Nrf2 (Nfe2l2) governs cellular defenses against oxidative and electrophilic stresses and protects against chemical carcinogenesis. However, many cancers have been found to accumulate NRF2 protein, raising questions of precisely how Nrf2 contributes to carcinogenesis. In this report, we explored such questions in an established urethane-induced multistep model of lung carcinogenesis. Consistent with earlier observations, Nrf2-deficient (Nrf2−/−) mice exhibited a relative increase in tumor foci by 8 weeks after urethane administration. However, after 16 weeks, we observed a relative reduction in the number of tumors with more malignant characteristics in Nrf2−/− mice. Furthermore, all Nrf2+/+ tumors harbored activated mutations in Kras, whereas Nrf2−/− tumors were rarely associated with similar Kras mutations. Overall, our results established that Nrf2 has two roles during carcinogenesis, one of which is preventive during tumor initiation and the second that promotes malignant progression. These findings establish Nrf2 inhibitors as rational tools to prevent malignant progression in lung cancer, whereas Nrf2 activators are more suited for lung cancer prevention. Cancer Res; 73(13); 4158–68. ©2013 AACR.

Nrf2 (Nuclear factor-erythroid derived 2-like 2, Nfe2l2) is a leucine zipper transcription factor and plays an important role in the maintenance of redox balance and cytoprotection against chemical carcinogens (1, 2). When subjected to oxidative and electrophilic stress conditions, Nrf2 is released from Keap1 (Kelch-like ECH-associated protein 1)-mediated rapid degradation. Nrf2 is stabilized, accumulated, and translocates to the nucleus, where Nrf2 dimerizes with a small Maf protein (sMaf). The Nrf2–sMaf heterodimer binds to a specific DNA sequence, referred to as the antioxidant/electrophile response element (ARE/EpRE), and induces the expression of a cohort of cytoprotective enzyme genes, such as Nqo1, HO-1, Gclc, and Gstp1/p2 (1, 2).

Previous reports on chemically induced carcinogenesis have shown that Nrf2-deficient (Nrf2−/−) mice tend to form a larger number of tumors than wild-type (Nrf2+/+) mice, indicating that the perturbation in the carcinogen detoxification system in Nrf2−/− mice leads to cancer susceptibility in various tissues (3–6). However, recent studies have revealed that the NRF2 protein is significantly accumulated in many types of human cancers through several independent mechanisms. For instance, somatic mutations in the interface of KEAP1 and NRF2 (7) or epigenetic modifications in the promoter region of KEAP1 genes (8) gives rise to the accumulation of NRF2. As these changes are often related to accelerated cancer cell growth and poor clinical prognosis, the oncogenic side of NRF2 function has been attracting considerable attention. Furthermore, we recently showed that the Nrf2−/− mice transplanted with Lewis lung carcinoma (3LL) cells provide a more tumor-permissive immune microenvironment than the Nrf2+/+ mice (9). These observations raise a fundamental question of whether Nrf2 exerts cancer-preventive or -promotive activity in each stage of the carcinogenic process. To address this issue, we used the urethane (ethyl carbamate)-induced lung carcinogenesis model, a well-known multistep murine carcinogenesis model (10).

Urethane has been extensively used as an inducer of chemical lung carcinogenesis (11). Administration of urethane evokes hyperplasia or adenoma formation in the lung and eventually leads to adenocarcinomas in a time-dependent manner (10). Through cytochrome P450 2E1 (Cyp2e1)-mediated oxidation, urethane is converted to vinyl carbamate epoxide, which serves as an ultimate carcinogen by inducing DNA-, RNA-, or protein–adduct formation in airway epithelial cells (12). In the detoxification process, microsomal epoxide hydrolase (mEH) converts vinyl carbamate epoxide into 1,2-dihydroxyethyl carbamate, which subsequently undergoes the glutathione conjugation catalyzed by Gstp1/p2 (13) and is excreted into urine (14).

In the present study, we found that Nrf2−/−-mutant mice developed a large number of urethane-induced lung micronodules in the early phase after urethane administration. However, in the later stages, Nrf2+/+ mice developed a higher number of Kras-mutated adenocarcinomas than did Nrf2−/− mice. These results show that Nrf2 deficiency leads to an increased susceptibility to chemical carcinogens and resultant high-level tumor initiations, whereas Nrf2 serves as an oncogenic factor that accelerates malignant progression of Kras-mutated adenocarcinomas in the later stages of lung carcinogenesis.

Experimental animals

Nrf2−/− mice with an ICR/CD-1 genetic background (outbred) were used in this study (2, 15). Age-matched (5–9 weeks) Nrf2+/+ mice were used as concurrent controls. The mice were maintained in a facility free of specific pathogens (SPF). Nude mice (8- to 9-weeks old) were purchased from CLEA Japan. All animal experiments were conducted with the approval of the Tohoku University Animal Care Committee.

Lung carcinogenesis experiments

Mouse lung tumors were induced by the intraperitoneal injection of urethane (1 g/kg body weight; refs. 16, 17). For the enumeration of lung surface tumors, the lungs were removed and the total number of lung surface nodules per mouse was counted macroscopically.

Kras mutation analysis

PCR-amplified DNA samples from the urethane-induced lung tumors and intact stromal tissues were subjected to sequencing analysis to detect Kras mutations. The primers amplifying the nucleotide sequences in the second exon of Kras gene, which contains codon 61, are listed in Supplementary Table S1.

Microarray analysis

Surface lung tumors were excised, and surrounding tissues were carefully removed under a observation via a stereo-microscope. The lung tumors and nontumor regions of Nrf2+/+ and Nrf2−/− mice that had been treated with urethane (8 mice/group) were pooled and subjected to a whole-mouse genome microarray analysis (4 × 44 k; Agilent Technologies). The expression data were analyzed with GeneSpring software (Silicon Genetics). Heatmaps were generated using Cluster 3.0 (http://bonsai.hgc.jp/~mdehoon/software/cluster/) and JAVA Treeview 159 (http://jtreeview.sourceforge.net/). The classification of the selected genes according to their biologic and toxicologic functions was done using Ingenuity Pathway Analysis (IPA) software (Ingenuity system). P value, represented as the negative log ratio of the IPA results, is the probability based on Fisher exact test. The GEO accession number for the microarray data is GSE46048.

Flow cytometry

Analyses of the bone marrow cells were conducted using FACS-Caliber (BD Pharmingen). Quantification of reactive oxygen series (ROS) level with 2,7-dichlorodihydrofluorescein diacetate (DCFDA), separation of myeloid-derived suppressor cells (MDSC), and T cells has been described previously (9).

Immunoblotting analysis

Nuclear extracts were prepared from NIH3T3 cells that were treated with the indicated concentrations of urethane for 6 hours. The mouse lung nuclear extracts were prepared from the Nrf2+/+ mice administered either vehicle (PBS) or urethane (1 g/kg body weight) for the indicated time periods. Immunoblotting analysis was conducted using anti-Nrf2 and anti-lamin B antibodies (Santa Cruz Biotechnology) as described previously (9).

Statistical analyses

The data are expressed as the mean ± SD. The statistical differences were determined using Student t test or the Mann–Whitney U test. The values for either the incidence of lung nodules or large tumors were analyzed using the Fisher exact probability test. P values less than 0.05 were considered significant.

See Supplementary Materials and Methods for further details.

