Elsevier

Free Radical Biology and Medicine

Volume 65, December 2013, Pages 750-764
Free Radical Biology and Medicine

Review Article
Oncogenic functions of the transcription factor Nrf2

https://doi.org/10.1016/j.freeradbiomed.2013.06.041Get rights and content

Highlights

  • Nrf2 is a cytoprotective transcription factor regulating antioxidant defense.

  • Upregulation or hyperactivation of Nrf2 is frequent in cancer.

  • Nrf2 can participate in tumorigenesis and cancer progression.

  • High Nrf2 activity induces resistance of cancer cells to chemotherapy.

  • Nrf2 is a potential biomarker and pharmacological target of interest in cancer.

Abstract

Nuclear factor E2-related factor 2 (Nrf2) is a transcription factor that controls the expression of a large pool of antioxidant and cytoprotective genes regulating the cellular response to oxidative and electrophilic stress. Nrf2 is negatively regulated by Kelch-like ECH-associated protein 1 (Keap1) and, upon stimulation by an oxidative or electrophilic insult, is rapidly activated by protein stabilization. Owing to its cytoprotective functions, Nrf2 has been traditionally studied in the field of chemoprevention; however, there is accumulated evidence that Keap1/Nrf2 mutations or unbalanced regulation that leads to overexpression or hyperactivation of Nrf2 may participate in tumorigenesis and be involved in chemoresistance of a wide number of solid cancers and leukemias. In addition to protecting cells from reactive oxygen species, Nrf2 seems to play a direct role in cell growth control and is related to apoptosis-regulating pathways. Moreover, Nrf2 activity is connected with oncogenic kinase pathways, structural proteins, hormonal regulation, other transcription factors, and epigenetic enzymes involved in the pathogenesis of various types of tumors. The aim of this review is to compile and summarize existing knowledge of the oncogenic functions of Nrf2 to provide a solid basis for its potential use as a molecular marker and pharmacological target in cancer.

Introduction

Nrf2, or nuclear factor E2-related factor 2, is a transcription factor belonging to the cap′n’collar family of leucine-zipper (b-ZIP) proteins [1], which regulates cell response to oxidative and electrophilic insults.

Under homeostatic conditions, Nrf2 is generally localized in the cytoplasm, where it is sequestered by its inhibitor, Keap1, or Kelch-like ECH-associated protein 1 (Fig. 1A). Keap1 interacts with the Nrf2 N-terminal domain Neh2 [2], [3] through two binding sites located on its Kelch-like domains and facilitates the association of Cul3, a Cullin-3-based E3 ubiquitin ligase [4], [5], [6]. Cul3 then mediates the ubiquitination and subsequent degradation of Nrf2 by 26 S proteasome [7], [8]. Thus, under basal conditions, Nrf2 undergoes a rapid physiological turnover triggered by Keap1.

Under oxidative or electrophilic stress conditions (Fig. 1B), however, Keap1 acts as a molecular sensor and undergoes chemical modifications in a series of reactive cysteine residues (reviewed in [9], [10]), allowing the release of Nrf2 [11], [12], which escapes from degradation and translocates to the nucleus. Therefore, protein stabilization is the main mechanism for the activation of the Nrf2 response [13], [14].

Once in the nucleus, Nrf2 heterodimerizes with proteins from a family of b-ZIP oncogenes called small Maf (musculoaponeurotic fibrosarcoma) proteins [15] and binds antioxidant response elements (AREs) localized in the promoter region of its target genes. Nrf2 target genes are mainly antioxidant and phase II enzymes such as heme oxygenase-1 (HO-1) [16], NAD(P)H–quinone oxidoreductase 1 (NQO1) [15], [17], glutathione S-transferases (GSTs) [18], γ-glutamylcysteinyl synthetase [19], glutathione peroxidases [20], thioredoxin reductase 1 (TrxR1) [21], [22], peroxiredoxin 1 [23], aldehyde oxygenase [24], and other genes regulating the response to oxidative stress [25]. Nrf2 also activates the transcription of some genes of the multidrug resistance (MDR) family such as MRP1 [26], MRP2 [27], [28], MRP3, and MRP4 [28]. Overall, Nrf2 regulates the expression, both basal and inducible [29], of enzymes and other proteins involved in cell protection from physical/chemical insults, detoxification, and restoration of homeostasis. For this reason, Nrf2 is considered a cytoprotective transcription factor.

