Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Feb 28.
Published in final edited form as: Toxicol Appl Pharmacol. 2009 Sep 2;244(1):66–76. doi: 10.1016/j.taap.2009.08.028

Targeting NRF2 Signaling for Cancer Chemoprevention

Mi-Kyoung Kwak 1, Thomas W Kensler 2
PMCID: PMC3584341  NIHMSID: NIHMS148331  PMID: 19732782

Abstract

Modulation of the metabolism and disposition of carcinogens through induction of cytoprotective enzymes is one of several promising strategies to prevent cancer. Chemopreventive efficacies of inducers such as dithiolethiones and sulforaphane have been extensively studied in animals as well as in humans. The KEAP1-NRF2 system is a key, but not unilateral, molecular target for these chemopreventive agents. The transcription factor NRF2 (NF-E2-related factor 2) is a master regulator of the expression of a subset of genes, which produce proteins responsible for the detoxication of electrophiles and reactive oxygen species as well as the removal or repair of some of their damage products. It is believed that chemopreventive enzyme inducers affect the interaction between KEAP1 and NRF2 through either mediating conformational changes of the KEAP1 protein or activating phosphorylation cascades targeting the KEAP1-NRF2 complex. These events in turn affect NRF2 stability and trafficking. Recent advances elucidating the underlying structural biology of KEAP1-NRF2 signaling and identification of the gene clusters under the transcriptional control of NRF2 are facilitating understanding of the potential pleiotropic effects of NRF2 activators and discovery of novel classes of potent chemopreventive agents such as the triterpenoids. Although there is, appropriately a concern regarding a deleterious role of the KEAP1-NRF2 system in cancer cell biology, especially as the pathway affects cell survival and drug resistance, the development and the use of NRF2 activators as chemopreventive agents still holds a great promise for protection of normal cells from a diversity of environmental stresses that contribute to the burden of cancer and other chronic, degenerative diseases.

Keywords: Chemoprevention, NRF2, KEAP1, phase 2 enzymes, antioxidant proteins, dithiolethiones, sulforaphane

Cancer chemoprevention and phase 2 detoxifying enzymes

Phase 2 response enzymes and cancer

Rates of morbidity and mortality from malignancies are only beginning to decline in the United States, while the epidemiologic transitions from acute infectious diseases to chronic diseases in developing countries presage a burgeoning burden of cancer globally in the next decades. Approaches that focus on prevention during early stages of cancer development and progression are essential components of strategies to reduce cancer incidence as well as reduce impacts on health care costs. Cancer prevention is defined as the use of dietary or pharmacological interventions to prevent, block, or even reverse the process of carcinogenesis before clinical manifestation of the diseases termed cancer (Wattenberg, 1985; Hong and Sporn, 1997). Common features of human carcinogenesis amendable to preventive interventions include mutagenesis, oxidative stress, and inflammation (Sporn and Liby, 2005). Mutagenesis, which is an initiator of carcinogenesis, reflects DNA damage mediated by electrophilic insults arising from exogenous carcinogens as well endogenous processes that amplify the formation of and damage by reactive oxygen species (ROS). In particular, chronic inflammation is strongly associated with carcinogenesis from the perspective that elevated levels of ROS, reactive nitrogen species (RNS), cytokines, and growth factors, which are released from activated immune cells, can lead to DNA damage, cell proliferation, and cell invasiveness (Sporn and Roberts, 1986; Coussens and Werb, 2002). Based on these concepts, inhibition of these processes by increasing the expression of electrophile detoxifying enzymes and antioxidant proteins has long been postulated to be an effective way to prevent carcinogenesis (Prestera et al., 1993; Hong and Sporn, 1997; Kensler, 1997).

Many carcinogens undergo metabolic activation to generate electrophilic intermediates that can directly damage DNA by forming adducts. Phase 1 drug metabolizing enzymes often mediate the metabolic activation of carcinogens, while phase 2 enzymes facilitate elimination of carcinogens by making conjugates with hydrophilic molecules including glutathione (GSH) and glucuronic acid. Glutathione S-transferases (GSTs), NAD(P)H quinine oxidoreductases (NQOs), and UDP-glucuronosyl transferases (UGTs) are some examples of phase 2 enzymes. These co-regulated enzymes sometimes enhance the bioactivation of carcinogens, a feature that tempers considerations about how to manipulate them for chemoprevention. Nonetheless, the importance of these enzymes in enhancing resistance to carcinogenesis has been highlighted in several studies with mice lacking specific enzymes and in analyses of human polymorphisms. For instance, deletion of GSTP1/P2 in mice increased skin tumor incidence in a multistage carcinogenesis model with 7, 12-dimethylbenz[a]anthracene (DMBA) and 12-O-tetradecanoylphorbol-13-acetate (TPA) (Henderson et al., 1998). Similarly, GSTP1-null mice developed profoundly increased numbers of lung cancer following exposure to tobacco-related carcinogen benzo[a]pyrene (B[a]P) (Ritchie et al., 2007). Consistent with these results from animals, several human studies confirmed the important role of GST enzymes in carcinogenesis. Meta-analysis of GST polymorphisms and human cancers concluded that GSTM1 and GSTT1 null genotypes are significantly associated with increased risk of acute lymphoblastic leukemia (Ye and Song, 2005). Other reports also found positive associations between both risk of prostate cancer and GSTM1 polymorphism (Mo et al., 2009), and risk of lung cancer in Asians and GSTT1 deletion (Raimondi et al., 2006). Silencing of GSTP expression by CpG island hypermethylation is a hallmark of early prostate carcinogenesis (Nakayama et al., 2004).

NQO1 is a key enzyme that protects against quinone-derived reactive intermediates and maintains cellular pool of antioxidants such as tocopherol (Nioi and Hayes, 2004). Targeted disruption of the NQO1 gene sensitized mice to B[a]P-induced skin tumorigenesis and increased susceptibility to benzene-induced toxicity (Long et al., 2000; Iskander and Jaiswal, 2005). Furthermore, an investigation of a benzene-exposed population found that individuals with the NQO1*2 allele (P187S), which has a negligible NQO1 activity, showed a 7-fold greater risk of bone marrow toxicity (Nebert et al., 2002). Leukemia patients with MLL translocation showed a significant positive association with heterozygosity at NQO1 C609T (Smith et al., 2002). These experimental and epidemiological lines of evidence indicate that phase 2 enzymes play an important role in susceptibility to carcinogenesis in both animals and humans.

Cancer chemopreventive agents and induction of phase 2 enzymes

Multiple strategies to impede mutagenesis, oxidative stress and inflammation have been evaluated in preclinical and clinical studies. They include administration of inducers of phase 2 enzymes to reduce genotoxicity, antioxidants to scavenge ROS and inhibitors of cyclooxygenase-2 (COX-2) to attenuate inflammation (Surh, 2003; Sporn and Liby, 2005). An uneven balance of benefits and risks has been seen with each approach. COX-2 inhibitors such as celebrex profoundly reduce the burden of adenomas in the colon, but appreciably increase the risk of cardiovascular events (Ulrich et al., 2006). The clinical trials using simple ROS scavengers such as vitamin C and ß-carotene as chemopreventive agents have been unsuccessful, and in some cases exacerbated cancer risk, perhaps due to these “anti”-oxidants facilitating a “pro”-oxidant state (Omenn et al., 1996; Greenwald et al., 2002). These strategies have sought to impede the formation or flux of signaling molecules. Although contrary to the current pharmacologic precept of precise molecular targeting, activation of the phase 2 enzyme response may provide a broad mechanistic basis for cancer chemoprevention that impacts on multiple components of the carcinogenic process including mutagenesis, oxidative stress and inflammation. Clearly, cancer prevention can be achieved through the induction of phase 2 enzymes by the use of naturally occurring or synthetic agents. Sulfurcontaining molecules such as the isothiocyanate sulforaphane (SFN) and synthetic dithiolethiones such as oltipraz and 1,2-dithiole-3-thione have been studied extensively as representative cancer chemopreventive agents. SFN was first isolated from broccoli by monitoring bioassay-guided induction of NQO1 in murine hepatoma cells (Prochaska et al., 1992; Zhang et al., 1992). Various experimental animal studies have demonstrated that cruciferous-derived isothiocyanates including SFN, inhibited lung carcinogenesis by tobacco carcinogens (Sticha et al., 2002; Conaway et al., 2005), and reduced formation of colonic aberrant crypt foci and pancreatic carcinogenesis following carcinogen challenges (Chung et al., 2000; Kuroiwa et al., 2006). In humans, consumption of broccoli and cruciferous vegetables has been correlated with the reduction of cancer risk in colon, lung, breast, and prostate (Spitz et al., 2000; Seow et al., 2002; Ambrosone et al., 2004; Joseph et al., 2004). Lung cancer risk is reduced especially by dietary intake of isothiocyanates or cruciferous vegetables in persons with genotypes of GSTM1-null and GSTT1-null, highlighting an important interplay between genetic susceptibility factors and chemopreventive agents (Spitz et al., 2000; Wang et al., 2004).

Dithiolethiones are a large class of organosulfur compounds, of which the parent compound of this class, 3H-1,2-dithiole-3-thione (D3T) was first synthesized in 1948 (Zhang and Munday, 2008). Among these dithiolethiones, 4-methyl-5-pyrazinyl-3H-1,2-dithiole-3-thione (oltipraz) was developed for the treatment of schistosomiasis and 5-(4-methyoxyphenyl)-3H-1,2-dithiole-3-thione (ADT) was used to stimulate salivation (Bella et al., 1982; Epstein et al., 1983). Since the first demonstration that administration of oltipraz and ADT to mice protected against the hepatotoxicities of acetaminophen and carbon tetrachloride with a concomitant increase in levels for GSH and GST (Ansher et al., 1983), protective effects of dithiolethiones have been observed in multiple animal studies (Kensler et al., 1985; Wattenberg and Bueding, 1986; Kensler et al., 1999). For instance, oltipraz reduced the number of both pulmonary adenoma and forestomach tumors following B[a]P exposure (Wattenberg and Bueding, 1986). Other studies consistently showed that oltipraz inhibit carcinogenesis in a variety of organs in rodents, including colon, kidney, liver, stomach, and bladder, which were induced by divergent chemical carcinogens (Rao et al., 1991; Roebuck et al., 1991; Rao et al., 1993; Moon et al., 1994; Clapper et al., 1995; Rao et al., 1996; Nishikawa et al., 1998; Iida et al., 2004; Sharma et al., 2006; Zhang and Munday, 2008). These promising preclinical efficacies of dithiolethiones led to Phase I and II clinical trials in humans, however the results were not incontrovertible. For instance, in a randomized, double-blind Phase IIa trial in China, where dietary aflatoxin exposure is high, one month weekly administration of 500 mg oltipraz reduced levels of a phase 1 hydroxylated metabolite of aflatoxin excreted in urine, while daily dosing of 125 mg oltipraz significantly increased excreted levels of the phase 2 metabolite aflatoxin-mercapturic acid (Kensler et al., 1998; Wang et al., 1999). However, no effects were seen in this intervention on biomarkers of oxidative stress (Glintborg et al., 2006) or genotoxicity (Camoirano et al., 2001). Another randomized, double-blind trial in smokers did not show significant differences among trial groups with the measurement of polyaromatic hydrocarbon-DNA adduct levels in lung epithelial cells and blood (Kelley et al., 2005). Bothersome side effects including flatulence, gastrointestinal irritation and paresthesia in fingertips precluded the continued evaluation of this agent. Another dithiolethione, ADT, was shown to decrease the progression of preexisting dysplastic lesions of smokers with daily dosing of 25 mg in a Phase IIb trial. Only minor and tolerable gastrointestinal effects were reported, implying a safer profile than oltipraz (Lam et al., 2002).

Transcription factor NRF2 as a target of chemopreventive enzyme inducers

NRF2 regulates ARE-containing gene expression

Given the profound efficacy of phase 2 inducers as anticarcinogens in animals and indications of pharmacodynamic action in humans, a focused effort has been made to identify the molecular mechanism underlying enzyme induction by these agents. It is now accepted that the transcription factor NRF2 (NF-E2-related factor 2) is a critical element in transactivating phase 2 enzyme expression through the cis-acting element termed the antioxidant response element (ARE) (Kwak et al., 2004b; Kobayashi and Yamamoto, 2005; Cho et al., 2006). One or more AREs (A/G)TGA(C/T)nnnGC(A/G) are found in the 5′-flanking regions of many NRF2 target genes, including the prototypic inducible genes GSTA1 and NQO1 (Friling et al., 1990; Rushmore and Pickett, 1990; Jaiswal, 1994; Nguyen et al., 1994; Wang and Williamson, 1994; Xie et al., 1995; Wasserman and Fahl, 1997; Moinova and Mulcahy, 1998). Later, homology of the ARE to the TRE-like MAF recognition element (T-MARE, TGCTGAGTCAGCA), which had been identified as a binding site of small MAF proteins and the bZIP CNC family transcription factors, raised the question as to whether these factors can mediate the regulation of the ARE as well (Itoh et al., 1995; Venugopal and Jaiswal, 1996). Among members of the bZIP CNC family, NF-E2 (nuclear factor-erythroid 2) p45, NRF1, NRF2, NRF3, and BACHs, the crucial role of NRF2 in ARE regulation was firmly proven by a study with NRF2-disrupted mice. Yamamoto and colleagues observed that, following treatment with t-butylhydroxy anisole (BHA), inducible expression of GSTs and NQO1 was completely abrogated in the absence of NRF2 (Itoh et al., 1997). Constitutive expression was partially attenuated indicating other factors influence basal levels of expression. Since this seminal finding, the use of NRF2-null mice has been an invaluable tool for probing the molecular mechanisms of action of cancer chemopreventive agents, as well as the underlying adaptive response system now known to protect against a myriad of environmental stresses.