Urethane induces accumulation of Nrf2 and detoxification enzymes in the lung

Although urethane exerts carcinogenic activity through electrophilicity of its metabolites (18), it remains unclear whether urethane induces Nrf2 accumulation. To address this question, we examined Nrf2 accumulation in NIH/3T3 cells treated with urethane (10 or 50 μmol/L for 6 hours). Upon treatment with urethane, Nrf2 accumulated in nuclear fraction of NIH/3T3 cells (Fig. 1A). In addition, we found that Nrf2 accumulated in the lung tissues 3 hours after intraperitoneal injection of urethane (1 g/kg body weight) into Nrf2+/+ mice (Fig. 1B). As the Nrf2 accumulation could be monitored by the immunohistochemistry for Nrf2–β-galactosidase fusion protein expressed from the Nrf2-targeted allele (9), we conducted anti-β-galactosidase antibody staining with paraffin-embedded lung sections. The Nrf2–β-galactosidase fusion protein predominantly accumulated in the bronchial epithelium of the urethane-treated mice (arrows in Fig. 1C). These results show that urethane induced Nrf2 accumulation in the nucleus of airway epithelial cells.

Figure 1.

Urethane administration activates Nrf2 and its downstream genes in vitro and in vivo. A and B, Nrf2 expression was detected by Western blotting in NIH3T3 cells and urethane-treated mouse lungs. Diethyl maleate (DEM) and PBS were used as positive and negative controls, respectively. Anti-lamin B antibody staining was used for equal nuclear protein loading control. C, induction of Nrf2-LacZ protein by urethane treatment in lung epithelial cells in Nrf2−/− and Nrf2+/+ mice. Arrows indicate positive cells. The black box indicates a high magnification region. Black scale bar, 20 μm; red scale bar; 5 μm. D, induction of Nrf2-target genes by urethane treatment in lung. mRNA expression of Nrf2-target genes was examined by qRT-PCR analysis using lung tissues from Nrf2−/− and Nrf2+/+ mice after the urethane treatment. The expression level of each mRNA was normalized to the β-actin abundance. The data are presented as the mean ± SD. The significant differences determined by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Figure 1.

Urethane administration activates Nrf2 and its downstream genes in vitro and in vivo. A and B, Nrf2 expression was detected by Western blotting in NIH3T3 cells and urethane-treated mouse lungs. Diethyl maleate (DEM) and PBS were used as positive and negative controls, respectively. Anti-lamin B antibody staining was used for equal nuclear protein loading control. C, induction of Nrf2-LacZ protein by urethane treatment in lung epithelial cells in Nrf2−/− and Nrf2+/+ mice. Arrows indicate positive cells. The black box indicates a high magnification region. Black scale bar, 20 μm; red scale bar; 5 μm. D, induction of Nrf2-target genes by urethane treatment in lung. mRNA expression of Nrf2-target genes was examined by qRT-PCR analysis using lung tissues from Nrf2−/− and Nrf2+/+ mice after the urethane treatment. The expression level of each mRNA was normalized to the β-actin abundance. The data are presented as the mean ± SD. The significant differences determined by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Close modal

To clarify downstream events upon the urethane treatment, we next examined mRNA expression of Nrf2 target genes in the lung. Expression levels of Nrf2 target genes, i.e., Nqo1, HO-1, and Gclc, were markedly induced in the Nrf2+/+ mice in a time-dependent manner (Fig. 1D). In contrast, inducible expression of the Nrf2 target genes was attenuated in the Nrf2−/− mice. These observations show that urethane-treatment increased the expression of Nrf2-target genes through the induction of Nrf2 protein accumulation in normal lung tissue.

Cyp2e1 expression is independent of Nrf2 activity

It has been shown that Cyp2e1-mediated oxidation plays an essential role in urethane-induced carcinogenicity (Supplementary Fig. S1A) and, indeed, Cyp2e1-deficient mice are resistant to urethane-induced lung carcinogenesis (14). To clarify whether the Nrf2-deficiency affects the urethane bioactivation to its carcinogenic metabolite, we examined the Cyp2e1 level in Nrf2−/− mouse lungs. Basal Cyp2e1 mRNA level did not differ substantially between Nrf2−/− and Nrf2+/+ mice, and induced Cyp2e1 mRNA expression after the urethane administration was comparable between the 2 genotypes (Supplementary Fig. S1B). Cyp2e1 immunoreactivity in the airway epithelial cells was also comparable between the 2 genotypes, regardless of the urethane treatment (Supplementary Fig. S1C). These results indicate that the basal and induced expression of Cyp2e1 is independent of the Nrf2 activity. In addition, we examined mRNA expression of the enzymes involved in the detoxification process of vinyl carbamate epoxide. We found that urethane-induced expression of mEH was diminished in Nrf2−/− mice in comparison with those of Nrf2+/+ mice (Supplementary Fig. S1D). Gstp1/p2 expression in Nrf2−/− mice at both basal and urethane-induced states was lower than that in Nrf2+/+ mice. Together, these observations support our contention that both, maintained Cyp2e1-mediated oxidation and attenuated mEH-Gst detoxification in Nrf2−/− mice, lead to the accumulation of a higher amount of vinyl carbamate epoxide than that in Nrf2+/+ mice upon the urethane treatment.

Urethane elicits acute inflammatory response in Nrf2-deficient mouse lung

Nrf2−/− mice are susceptible to a number of oxidative or electrophilic insults, including butylated hydroxytoluene and bleomycin, and the mice are more susceptible to pneumonia, fibrosis, and inflammatory cell infiltration than Nrf2+/+ mice (19). Given this, we next examined inflammatory status of the lungs after the urethane treatment. We observed a large number of inflammatory cell foci in the pulmonary perivascular region of the Nrf2−/− mice 24 hours after urethane administration, whereas such inflammatory cell infiltration was not observed in Nrf2+/+ mouse lungs (Supplementary Fig. S2A). These cell clusters mainly consisted of CD3+ T-lymphocytes. Pan-cytokeratin+ epithelial cells were also involved frequently (Supplementary Fig. S2B). Notably, we observed a greater increase in Ki-67–positive cells in the cell clusters of Nrf2−/− mice, which presumably represent increased proliferation of the epithelial cells (Supplementary Fig. S2B). Therefore, we surmised that Nrf2−/− mice were susceptible to the urethane-induced inflammatory lung injury; thereafter, the inflammation-induced proliferating pulmonary epithelial cells might give rise to high frequency of lung tumorigenesis.

Nrf2−/− mice are susceptible to urethane-induced lung carcinogenesis in early stage

To examine susceptibility to lung cancer, we adopted urethane-induced carcinogenesis experiments with Nrf2−/− and Nrf2+/+ mice in the following 4 different observation periods: (i) very short-term observation (4 weeks); (ii) short-term observation (8 weeks); (iii) middle-term observation (16 weeks); and (iv) long-term observation (24 weeks; Supplementary Fig. S3). At 4 weeks after the intraperitoneal urethane injection, Nrf2−/− mice showed a much larger number of microscopic nodules (average 11.7, n = 3; P < 0.05) than the Nrf2+/+ mice (average 1.67, n = 3; Fig. 2A and B), whereas both groups rarely showed gross surface tumors. At 8 weeks after urethane administration, all the urethane-treated Nrf2−/− mice developed macroscopic (φ > 0.5 mm) lung surface tumors, whereas only half of the Nrf2+/+ mice developed gross surface tumors (Fig. 2C and D, Table 1, A). Furthermore, Nrf2−/− mice had a much higher number of lung surface nodules than Nrf2+/+ mice. These results indicate that Nrf2 contributes to the prevention of urethane-induced carcinogenesis in the early tumorigenic stages (4 or 8 weeks).

