  16. Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
  17. Case Reports
  18. A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
  19. Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
  20. Rapid Communication
  21. ROMO1 – a potential immunohistochemical prognostic marker for cancer development
  22. Article Commentary
  23. A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism
Downloaded on 10.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2023-0262/html
Scroll to top button