Under quiescent conditions, NRF2 is anchored in the cytoplasm through binding to Kelch-like ECH-associated protein 1 (KEAP1), which in turn facilitates the ubiquitylation and subsequent proteolysis of NRF2 (Itoh et al., 1999; Zipper and Mulcahy, 2002; Zhang and Hannink, 2003; Cullinan et al., 2004; Tong et al., 2006). NRF2 has some highly conserved domains called NRF2-ECH homology (Neh) domains and among them, the Neh2 domain mediates the binding of NRF2 with KEAP1, while other domains such as Neh4 and Neh5 are known to mediate the transacriptional activity of NRF2 (Itoh et al., 1999; Kobayashi et al., 2002; McMahon et al., 2004; Katoh et al., 2005; Nioi et al., 2005). KEAP1 was first identified in a yeast two hybrid analysis using the Neh2 domain as bait (Itoh et al., 1999). KEAP1 serves as an adaptor protein between NRF2 and the Cullin3-based E3-ligase ubiquitylation complex, leading to ubiquitylation of NRF2 and subsequent degradation by the 26S proteasome (Cullinan et al., 2004; Kobayashi et al., 2004; Zhang et al., 2004). Cytoplasmic localization of KEAP1 protein is known to be determined by both the Kelch/double glycine repeat (DGR) domain, which mediates binding of KEAP1 to actin cytoskeleton, and the IVR domain, which contains a nuclear export signal (NES) (Li and Kong, 2009). Therefore, sequestration and further degradation of NRF2 in the cytoplasm are provided mechanisms for the repressive effects of KEAP1 on NRF2 function. Indeed, gene deletion of KEAP1 in mice resulted in constitutive overexpression of GSTs and NQO1 with a concomitant accumulation of NRF2 within the nucleus (Wakabayashi et al., 2003).

Various oxidizing conditions and treatment with cancer preventive agents can lead to the accumulation of NRF2 into the nucleus, where it can react with the AREs of many cytoprotective genes. A nuclear localization signal (NLS) in the NRF2 protein is known to facilitate its translocation from the cytoplasm into the nucleus (Jain et al., 2005; Li et al., 2006). Within the nucleus, NRF2 protein dimerizes with small MAF proteins to bind to the ARE (Itoh et al., 1997). It has been suggested that NRF2 can also form heterodimers with other bZIP transcription factors such as ATF4 for the binding to the ARE (Venugopal and Jaiswal, 1996; Venugopal and Jaiswal, 1998; He et al., 2001); however, findings from studies with MAF-null mice support an essential role of MAF proteins in ARE regulation. Although single knockouts of each small MAF gene, including MAFG, MAFK, and MAFF, did not affect inducibility of phase 2 genes, murine embryonic fibrobalsts from compound knockout mice with MAFG-/-∷MAFK-/-∷MAFF-/- genotype lost their inducibility for most ARE-containing genes and were much more susceptible to oxidative stress (Katsuoka et al., 2005; Blank, 2008). This result indicates that small MAF proteins are the major binding partners of NRF2 for binding to the ARE and subsequent transactivation.

NRF2 is a molecular target of chemopreventive dithiolethiones and SFN

At this point in time, numerous studies have demonstrated that cancer preventive phytochemicals and synthetic chemicals can act on the KEAP1-NRF2 system to enhance ARE-regulated gene expression (Surh, 2003; Kwak et al., 2004a; Yates and Kensler, 2007; Eggler et al., 2008; Tan and Spivack, 2009). As prime examples, induction of various protective genes by the dithiolethiones D3T and oltipraz as well as SFN was largely abrogated in NRF2-null mice (Kwak et al., 2001; Ramos-Gomez et al., 2001; Fahey et al., 2002). Moreover, the inhibitory effects of oltipraz and SFN on B[a]P-induced gastric tumor formation were completely lost in the absence of NRF2, indicating that the KEAP1-NRF2 system is a molecular target of enzyme inducing chemopreventive agents (Ramos-Gomez et al., 2001; Fahey et al., 2002). At the same time, this key pair of studies highlighted the importance of NRF2 in chemical carcinogenesis. Gastric neoplasia evoked by B[a]P administration was significantly increased in NRF2-null mice compared to wild-type mice (Ramos-Gomez et al., 2001; Fahey et al., 2002). Concordantly, higher levels of B[a]P-DNA adducts were formed in the forestomach mucosa of NRF2-null mice than in wild-type mice (Ramos-Gomez et al., 2003). DNA damage has also been reported to be elevated in NRF2-null mice exposed to diesel exhaust or aflatoxin B1 (Aoki et al., 2001; Kwak et al., 2004a).

Follow-up studies, listed in table 1, have extended this view of the central role of KEAP1-NRF2 signaling in carcinogenesis and the potential benefit of NRF2-targeting enzyme inducers. The incidence of urinary bladder carcinoma induced by N-nitrosobutyl(4-hydroxybutyl)amine (BBN) was significantly higher in NRF2-null mice than wild-type mice (Iida et al., 2004; Iida et al., 2007). The incidence and numbers of skin tumors per mouse following the exposure to DMBA-TPA were markedly increased in NRF2-null mice compared to wild-type mice (Xu et al., 2006). Moreover, the onset, incidence, and multiplicity of DMBA/TPA-induced skin papillomas was greatly enhanced in transgenic mice overexpressing a dominant-negative NRF2 mutant in the epidermis (auf dem Keller et al., 2006). In addition, in a model of inflammation-promoted colorectal cancer, loss of NRF2 facilitated carcinogenesis. Oral administration of dextran sulfate sodium (DSS) after an initiating dose of azoxymethane (AOM) significantly increased numbers of presumptive preneoplastic aberrant cryptic foci in NRF2-null mice, but not in wild-type (Osburn et al., 2007). A similar observation was reported by Khor et al (Khor et al., 2008) in which the incidence, multiplicity, and size of total tumors were enhanced by the loss of NRF2: in particular, those of adenomas were remarkably increased in NRF2-null mice in the AOM-DDS colon cancer model (Khor et al., 2008). As for hepatocarcinogenesis, administration of mutagenic heterocyclic compound 2-amino-3-methylimidazol[4,5-f]quinoline significantly increased the multiplicity and incidence of liver tumors in NRF2-null mice compared to wild-type mice (Kitamura et al., 2007). On the other hand, NRF2 has been shown to protect genomic DNA from spontaneous mutation in the lung (Aoki et al., 2007). In their study, transgenic mice with guanine phosphoribosyltransferase gene (GPT) gene were used to assess genotoxicity in vivo and obtained results indicate that the lung mutation frequencies of the GPT gene were much higher in NRF2-null mice compared to NRF2 heterozygous.

Table 1. NRF2-dependent susceptibility to chemical carcinogenesis.

Carcinogen Phenotype Ref
Benzo[a]pyrene (B[a]P) Increased number of gastric neoplasia in NRF2-null mice
Increased levels of B[a]P-DNA adduct in the forestomach of NRF2-null mice
Ramos-Gomez et al., 2001
Ramos-Gomez et al., 2003
Diesel exhaust Increased DNA adduct formation in the lung and severe epithelial hyperplasia and DNA oxidation adducts in the bronchial epidermis of NRF2-null mice Aoki et al., 2001
Aflatoxin B1 Increased levels of aflatoxin B1-DNA adduct in the liver of NRF2-null mice Kwak et al., 2004a
N-nitrosobutyl(4-hydroxybutyl)amine (BBN) Increased incidence of urinary bladder carcinoma and invasive carcinoma in NRF2-null mice Iida et al, 2004
7, 12-Dimethylbenz[a]anthracene (DMBA) and 12-O- tetradecanoylphorbol-13-acetate (TPA) Increased incidence and numbers of skin tumors in NRF2-null mice
Increased onset, incidence, and numbers of skin papilloma in transgenic mice with overexpression of dominant-negative NRF2 mutant
Xu et al., 2006
auf dem Keller et al., 2006
Azoxymethane (AOM) and dextran sulfate sodium (DSS) Increased numbers of aberrant cryptic foci in NRF2-null mice
Increased incidence, numbers, and size of colon tumors in NRF2-null mice
Osburn et al., 2007
Khor et al., 2008
2-Amino-3-methylimidazol[4,5-f]quinoline Increased incidence and numbers of hepatocellular adenoma and carcinoma in NRF2-null mice Kitamura et al., 2007
Guanine phosphoribosyltransferase (GPT) gene transgenic mice Increased spontaneous mutation of the lung GPT gene in NRF2-null mice Aoki et al., 2007

Activation of the KEAP1-NRF2 system by chemopreventive inducers

The impact of disruption of NRF2 on carcinogenesis and the broad efficacy of NRF2 activators as chemopreventive agents provide striking prospects for the utility of targeting this pathway in prevention. However, a number of critical questions remain; how do these agents activate the pathway? and what is the full range of consequences, both beneficial and harmful? It is becoming clear that activators of NRF2 signaling can facilitate the nuclear accumulation of NRF2 through different mechanisms, some of which entail direct sensing by KEAP1 while others may function through altering other cellular signaling modalities such as phosphorylation (Li and Kong, 2009). As KEAP1 is a cysteine-rich protein, for instance human KEAP1 contains 27 cysteines, thiol-modification of this protein has long been speculated as a primary sensing mechanism (Talalay et al., 1988; Dinkova-Kostova et al., 2002). Recent advances in understanding how the KEAP1 molecule interacts with NRF2 have provided some clarity to this process. NRF2 has two KEAP1-binding sites in the Neh2 domain, which are called ETGE motif (D/N-X-E-T/S-G-E) and DLG motif (L-X-X-Q-D-X-D-L-G), leading to binding with two molecules of KEAP1 (Kobayashi et al., 2002; McMahon et al., 2004). These two sites have different binding affinities to KEAP1: the binding affinity of ETGE (Ka=20 ×107 M−1) to KEAP1 is much stronger than that of DLG (Ka=0.1 ×107 M−1) (Tong et al., 2006; Tong et al., 2007). Based on this observation, a “hinge & latch” two binding sites model has been proposed. The “latch” binding of the DLG motif to KEAP1 is thought to be easily disrupted by conformational changes within the two interacting KEAP1 molecules. Following deletion of the DLG motif from NRF2, this mutant protein has a longer half-life within the cell, indicating that binding of DLG to KEAP1 is associated with Cul3-proteasome degradation. Direct thiol-modifications of KEAP1 protein are likely triggering destabilization and disruption of the “latch” binding and perhaps complete dissociation of NRF2 from KEAP1.

It has been of keen interest to identify the reactive cysteine residues in KEAP1 which are responsible for the sensing of chemopreventive agents or other activating signals. Several independent studies have demonstrated that direct modification of sulfhydryl groups of multiple KEAP1 cysteines can be mediated by oxidation, reduction, or alkylation. Among these cysteine residues, Cys151, 273, and 288 were known to be essential for regulating NRF2 (Yamamoto et al., 2008; Kobayashi et al., 2009; Li and Kong, 2009). Mutation of either Cys273 or Cys288 residues of KEAP1 could not repress NRF2 activity and, therefore, NRF2 accumulated within the cell, indicating these residues are required for KEAP1-dependent ubiquitylation of NRF2 (Zhang and Hannink, 2003; Tong et al., 2007). Another study has demonstrated that D3T or SFN led to a formation of intermolecular disulfide bonds between Cys273 of one KEAP1 and Cys288 of the other KEAP1 molecule, which can mediate conformational changes of KEAP1 dimers (Wakabayashi et al., 2004). A recent independent study has confirmed that the Cys 273 and Cys 288 amino acids are essential for degradation of NRF2, whereas, these residues did not modulate the association or dissociation of NRF2 and KEAP1 (Kobayashi et al., 2006). In addition to Cys273 and Cys288, Cys 151 was reported to be required for inhibition of KEAP1-dependent degradation of NRF2 by SFN and t-butylhydroxy toluene (tBHQ) (Zhang and Hannink, 2003; Eggler et al., 2005; Eggler et al., 2007). A recent study by Kobayashi et al (Kobayashi et al., 2009), has classified eleven NRF2 activators into six classes based on sensor requirement using zebrafish as an elegant experimental system. They could observe that SFN, D3T, and GSH-depleting diethylmaleate (DEM) require Cys151 of KEAP1 to activate NRF2 signaling, whereas tBHQ and 15-deoxy-Δ12,14-prostaglandin J2 target Cys273 of zebrafish KEAP1. Holland et al. (Holland et al., 2008) have recently observed that H2O2, which may be the actual signaling mediator for inducers such as oltipraz, modify multiple cysteines in KEAP1. Cysteines most sensitive to S-glutathionylation include Cys77, Cys297, Cys319, Cys368, and Cys434, while cysteine disulfides most readily formed are Cys23-Cys38 and Cys257-Cys297. Collectively, these reports are suggesting that multiple cysteine residues of KEAP1 collectively contribute to transduce NRF2 signaling through conformational changes of KEAP1. High resolution X-ray crystallography or other techniques should define the juxtaposition of the reactive cysteines to the NRF2 binding domains within KEAP1.