Figure 2.

Urethane-induced lung tumorigenesis in the short- and middle-term observation protocols. A–D, micro- and macroscopic examination of precancerous nodules at 4 and 8 weeks after urethane administration. A, histologic examination of precancerous lesions in the lung of Nrf2+/+ and Nrf2−/− mice. Lung sections from mice at 4 weeks after urethane administration were stained with hematoxylin and eosin. Scale bar, 100 μm. B, number of microscopic nodules in Nrf2+/+ and Nrf2−/− mice four weeks after urethane administration. C, representative gross observation of lung surface tumors in Nrf2+/+ and Nrf2−/− mice. Lungs from mice at 8 weeks after urethane administration were examined. Arrowheads show the surface tumors. Scale bar, 10 mm. Bottom, representative hematoxylin and eosin-stained sections. Scale bar, 100 μm. D, number of lung surface tumors in each mouse of both genotypes. E–H, macroscopic examination of surface tumors at 16 weeks after urethane administration. E, representative gross pictures of lung surface tumors in Nrf2+/+ and Nrf2−/− mice. Arrows indicate the lung surface tumors. Scale bars, 10 mm. F, numbers of surface tumors in lungs of Nrf2+/+ and Nrf2−/− mice. Each dot represents total number of macroscopic tumors (φ > 0.5 mm) in individual mouse. G, number of large-size nodules (φ > 1.0 mm) in lungs of Nrf2+/+ and Nrf2−/− mice. The color of dots indicates size of the largest tumor in each mouse lung. H, average of tumor diameters in lungs of Nrf2+/+ and Nrf2−/− mice. The significant differences by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Figure 2.

Urethane-induced lung tumorigenesis in the short- and middle-term observation protocols. A–D, micro- and macroscopic examination of precancerous nodules at 4 and 8 weeks after urethane administration. A, histologic examination of precancerous lesions in the lung of Nrf2+/+ and Nrf2−/− mice. Lung sections from mice at 4 weeks after urethane administration were stained with hematoxylin and eosin. Scale bar, 100 μm. B, number of microscopic nodules in Nrf2+/+ and Nrf2−/− mice four weeks after urethane administration. C, representative gross observation of lung surface tumors in Nrf2+/+ and Nrf2−/− mice. Lungs from mice at 8 weeks after urethane administration were examined. Arrowheads show the surface tumors. Scale bar, 10 mm. Bottom, representative hematoxylin and eosin-stained sections. Scale bar, 100 μm. D, number of lung surface tumors in each mouse of both genotypes. E–H, macroscopic examination of surface tumors at 16 weeks after urethane administration. E, representative gross pictures of lung surface tumors in Nrf2+/+ and Nrf2−/− mice. Arrows indicate the lung surface tumors. Scale bars, 10 mm. F, numbers of surface tumors in lungs of Nrf2+/+ and Nrf2−/− mice. Each dot represents total number of macroscopic tumors (φ > 0.5 mm) in individual mouse. G, number of large-size nodules (φ > 1.0 mm) in lungs of Nrf2+/+ and Nrf2−/− mice. The color of dots indicates size of the largest tumor in each mouse lung. H, average of tumor diameters in lungs of Nrf2+/+ and Nrf2−/− mice. The significant differences by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Close modal
Table 1.

Summary of urethane-induced lung carcinogenesis experiments

A. Short-term observation (8 weeks) after single urethane administration in Nrf2+/+ and Nrf2−/− mice 
 Incidence of lung surface tumors Average number of lung surface tumors per mouse 
Tumor size (mm) 0.5 < φ 
Nrf2+/+ (n = 10) 5/10 (50.0%) 0.8 ± 0.5 (n = 10) 
Nrf2−/− (n = 9) 9/9b (100.0%) 3.1 ± 1.1a (n = 9) 
B. Middle-term observation (16 weeks) after single urethane treatment in Nrf2+/+ and Nrf2−/− mice 
 Incidence of lung surface tumors Average number of lung surface tumors per mouse 
Tumor size (mm) 0.5 < φ 1.5 < φ 0.5 < φ 1.5 < φ 
Nrf2+/+ (n = 7) 7/7 (100.0%) 7/7 (100.0%) 18.1 ± 13.3 4.7 ± 3.4 
Nrf2−/− (n = 8) 8/8 (100.0%) 1/8a (12.5%) 11.3 ± 7.0 0.125 ± 0.3a 
C. Long-term observation (24 weeks) after four weekly contiguous urethane-treatment in Nrf2+/+ and Nrf2−/− mice 
 Incidence of lung surface tumors Number of lung surface tumors per mouse 
Tumor size (mm) 0.5 < φ < 2.0 2.0 < φ 0.5 < φ 2.0 < φ 
Nrf2+/+ (n = 10) 10/10 (100.0%) 6/10a (60.0%) 35.7 ± 22.5a 4.9 ± 6.59a 
Nrf2+/− (n = 5) 5/5 (100.0%) 2/5b (40.0%) 37.0 ± 10.9a 0.4 ± 0.55b 
Nrf2−/− (n = 10) 10/10 (100.0%) 0/10 (0.0%) 16.4 ± 12.6 0.0 ± 0.0 
A. Short-term observation (8 weeks) after single urethane administration in Nrf2+/+ and Nrf2−/− mice 
 Incidence of lung surface tumors Average number of lung surface tumors per mouse 
Tumor size (mm) 0.5 < φ 
Nrf2+/+ (n = 10) 5/10 (50.0%) 0.8 ± 0.5 (n = 10) 
Nrf2−/− (n = 9) 9/9b (100.0%) 3.1 ± 1.1a (n = 9) 
B. Middle-term observation (16 weeks) after single urethane treatment in Nrf2+/+ and Nrf2−/− mice 
 Incidence of lung surface tumors Average number of lung surface tumors per mouse 
Tumor size (mm) 0.5 < φ 1.5 < φ 0.5 < φ 1.5 < φ 
Nrf2+/+ (n = 7) 7/7 (100.0%) 7/7 (100.0%) 18.1 ± 13.3 4.7 ± 3.4 
Nrf2−/− (n = 8) 8/8 (100.0%) 1/8a (12.5%) 11.3 ± 7.0 0.125 ± 0.3a 
C. Long-term observation (24 weeks) after four weekly contiguous urethane-treatment in Nrf2+/+ and Nrf2−/− mice 
 Incidence of lung surface tumors Number of lung surface tumors per mouse 
Tumor size (mm) 0.5 < φ < 2.0 2.0 < φ 0.5 < φ 2.0 < φ 
Nrf2+/+ (n = 10) 10/10 (100.0%) 6/10a (60.0%) 35.7 ± 22.5a 4.9 ± 6.59a 
Nrf2+/− (n = 5) 5/5 (100.0%) 2/5b (40.0%) 37.0 ± 10.9a 0.4 ± 0.55b 
Nrf2−/− (n = 10) 10/10 (100.0%) 0/10 (0.0%) 16.4 ± 12.6 0.0 ± 0.0 

aP < 0.01 compared with wild-type mice.

bP < 0.05 compared with wild-type mice.