Owing to its role in protecting the cell from cytotoxicity associated with reactive oxygen species (ROS) and electrophilic stressors, Nrf2 is especially important in chemoprevention of diseases. The role of Nrf2 in this field has been widely studied and a great number of Nrf2 inductors, mainly natural compounds present in vegetables, have been described (reviewed in [30], [31]). Moreover, there are currently several active clinical trials for activators of the Nrf2 pathway with potential utility in chemoprevention of various pathologies that are generally characterized by the production of intracellular ROS and eventual cell death [32]. Among those pathologies, the role of Nrf2 has been deeply studied in cancer, in which the response of cells to physical (radiation) and chemical (pollution, toxins, drugs) insults is especially important. Consequently, the number of citations relating Nrf2 and cancer has exponentially increased in the past decade [33].

As a cytoprotective gene, Nrf2 has been traditionally considered to be a tumor suppressor. For instance, Nrf2-deficient mice seem to be more sensitive to carcinogenesis [34], [35] and Nrf2 loss has been related to enhanced metastasis [36], [37]. Accordingly, there are multiple reports describing the beneficial effects of Nrf2 signaling in cancer chemoprevention (reviewed in [30]). However, in the past few years, mounting evidence that the activation of the Nrf2 pathway might not be beneficial in all cancer types and stages has started to arise. In fact, there are many reports that support the idea that Nrf2 activation in malignant cells could be detrimental for the evolution of the disease as well as for the outcomes of the treatment, and findings of several mutations and aberrant signaling of the Nrf2 pathway in cancer reveal a new role for this factor beyond its functions in chemoprevention. Thus, the beneficial effects of the activation of Nrf2 signaling in cancer have become a controversial issue (reviewed in [38]).

From either perspective, it seems to be clear that Nrf2 is an interesting pharmacological target for the prevention or treatment of malignant diseases. There is therefore an increasing need to define the limits between Nrf2's positive and negative effects in cancer and establish the basis for rational Nrf2-targeted therapies. In this article, we attempt to address this issue, focusing on Nrf2 as a potential oncogene and reviewing the most recent advances in this field to help provide a solid basis for the use of Nrf2 as a molecular marker and pharmacological target in cancer.

Section snippets

Nrf2 as a proto-oncogene

Nrf2 signaling in physiological conditions acts as a switch that is turned on by the presence of stressors in the cellular microenvironment and that is rapidly deactivated when the insult is withdrawn and homeostasis is restored. However, under pathological conditions, the tight regulation of Nrf2 by rapid protein turnover is highly susceptible to being altered. This could result in the loss of responsiveness to cell stressors and subsequent vulnerability of the cell to various insults. For

Nrf2 in tumorigenesis

The participation of Nrf2 in cancer pathogenesis is a controversial topic, provided a number of reports that still assign Nrf2 a role in cancer chemoprevention from genotoxic agents [43], [44], [45] or inflammation [46]. However, some reports have shown that drugs that activate Nrf2 can promote cell growth [47], [48], [49] and an increasing number of works point to a potential role for Nrf2 and its transcriptional target genes in tumorigenesis.

Nrf2 in chemoresistance

The aforementioned ability of Nrf2 to counterbalance proapoptotic signals and favor cell survival, along with its cytoprotective nature, makes this factor not only a good promoter of tumorigenesis but also an important antagonist of the effects of chemotherapy.

High expression of some antioxidant Nrf2 target genes has been related to chemoresistance before. In myeloid leukemias, both acute and chronic, HO-1 seems to be an especially important effector of Nrf2-induced chemoresistance and the

Deregulation of the Nrf2 pathway in cancer

Consistent with its role in tumorigenesis and chemoresistance, the hyperactivation and/or aberrant expression of Nrf2 and its transcriptional targets is well documented in cancer (summarized in Fig. 2, Fig. 3).