On the other hand, numerous studies have demonstrated that activation of NRF2 signaling involves phosphorylation by multiple cellular kinase pathways: mitogen-activated protein kinases (MAPKs), protein kinase C (PKC), and phosphatidylinositol 3-kinase (PI3K) (Jeong et al., 2006; Surh et al., 2008). Although phosphorylation of NRF2 has been demonstrated in several studies (Huang et al., 2002; Bloom and Jaiswal, 2003; Cullinan et al., 2003), further studies will be required to explain how these signaling cascades, which participate in a broad range of cell signaling pathways, are specifically associated with the KEAP1-NRF2 adaptive system in response to stresses as well as chemopreventive agents. For instance, inhibition of p38 MAPK was found to contribute to induction of ARE-containing genes by SFN in human hepatoma cells (Keum et al., 2006). Activation of extracellular signal-regulated kinase (ERK) activation was associated with NRF2 activation by D3T in murine keratinocytes; however ERK signaling was not involved in D3T-mediated NRF2 activation in murine hepatoma cells (Manandhar et al., 2007). These reports imply that the different cell types may utilize distinct signaling pathways to activate the NRF2 system in response to chemopreventive agents.

Genes regulated by the chemopreventive agent-NRF2 pathway

NRF2-dependent, chemopreventive inducible genes

Given that NRF2 signaling exerts a wide spectrum of protection against divergent stresses, identification of key NRF2-regulated genes can define target genes for achieving cancer prevention. For this purpose, comparative analyses of global gene expression changes in wild-type and NRF2-null mice treated with chemopreventive agents have been performed by several laboratories as summarized in table 2. This approach has been used to indentify functional gene clusters, which are regulated by chemopreventive inducers through the NRF2 signaling. Many of these clusters are found to be common between different treatment agents. As a first case, the dithiolethione D3T was administered to wild-type and NRF2-null mice and global gene expression patterns were analyzed in the liver (Kwak et al., 2003). In this analysis, 300 genes were screened as D3T-inducible genes in wild-type mice, while 77% of these genes were not increased in the absence of NRF2, indicating most of D3T-inducible gene expression is mediated by NRF2. These genes could be classified into several major categories: xenobiotic-metabolizing enzymes, antioxidants, general enzymes, molecular chasperone-26S proteasome, and genes associated signal transmission. Several enzymes belonging to cytochrome P450s, a number of phase 2 metabolizing enzymes, and atypical enzymes such as carbonyl reductase were elevated 24 h after D3T administration. In particular, it was of interest that 24 subunits of the 26S proteasome were coordinately increased by D3T in the liver and 80% of these inducible proteasome subunits were only seen in wild-type mice. A recent report by Tran et al (Tran et al., 2009), could confirm that many of these NRF2-dependent genes, particularly detoxifying and antioxidant proteins, are increased in rat liver following D3T treatment. Furthermore, in their gene array analysis of a pharmacological structure-activity relationship, 226 differentially expressed genes are common to D3T and two of its analogs: oltipraz and 5-tert-butyl-3H-1,2-dithiole-3-thione (TBD).

Table 2.

Studies evaluating NRF2-dependent gene expression by chemopreventive agents.

Chemical class Agent Animal/ tissue Dosing NRF2-dependently increased gene numbers Major gene clusters Ref
Dithiolethiones D3T Mouse/liver 6 and 24 h after a single administration (po) 106 and 231 genes were increased NRF2-dependently at 6 h and 24 h post treatment Xenobiotic-metabolizing enzymes (Phase 2 enzymes)/Antioxidant proteins/ Ubproteasome Kwak et al., 2003
D3T, oltipra z, TBD Rat/liver 24 h after three dosing 226 genes were increased in three treatment groups Xenobiotic metabolizing enzymes/GSH metabolism/Antioxidant proteins Tran et al., 2009

Isothiocyanates SFN Mouse/small intestine 24 h after 7 consecutive days (po) 50 genes were increased as NRF2-dependent genes Xenobiotic metabolizing enzymes/Antioxidant proteins/ GSH synthesis Thimmulappa et al., 2002
SFN Mouse/liver 3 and 12 h after a single administration 1427 and 2353 genes were increased NRF2-dependently at 3 h and 12 h post treatment Xenobiotic metabolizing enzymes/Antioxidants/ Ubproteasome/Metabolism/Stress response/Cell cycle-growth Hu et al., 2006

Phenolic Antioxidants tBHQ Mouse/primary cortical astrocytes Incubation with 50 μM tBHQ for 24 h 202 genes were increased in an NRF2-dependent manner Detoxification/Antioxidants/Reducing potential/Immune-inflammation Lee et al., 2003
BHA Mouse/small intestine Mouse/liver 3 h after a single dose of BHA (200 mg/kg) 1,490 genes were increased NRF2-dependently
493 genes were increased NRF2-dependently
Detoxificationenzymes/Biosynthesis/Metabolism/ Transcription factors/Transporters Nair et al., 2006

Triterpenoids CDDO-Im Mouse/liver 24 h after a single dose of CDDO-Im 225 genes were increased NRF2-dependently Xenobiotic-metabolizing enzymes (Phase 2 enzymes)/Antioxidant proteins/Transporters/Lipid-fatty acid metabolism/Carbohydrate metabolism Yates et al., 2006
Yates et al., 2009

As for identification of NRF2-dependent isothiocyanates-inducible genes, Thimmulappa et al (Thimmulappa et al., 2002), analyzed altered gene expression in the small intestine of wild-type and NRF2-null mice following treatment with SFN for seven consecutive days. They observed fifty genes enhanced by SFN in wild-type mice and among these, 26 genes were elevated by SFN in an NRF2-dependent manner: most of genes are well-known detoxifying enzymes such as GSTs and NQO1, and enzymes associated with GSH and NADPH generation. A study by Hu et al (Hu et al., 2006a), demonstrated that 2,300 genes, which were increased at 12 h after a single dose of SFN in an NRF2-dependent manner, can account for 70% of total SFN-inducible genes in wild-type mice. These gene products can be functionally categorized into xenobiotic metabolizing enzymes, antioxidant proteins, stress response proteins, transporters, ubiquitin-26S proteasome, growth arrest-related proteins, and transcription factors. This group also used phenethyl isothiocyanate (PEITC), another isothiocyanate found in cruciferous vegetables, to identify NRF2-inducible genes and very similar results were obtained (Hu et al., 2006b).

The phenolic antioxidants BHA and tBHQ have long been of interest as effective, albeit not potent phase 2 enzyme inducers. As the protective effect of tBHQ against hydrogen peroxide (H2O2) was only observed in wild-type astrocytes, but not in NRF2-deficient cells, Lee et al (Lee et al., 2003), have hypothesized that NRF2-dependent genes are responsible. They analyzed differential gene expressions in tBHQ-treated cortical astrocytes from wild-type and NRF2-null mice and observed that 98% of tBHQ-inducible genes in wild-type cells are not altered in NRF2-null astrocytes. Similarly, these gene clusters include detoxifying enzymes, antioxidant proteins, NADPH-generating enzymes, and anti-inflammatory proteins, confirming the essential role of NRF2-regulated gene clusters in the cellular defense system. Similar gene clusters were also identified as NRF2-dependent BHA-inducible genes from a study by Nair et al (Nair et al., 2006), wherein they analyzed differential gene expression in the liver and small intestine at 3 h after a single dose of BHA. Although, expression of NRF2-target genes were more profoundly increased in small intestine than liver at this time point, detoxification enzymes such as GSTs, proteins related to GSH biosynthesis and metabolism, and transcription factors were elevated in both tissues in an NRF2-dependent manner.

Coupling these comprehensive studies of global gene expression analyses together with animal carcinogenesis studies, now it appears to be clear that NRF2-target genes, which can be increased in response to enzyme inducers such as dithiolethiones and SFN, are primarily responsible for their chemopreventive efficacy. This notion can be expanded into other types of known chemopreventive agents that have not been recognized as enzyme inducers. For instance, the chemopreventive efficacy of curcumin, a naturally occurring flavonoid from the spice turmeric, has been explained by regulation of multiple signaling pathways associated with cancer cell proliferation, apoptosis, and inflammation: NFκB, MAPK pathway, and epidermal growth factor receptors (Aggarwal and Shishodia, 2006; Khan et al., 2008). However, several recent studies raised a potential role of NRF2-target gene expression in the mechanism of action of curcumin (Balogun et al., 2003; Lee and Surh, 2005), and indeed, expression of multiple NRF2-regulated genes were altered following curcumin treatment in livers and small intestines: 660 genes encoding detoxifying enzymes, transporters, ubiquitin-proteasome, and multiple transcription factors were only increased in wild-type mice, but not in NRF2-null mice (Shen et al., 2006).

Another example is a series of synthetic oleanane triterpenoids. Initially, a strong inhibitory effect on an inflammatory response was postulated as a mechanism of action of these chemicals. However, the demonstration of a potent capacity to activate NRF2 has led to the application of triterpenoids into chemoprevention against chemical carcinogenesis (Dinkova-Kostova et al., 2005; Liby et al., 2005; Yates et al., 2006; Yates et al., 2007). In fact, a synthetic triterpenoid analogue 1-[2-cyano-3-,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im) effectively inhibited aflatoxin B1-induced preneoplastic lesion formation in rats at doses of μmol/kg body weight (Yates et al., 2006). Interestingly, the anti-inflammatory effect of triterpenoids is strongly correlated with the potency of NRF2 activation, indicating that common molecular mechanism might involve in NRF2 activation and anti-inflammatory effect (Dinkova-Kostova et al., 2005). These examples support that the KEAP1-NRF2 system can be a promising target for developing novel cancer preventive agents through up-regulation of an adaptive cell survival pathway.

Genes regulated by genetic activation of NRF2

Wakabayashi et al., developed KEAP1-null mice and observed that these mice died within 3 weeks because of hyperkeratocytosis in the forestomach and esophagus. This phenotype was rescued in KEAP1∷NRF2 double knockout mice (Wakabayashi et al., 2003). To circumvent this post-natal lethality Okawa et al (Okawa et al., 2006) created hepatocyte-specific KEAP1-disrupted mice. They exhibit a normal phenotype and express high levels of prototypic NRF2-regulated genes including GSTs and NQO1 in their livers. As expected, these mutant mice are considerably more resistant to the acute hepatotoxicity of acetaminophen (Okawa et al., 2006) as well as an immune hepatitis induced by conconavalin A (Osburn et al., 2008). Gene expression changes have been characterized in the livers of these KEAP1-disrupted mice. Recently, Yates et al (Yates et al., 2009) compared the patterns and magnitude of response between genetic activation of the pathway to that afforded by activation by a very potent pharmacologic activator of NRF2 signaling, CDDO-Im. Both means of activating the pathway yielded similar patterns of altered gene expressions. However, the magnitude of gene expression changes was substantially higher in the genetic model than the pharmacologic one. In addition, beyond looking a relative expression level of individual genes, it was clear that additional genes within functional classes were elevated in the genetic as opposed to the pharmacologic model. Clearly, genetic disruption of KEAP1 provides a more vigorous activation of NRF2 target genes and, of course, more sustained duration of NRF2 targeted responses.