Nrf2−/− mice are resistant to urethane carcinogenesis in the middle-term observation period

Given the oncogenic function of NRF2 in human cancers (20), we hypothesized that Nrf2−/− tumor cells might have a lower proliferative potency than Nrf2+/+ tumor cells. To test this hypothesis, we conducted a middle-term observation (16 weeks; Supplementary Fig. S3). The total number of gross surface tumors (φ > 0.5 mm) per mouse was comparable between 2 genotypes (Fig. 2F and Table 1, B). However, when we determined the diameter of the largest tumors in the individual lung, we found that, while all Nrf2+/+ mice developed large tumors (φ > 1.5 mm), only 1 out of the 8 Nrf2−/− mice harbored such large tumors (Fig. 2E and G). Furthermore, when we measured all tumors in the individual lung, the average diameter of Nrf2+/+ tumors tended to be larger (average 1.2 mm) than those of Nrf2−/− mice (average 0.9 mm; Fig. 2H). These results show that the Nrf2−/− mice showed a lower susceptibility to urethane-induced carcinogenesis than the Nrf2+/+ mice at 16 weeks after the urethane administration.

Nrf2-deficient mice are resistant to urethane carcinogenesis in the long-term observation period

An observation at an even late stage revealed more significant difference in the cancer progression between Nrf2−/− and Nrf2+/+ mice. At 24 weeks after the urethane administration (Supplementary Fig. S3D), Nrf2−/− mice exhibited a markedly reduced number of tumors compared with the Nrf2+/+ mice (Fig. 3A and B and Table 1, C). In addition, the size of the tumors was larger in Nrf2+/+ mice than in Nrf2−/− mice. Indeed, 60% of mice in the Nrf2+/+ group exhibited large palpable nodules (φ > 2 mm), including some that were very large (φ > 5 mm), while no Nrf2−/− mice formed such large tumors (Fig. 3B). Consistent with this observation, the average tumor diameter of Nrf2+/+ tumors was larger than that of Nrf2−/− mice (1.2 vs. 0.8 mm, respectively; Fig. 3C). Importantly, the heterozygous (Nrf2+/−) mice showed an increased number of lung surface tumors compared with the Nrf2−/− mice (Fig. 3B), suggesting that the Nrf2 abundance is a critical determinant of the lung cancer growth.

Figure 3.

Urethane-induced lung carcinogenesis in the long-term observation protocol. A, gross observation of lungs (left) and hematoxylin and eosin staining of lung sections (right) of Nrf2+/+ and Nrf2−/− mice. Black scale bar, 10 mm; blue scale bar, 400 μm. B, number of lung surface tumors (φ > 1 mm) in the Nrf2+/+, Nrf2+/−, and Nrf2−/−-mice. The color of dots indicates size of the largest tumor in each mouse lung. C, average tumor diameter in each mouse group. D, representative pathologic photomicrographs of the urethane-induced lung tumors in the Nrf2+/+ and Nrf2−/− mice by hematoxylin and eosin (top), Ki-67 (middle), and PAS staining (bottom). Higher magnification images of each genotype are in the inlets. Black scale bars, 50 μm; blue scale bars, 10 μm. E, number of total adenocarcinomas in a section from each mouse. F, number of Ki67-positive cells in similar size tumors (φ 100 μm) of Nrf2+/+ and Nrf2−/− mice. G and H, Nrf2 is necessary for colonization and growth of urethane-induced tumors in nude mice. G, reduced growth of Nrf2−/− tumors compared with Nrf2+/+ tumors subcutaneously transplanted in nude mice. H, representative photographs (top) and photomicrographs (middle and bottom) from each genotype. Black scale bar, 10 mm; blue scale bar, 1 mm; white scale bar, 20 μm. The significant differences by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Figure 3.

Urethane-induced lung carcinogenesis in the long-term observation protocol. A, gross observation of lungs (left) and hematoxylin and eosin staining of lung sections (right) of Nrf2+/+ and Nrf2−/− mice. Black scale bar, 10 mm; blue scale bar, 400 μm. B, number of lung surface tumors (φ > 1 mm) in the Nrf2+/+, Nrf2+/−, and Nrf2−/−-mice. The color of dots indicates size of the largest tumor in each mouse lung. C, average tumor diameter in each mouse group. D, representative pathologic photomicrographs of the urethane-induced lung tumors in the Nrf2+/+ and Nrf2−/− mice by hematoxylin and eosin (top), Ki-67 (middle), and PAS staining (bottom). Higher magnification images of each genotype are in the inlets. Black scale bars, 50 μm; blue scale bars, 10 μm. E, number of total adenocarcinomas in a section from each mouse. F, number of Ki67-positive cells in similar size tumors (φ 100 μm) of Nrf2+/+ and Nrf2−/− mice. G and H, Nrf2 is necessary for colonization and growth of urethane-induced tumors in nude mice. G, reduced growth of Nrf2−/− tumors compared with Nrf2+/+ tumors subcutaneously transplanted in nude mice. H, representative photographs (top) and photomicrographs (middle and bottom) from each genotype. Black scale bar, 10 mm; blue scale bar, 1 mm; white scale bar, 20 μm. The significant differences by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Close modal

Showing very good agreement with the macroscopic observations, histologic examinations revealed that the Nrf2+/+ mice developed malignant adenocarcinomas at a higher frequency than the Nrf2−/− mice (Fig. 3D and E). Tumors in the Nrf2+/+ mice exhibited extensive invasion into the surrounding tissues, but Nrf2−/− tumors were minimally invasive with a clear border. Tumors in the Nrf2+/+ mice seemed to show a higher number of Ki-67–positive cells than those in the Nrf2−/− mice (Fig. 3D and F). Nrf2+/+ tumors exhibited accumulation of mucosubstances stained with periodic acid-Schiff (PAS; Fig. 3D), which correlates with progression to adenocarcinoma (21). Taken together, these observations show that Nrf2 plays important roles for the malignant transformation of lung adenomas in the late stage of carcinogenesis.

Urethane-induced lung tumors of Nrf2−/− mice failed to engraft in nude mice

One of the standard approaches in testing the tumorigenicity of cancer cells is to transplant the cells into immunodeficient mice (22). When highly metastatic 3LL cells were transplanted into Nrf2−/− mice, Nrf2 deficiency generated a more permissive microenvironment for cancer cell growth than in Nrf2+/+ mice (9). We, therefore, hypothesized that the less proliferative Nrf2-deficient tumor cells might be able to grow in a tumor-permissive microenvironment in the Nrf2−/− mice. To compare the cell-autonomous proliferative ability of Nrf2+/+ and Nrf2−/− tumor cells by excluding the host environmental factors, we excised lung tumors of approximately equal sizes (φ = 1 mm) from the Nrf2−/− and Nrf2+/+ mice and transplanted these tumors into nude mice. During a 5-month observation period, Nrf2−/− tumors failed to engraft and grow in the nude mice, whereas Nrf2+/+ tumors grew progressively to an approximately 50-fold increase in volume (Fig. 3G and H). These results show that Nrf2−/− tumor cells suffer from a cell-autonomous growth defect.