Conclusions and perspectives

In this review, we have presented compelling evidence that the transcription factor Nrf2 can function as a proto-oncogene in plenty of solid tumors and leukemias.

Nrf2 can be activated by numerous compounds and is also frequently deregulated in a wide variety of cancers by mutations, aberrant epigenetic or posttranslational regulation, or hyperactivation of oncogenic signaling pathways involving other transcription factors such as NF-κB, various protein kinases, structural proteins such as

Acknowledgments

This work was supported by a research personnel training (Formación de Personal Investigador) grant from the Human Resources Promotion Program of the Regional Plan for Scientific Research, Technological Development, and Innovation 2005–2010 (Plan Regional para la Investigación Científica, Desarrollo Tecnológico e Innovación) from the Junta de Comunidades de Castilla–la Mancha, Spain, and the European Social Fund 2007/2013.

References (204)

  • B. Yang et al.

    Pi, J. Deficiency in the nuclear factor E2-related factor 2 renders pancreatic β-cells vulnerable to arsenic-induced cell damage

    Toxicol. Appl. Pharmacol.

    (2012)
  • C.J. Powell et al.

    Hepatic responses to the administration of high doses of BHT to the rat: their relevance to hepatocarcinogenicity

    Food Chem. Toxicol.

    (1986)
  • E. Hara et al.

    Expression of heme oxygenase and inducible nitric oxide synthase mRNA in human brain tumors

    Biochem. Biophys. Res. Commun.

    (1996)
  • M.D. Maines et al.

    Expression of heme oxygenase-1 (HSP32) in human prostate: normal, hyperplastic, and tumor tissue distribution

    Urology

    (1996)
  • T. Cresteil et al.

    High levels of expression of the NAD(P)H:quinone oxidoreductase (NQO1) gene in tumor cells compared to normal cells of the same origin

    Biochem. Pharmacol

    (1991)
  • A.T. Dinkova-Kostova et al.

    NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1), a multifunctional antioxidant enzyme and exceptionally versatile cytoprotector

    Arch. Biochem. Biophys.

    (2010)
  • C.M. Cabello et al.

    3rd; Bause, A. S.; Wondrak, G. T. Antimelanoma activity of the redox dye DCPIP (2,6-dichlorophenolindophenol) is antagonized by NQO1

    Biochem. Pharmacol.

    (2009)
  • M. Garate et al.

    The NAD(P)H:quinone oxidoreductase 1 induces cell cycle progression and proliferation of melanoma cells

    Free Radic. Biol. Med.

    (2010)
  • M.H. Yoo et al.

    Thioredoxin reductase 1 deficiency reverses tumor phenotype and tumorigenicity of lung carcinoma cells

    J. Biol. Chem.

    (2006)
  • K. Miyanishi et al.

    Glutathione S-transferase-pi overexpression is closely associated with K-ras mutation during human colon carcinogenesis

    Gastroenterology

    (2001)
  • Y. Mitsuishi et al.

    Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming

    Cancer Cell

    (2012)
  • R.J. DeBerardinis et al.

    The biology of cancer: metabolic reprogramming fuels cell growth and proliferation

    Cell Metab.

    (2008)
  • N.R. Kitteringham et al.

    Proteomic analysis of Nrf2 deficient transgenic mice reveals cellular defence and lipid metabolism as primary Nrf2-dependent pathways in the liver

    J. Proteomics

    (2010)
  • R. Faraonio et al.

    p53 suppresses the Nrf2-dependent transcription of antioxidant response genes

    J. Biol. Chem.

    (2006)
  • W.S. el-Deiry et al.

    WAF1, a potential mediator of p53 tumor suppression

    Cell

    (1993)
  • G. Filomeni et al.

    p38(MAPK) and ERK1/2 dictate cell death/survival response to different pro-oxidant stimuli via p53 and Nrf2 in neuroblastoma cells SH-SY5Y

    Biochem. Pharmacol

    (2012)
  • S.K. Niture et al.