Dysregulation of NRF2 in cancer and chemopreventive agents

Constitutive activation of NRF2 in cancer

As recently reviewed by Hayes and McMahon (Hayes and McMahon, 2009), evidence is accumulating for the frequent mutation of KEAP1 and NRF2 in human cancers. Such mutations lead to constitutive expression of pro-survival cytoprotective genes. While perhaps providing intrinsic growth advantages, hyperactivation of the pathway also contributes to chemoresistance during therapy. Initially, Padmanabhan et al (Padmanabhan et al., 2006) have identified mutations of KEAP1 in the DGR domain of KEAP1, which involving glycine to cysteine substitution, in tissues or cell lines derived from lung cancer patients. Because of the reduced affinity to NRF2, these mutant KEAP1 proteins could not repress NRF2 activity and consequently, NRF2 is constitutively activated in these cancer cells. Similarly, multiple somatic mutations have been identified in the Kelch or intervening region domain of the KEAP1 protein in lung cancer cell lines and non-small-cell lung cancer samples at a frequency of 50% and 19% respectively (Singh et al., 2006). Decreased KEAP1 activity in these cancer cells induced greater nuclear accumulation of NRF2 and constitutive overexpression of ARE-containing genes including drug efflux pumps, which facilitates resistance of tumor cells to chemotherapy. Furthermore, KEAP1 mutation (C23Y) found in tumors from breast cancer patients has been associated with impaired ubiquitylation of NRF2 (Nioi and Nguyen, 2007) and recurrent KEAP1 gene alterations were observed in gallbladder cancer with a frequency of 30% (Shibata et al., 2008a).

The causal link of aberrant activation of NRF2 to tumor growth and resistance can be further supported by additional findings showing NRF2 somatic mutations and epigenetic change of the KEAP1 gene in tumors. Shibata et al (Shibata et al., 2008b) identified NRF2 somatic mutations in 11 of 103 patients with primary lung cancers and in 3 of 11 patients with primary head and neck tumors. All of these mutations led to missense amino acid substitutions and are found in the DLG and the ETGE motifs of NRF2. As described earlier, these motifs are responsible for the binding to KEAP1, therefore mutations in this region impair the two-site substrate recognition of KEAP1 and mediate constitutive induction of cytoprotective genes and drug efflux pumps. Wang et al (Wang et al., 2008) showed that KEAP1 expression was down-regulated in lung cancer cell lines and tissues compared to a normal bronchial epithelial cell line and treatment with the methylation inhibitor 5′-aza-2′-deoxycytidine restored KEAP1 mRNA levels in A549, H460 and SPC-A1 lung cancer cell lines. They could identify that the CpG island of the KEAP1 promoter (-291 to 337) was highly methylated in lung cancer cells and tissues, but not in normal cells, indicating that epigenetic regulation of KEAP1 might contribute to tumorigenesis.

It is apparent that an aberrant continuous activation of NRF2 in premalignant cells can promote cancer cell survival in response to an oxidizing tumor environment, which can be encountered by metabolic activation, mitochondrial dysfunction and activation of oncogenic signals such as Ras in cancer cells, as well as treatment with anticancer agents. Indeed, it has been noted that patients with lung tumors containing mutant KEAP1 or NRF2 showed a poorer prognosis than patients with non-mutant tumors (Shibata et al., 2008b). Therefore, in tumors, inhibition of NRF2 can be expected to repress tumor cell proliferation and enhance apoptosis. Several reports have demonstrated that administration of NRF2-specific siRNA into cancer cells could decrease the growth rate of cells and enhanced sensitivity to chemotherapeutic agents such as platinum-based anticancer agents, 5-fluorouracil, and topoisomerase inhibitors in cancer cells from lung, gallbladder, and ovariantumors (Cho et al., 2008; Ohta et al., 2008; Shibata et al., 2008a; Shibata et al., 2008b). Consistently, the intratumoral injection of NRF2 shRNA significantly suppressed tumor growth rate in xenograft model of mice with lung cancer cell lines A549 and H460 (Singh et al., 2008). At this time point, it can be hypothesized that growth advantage of NRF2 overexpression in cancer cells can be mediated by the increase in general NRF2-target antioxidant proteins, which can counteract oxidative stress; however several recent findings raised a possibility that specific cell signaling pathways can be governed by NRF2. For instance, it has been shown that a knockdown of NRF2 in lung cancer cells reduced phosphorylated retinoblastoma (pRb) protein level, which in turn led to a cell-cycle arrest at G1 phase (Homma et al., 2009). In the study by Beyer et al., after partial hepatectomy, liver regeneration was significantly delayed in NRF2-null mice and a defect in insulin/insulin-like growth factor-1 (IGF-1) signaling has been identified to be a cause of enhanced cell death and repressed proliferation of hepatocytes (Beyer et al., 2008). These reports are unraveling a novel role of NRF2 in cell proliferation and growth, which can account for a positive correlation of NRF2 overexpression and tumor growth. Furthermore, it can be speculated that activation of the KEAP1-NRF2 contributes to the development of acquired resistance to chemotherapy. In ovarian cancer cells, acquired resistance to doxorubicin was associated with increased NRF2 signaling and a subsequent increase in the GSH pool (Shim et al., 2009). Activation of the NRF2-ARE pathway has been also observed in breast cancer cells, which acquired resistance to tamoxifen following a prolonged incubation (Kim et al., 2008).

Chemoprevention versus tumor growth: a tipping point?

These findings raise an intriguing and critical question regarding the impact of enzyme inducing chemopreventive agents on premalignant cancer cells. To what extent might unabated exposure to enzyme inducers enhance a cancer phenotype? A partial answer can be developed by considering the similarities and differences in the gene expression patterns, amplitudes and durations of response between pharmacologic and genetic (e.g., mutation, deletion) modes of activating the pathway. The recent study by Yates et al (Yates et al., 2009) comparing the expression levels and patterns in hepatocyte-specific KEAP1 knockout mice to those imparted in the liver by the potent NRF2 activator, CDDO-Im, provides some insight. The overall pathways influenced by either pharmacologic or genetic activation of NRF2 signaling appear quite similar, although the magnitudes of gene expression changes in the genetic model are substantially higher. Not surprisingly, a pharmacological challenge in the genetic model does not result in any significant increase in expression of NRF2-regulated genes over that imparted by the disruption of KEAP1 itself. Not only does genetic disruption of the pathway impart a stronger signal, the kinetics of the response also quite distinct from typical pharmacological activation. With genetic disruption, the signal is persistent in the absence of any corrective gene therapy intervention. By contrast, pharmacological interventions cause transient fluctuations in the expression of NRF2 target genes. The pharmacokinetic half-lives of most inducers are measured in hours and the half-lives of most of the induced proteins measured in hours to days. As a result, intermittent dosings with chemopreventive agents have been shown to be sufficient to elevate response genes and to achieve chemoprevention in the face of chronic exposures to carcinogens. Considering magnitude and duration of responses together, the relative “areas under the curve” for the pharmacodynamic responses to pathway activation are substantially smaller than for genetic activation

Of course, the pharmacological agents are not necessarily specific activators of NRF2 signaling. Given the general propensity for these chemopreventive agents to react with cysteines in their targets, multiple pathways can conceivably be modulated. However, dose of the chemopreventive agent appears to go a long way to defining which pathways will be activated. There appears to be a cysteine code exhibiting differential reactivity towards the inducers that go a long way in defining the type of response that results. For chemopreventive agents such as CDDO-Im, sulforaphane and dithiolethione, it is clear that multiple pathways can be affected at high, but nonetheless non-cytotoxic doses. However, it is also possible to define low doses where activation of NRF2 is the primary response. Under these low-dose conditions, upwards of 80% of the altered gene expression seen in vivo (by comparing treated wild-type and NRF2 knockout mice) is mediated through the NRF2 pathway. There are very limited “off-target” effects under these conditions.

Do NRF2 activators enhance tumor growth? As a number of NRF2 activators either are or have been evaluated for efficacy in humans (e.g., dithiolethiones, isothiocyanates and triterpenoids), there has been a substantial investment in characterizing their preclinical toxicology. There is no evidence for direct genotoxicity of these agents, thus they are unlikely to induce mutations in the pathway or elsewhere. While no carcinogenicity studies have been undertaken with any of these agents, several have been evaluated as modifiers of multistage carcinogenesis in animal models. No tumor promoting or enhancing effects have been observed. As examples, administration of oltipraz following treatment of rats with multiple doses of aflatoxin B1 has no effect on hepatic tumor yield or burden (Maxuitenko et al., 1993). A similar outcome is seen with triterpenoids (M.S. Yates, T.W. Kensler, and B.D. Roebuck, unpublished observations). Post-initiation treatment with CDDO-Me of mice challenged with the pulmonary carcinogen vinyl carbamate led to decreased tumor burden (Liby et al., 2007). Six months of feeding CDDO-Im to mice chronically exposed to cigarette smoke led to substantial protection against the development of emphysema; no effects were observed on sham-exposed mice fed the triterpenoid (Sussan et al., 2009). Thus, there is no evidence to date to suggest that the agents used to date to activate the NRF2 pathway have adverse impacts on tumor growth. That genetic disruption of pathway components can profoundly enhance or impede tumor development is well established. Thus, pharmacological (or food-based) interventions in healthy, but at-risk populations offer the prospect of a very favorable benefit-risk outcome. By contrast, their inadvertent use in individuals harboring mutations in NRF2 or KEAP1 would provide no benefit, but perhaps more importantly, would not appear to impart any increased risk. While it appears there might not be a pharmacological tipping point at which either dose or schedule (duration) enhances risk of an adverse tumorigenic outcome, these interpretations need more rigorous experimental validation.

Acknowledgments

This work was supported by the Korea Science and Engineering Foundation grant (MOST, R01-2007-000-10890-0) funded by the Korea government (M.Kwak) and NIH grants CA39416 and CA94076 (T.W.Kensler).