Nrf2-deficiency decreased the malignancy-risk gene signature

To clarify whether Nrf2 is functionally activated in urethane-induced tumors, we examined the expression of glutathione peroxide (Gpx2) and multidrug resistance–associated protein 4 (Mrp4) mRNAs, as both Gpx2 and Mrp4 contribute to the promotion of cancer (23, 24). The Gpx2 and Mrp4 mRNA expressions were significantly increased in the tumors compared with the normal lung tissues in Nrf2+/+ mice (Fig. 4A), supporting the notion that accumulated Nrf2 contributes to the proliferation of urethane-induced adenocarcinomas. To comprehensively examine changes in the gene expression profile, we conducted microarray analyses using the comparable size of lung tumors (1.0 mm < φ < 1.5 mm) and intact nontumor lung tissues derived either from Nrf2+/+ (n = 8) or Nrf2−/− mice (n = 8) at 16 weeks after the urethane treatment.

Figure 4.

Differential gene expression in the urethane-induced lung tumors (4 months after urethane treatment) of Nrf2−/− mice. A, expression of Gpx2 and Mrp4 in the tumors and nontumor regions of Nrf2−/− and Nrf2+/+ mice. B, Venn diagram of the differentially expressed genes in the Nrf2+/+ and Nrf2−/− tumors. The numbers of genes highly expressed in tumors compared with nontumor tissues of both genotype mice are shown together with the overlap in both genotypes of mice. C, clustering analysis of differentially expressed genes in the lung tumors of Nrf2+/+ and Nrf2−/− mice. Gene expression values (shown below B) using the color scheme (green-black-red) indicate low-moderate-high gene expression levels compared with the corresponding nontumor tissues. Heat map comparisons of genes preferentially expressed in Nrf2+/+ mouse tumors (left) and those preferentially expressed in Nrf2−/− tumors (right) are shown. D, qRT-PCR analysis of representative mRNAs for cancer-related and lung development-related genes. Data are presented as mean ± SD. The significant differences by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Figure 4.

Differential gene expression in the urethane-induced lung tumors (4 months after urethane treatment) of Nrf2−/− mice. A, expression of Gpx2 and Mrp4 in the tumors and nontumor regions of Nrf2−/− and Nrf2+/+ mice. B, Venn diagram of the differentially expressed genes in the Nrf2+/+ and Nrf2−/− tumors. The numbers of genes highly expressed in tumors compared with nontumor tissues of both genotype mice are shown together with the overlap in both genotypes of mice. C, clustering analysis of differentially expressed genes in the lung tumors of Nrf2+/+ and Nrf2−/− mice. Gene expression values (shown below B) using the color scheme (green-black-red) indicate low-moderate-high gene expression levels compared with the corresponding nontumor tissues. Heat map comparisons of genes preferentially expressed in Nrf2+/+ mouse tumors (left) and those preferentially expressed in Nrf2−/− tumors (right) are shown. D, qRT-PCR analysis of representative mRNAs for cancer-related and lung development-related genes. Data are presented as mean ± SD. The significant differences by Student t test are indicated (*, P < 0.05 and **, P < 0.01).

Close modal

Of the 114 genes increased in the lung tumor tissues, 57 genes were increased solely in the Nrf2+/+ tumors and 25 genes were increased solely in the Nrf2−/− tumors, whereas 32 genes were increased in both tumor genotypes (Fig. 4B and C; Supplementary Tables S2A–S2C for details). Lung adenocarcinomas tend to be associated with increased expression of a variety of lung development-related genes (21, 25). In the expression array data, we noticed upregulation of a series of lung development-related genes, including Sox9 (21, 26), Id2 (21), Nkx2-1 (27), Foxa2 (28), and N-myc (29) in the Nrf2+/+ tumors compared with the Nrf2−/− tumors (Fig. 4D). It has been reported that Sox9 accelerates malignant transformation through an increase in Cdk4 expression (26). Indeed, the increased expression of Sox9 in the Nrf2+/+ tumor was accompanied by an increase in Cdk4 expression. Similarly, cyclin D1 expression was more significantly increased in the Nrf2+/+ than in the Nrf2−/− tumors, and this observation is consistent with the theory that many oncogenic signaling pathways converge to elevate cyclin D1 expression at the transcription level (30).

Consistent with these results, we noticed that majority of the genes preferentially expressed in the Nrf2+/+ rather than in the Nrf2−/− tumor are classified into the Cellular Development, Cell Growth and Proliferation, and Embryonic Development categories by the Ingenuity Pathway Analysis (IPA) software analysis. Furthermore, this pathway analysis revealed that the Nrf2+/+ tumors were associated with more prominent activation of a number of carcinogenic or stem cell–related pathways, including Wnt/β-catenin signaling and Notch signaling compared with the Nrf2−/− tumors, suggesting substantial contribution of Nrf2 to cancer progression (Supplementary Fig. S4A and S4B).

Low frequency of constitutively active Kras mutations in Nrf2-deficient mice

Kras mutations are strongly associated with the progression of adenocarcinomas (29, 31) and Kras activation induces Nrf2 mRNA expression to exert its oncogenic activity as summarized in Fig. 5A (32). However, frequency of the Kras mutations in the Nrf2−/− cancer cells has not been evaluated. As urethane is known to evoke constitutively active mutations in Kras, particularly at codon 61 (CAA→CGA; Gln→Arg) in the second exon (33), we sequenced the second exon of the Kras gene in the Nrf2−/− lung tumors to clarify the somatic Kras mutation status (Fig. 5C). Importantly, all the Nrf2+/+ tumors exhibited codon 61 substitution, whereas only 1 out of 13 Nrf2−/− tumors showed a Kras mutation (Fig. 5D). We concomitantly observed an increase of mRNA abundance of Kras signaling pathway genes, including Erk1 and c-Myc, in the Nrf2+/+ lung tumors, whereas the mRNA levels of these genes were not significantly increased in the Nrf2−/− tumors (Fig. 5B). These results revealed that the Nrf2-deficient tumors were associated with a decreased frequency of Kras-activating mutations and diminished expression of the Kras pathway genes.

Figure 5.

Constitutively active Kras mutation frequently observed in the lung tumors of urethane-treated mice. A, schematic diagram of the Kras signaling pathway. Blue arrows indicate the genes highly induced in the Nrf2+/+ tumor. B, expression of mRNAs coding for representative Kras downstream factors. mRNAs were quantified by qRT-PCR analysis using lung tumors from Nrf2−/− and Nrf2+/+ mice 16 weeks after urethane treatment. C, left, the A-to-G substitution at the second nucleotide of codon 61 in the second exon of the Kras gene in the Nrf2+/+ tumors. Right, the Nrf2−/− tumors showed intact nucleotide sequences of codon 61. D, frequency of Kras codon-61 mutation (CAA→CGA; Gln→Arg) detected in the tumors from Nrf2−/− or Nrf2+/+ mice. Significant differences were determined using Fisher exact test (**, P < 0.01).

Figure 5.

Constitutively active Kras mutation frequently observed in the lung tumors of urethane-treated mice. A, schematic diagram of the Kras signaling pathway. Blue arrows indicate the genes highly induced in the Nrf2+/+ tumor. B, expression of mRNAs coding for representative Kras downstream factors. mRNAs were quantified by qRT-PCR analysis using lung tumors from Nrf2−/− and Nrf2+/+ mice 16 weeks after urethane treatment. C, left, the A-to-G substitution at the second nucleotide of codon 61 in the second exon of the Kras gene in the Nrf2+/+ tumors. Right, the Nrf2−/− tumors showed intact nucleotide sequences of codon 61. D, frequency of Kras codon-61 mutation (CAA→CGA; Gln→Arg) detected in the tumors from Nrf2−/− or Nrf2+/+ mice. Significant differences were determined using Fisher exact test (**, P < 0.01).