    Nrf2 protein up-regulates antiapoptotic protein bcl-2 and prevents cellular apoptosis

    J. Biol. Chem.

    (2012)
  • D.F. Lee et al.

    KEAP1 E3 ligase-mediated downregulation of NF-kappaB signaling by targeting IKKbeta

    Mol. Cell

    (2009)
  • P. Moi et al.

    Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region

    Proc. Natl. Acad. Sci. USA

    (1994)
  • K. Itoh et al.

    Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain

    Genes Dev.

    (1999)
  • S.B. Cullinan et al.

    The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: oxidative stress sensing by a Cul3–Keap1 ligase

    Mol. Cell. Biol.

    (2004)
  • M. Furukawa et al.

    BTB protein Keap1 targets antioxidant transcription factor Nrf2 for ubiquitination by the cullin 3–Roc1 ligase

    Mol. Cell. Biol

    (2005)
  • K.I. Tong et al.

    Keap1 recruits Neh2 through binding to ETGE and DLG motifs: characterization of the two-site molecular recognition model

    Mol. Cell. Biol.

    (2006)
  • A. Kobayashi et al.

    Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2

    Mol. Cell. Biol.

    (2004)
  • W. Li et al.

    Molecular mechanisms of Nrf2-mediated antioxidant response

    Mol. Carcinog.

    (2009)
  • K. Taguchi et al.

    Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution

    Genes Cells

    (2011)
  • A. Kobayashi et al.

    Oxidative and electrophilic stresses activate Nrf2 through inhibition of ubiquitination activity of Keap1

    Mol. Cell. Biol.

    (2006)
  • G. Rachakonda et al.

    Covalent modification at Cys151 dissociates the electrophile sensor Keap1 from the ubiquitin ligase CUL3

    Chem. Res. Toxicol.

    (2008)
  • K. Itoh et al.

    Keap1 regulates both cytoplasmic–nuclear shuttling and degradation of Nrf2 in response to electrophiles

    Genes Cells

    (2003)
  • R. Venugopal et al.

    Nrf1 and Nrf2 positively and c-fos and Fra1 negatively regulate the human antioxidant response element-mediated expression of NAD(P)H:quinone oxidoreductase 1 gene

    Proc. Natl. Acad. Sci. USA

    (1996)
  • M. McMahon et al.

    The cap'n'collar basic leucine zipper transcription factor Nrf2 (NF-E2 p45-related factor 2) controls both constitutive and inducible expression of intestinal detoxification and glutathione biosynthetic enzymes

    Cancer Res.

    (2001)
  • A. Banning et al.

    The GI-GPx gene is a target for Nrf2

    Mol. Cell. Biol.

    (2005)
  • J. Zhang et al.

    Synergy between sulforaphane and selenium in the induction of thioredoxin reductase 1 requires both transcriptional and translational modulation

    Carcinogenesis

    (2003)
  • Y.J. Kim et al.

    Human prx1 gene is a target of Nrf2 and is up-regulated by hypoxia/reoxygenation: implication to tumor biology

    Cancer Res.

    (2007)
  • R.K. Thimmulappa et al.

    Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray

    Cancer Res.

    (2002)
  • V. Vollrath et al.

    Role of Nrf2 in the regulation of the Mrp2 (ABCC2) gene

    Biochem. J.

    (2006)
  • K. Okada et al.

    Ursodeoxycholic acid stimulates Nrf2-mediated hepatocellular transport, detoxification, and antioxidative stress systems in mice

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2008)
  • K.C. Wu et al.

    Effect of graded Nrf2 activation on phase-I and -II drug metabolizing enzymes and transporters in mouse liver

    PLoS One

    (2012)
  • J.D. Hayes et al.

    Cancer chemoprevention mechanisms mediated through the Keap1–Nrf2 pathway

    Antioxid. Redox Signaling

    (2010)
  • S. Magesh et al.

    Small molecule modulators of Keap1–Nrf2–ARE pathway as potential preventive and therapeutic agents

    Med. Res. Rev.

    (2012)
  • Cited by (171)

    View all citing articles on Scopus
    View full text