Abbreviations

NRF2

NF-E2-related factor 2

KEAP1

Kelch-like ECH-associated protein 1

GSTs

Glutathione S-transferases

NQO1

NAD(P)H quinine oxidoreductases

UGTs

UDP-glucuronosyl transferases

ROS

reactive oxygen species

DMBA

7, 12-dimethylbenz[a]anthracene

TPA

12-O-tetradecanoylphorbol-13-acetate

B[a]p

benzo[a]pyrene

D3T

3H-1,2-dithiole-3-thione

oltipraz

4-methyl-5-pyrazinyl-3H-1,2-dithiole-3-thione

ADT

5-(4-methyoxyphenyl)-3H-1,2-dithiole-3-thione

TBD

5-tert-butyl-3H-1,2-dithiole-3-thione

tBHQ

t-butylhydroxy toluene

BHA

butylhydroxy anisole

SFN

sulforaphane

ARE

Antioxidant Response Element

NES

nuclear export signal

NLS

nuclear localization signal

AOM

azoxymethane

DSS

dextran sulfate sodium

MAPKs

mitogen-activated protein kinases

PKC

protein kinase C

PI3K

phosphatidylinositol 3-kinase

ERK

extracellular signal-regulated kinase

CDDO-Im

1-[2-cyano-3-,12 dioxooleana-19(11)-dien-28-oyl]imidazole

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Aggarwal BB, Shishodia S. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem Pharmacol. 2006;71:1397–1421. doi: 10.1016/j.bcp.2006.02.009. [DOI] [PubMed] [Google Scholar]
  2. Ambrosone CB, McCann SE, Freudenheim JL, Marshall JR, Zhang Y, Shields PG. Breast cancer risk in premenopausal women is inversely associated with consumption of broccoli, a source of isothiocyanates, but is not modified by GST genotype. J Nutr. 2004;134:1134–1138. doi: 10.1093/jn/134.5.1134. [DOI] [PubMed] [Google Scholar]
  3. Ansher SS, Dolan P, Bueding E. Chemoprotective effects of two dithiolthiones and of butylhydroxyanisole against carbon tetrachloride and acetaminophen toxicity. Hepatology. 1983;3:932–935. doi: 10.1002/hep.1840030608. [DOI] [PubMed] [Google Scholar]
  4. Aoki Y, Hashimoto AH, Amanuma K, Matsumoto M, Hiyoshi K, Takano H, Masumura K, Itoh K, Nohmi T, Yamamoto M. Enhanced spontaneous and benzo(a)pyrene-induced mutations in the lung of Nrf2-deficient gpt delta mice. Cancer Res. 2007;67:5643–5648. doi: 10.1158/0008-5472.CAN-06-3355. [DOI] [PubMed] [Google Scholar]
  5. Aoki Y, Sato H, Nishimura N, Takahashi S, Itoh K, Yamamoto M. Accelerated DNA adduct formation in the lung of the Nrf2 knockout mouse exposed to diesel exhaust. Toxicol Appl Pharmacol. 2001;173:154–160. doi: 10.1006/taap.2001.9176. [DOI] [PubMed] [Google Scholar]
  6. auf dem Keller U, Huber M, Beyer TA, Kumin A, Siemes C, Braun S, Bugnon P, Mitropoulos V, Johnson DA, Johnson JA, Hohl D, Werner S. Nrf transcription factors in keratinocytes are essential for skin tumor prevention but not for wound healing. Mol Cell Biol. 2006;26:3773–3784. doi: 10.1128/MCB.26.10.3773-3784.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Balogun E, Hoque M, Gong P, Killeen E, Green CJ, Foresti R, Alam J, Motterlini R. Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Biochem J. 2003;371:887–895. doi: 10.1042/BJ20021619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bella H, Rahim AG, Mustafa MD, Ahmed MA, Wasfi S, Bennett JL. Oltipraz--antischistosomal efficacy in Sudanese infected with Schistosoma mansoni. Am J Trop Med Hyg. 1982;31:775–778. doi: 10.4269/ajtmh.1982.31.775. [DOI] [PubMed] [Google Scholar]
  9. Beyer TA, Xu W, Teupser D, auf dem Keller U, Bugnon P, Hildt E, Thiery J, Kan YW, Werner S. Impaired liver regeneration in Nrf2 knockout mice: role of ROS-mediated insulin/IGF-1 resistance. Embo J. 2008;27:212–223. doi: 10.1038/sj.emboj.7601950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Blank V. Small Maf proteins in mammalian gene control: mere dimerization partners or dynamic transcriptional regulators? J Mol Biol. 2008;376:913–925. doi: 10.1016/j.jmb.2007.11.074. [DOI] [PubMed] [Google Scholar]
  11. Bloom DA, Jaiswal AK. Phosphorylation of Nrf2S40 by PKC in response to antioxidants leads to the release of Nrf2 from INrf2 but not required for Nrf2 stabilization/accumulation in the nucleus and transcriptional activation of ARE-mediated NQO1 gene expression. J Biol Chem. 2003;278:44675–44682. doi: 10.1074/jbc.M307633200. [DOI] [PubMed] [Google Scholar]
  12. Camoirano A, Bagnasco M, Bennicelli C, Cartiglia C, Wang JB, Zhang BC, Zhu YR, Qian GS, Egner PA, Jacobson LP, Kensler TW, De Flora S. Oltipraz chemoprevention trial in Qidong, People's Republic of China: results of urine genotoxicity assays as related to smoking habits. Cancer Epidemiol Biomarkers Prev. 2001;10:775–783. [PubMed] [Google Scholar]
  13. Cho HY, Reddy SP, Kleeberger SR. Nrf2 defends the lung from oxidative stress. Antioxid Redox Signal. 2006;8:76–87. doi: 10.1089/ars.2006.8.76. [DOI] [PubMed] [Google Scholar]
  14. Cho JM, Manandhar S, Lee HR, Park HM, Kwak MK. Role of the Nrf2-antioxidant system in cytotoxicity mediated by anticancer cisplatin: implication to cancer cell resistance. Cancer Lett. 2008;260:96–108. doi: 10.1016/j.canlet.2007.10.022. [DOI] [PubMed] [Google Scholar]
  15. Chung FL, Conaway CC, Rao CV, Reddy BS. Chemoprevention of colonic aberrant crypt foci in Fischer rats by sulforaphane and phenethyl isothiocyanate. Carcinogenesis. 2000;21:2287–2291. doi: 10.1093/carcin/21.12.2287. [DOI] [PubMed] [Google Scholar]
  16. Clapper ML, Wood M, Leahy K, Lang D, Miknyoczki S, Ruggeri BA. Chemopreventive activity of Oltipraz against N-nitrosobis(2-oxopropyl)amine (BOP)-induced ductal pancreatic carcinoma development and effects on survival of Syrian golden hamsters. Carcinogenesis. 1995;16:2159–2165. doi: 10.1093/carcin/16.9.2159. [DOI] [PubMed] [Google Scholar]
  17. Conaway CC, Wang CX, Pittman B, Yang YM, Schwartz JE, Tian D, McIntee EJ, Hecht SS, Chung FL. Phenethyl isothiocyanate and sulforaphane and their N-acetylcysteine conjugates inhibit malignant progression of lung adenomas induced by tobacco carcinogens in A/J mice. Cancer Res. 2005;65:8548–8557. doi: 10.1158/0008-5472.CAN-05-0237. [DOI] [PubMed] [Google Scholar]
  18. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cullinan SB, Gordan JD, Jin J, Harper JW, Diehl JA. 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;24:8477–8486. doi: 10.1128/MCB.24.19.8477-8486.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cullinan SB, Zhang D, Hannink M, Arvisais E, Kaufman RJ, Diehl JA. Nrf2 is a direct PERK substrate and effector of PERK-dependent cell survival. Mol Cell Biol. 2003;23:7198–7209. doi: 10.1128/MCB.23.20.7198-7209.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Dinkova-Kostova AT, Holtzclaw WD, Cole RN, Itoh K, Wakabayashi N, Katoh Y, Yamamoto M, Talalay P. Direct evidence that sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants. Proc Natl Acad Sci U S A. 2002;99:11908–11913. doi: 10.1073/pnas.172398899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Dinkova-Kostova AT, Liby KT, Stephenson KK, Holtzclaw WD, Gao X, Suh N, Williams C, Risingsong R, Honda T, Gribble GW, Sporn MB, Talalay P. Extremely potent triterpenoid inducers of the phase 2 response: correlations of protection against oxidant and inflammatory stress. Proc Natl Acad Sci U S A. 2005;102:4584–4589. doi: 10.1073/pnas.0500815102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Eggler AL, Gay KA, Mesecar AD. Molecular mechanisms of natural products in chemoprevention: induction of cytoprotective enzymes by Nrf2. Mol Nutr Food Res. 2008;52(l):S84–94. doi: 10.1002/mnfr.200700249. [DOI] [PubMed] [Google Scholar]
  24. Eggler AL, Liu G, Pezzuto JM, van Breemen RB, Mesecar AD. Modifying specific cysteines of the electrophile-sensing human Keap1 protein is insufficient to disrupt binding to the Nrf2 domain Neh2. Proc Natl Acad Sci U S A. 2005;102:10070–10075. doi: 10.1073/pnas.0502402102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Eggler AL, Luo Y, van Breemen RB, Mesecar AD. Identification of the highly reactive cysteine 151 in the chemopreventive agent-sensor Keap1 protein is method-dependent. Chem Res Toxicol. 2007;20:1878–1884. doi: 10.1021/tx700217c. [DOI] [PubMed] [Google Scholar]
  26. Epstein JB, Decoteau WE, Wilkinson A. Effect of Sialor in treatment of xerostomia in Sjogren's syndrome. Oral Surg Oral Med Oral Pathol. 1983;56:495–499. doi: 10.1016/0030-4220(83)90096-8. [DOI] [PubMed] [Google Scholar]
  27. Fahey JW, Haristoy X, Dolan PM, Kensler TW, Scholtus I, Stephenson KK, Talalay P, Lozniewski A. Sulforaphane inhibits extracellular, intracellular, and antibiotic-resistant strains of Helicobacter pylori and prevents benzo[a]pyrene-induced stomach tumors. Proc Natl Acad Sci U S A. 2002;99:7610–7615. doi: 10.1073/pnas.112203099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Friling RS, Bensimon A, Tichauer Y, Daniel V. Xenobiotic-inducible expression of murine glutathione S-transferase Ya subunit gene is controlled by an electrophile-responsive element. Proc Natl Acad Sci U S A. 1990;87:6258–6262. doi: 10.1073/pnas.87.16.6258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Glintborg B, Weimann A, Kensler TW, Poulsen HE. Oltipraz chemoprevention trial in Qidong, People's Republic of China: unaltered oxidative biomarkers. Free Radic Biol Med. 2006;41:1010–1014. doi: 10.1016/j.freeradbiomed.2006.06.015. [DOI] [PubMed] [Google Scholar]
  30. Greenwald P, McDonald SS, Anderson DE. An evidence-based approach to cancer prevention clinical trials. Eur J Cancer Prev. 2002;11(2):S43–47. [PubMed] [Google Scholar]
  31. Hayes JD, McMahon M. NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer. Trends Biochem Sci. 2009;34:176–188. doi: 10.1016/j.tibs.2008.12.008. [DOI] [PubMed] [Google Scholar]
  32. He CH, Gong P, Hu B, Stewart D, Choi ME, Choi AM, Alam J. Identification of activating transcription factor 4 (ATF4) as an Nrf2-interacting protein. Implication for heme oxygenase-1 gene regulation. J Biol Chem. 2001;276:20858–20865. doi: 10.1074/jbc.M101198200. [DOI] [PubMed] [Google Scholar]
  33. Henderson CJ, Smith AG, Ure J, Brown K, Bacon EJ, Wolf CR. Increased skin tumorigenesis in mice lacking pi class glutathione S-transferases. Proc Natl Acad Sci U S A. 1998;95:5275–5280. doi: 10.1073/pnas.95.9.5275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Holland R, Hawkins AE, Eggler AL, Mesecar AD, Fabris D, Fishbein JC. Prospective type 1 and type 2 disulfides of Keap1 protein. Chem Res Toxicol. 2008;21:2051–2060. doi: 10.1021/tx800226m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Homma S, Ishii Y, Morishima Y, Yamadori T, Matsuno Y, Haraguchi N, Kikuchi N, Satoh H, Sakamoto T, Hizawa N, Itoh K, Yamamoto M. Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer. Clin Cancer Res. 2009;15:3423–3432. doi: 10.1158/1078-0432.CCR-08-2822. [DOI] [PubMed] [Google Scholar]
  36. Hong WK, Sporn MB. Recent advances in chemoprevention of cancer. Science. 1997;278:1073–1077. doi: 10.1126/science.278.5340.1073. [DOI] [PubMed] [Google Scholar]
  37. Hu R, Xu C, Shen G, Jain MR, Khor TO, Gopalkrishnan A, Lin W, Reddy B, Chan JY, Kong AN. Gene expression profiles induced by cancer chemopreventive isothiocyanate sulforaphane in the liver of C57BL/6J mice and C57BL/6J/Nrf2 (-/-) mice. Cancer Lett. 2006a;243:170–192. doi: 10.1016/j.canlet.2005.11.050. [DOI] [PubMed] [Google Scholar]
  38. Hu R, Xu C, Shen G, Jain MR, Khor TO, Gopalkrishnan A, Lin W, Reddy B, Chan JY, Kong AN. Identification of Nrf2-regulated genes induced by chemopreventive isothiocyanate PEITC by oligonucleotide microarray. Life Sci. 2006b;79:1944–1955. doi: 10.1016/j.lfs.2006.06.019. [DOI] [PubMed] [Google Scholar]