Close modal

Secreted phosphoprotein 1 or osteopontin (Spp1) is a secreted glycoprotein highly expressed in several types of cancers and precancerous lesions. The high-level expression of Spp1 is frequently associated with a high-grade malignancy of lung adenocarcinomas and poor clinical prognosis of the patients (34). Spp1-deficient mice are resistant to chemically induced skin tumorigenesis, suggesting the oncogenic function of Spp1 (35). We found that Spp1 expression was 2.5-fold higher in Nrf2−/− tumors than in Nrf2+/+ tumors (Fig. 4D). A microarray analysis revealed that a number of other lung cancer–related genes, such as Grin2A (36), Itgav (37), and Ptk6 (38), were preferentially induced in the Nrf2−/− tumors. These data suggest that the Nrf2−/− cancer cells develop through Kras-independent oncogenic pathways.

Nrf2-deficient cancers show reduced potency for activation of MDSCs

MDSCs are potent immunosuppressor cells, which are increased in many types of cancer hosts and create immune tolerance to cancers (39). The intracellular ROS level primarily determines the immunosuppressive activity of MDSCs, which decreases the CD8-mediated cancer immune response through peroxynitrite modification of the T-cell receptor (TCR)–CD8 complex (40). We previously found that transplantation of 3LL cells induced abundant ROS accumulation in the MDSCs population of the Nrf2−/− host mice, which led to the generation of a tumor-permissive microenvironment (9). To delineate changes in the host immune microenvironment after urethane treatment, we examined ROS levels in the MDSCs' fraction (MDSCs-ROS) and population of CD8+-T-cells at 16 weeks after the urethane administration, when the urethane-induced tumor numbers were comparable between Nrf2−/− and Nrf2+/+ mice. The tumor-bearing Nrf2+/+ mice showed increased MDSC-ROS levels compared with the concurrently vehicle-treated mice (Fig. 6A). However, the MDSC-ROS levels in the tumor-bearing Nrf2−/− mice were only slightly increased relative to the vehicle-treated mice.

Figure 6.

Nrf2-deficient cancers show reduced potency for activation of MDSCs. Nrf2−/− mice harboring urethane-induced lung tumors exhibit a lower level of ROS accumulation in MDSCs. A, quantification of ROS levels of the Mac1+Gr1+ MDSCs fraction in the bone marrow cells from Nrf2−/− or Nrf2+/+ mice harboring lung tumors (left). Data are presented as the median ± SD (**, P < 0.01 by Student t test; n = 3 in each group). Representative histogram of ROS level analysis with DCFDA in MDSCs from bone marrow in each group (right). B, GM-CSF expression level was significantly repressed in the Nrf2−/− tumors in comparison with the Nrf2+/+ tumors. C–F, splenic CD8+-T-cell population was decreased in the tumor-bearing Nrf2+/+ mice compared with the tumor-bearing Nrf2−/− mice. C, representative flow cytometric dot plots of spleen cells using CD4 and CD8 antibodies. The green frames indicate the CD8+-T-cell population. D, the CD4+-T-cell population was comparable between both genotype groups. E, splenic CD8+-T-cell population was decreased in the Nrf2+/+ mice bearing tumors more significantly than in the vehicle-treated Nrf2+/+ mice. F, population of CD8+-T cells are normalized with that of CD4+-T cells. Data are presented as the median ± SD (*, P < 0.05 using Student t test; n = 3 in each group).

Figure 6.

Nrf2-deficient cancers show reduced potency for activation of MDSCs. Nrf2−/− mice harboring urethane-induced lung tumors exhibit a lower level of ROS accumulation in MDSCs. A, quantification of ROS levels of the Mac1+Gr1+ MDSCs fraction in the bone marrow cells from Nrf2−/− or Nrf2+/+ mice harboring lung tumors (left). Data are presented as the median ± SD (**, P < 0.01 by Student t test; n = 3 in each group). Representative histogram of ROS level analysis with DCFDA in MDSCs from bone marrow in each group (right). B, GM-CSF expression level was significantly repressed in the Nrf2−/− tumors in comparison with the Nrf2+/+ tumors. C–F, splenic CD8+-T-cell population was decreased in the tumor-bearing Nrf2+/+ mice compared with the tumor-bearing Nrf2−/− mice. C, representative flow cytometric dot plots of spleen cells using CD4 and CD8 antibodies. The green frames indicate the CD8+-T-cell population. D, the CD4+-T-cell population was comparable between both genotype groups. E, splenic CD8+-T-cell population was decreased in the Nrf2+/+ mice bearing tumors more significantly than in the vehicle-treated Nrf2+/+ mice. F, population of CD8+-T cells are normalized with that of CD4+-T cells. Data are presented as the median ± SD (*, P < 0.05 using Student t test; n = 3 in each group).

Close modal

Consistent with the increase of MDSC-ROS levels, the number of splenic CD8+-T-cells was markedly decreased in the tumor-bearing Nrf2+/+ mice (Fig. 6C and E). In contrast, the CD8+-T-cell population was not changed substantially in the Nrf2−/− mice regardless of tumor progression. The number of CD4+-T-cells was almost equivalent, irrespective of tumor bearing in both mouse genotypes (Fig. 6C and D). We normalized the population of CD8+-T-cells with that of CD4+-T-cells and further confirmed the preferential reduction of CD8+-T-cell population in the tumor-bearing Nrf2+/+ mice (Fig. 6F). These observations emphasize that MDSCs predominantly suppress CD8+-T-cell proliferation in the tumor-bearing Nrf2+/+ mice (9).

It has been reported that malignant cancers release a set of soluble factors, including granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), interleukin (IL-3), IL-6, and VEGF, which facilitate the recruitment and ROS accumulation in MDSCs (41). We observed that GM-CSF expression was more highly increased in Nrf2+/+ tumors than in Nrf2−/− tumors (Fig. 6B). Overall, these results argue that Nrf2−/− tumors have reduced potency for the induction of ROS accumulation in MDSCs and, therefore, the CD8+-T-cell immunity is barely diminished in the tumor-bearing Nrf2−/− mice.

In this study, we have shown that Nrf2−/− mice are sensitive to chemical carcinogens and exhibit a high-level of cancer initiation in the early stages of urethane-induced lung carcinogenesis. In stark contrast, in the late stages of urethane carcinogenesis, the Nrf2−/− mice developed cancers that were significantly less malignant than did the control Nrf2+/+ mice. These results indicate that the Nrf2 activity accelerates the malignant transformation of benign adenoma to adenocarcinoma. The molecular basis of the Nrf2 contribution to the urethane carcinogenesis is summarized in Supplementary Fig. S5.

The constitutively active Kras mutations have been identified in many types of human cancers, including those of the lung, pancreas, and colon, as well as in experimental cancer models of rodents (42). Urethane is known to cause Kras codon 61 mutations in multiple mouse strains (33). An important observation is that Kras activates Nrf2 and leads to the cancer by malignant transformation (32). Therefore, in the Nrf2−/− mice, even if lung cells suffer a large number of initial genetic hits by the urethane metabolites, subsequent progression of malignant transformation is markedly attenuated because of the lack of Nrf2 activity.