  39. Huang HC, Nguyen T, Pickett CB. Phosphorylation of Nrf2 at Ser-40 by protein kinase C regulates antioxidant response element-mediated transcription. J Biol Chem. 2002;277:42769–42774. doi: 10.1074/jbc.M206911200. [DOI] [PubMed] [Google Scholar]
  40. Iida K, Itoh K, Kumagai Y, Oyasu R, Hattori K, Kawai K, Shimazui T, Akaza H, Yamamoto M. Nrf2 is essential for the chemopreventive efficacy of oltipraz against urinary bladder carcinogenesis. Cancer Res. 2004;64:6424–6431. doi: 10.1158/0008-5472.CAN-04-1906. [DOI] [PubMed] [Google Scholar]
  41. Iida K, Itoh K, Maher JM, Kumagai Y, Oyasu R, Mori Y, Shimazui T, Akaza H, Yamamoto M. Nrf2 and p53 cooperatively protect against BBN-induced urinary bladder carcinogenesis. Carcinogenesis. 2007;28:2398–2403. doi: 10.1093/carcin/bgm146. [DOI] [PubMed] [Google Scholar]
  42. Iskander K, Jaiswal AK. Quinone oxidoreductases in protection against myelogenous hyperplasia and benzene toxicity. Chem Biol Interact. 2005;153-154:147–157. doi: 10.1016/j.cbi.2005.03.019. [DOI] [PubMed] [Google Scholar]
  43. Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, Yamamoto M, Nabeshima Y. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 1997;236:313–322. doi: 10.1006/bbrc.1997.6943. [DOI] [PubMed] [Google Scholar]
  44. Itoh K, Igarashi K, Hayashi N, Nishizawa M, Yamamoto M. Cloning and characterization of a novel erythroid cell-derived CNC family transcription factor heterodimerizing with the small Maf family proteins. Mol Cell Biol. 1995;15:4184–4193. doi: 10.1128/mcb.15.8.4184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, Yamamoto M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999;13:76–86. doi: 10.1101/gad.13.1.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Jain AK, Bloom DA, Jaiswal AK. Nuclear import and export signals in control of Nrf2. J Biol Chem. 2005;280:29158–29168. doi: 10.1074/jbc.M502083200. [DOI] [PubMed] [Google Scholar]
  47. Jaiswal AK. Antioxidant response element. Biochem Pharmacol. 1994;48:439–444. doi: 10.1016/0006-2952(94)90272-0. [DOI] [PubMed] [Google Scholar]
  48. Jeong WS, Jun M, Kong AN. Nrf2: a potential molecular target for cancer chemoprevention by natural compounds. Antioxid Redox Signal. 2006;8:99–106. doi: 10.1089/ars.2006.8.99. [DOI] [PubMed] [Google Scholar]
  49. Joseph MA, Moysich KB, Freudenheim JL, Shields PG, Bowman ED, Zhang Y, Marshall JR, Ambrosone CB. Cruciferous vegetables, genetic polymorphisms in glutathione S-transferases M1 and T1, and prostate cancer risk. Nutr Cancer. 2004;50:206–213. doi: 10.1207/s15327914nc5002_11. [DOI] [PubMed] [Google Scholar]
  50. Katoh Y, Iida K, Kang MI, Kobayashi A, Mizukami M, Tong KI, McMahon M, Hayes JD, Itoh K, Yamamoto M. Evolutionary conserved N-terminal domain of Nrf2 is essential for the Keap1-mediated degradation of the protein by proteasome. Arch Biochem Biophys. 2005;433:342–350. doi: 10.1016/j.abb.2004.10.012. [DOI] [PubMed] [Google Scholar]
  51. Katsuoka F, Motohashi H, Ishii T, Aburatani H, Engel JD, Yamamoto M. Genetic evidence that small maf proteins are essential for the activation of antioxidant response element-dependent genes. Mol Cell Biol. 2005;25:8044–8051. doi: 10.1128/MCB.25.18.8044-8051.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Kelley MJ, Glaser EM, Herndon JE, 2nd, Becker F, Bhagat R, Zhang YJ, Santella RM, Carmella SG, Hecht SS, Gallot L, Schilder L, Crowell JA, Perloff M, Folz RJ, Bergan RC. Safety and efficacy of weekly oral oltipraz in chronic smokers. Cancer Epidemiol Biomarkers Prev. 2005;14:892–899. doi: 10.1158/1055-9965.EPI-04-0585. [DOI] [PubMed] [Google Scholar]
  53. Kensler TW. Chemoprevention by inducers of carcinogen detoxication enzymes. Environ Health Perspect. 1997;105:965–970. doi: 10.1289/ehp.97105s4965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kensler TW, Egner PA, Trush MA, Bueding E, Groopman JD. Modification of aflatoxin B1 binding to DNA in vivo in rats fed phenolic antioxidants, ethoxyquin and a dithiothione. Carcinogenesis. 1985;6:759–763. doi: 10.1093/carcin/6.5.759. [DOI] [PubMed] [Google Scholar]
  55. Kensler TW, Groopman JD, Sutter TR, Curphey TJ, Roebuck BD. Development of cancer chemopreventive agents: oltipraz as a paradigm. Chem Res Toxicol. 1999;12:113–126. doi: 10.1021/tx980185b. [DOI] [PubMed] [Google Scholar]
  56. Kensler TW, He X, Otieno M, Egner PA, Jacobson LP, Chen B, Wang JS, Zhu YR, Zhang BC, Wang JB, Wu Y, Zhang QN, Qian GS, Kuang SY, Fang X, Li YF, Yu LY, Prochaska HJ, Davidson NE, Gordon GB, Gorman MB, Zarba A, Enger C, Munoz A, Helzlsouer KJ, et al. Oltipraz chemoprevention trial in Qidong, People's Republic of China: modulation of serum aflatoxin albumin adduct biomarkers. Cancer Epidemiol Biomarkers Prev. 1998;7:127–134. [PubMed] [Google Scholar]
  57. Keum YS, Yu S, Chang PP, Yuan X, Kim JH, Xu C, Han J, Agarwal A, Kong AN. Mechanism of action of sulforaphane: inhibition of p38 mitogen-activated protein kinase isoforms contributing to the induction of antioxidant response element-mediated heme oxygenase-1 in human hepatoma HepG2 cells. Cancer Res. 2006;66:8804–8813. doi: 10.1158/0008-5472.CAN-05-3513. [DOI] [PubMed] [Google Scholar]
  58. Khan N, Afaq F, Mukhtar H. Cancer chemoprevention through dietary antioxidants: progress and promise. Antioxid Redox Signal. 2008;10:475–510. doi: 10.1089/ars.2007.1740. [DOI] [PubMed] [Google Scholar]
  59. Khor TO, Huang MT, Prawan A, Liu Y, Hao X, Yu S, Cheung WK, Chan JY, Reddy BS, Yang CS, Kong AN. Increased susceptibility of Nrf2 knockout mice to colitis-associated colorectal cancer. Cancer Prev Res (Phila Pa) 2008;1:187–191. doi: 10.1158/1940-6207.CAPR-08-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kim SK, Yang JW, Kim MR, Roh SH, Kim HG, Lee KY, Jeong HG, Kang KW. Increased expression of Nrf2/ARE-dependent anti-oxidant proteins in tamoxifen-resistant breast cancer cells. Free Radic Biol Med. 2008;45:537–546. doi: 10.1016/j.freeradbiomed.2008.05.011. [DOI] [PubMed] [Google Scholar]
  61. Kitamura Y, Umemura T, Kanki K, Kodama Y, Kitamoto S, Saito K, Itoh K, Yamamoto M, Masegi T, Nishikawa A, Hirose M. Increased susceptibility to hepatocarcinogenicity of Nrf2-deficient mice exposed to 2-amino-3-methylimidazo[4,5-f]quinoline. Cancer Sci. 2007;98:19–24. doi: 10.1111/j.1349-7006.2006.00352.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, Igarashi K, Yamamoto M. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol. 2004;24:7130–7139. doi: 10.1128/MCB.24.16.7130-7139.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Kobayashi A, Kang MI, Watai Y, Tong KI, Shibata T, Uchida K, Yamamoto M. Oxidative and electrophilic stresses activate Nrf2 through inhibition of ubiquitination activity of Keap1. Mol Cell Biol. 2006;26:221–229. doi: 10.1128/MCB.26.1.221-229.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Kobayashi M, Itoh K, Suzuki T, Osanai H, Nishikawa K, Katoh Y, Takagi Y, Yamamoto M. Identification of the interactive interface and phylogenic conservation of the Nrf2-Keap1 system. Genes Cells. 2002;7:807–820. doi: 10.1046/j.1365-2443.2002.00561.x. [DOI] [PubMed] [Google Scholar]
  65. Kobayashi M, Li L, Iwamoto N, Nakajima-Takagi Y, Kaneko H, Nakayama Y, Eguchi M, Wada Y, Kumagai Y, Yamamoto M. The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds. Mol Cell Biol. 2009;29:493–502. doi: 10.1128/MCB.01080-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Kobayashi M, Yamamoto M. Molecular mechanisms activating the Nrf2-Keap1 pathway of antioxidant gene regulation. Antioxid Redox Signal. 2005;7:385–394. doi: 10.1089/ars.2005.7.385. [DOI] [PubMed] [Google Scholar]
  67. Kuroiwa Y, Nishikawa A, Kitamura Y, Kanki K, Ishii Y, Umemura T, Hirose M. Protective effects of benzyl isothiocyanate and sulforaphane but not resveratrol against initiation of pancreatic carcinogenesis in hamsters. Cancer Lett. 2006;241:275–280. doi: 10.1016/j.canlet.2005.10.028. [DOI] [PubMed] [Google Scholar]
  68. Kwak MK, Egner PA, Dolan PM, Ramos-Gomez M, Groopman JD, Itoh K, Yamamoto M, Kensler TW. Role of phase 2 enzyme induction in chemoprotection by dithiolethiones. Mutat Res. 2001;480-481:305–315. doi: 10.1016/s0027-5107(01)00190-7. [DOI] [PubMed] [Google Scholar]
  69. Kwak MK, Ramos-Gomez M, Wakabayashi N, Kensler TW. Chemoprevention by 1,2-dithiole-3-thiones through induction of NQO1 and other phase 2 enzymes. Methods Enzymol. 2004a;382:414–423. doi: 10.1016/S0076-6879(04)82022-6. [DOI] [PubMed] [Google Scholar]
  70. Kwak MK, Wakabayashi N, Itoh K, Motohashi H, Yamamoto M, Kensler TW. Modulation of gene expression by cancer chemopreventive dithiolethiones through the Keap1-Nrf2 pathway. Identification of novel gene clusters for cell survival. J Biol Chem. 2003;278:8135–8145. doi: 10.1074/jbc.M211898200. [DOI] [PubMed] [Google Scholar]
  71. Kwak MK, Wakabayashi N, Kensler TW. Chemoprevention through the Keap1-Nrf2 signaling pathway by phase 2 enzyme inducers. Mutat Res. 2004b;555:133–148. doi: 10.1016/j.mrfmmm.2004.06.041. [DOI] [PubMed] [Google Scholar]
  72. Lam S, MacAulay C, Le Riche JC, Dyachkova Y, Coldman A, Guillaud M, Hawk E, Christen MO, Gazdar AF. A randomized phase IIb trial of anethole dithiolethione in smokers with bronchial dysplasia. J Natl Cancer Inst. 2002;94:1001–1009. doi: 10.1093/jnci/94.13.1001. [DOI] [PubMed] [Google Scholar]
  73. Lee JM, Calkins MJ, Chan K, Kan YW, Johnson JA. Identification of the NF-E2-related factor-2-dependent genes conferring protection against oxidative stress in primary cortical astrocytes using oligonucleotide microarray analysis. J Biol Chem. 2003;278:12029–12038. doi: 10.1074/jbc.M211558200. [DOI] [PubMed] [Google Scholar]
  74. Lee JS, Surh YJ. Nrf2 as a novel molecular target for chemoprevention. Cancer Lett. 2005;224:171–184. doi: 10.1016/j.canlet.2004.09.042. [DOI] [PubMed] [Google Scholar]
  75. Li W, Kong AN. Molecular mechanisms of Nrf2-mediated antioxidant response. Mol Carcinog. 2009;48:91–104. doi: 10.1002/mc.20465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Li W, Yu SW, Kong AN. Nrf2 possesses a redox-sensitive nuclear exporting signal in the Neh5 transactivation domain. J Biol Chem. 2006;281:27251–27263. doi: 10.1074/jbc.M602746200. [DOI] [PubMed] [Google Scholar]
  77. Liby K, Hock T, Yore MM, Suh N, Place AE, Risingsong R, Williams CR, Royce DB, Honda T, Honda Y, Gribble GW, Hill-Kapturczak N, Agarwal A, Sporn MB. The synthetic triterpenoids, CDDO and CDDO-imidazolide, are potent inducers of heme oxygenase-1 and Nrf2/ARE signaling. Cancer Res. 2005;65:4789–4798. doi: 10.1158/0008-5472.CAN-04-4539. [DOI] [PubMed] [Google Scholar]
  78. Liby K, Royce DB, Williams CR, Risingsong R, Yore MM, Honda T, Gribble GW, Dmitrovsky E, Sporn TA, Sporn MB. The synthetic triterpenoids CDDO-methyl ester and CDDO-ethyl amide prevent lung cancer induced by vinyl carbamate in A/J mice. Cancer Res. 2007;67:2414–2419. doi: 10.1158/0008-5472.CAN-06-4534. [DOI] [PubMed] [Google Scholar]
  79. Long DJ, 2nd, Waikel RL, Wang XJ, Perlaky L, Roop DR, Jaiswal AK. NAD(P)H:quinone oxidoreductase 1 deficiency increases susceptibility to benzo(a)pyrene-induced mouse skin carcinogenesis. Cancer Res. 2000;60:5913–5915. [PubMed] [Google Scholar]
  80. Manandhar S, Cho JM, Kim JA, Kensler TW, Kwak MK. Induction of Nrf2-regulated genes by 3H-1, 2-dithiole-3-thione through the ERK signaling pathway in murine keratinocytes. Eur J Pharmacol. 2007;577:17–27. doi: 10.1016/j.ejphar.2007.08.018. [DOI] [PubMed] [Google Scholar]
  81. Maxuitenko YY, MacMillan DL, Kensler TW, Roebuck BD. Evaluation of the post-initiation effects of oltipraz on aflatoxin B1-induced preneoplastic foci in a rat model of hepatic tumorigenesis. Carcinogenesis. 1993;14:2423–2425. doi: 10.1093/carcin/14.11.2423. [DOI] [PubMed] [Google Scholar]
  82. McMahon M, Thomas N, Itoh K, Yamamoto M, Hayes JD. Redox-regulated turnover of Nrf2 is determined by at least two separate protein domains, the redox-sensitive Neh2 degron and the redox-insensitive Neh6 degron. J Biol Chem. 2004;279:31556–31567. doi: 10.1074/jbc.M403061200. [DOI] [PubMed] [Google Scholar]
  83. Mo Z, Gao Y, Cao Y, Gao F, Jian L. An updating meta-analysis of the GSTM1, GSTT1, and GSTP1 polymorphisms and prostate cancer: a HuGE review. Prostate. 2009;69:662–688. doi: 10.1002/pros.20907. [DOI] [PubMed] [Google Scholar]
  84. Moinova HR, Mulcahy RT. An electrophile responsive element (EpRE) regulates beta-naphthoflavone induction of the human gamma-glutamylcysteine synthetase regulatory subunit gene. Constitutive expression is mediated by an adjacent AP-1 site. J Biol Chem. 1998;273:14683–14689. doi: 10.1074/jbc.273.24.14683. [DOI] [PubMed] [Google Scholar]
  85. Moon RC, Kelloff GJ, Detrisac CJ, Steele VE, Thomas CF, Sigman CC. Chemoprevention of OH-BBN-induced bladder cancer in mice by oltipraz, alone and in combination with 4-HPR and DFMO. Anticancer Res. 1994;14:5–11. [PubMed] [Google Scholar]
  86. Nair S, Xu C, Shen G, Hebbar V, Gopalakrishnan A, Hu R, Jain MR, Lin W, Keum YS, Liew C, Chan JY, Kong AN. Pharmacogenomics of phenolic antioxidant butylated hydroxyanisole (BHA) in the small intestine and liver of Nrf2 knockout and C57BL/6J mice. Pharm Res. 2006;23:2621–2637. doi: 10.1007/s11095-006-9099-x. [DOI] [PubMed] [Google Scholar]
  87. Nakayama M, Gonzalgo ML, Yegnasubramanian S, Lin X, De Marzo AM, Nelson WG. GSTP1 CpG island hypermethylation as a molecular biomarker for prostate cancer. J Cell Biochem. 2004;91:540–552. doi: 10.1002/jcb.10740. [DOI] [PubMed] [Google Scholar]
  88. Nebert DW, Roe AL, Vandale SE, Bingham E, Oakley GG. NAD(P)H:quinone oxidoreductase (NQO1) polymorphism, exposure to benzene, and predisposition to disease: a HuGE review. Genet Med. 2002;4:62–70. doi: 10.1097/00125817-200203000-00003. [DOI] [PubMed] [Google Scholar]
  89. Nguyen T, Rushmore TH, Pickett CB. Transcriptional regulation of a rat liver glutathione S-transferase Ya subunit gene. Analysis of the antioxidant response element and its activation by the phorbol ester 12-O-tetradecanoylphorbol-13-acetate. J Biol Chem. 1994;269:13656–13662. [PubMed] [Google Scholar]
  90. Nioi P, Hayes JD. Contribution of NAD(P)H:quinone oxidoreductase 1 to protection against carcinogenesis, and regulation of its gene by the Nrf2 basic-region leucine zipper and the arylhydrocarbon receptor basic helix-loop-helix transcription factors. Mutat Res. 2004;555:149–171. doi: 10.1016/j.mrfmmm.2004.05.023. [DOI] [PubMed] [Google Scholar]
  91. Nioi P, Nguyen T. A mutation of Keap1 found in breast cancer impairs its ability to repress Nrf2 activity. Biochem Biophys Res Commun. 2007;362:816–821. doi: 10.1016/j.bbrc.2007.08.051. [DOI] [PubMed] [Google Scholar]
  92. Nioi P, Nguyen T, Sherratt PJ, Pickett CB. The carboxy-terminal Neh3 domain of Nrf2 is required for transcriptional activation. Mol Cell Biol. 2005;25:10895–10906. doi: 10.1128/MCB.25.24.10895-10906.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Nishikawa A, Tanakamura Z, Furukawa F, Lee IS, Kasahara K, Ikezaki S, Takahashi M. Chemopreventive activity of oltipraz against induction of glandular stomach carcinogenesis in rats by N-methyl-N′-nitro-N-nitrosoguanidine. Carcinogenesis. 1998;19:365–368. doi: 10.1093/carcin/19.2.365. [DOI] [PubMed] [Google Scholar]
  94. Ohta T, Iijima K, Miyamoto M, Nakahara I, Tanaka H, Ohtsuji M, Suzuki T, Kobayashi A, Yokota J, Sakiyama T, Shibata T, Yamamoto M, Hirohashi S. Loss of Keap1 function activates Nrf2 and provides advantages for lung cancer cell growth. Cancer Res. 2008;68:1303–1309. doi: 10.1158/0008-5472.CAN-07-5003. [DOI] [PubMed] [Google Scholar]
  95. Okawa H, Motohashi H, Kobayashi A, Aburatani H, Kensler TW, Yamamoto M. Hepatocyte-specific deletion of the keap1 gene activates Nrf2 and confers potent resistance against acute drug toxicity. Biochem Biophys Res Commun. 2006;339:79–88. doi: 10.1016/j.bbrc.2005.10.185. [DOI] [PubMed] [Google Scholar]
  96. Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A, Keogh JP, Meyskens FL, Jr, Valanis B, Williams JH, Jr, Barnhart S, Cherniack MG, Brodkin CA, Hammar S. Risk factors for lung cancer and for intervention effects in CARET, the Beta-Carotene and Retinol Efficacy Trial. J Natl Cancer Inst. 1996;88:1550–1559. doi: 10.1093/jnci/88.21.1550. [DOI] [PubMed] [Google Scholar]
  97. Osburn WO, Karim B, Dolan PM, Liu G, Yamamoto M, Huso DL, Kensler TW. Increased colonic inflammatory injury and formation of aberrant crypt foci in Nrf2-deficient mice upon dextran sulfate treatment. Int J Cancer. 2007;121:1883–1891. doi: 10.1002/ijc.22943. [DOI] [PubMed] [Google Scholar]
  98. Osburn WO, Yates MS, Dolan PD, Chen S, Liby KT, Sporn MB, Taguchi K, Yamamoto M, Kensler TW. Genetic or pharmacologic amplification of nrf2 signaling inhibits acute inflammatory liver injury in mice. Toxicol Sci. 2008;104:218–227. doi: 10.1093/toxsci/kfn079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Padmanabhan B, Tong KI, Ohta T, Nakamura Y, Scharlock M, Ohtsuji M, Kang MI, Kobayashi A, Yokoyama S, Yamamoto M. Structural basis for defects of Keap1 activity provoked by its point mutations in lung cancer. Mol Cell. 2006;21:689–700. doi: 10.1016/j.molcel.2006.01.013. [DOI] [PubMed] [Google Scholar]
  100. Prestera T, Zhang Y, Spencer SR, Wilczak CA, Talalay P. The electrophile counterattack response: protection against neoplasia and toxicity. Adv Enzyme Regul. 1993;33:281–296. doi: 10.1016/0065-2571(93)90024-8. [DOI] [PubMed] [Google Scholar]
  101. Prochaska HJ, Santamaria AB, Talalay P. Rapid detection of inducers of enzymes that protect against carcinogens. Proc Natl Acad Sci U S A. 1992;89:2394–2398. doi: 10.1073/pnas.89.6.2394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Raimondi S, Paracchini V, Autrup H, Barros-Dios JM, Benhamou S, Boffetta P, Cote ML, Dialyna IA, Dolzan V, Filiberti R, Garte S, Hirvonen A, Husgafvel-Pursiainen K, Imyanitov EN, Kalina I, Kang D, Kiyohara C, Kohno T, Kremers P, Lan Q, London S, Povey AC, Rannug A, Reszka E, Risch A, Romkes M, Schneider J, Seow A, Shields PG, Sobti RC, Sorensen M, Spinola M, Spitz MR, Strange RC, Stucker I, Sugimura H, To-Figueras J, Tokudome S, Yang P, Yuan JM, Warholm M, Taioli E. Meta-and pooled analysis of GSTT1 and lung cancer: a HuGE-GSEC review. Am J Epidemiol. 2006;164:1027–1042. doi: 10.1093/aje/kwj321. [DOI] [PubMed] [Google Scholar]
  103. Ramos-Gomez M, Dolan PM, Itoh K, Yamamoto M, Kensler TW. Interactive effects of nrf2 genotype and oltipraz on benzo[a]pyrene-DNA adducts and tumor yield in mice. Carcinogenesis. 2003;24:461–467. doi: 10.1093/carcin/24.3.461. [DOI] [PubMed] [Google Scholar]
  104. Ramos-Gomez M, Kwak MK, Dolan PM, Itoh K, Yamamoto M, Talalay P, Kensler TW. Sensitivity to carcinogenesis is increased and chemoprotective efficacy of enzyme inducers is lost in nrf2 transcription factor-deficient mice. Proc Natl Acad Sci U S A. 2001;98:3410–3415. doi: 10.1073/pnas.051618798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Rao CV, Rivenson A, Katiwalla M, Kelloff GJ, Reddy BS. Chemopreventive effect of oltipraz during different stages of experimental colon carcinogenesis induced by azoxymethane in male F344 rats. Cancer Res. 1993;53:2502–2506. [PubMed] [Google Scholar]
  106. Rao CV, Rivenson A, Zang E, Steele V, Kelloff G, Reddy BS. Inhibition of 2-Amino-1-methyl-6-phenylimidazo[4,5]pyridine-induced lymphoma formation by oltipraz. Cancer Res. 1996;56:3395–3398. [PubMed] [Google Scholar]
  107. Rao CV, Tokomo K, Kelloff G, Reddy BS. Inhibition by dietary oltipraz of experimental intestinal carcinogenesis induced by azoxymethane in male F344 rats. Carcinogenesis. 1991;12:1051–1055. doi: 10.1093/carcin/12.6.1051. [DOI] [PubMed] [Google Scholar]
  108. Ritchie KJ, Henderson CJ, Wang XJ, Vassieva O, Carrie D, Farmer PB, Gaskell M, Park K, Wolf CR. Glutathione transferase pi plays a critical role in the development of lung carcinogenesis following exposure to tobacco-related carcinogens and urethane. Cancer Res. 2007;67:9248–9257. doi: 10.1158/0008-5472.CAN-07-1764. [DOI] [PubMed] [Google Scholar]
  109. Roebuck BD, Liu YL, Rogers AE, Groopman JD, Kensler TW. Protection against aflatoxin B1-induced hepatocarcinogenesis in F344 rats by 5-(2-pyrazinyl)-4-methyl-1,2-dithiole-3-thione (oltipraz): predictive role for short-term molecular dosimetry. Cancer Res. 1991;51:5501–5506. [PubMed] [Google Scholar]
  110. Rushmore TH, Pickett CB. Transcriptional regulation of the rat glutathione S-transferase Ya subunit gene. Characterization of a xenobiotic-responsive element controlling inducible expression by phenolic antioxidants. J Biol Chem. 1990;265:14648–14653. [PubMed] [Google Scholar]
  111. Seow A, Yuan JM, Sun CL, Van Den Berg D, Lee HP, Yu MC. Dietary isothiocyanates, glutathione S-transferase polymorphisms and colorectal cancer risk in the Singapore Chinese Health Study. Carcinogenesis. 2002;23:2055–2061. doi: 10.1093/carcin/23.12.2055. [DOI] [PubMed] [Google Scholar]
  112. Sharma S, Gao P, Steele VE. The chemopreventive efficacy of inhaled oltipraz particulates in the B[a]P-induced A/J mouse lung adenoma model. Carcinogenesis. 2006;27:1721–1727. doi: 10.1093/carcin/bgl052. [DOI] [PubMed] [Google Scholar]
  113. Shen G, Xu C, Hu R, Jain MR, Gopalkrishnan A, Nair S, Huang MT, Chan JY, Kong AN. Modulation of nuclear factor E2-related factor 2-mediated gene expression in mice liver and small intestine by cancer chemopreventive agent curcumin. Mol Cancer Ther. 2006;5:39–51. doi: 10.1158/1535-7163.MCT-05-0293. [DOI] [PubMed] [Google Scholar]
  114. Shibata T, Kokubu A, Gotoh M, Ojima H, Ohta T, Yamamoto M, Hirohashi S. Genetic alteration of Keap1 confers constitutive Nrf2 activation and resistance to chemotherapy in gallbladder cancer. Gastroenterology. 2008a;135:1358–1368. 1368 e1351–1354. doi: 10.1053/j.gastro.2008.06.082. [DOI] [PubMed] [Google Scholar]
  115. Shibata T, Ohta T, Tong KI, Kokubu A, Odogawa R, Tsuta K, Asamura H, Yamamoto M, Hirohashi S. Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc Natl Acad Sci U S A. 2008b;105:13568–13573. doi: 10.1073/pnas.0806268105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Shim GS, Manandhar S, Shin DH, Kim TH, Kwak MK. Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the NRF2-GSH pathway. Free Radic Biol Med. 2009 doi: 10.1016/j.freeradbiomed.2009.09.006. Submitted. [DOI] [PubMed] [Google Scholar]