In contrast, several chemical carcinogens are known to rarely induce activating Kras mutations. Such carcinogens include N-nitrosobutyl (4-hydroxybutyl) amine, azoxymethane, and 7,12-dimethyl-benz[a]anthracene. Notably, these carcinogens are found to provoke cancers more abundantly in the Nrf2−/− mice than Nrf2+/+ mice (3, 6, 43). We surmise that these Kras-independent carcinogens evoke a higher number of cancers in the Nrf2−/− mice as a direct consequence of the increased chemical susceptibility and activation of other oncogenic pathways.

Spp1, an integrin-binding and cell transformation-related protein, highly associates with the malignancy of non–small cell lung carcinoma (34). We found that Spp1 is much more abundantly expressed in Nrf2−/− than in Nrf2+/+ tumors, suggesting that Spp1 contributes to lung tumorigenesis in Nrf2−/− mice. Because forced expression of Nrf2 in a chondrocyte cell line significantly decreased Spp1 mRNA expression (44), Nrf2 may act as a negative regulator for Spp1 gene expression. These results, thus, imply that Spp1 may comprise one alternative oncogenic pathway that replaces the Kras-Nrf2 pathway.

Nrf2 activators have been shown to exert preventive effects against various types of carcinogens in animal models and humans (45, 46). In addition, therapeutic efficacy of a potent Nrf2 activator, bardoxolone methyl (CDDO-Me), was shown for the treatment of chronic kidney disease and diabetic nephropathy in a large clinical trial (47). Our results unequivocally showed that Nrf2 is critical for the prevention of the initiation step of lung carcinogenesis. Therefore, preventive treatment that attains Nrf2 activation seems to reduce the cancer initiation and would be of particular value for those who are at high risk of lung cancers, such as those with a history of heavy smoking and inhalation of asbestos. On the other hand, treatment with Nrf2 inhibitors through the lung cancer-targeted drug delivery system would reduce the cancer lesions associated with oncogenic Kras mutation. Overall, this study has revealed that Nrf2 is a prime candidate of personalized cancer treatment in the near future, and development of Nrf2 inhibitors and activators, as well as accurate diagnostic procedures quantifying Nrf2 expression levels in lung cancer cells, are critically important for this purpose.

No conflicts of interest were disclosed.

Conception and design: H. Satoh, T. Moriguchi, M. Yamamoto

Development of methodology: H. Satoh, T. Moriguchi, J. Takai

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Satoh, T. Moriguchi, J. Takai

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Satoh, T. Moriguchi, J. Takai

Writing, review, and/or revision of the manuscript: H. Satoh, T. Moriguchi, M. Yamamoto

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Satoh, T. Moriguchi

Study supervision: T. Moriguchi, M. Ebina, M. Yamamoto

The authors thank Drs. Hozumi Motohashi, Michito Hamada, and Jon Maher for their insightful advice and helpful discussions.