  117. Singh A, Boldin-Adamsky S, Thimmulappa RK, Rath SK, Ashush H, Coulter J, Blackford A, Goodman SN, Bunz F, Watson WH, Gabrielson E, Feinstein E, Biswal S. RNAi-mediated silencing of nuclear factor erythroid-2-related factor 2 gene expression in non-small cell lung cancer inhibits tumor growth and increases efficacy of chemotherapy. Cancer Res. 2008;68:7975–7984. doi: 10.1158/0008-5472.CAN-08-1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, Herman JG, Baylin SB, Sidransky D, Gabrielson E, Brock MV, Biswal S. Dysfunctional KEAP1-NRF2 interaction in non-small-cell lung cancer. PLoS Med. 2006;3:e420. doi: 10.1371/journal.pmed.0030420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Smith MT, Wang Y, Skibola CF, Slater DJ, Lo Nigro L, Nowell PC, Lange BJ, Felix CA. Low NAD(P)H:quinone oxidoreductase activity is associated with increased risk of leukemia with MLL translocations in infants and children. Blood. 2002;100:4590–4593. doi: 10.1182/blood-2001-12-0264. [DOI] [PubMed] [Google Scholar]
  120. Spitz MR, Duphorne CM, Detry MA, Pillow PC, Amos CI, Lei L, de Andrade M, Gu X, Hong WK, Wu X. Dietary intake of isothiocyanates: evidence of a joint effect with glutathione S-transferase polymorphisms in lung cancer risk. Cancer Epidemiol Biomarkers Prev. 2000;9:1017–1020. [PubMed] [Google Scholar]
  121. Sporn MB, Liby KT. Cancer chemoprevention: scientific promise, clinical uncertainty. Nat Clin Pract Oncol. 2005;2:518–525. doi: 10.1038/ncponc0319. [DOI] [PubMed] [Google Scholar]
  122. Sporn MB, Roberts AB. Peptide growth factors and inflammation, tissue repair, and cancer. J Clin Invest. 1986;78:329–332. doi: 10.1172/JCI112580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Sticha KR, Kenney PM, Boysen G, Liang H, Su X, Wang M, Upadhyaya P, Hecht SS. Effects of benzyl isothiocyanate and phenethyl isothiocyanate on DNA adduct formation by a mixture of benzo[a]pyrene and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone in A/J mouse lung. Carcinogenesis. 2002;23:1433–1439. doi: 10.1093/carcin/23.9.1433. [DOI] [PubMed] [Google Scholar]
  124. Surh YJ. Cancer chemoprevention with dietary phytochemicals. Nat Rev Cancer. 2003;3:768–780. doi: 10.1038/nrc1189. [DOI] [PubMed] [Google Scholar]
  125. Surh YJ, Kundu JK, Na HK. Nrf2 as a master redox switch in turning on the cellular signaling involved in the induction of cytoprotective genes by some chemopreventive phytochemicals. Planta Med. 2008;74:1526–1539. doi: 10.1055/s-0028-1088302. [DOI] [PubMed] [Google Scholar]
  126. Sussan TE, Rangasamy T, Blake DJ, Malhotra D, El-Haddad H, Bedja D, Yates MS, Kombairaju P, Yamamoto M, Liby KT, Sporn MB, Gabrielson KL, Champion HC, Tuder RM, Kensler TW, Biswal S. Targeting Nrf2 with the triterpenoid CDDO-imidazolide attenuates cigarette smoke-induced emphysema and cardiac dysfunction in mice. Proc Natl Acad Sci U S A. 2009;106:250–255. doi: 10.1073/pnas.0804333106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Talalay P, De Long MJ, Prochaska HJ. Identification of a common chemical signal regulating the induction of enzymes that protect against chemical carcinogenesis. Proc Natl Acad Sci U S A. 1988;85:8261–8265. doi: 10.1073/pnas.85.21.8261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Tan XL, Spivack SD. Dietary chemoprevention strategies for induction of phase II xenobiotic-metabolizing enzymes in lung carcinogenesis: A review. Lung Cancer. 2009 doi: 10.1016/j.lungcan.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Thimmulappa RK, Mai KH, Srisuma S, Kensler TW, Yamamoto M, Biswal S. Identification of Nrf2-regulated genes induced by the chemopreventive agent sulforaphane by oligonucleotide microarray. Cancer Res. 2002;62:5196–5203. [PubMed] [Google Scholar]
  130. Tong KI, Kobayashi A, Katsuoka F, Yamamoto M. Two-site substrate recognition model for the Keap1-Nrf2 system: a hinge and latch mechanism. Biol Chem. 2006;387:1311–1320. doi: 10.1515/BC.2006.164. [DOI] [PubMed] [Google Scholar]
  131. Tong KI, Padmanabhan B, Kobayashi A, Shang C, Hirotsu Y, Yokoyama S, Yamamoto M. Different electrostatic potentials define ETGE and DLG motifs as hinge and latch in oxidative stress response. Mol Cell Biol. 2007;27:7511–7521. doi: 10.1128/MCB.00753-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Tran QT, Xu L, Phan V, Goodwin SB, Rahman M, Jin VX, Sutter CH, Roebuck BD, Kensler TW, George EO, Sutter TR. Chemical genomics of cancer chemopreventive dithiolethiones. Carcinogenesis. 2009;30:480–486. doi: 10.1093/carcin/bgn292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Ulrich CM, Bigler J, Potter JD. Non-steroidal anti-inflammatory drugs for cancer prevention: promise, perils and pharmacogenetics. Nat Rev Cancer. 2006;6:130–140. doi: 10.1038/nrc1801. [DOI] [PubMed] [Google Scholar]
  134. Venugopal R, Jaiswal AK. Nrf1 and Nrf2 positively and c-Fos and Fra1 negatively regulate the human antioxidant response element-mediated expression of NAD(P)H:quinone oxidoreductase1 gene. Proc Natl Acad Sci U S A. 1996;93:14960–14965. doi: 10.1073/pnas.93.25.14960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Venugopal R, Jaiswal AK. Nrf2 and Nrf1 in association with Jun proteins regulate antioxidant response element-mediated expression and coordinated induction of genes encoding detoxifying enzymes. Oncogene. 1998;17:3145–3156. doi: 10.1038/sj.onc.1202237. [DOI] [PubMed] [Google Scholar]
  136. Wakabayashi N, Dinkova-Kostova AT, Holtzclaw WD, Kang ML, Kobayashi A, Yamamoto M, Kensler TW, Talalay P. Protection against electrophile and oxidant stress by induction of the phase 2 response: Fate of cysteines of the Keap1 sensor modified by inducers. Proc Natl Acad Sci U S A. 2004;101:2040–2045. doi: 10.1073/pnas.0307301101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Wakabayashi N, Itoh K, Wakabayashi J, Motohashi H, Noda S, Takahashi S, Imakado S, Kotsuji T, Otsuka F, Roop DR, Harada T, Engel JD, Yamamoto M. Keap1-null mutation leads to postnatal lethality due to constitutive Nrf2 activation. Nat Genet. 2003;35:238–245. doi: 10.1038/ng1248. [DOI] [PubMed] [Google Scholar]
  138. Wang B, Williamson G. Detection of a nuclear protein which binds specifically to the antioxidant responsive element (ARE) of the human NAD(P) H:quinone oxidoreductase gene. Biochim Biophys Acta. 1994;1219:645–652. doi: 10.1016/0167-4781(94)90223-2. [DOI] [PubMed] [Google Scholar]
  139. Wang JS, Shen X, He X, Zhu YR, Zhang BC, Wang JB, Qian GS, Kuang SY, Zarba A, Egner PA, Jacobson LP, Munoz A, Helzlsouer KJ, Groopman JD, Kensler TW. Protective alterations in phase 1 and 2 metabolism of aflatoxin B1 by oltipraz in residents of Qidong, People's Republic of China. J Natl Cancer Inst. 1999;91:347–354. doi: 10.1093/jnci/91.4.347. [DOI] [PubMed] [Google Scholar]
  140. Wang LI, Giovannucci EL, Hunter D, Neuberg D, Su L, Christiani DC. Dietary intake of Cruciferous vegetables, Glutathione S-transferase (GST) polymorphisms and lung cancer risk in a Caucasian population. Cancer Causes Control. 2004;15:977–985. doi: 10.1007/s10552-004-1093-1. [DOI] [PubMed] [Google Scholar]
  141. Wang R, An J, Ji F, Jiao H, Sun H, Zhou D. Hypermethylation of the Keap1 gene in human lung cancer cell lines and lung cancer tissues. Biochem Biophys Res Commun. 2008;373:151–154. doi: 10.1016/j.bbrc.2008.06.004. [DOI] [PubMed] [Google Scholar]
  142. Wasserman WW, Fahl WE. Functional antioxidant responsive elements. Proc Natl Acad Sci U S A. 1997;94:5361–5366. doi: 10.1073/pnas.94.10.5361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Wattenberg LW. Chemoprevention of cancer. Cancer Res. 1985;45:1–8. [PubMed] [Google Scholar]
  144. Wattenberg LW, Bueding E. Inhibitory effects of 5-(2-pyrazinyl)-4-methyl-1,2-dithiol-3-thione (Oltipraz) on carcinogenesis induced by benzo[a]pyrene, diethylnitrosamine and uracil mustard. Carcinogenesis. 1986;7:1379–1381. doi: 10.1093/carcin/7.8.1379. [DOI] [PubMed] [Google Scholar]
  145. Xie T, Belinsky M, Xu Y, Jaiswal AK. ARE- and TRE-mediated regulation of gene expression. Response to xenobiotics and antioxidants. J Biol Chem. 1995;270:6894–6900. doi: 10.1074/jbc.270.12.6894. [DOI] [PubMed] [Google Scholar]
  146. Xu C, Huang MT, Shen G, Yuan X, Lin W, Khor TO, Conney AH, Kong AN. Inhibition of 7,12-dimethylbenz(a)anthracene-induced skin tumorigenesis in C57BL/6 mice by sulforaphane is mediated by nuclear factor E2-related factor 2. Cancer Res. 2006;66:8293–8296. doi: 10.1158/0008-5472.CAN-06-0300. [DOI] [PubMed] [Google Scholar]
  147. Yamamoto T, Suzuki T, Kobayashi A, Wakabayashi J, Maher J, Motohashi H, Yamamoto M. Physiological significance of reactive cysteine residues of Keap1 in determining Nrf2 activity. Mol Cell Biol. 2008;28:2758–2770. doi: 10.1128/MCB.01704-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Yates MS, Kensler TW. Keap1 eye on the target: chemoprevention of liver cancer. Acta Pharmacol Sin. 2007;28:1331–1342. doi: 10.1111/j.1745-7254.2007.00688.x. [DOI] [PubMed] [Google Scholar]
  149. Yates MS, Kwak MK, Egner PA, Groopman JD, Bodreddigari S, Sutter TR, Baumgartner KJ, Roebuck BD, Liby KT, Yore MM, Honda T, Gribble GW, Sporn MB, Kensler TW. Potent protection against aflatoxin-induced tumorigenesis through induction of Nrf2-regulated pathways by the triterpenoid 1-[2-cyano-3-,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole. Cancer Res. 2006;66:2488–2494. doi: 10.1158/0008-5472.CAN-05-3823. [DOI] [PubMed] [Google Scholar]
  150. Yates MS, Tauchi M, Katsuoka F, Flanders KC, Liby KT, Honda T, Gribble GW, Johnson DA, Johnson JA, Burton NC, Guilarte TR, Yamamoto M, Sporn MB, Kensler TW. Pharmacodynamic characterization of chemopreventive triterpenoids as exceptionally potent inducers of Nrf2-regulated genes. Mol Cancer Ther. 2007;6:154–162. doi: 10.1158/1535-7163.MCT-06-0516. [DOI] [PubMed] [Google Scholar]
  151. Yates MS, Tran QT, Dolan PM, Osburn WO, Shin S, McCulloch CC, Silkworth JB, Taguchi K, Yamamoto M, Williams CR, Liby KT, Sporn MB, Sutter TR, Kensler TW. Genetic versus Chemoprotective Activation of Nrf2 Signaling: Overlapping yet Distinct Gene Expression Profiles between Keap1 Knockout and Triterpenoid Treated Mice. Carcinogenesis. 2009 doi: 10.1093/carcin/bgp100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Ye Z, Song H. Glutathione s-transferase polymorphisms (GSTM1, GSTP1 and GSTT1) and the risk of acute leukaemia: a systematic review and meta-analysis. Eur J Cancer. 2005;41:980–989. doi: 10.1016/j.ejca.2005.01.014. [DOI] [PubMed] [Google Scholar]
  153. Zhang DD, Hannink M. Distinct cysteine residues in keap1 are required for keap1-dependent ubiquitination of nrf2 and for stabilization of nrf2 by chemopreventive agents and oxidative stress. Mol Cell Biol. 2003;23:8137–8151. doi: 10.1128/MCB.23.22.8137-8151.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Zhang DD, Lo SC, Cross JV, Templeton DJ, Hannink M. Keap1 is a redox-regulated substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex. Mol Cell Biol. 2004;24:10941–10953. doi: 10.1128/MCB.24.24.10941-10953.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Zhang Y, Munday R. Dithiolethiones for cancer chemoprevention: where do we stand? Mol Cancer Ther. 2008;7:3470–3479. doi: 10.1158/1535-7163.MCT-08-0625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Zhang Y, Talalay P, Cho CG, Posner GH. A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. Proc Natl Acad Sci U S A. 1992;89:2399–2403. doi: 10.1073/pnas.89.6.2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Zipper LM, Mulcahy RT. The Keap1 BTB/POZ dimerization function is required to sequester Nrf2 in cytoplasm. J Biol Chem. 2002;277:36544–36552. doi: 10.1074/jbc.M206530200. [DOI] [PubMed] [Google Scholar]

RESOURCES