This work was financially supported in part by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology and the Japan Society for Promotion of Science (T. Moriguchi and M. Yamamoto), Scientific Research on Priority Areas (M. Yamamoto), and Specially Promoted Research (M. Yamamoto). H. Satoh was supported by Research Resident Fellowship from the Promotion of Cancer Research Japan for the Third Term Comprehensive 10-Year Strategy for Cancer Control.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Itoh
K
,
Mimura
J
,
Yamamoto
M
. 
Discovery of the negative regulator of Nrf2, Keap1: a historical overview
.
Antioxid Redox Signal
2010
;
13
:
1665
78
.
2.
Itoh
K
,
Chiba
T
,
Takahashi
S
,
Ishii
T
,
Igarashi
K
,
Katoh
Y
, et al
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
22
.
3.
Iida
K
,
Itoh
K
,
Kumagai
Y
,
Oyasu
R
,
Hattori
K
,
Kawai
K
, et al
Nrf2 is essential for the chemopreventive efficacy of oltipraz against urinary bladder carcinogenesis
.
Cancer Res
2004
;
64
:
6424
31
.
4.
Osburn
WO
,
Karim
B
,
Dolan
PM
,
Liu
G
,
Yamamoto
M
,
Huso
DL
, et al
Increased colonic inflammatory injury and formation of aberrant crypt foci in Nrf2-deficient mice upon dextran sulfate treatment
.
Int J Cancer
2007
;
121
:
1883
91
.
5.
Ramos-Gomez
M
,
Kwak
MK
,
Dolan
PM
,
Itoh
K
,
Yamamoto
M
,
Talalay
P
, et al
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
5
.
6.
Xu
C
,
Huang
MT
,
Shen
G
,
Yuan
X
,
Lin
W
,
Khor
TO
, et al
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
6
.
7.
Shibata
T
,
Ohta
T
,
Tong
KI
,
Kokubu
A
,
Odogawa
R
,
Tsuta
K
, et al
Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy
.
Proc Natl Acad Sci U S A
2008
;
105
:
13568
73
.
8.
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
4
.
9.
Satoh
H
,
Moriguchi
T
,
Taguchi
K
,
Takai
J
,
Maher
JM
,
Suzuki
T
, et al
Nrf2-deficiency creates a responsive microenvironment for metastasis to the lung
.
Carcinogenesis
2010
;
31
:
1833
43
.
10.
Horio
Y
,
Chen
A
,
Rice
P
,
Roth
JA
,
Malkinson
AM
,
Schrump
DS
. 
Ki-ras and p53 mutations are early and late events, respectively, in urethane-induced pulmonary carcinogenesis in A/J mice
.
Mol Carcinog
1996
;
17
:
217
23
.
11.
Anderson
N
,
Paul
SH
,
Henry
SM
. 
Induction of pulmonary tumors in mice with ethyl carbamate (urethane)
.
J Natl Cancer Inst
1943
;
4
:
309
19
.
12.
Ghanayem
BI
,
Hoffler
U
. 
Investigation of xenobiotics metabolism, genotoxicity, and carcinogenicity using Cyp2e1(−/−) mice
.
Curr Drug Metab
2007
;
8
:
728
49
.
13.
Kemper
RA
,
Myers
SR
,
Hurst
HE
. 
Detoxification of vinyl carbamate epoxide by glutathione: evidence for participation of glutathione S-transferases in metabolism of ethyl carbamate
.
Toxicol Appl Pharmacol
1995
;
135
:
110
8
.
14.
Hoffler
U
,
Ghanayem
BI
. 
Increased bioaccumulation of urethane in CYP2E1−/− versus CYP2E1+/+ mice
.
Drug Metab Dispos
2005
;
33
:
1144
50
.
15.
Gebel
S
,
Diehl
S
,
Pype
J
,
Friedrichs
B
,
Weiler
H
,
Schuller
J
, et al
The transcriptome of Nrf2−/− mice provides evidence for impaired cell cycle progression in the development of cigarette smoke-induced emphysematous changes
.
Toxicol Sci
2010
;
115
:
238
52
.
16.
Ihara
S
,
Kida
H
,
Arase
H
,
Tripathi
LP
,
Chen
YA
,
Kimura
T
, et al
Inhibitory roles of signal transducer and activator of transcription 3 in antitumor immunity during carcinogen-induced lung tumorigenesis
.
Cancer Res
2012
;
72
:
2990
9
.
17.
Stathopoulos
GT
,
Sherrill
TP
,
Cheng
DS
,
Scoggins
RM
,
Han
W
,
Polosukhin
VV
, et al
Epithelial NF-kappaB activation promotes urethane-induced lung carcinogenesis
.
Proc Natl Acad Sci U S A
2007
;
104
:
18514
9
.
18.
Forkert
PG
. 
Mechanisms of lung tumorigenesis by ethyl carbamate and vinyl carbamate
.
Drug Metab Rev
2010
;
42
:
355
78
.
19.
Cho
HY
,
Kleeberger
SR
. 
Nrf2 protects against airway disorders
.
Toxicol Appl Pharmacol
2010
;
244
:
43
56
.
20.
Mitsuishi
Y
,
Taguchi
K
,
Kawatani
Y
,
Shibata
T
,
Nukiwa
T
,
Aburatani
H
, et al
Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming
.
Cancer Cell
2012
;
22
:
66
79
.
21.
Pacheco-Pinedo
EC
,
Durham
AC
,
Stewart
KM
,
Goss
AM
,
Lu
MM
,
Demayo
FJ
, et al
Wnt/beta-catenin signaling accelerates mouse lung tumorigenesis by imposing an embryonic distal progenitor phenotype on lung epithelium
.
J Clin Invest
2011
;
121
:
1935
45
.
22.
Cardoso
CC
,
Bornstein
SR
,
Hornsby
PJ
. 
New methods for investigating experimental human adrenal tumorigenesis
.
Mol Cell Endocrinol
2009
;
300
:
175
9
.
23.
Brigelius-Flohe
R
,
Kipp
A
. 
Glutathione peroxidases in different stages of carcinogenesis
.
Biochim Biophys Acta
2009
;
1790
:
1555
68
.
24.
Borst
P
,
Evers
R
,
Kool
M
,
Wijnholds
J
. 
A family of drug transporters: the multidrug resistance-associated proteins
.
J Natl Cancer Inst
2000
;
92
:
1295
302
.
25.
Borczuk
AC
,
Gorenstein
L
,
Walter
KL
,
Assaad
AA
,
Wang
L
,
Powell
CA
. 
Non-small-cell lung cancer molecular signatures recapitulate lung developmental pathways
.
Am J Pathol
2003
;
163
:
1949
60
.
26.
Jiang
SS
,
Fang
WT
,
Hou
YH
,
Huang
SF
,
Yen
BL
,
Chang
JL
, et al
Upregulation of SOX9 in lung adenocarcinoma and its involvement in the regulation of cell growth and tumorigenicity
.
Clin Cancer Res
2010
;
16
:
4363
73
.
27.
Weir
BA
,
Woo
MS
,
Getz
G
,
Perner
S
,
Ding
L
,
Beroukhim
R
, et al
Characterizing the cancer genome in lung adenocarcinoma
.
Nature
2007
;
450
:
893
8
.
28.
Wan
H
,
Dingle
S
,
Xu
Y
,
Besnard
V
,
Kaestner
KH
,
Ang
SL
, et al
Compensatory roles of Foxa1 and Foxa2 during lung morphogenesis
.
J Biol Chem
2005
;
280
:
13809
16
.
29.
Meuwissen
R
,
Berns
A
. 
Mouse models for human lung cancer
.
Genes Dev
2005
;
19
:
643
64
.
30.
Pestell
RG
,
Albanese
C
,
Reutens
AT
,
Segall
JE
,
Lee
RJ
,
Arnold
A
. 
The cyclins and cyclin-dependent kinase inhibitors in hormonal regulation of proliferation and differentiation
.
Endocr Rev
1999
;
20
:
501
34
.
31.
Lee
GH
. 
The Kras2 oncogene and mouse lung carcinogenesis
.
Med Mol Morphol
2008
;
41
:
199
203
.
32.
DeNicola
GM
,
Karreth
FA
,
Humpton
TJ
,
Gopinathan
A
,
Wei
C
,
Frese
K
, et al
Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis
.
Nature
2011
;
475
:
106
9
.
33.
Dragani
TA
,
Manenti
G
,
Pierotti
MA
. 
Genetics of murine lung tumors
.
Adv Cancer Res
1995
;
67
:
83
112
.
34.
Chambers
AF
,
Wilson
SM
,
Kerkvliet
N
,
O'Malley
FP
,
Harris
JF
,
Casson
AG
. 
Osteopontin expression in lung cancer
.
Lung Cancer
1996
;
15
:
311
23
.
35.
Hsieh
YH
,
Juliana
MM
,
Hicks
PH
,
Feng
G
,
Elmets
C
,
Liaw
L
, et al
Papilloma development is delayed in osteopontin-null mice: implicating an antiapoptosis role for osteopontin
.
Cancer Res
2006
;
66
:
7119
27
.
36.
Okayama
H
,
Kohno
T
,
Ishii
Y
,
Shimada
Y
,
Shiraishi
K
,
Iwakawa
R
, et al
Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas
.
Cancer Res
2012
;
72
:
100
11
.
37.
Desgrosellier
JS
,
Cheresh
DA
. 
Integrins in cancer: biological implications and therapeutic opportunities
.
Nat Rev Cancer
2010
;
10
:
9
22
.
38.
Brauer
PM
,
Tyner
AL
. 
Building a better understanding of the intracellular tyrosine kinase PTK6 - BRK by BRK
.
Biochim Biophys Acta
2010
;
1806
:
66
73
.
39.
Gabrilovich
DI
,
Nagaraj
S
. 
Myeloid-derived suppressor cells as regulators of the immune system
.
Nat Rev Immunol
2009
;
9
:
162
74
.
40.
Nagaraj
S
,
Gupta
K
,
Pisarev
V
,
Kinarsky
L
,
Sherman
S
,
Kang
L
, et al
Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer
.
Nat Med
2007
;
13
:
828
35
.
41.
Liu
CY
,
Wang
YM
,
Wang
CL
,
Feng
PH
,
Ko
HW
,
Liu
YH
, et al
Population alterations of l-arginase- and inducible nitric oxide synthase-expressed CD11b+/CD14/CD15+/CD33+ myeloid-derived suppressor cells and CD8+ T lymphocytes in patients with advanced-stage non-small cell lung cancer
.
J Cancer Res Clin Oncol
2010
;
136
:
35
45
.
42.
Schubbert
S
,
Shannon
K
,
Bollag
G
. 
Hyperactive Ras in developmental disorders and cancer
.
Nat Rev Cancer
2007
;
7
:
295
308
.
43.
Khor
TO
,
Huang
MT
,
Prawan
A
,
Liu
Y
,
Hao
X
,
Yu
S
, et al
Increased susceptibility of Nrf2 knockout mice to colitis-associated colorectal cancer
.
Cancer Prev Res (Phila Pa)
2008
;
1
:
187
91
.
44.
Hinoi
E
,
Takarada
T
,
Fujimori
S
,
Wang
L
,
Iemata
M
,
Uno
K
, et al
Nuclear factor E2 p45-related factor 2 negatively regulates chondrogenesis
.
Bone
2007
;
40
:
337
44
.
45.
Kensler
TW
,
Wakabayashi
N
. 
Nrf2: friend or foe for chemoprevention?
Carcinogenesis
2010
;
31
:
90
9
.
46.
Zhang
Y
,
Munday
R
. 
Dithiolethiones for cancer chemoprevention: where do we stand?
Mol Cancer Ther
2008
;
7
:
3470
9
.
47.
Pergola
PE
,
Raskin
P
,
Toto
RD
,
Meyer
CJ
,
Huff
JW
,
Grossman
EB
, et al
Bardoxolone methyl and kidney function in CKD with type 2 diabetes
.
N Engl J Med
2011
;
365
:
327
36
.