Nrf2 has an anti-carcinogenic effect. However, an increase in Nrf2 activity is also implicated in cancer chemoresistance. A switch from E-cadherin to N-cadherin affects the transdifferentiation and metastasis of cancer cells. In view of the key role of this switch in cancer malignancy, we investigated the regulatory effect of E-cadherin on Nrf2. In HEK293 cells, overexpression of E-cadherin inhibited the nuclear accumulation of Nrf2, and prevented Nrf2-dependent gene induction. GST pull-down and immunocytochemical assays verified the interaction between E-cadherin and Nrf2: E-cadherin bound the C-terminus of Nrf2, but not its N-terminus, which comprises the Neh2 domain responsible for phosphorylation of Ser40. Our finding that the mutation of Ser40 to alanine in Nrf2 did not affect the ability of E-cadherin to bind Nrf2 and repress target gene transactivation suggests that E-cadherin might not disturb the phosphorylation. Studies using mutant constructs of E-cadherin suggested that the β-catenin-binding domain contributes to the inhibitory effect of E-cadherin on Nrf2. Consistently, knockdown of β-catenin attenuated not only the effect of E-cadherin binding to Nrf2, but also Keap1-dependent ubiquitylation of Nrf2, and thereby increased Nrf2 activity, supporting the involvement of β-catenin in the interactions. Collectively, E-cadherin recruits Nrf2 through β-catenin, and assists the function of Keap1 for the inhibition of nuclear localization and transcriptional activity of Nrf2. In HepG2 cells, the loss of E-cadherin by either siRNA knockdown or treatment with TGFβ1 enhanced the constitutive or inducible activity of Nrf2, implying that chemoresistance of cancer cells upon the loss of E-cadherin might be associated with Nrf2.

An excess of reactive oxygen species (ROS) or reactive nitrogen species (RNS) causes injuries on molecules and components of the cell. As an adaptive response, NF-E2-related factor-2 (Nrf2), which is a member of the cap’n’collar (CNC) family of basic leucine zipper proteins, regulates antioxidant gene expression. Therefore, Nrf2 might serve as a transcription factor responsible for the induction of detoxifying enzymes and consequently for the prevention of chemical carcinogenesis (Jeong et al., 2006; Satoh et al., 2010). However, there is controversy on the role of Nrf2 in the progression of cancer; a series of studies have shown that mutational activation of Nrf2 might cause malignancy and increase chemoresistance (Ikeda et al., 2004; Lau et al., 2008; Wang et al., 2008). Thus, Nrf2 is somewhat of a double-edged sword in cancer biology. Because the turnover and activation processes of Nrf2 are linked to cellular molecule(s) and component(s) that have differential binding affinities, the upstream regulators of Nrf2 expressed in a cell-type-specific manner need to be identified, particularly in association with cancer resistance.

Cadherins are the proteins responsible for cell–cell adhesion at the adherens junctions, and their expression affects cell morphogenesis, rearrangements, migration and epithelial–mesenchymal transition (EMT) (Gumbiner, 2005). Cadherin switching from E-cadherin to N-cadherin is a key feature of the EMT process: the loss of E-cadherin and/or transcriptional repression of bone morphogenetic protein 7 (BMP-7) and inhibitor of DNA binding 2 (Id2) represent EMT. Loss of E-cadherin might also be associated with the resistance of cancer cells to chemotherapy because it promotes cancer progression and metastasis (Arumugam et al., 2009; Huber et al., 2005; Wheelock et al., 2008). Oxidative stress disrupts the E-cadherin and catenin cell-adhesion complex (Parrish et al., 1999; Rao et al., 2002). Thus, it is plausible that E-cadherin might affect Nrf2 activity in response to oxidative stress. This concept is particularly important because a mutational increase in Nrf2 activity might stimulate tumor growth and metastasis. However, the link between E-cadherin and modulation of Nrf2 activity has never been explored.

Catenins link E-cadherin to the actin cytoskeleton in cell adhesion (Gumbiner, 2005). In addition, E-cadherin regulates actin assembly and organization at cadherin-mediated cell–cell adhesion sites through catenins (Drees et al., 2005). In particular, the N-terminal region of β-catenin binds with α-catenin, which interacts both indirectly and directly with the actin cytoskeleton (Wheelock and Johnson, 2003). β-Catenin interacts with the distal region of the cytoplasmic domain of E-cadherin. Previously, we reported that depolymerization of actin filaments in response to oxidative stress causes a complex of Nrf2 and actin to translocate into the nucleus to effect transactivation of target genes (Kang et al., 2002). Moreover, as a negative regulator of Nrf2, kelch-like ECH-associated protein 1 (Keap1) has the ability to bind actin filaments as a scaffold protein in the cytoplasm under unstressed conditions (Kang et al., 2004). Because E-cadherin regulates actin dynamics, it might affect the activity of Nrf2 through its binding to actin and/or Keap1.

In view of the importance of E-cadherin for the signaling of cell growth and transdifferentiation, and the lack of clear understanding of the link between E-cadherin and Nrf2, this study investigated whether E-cadherin affects Nrf2 activity and if so, what the molecular basis is. Our findings revealed a novel inhibitory effect of E-cadherin on Nrf2, also showing that E-cadherin might enhance the interaction between Nrf2 and Keap1. Moreover, the results of this study identified β-catenin as a molecule necessary for complex formation of E-cadherin with Nrf2 and Keap1. Our data presented here reveal the ability of E-cadherin to recruit Nrf2, which creates an inactive pool of Nrf2 in association with β-catenin binding.

Prevention of nuclear accumulation of Nrf2 by E-cadherin

First, we sought to determine the effect of enforced E-cadherin expression on the activity of Nrf2 in HEK293 cells. Immunoblot analysis showed that overexpression of E-cadherin prevented nuclear accumulation of Nrf2 (Fig. 1A). Overexpressed Nrf2 had a molecular mass of ∼110 kDa (supplementary material Fig. S1A); endogenous Nrf2 was detected at ∼66 kDa and ∼110 kDa, although its predicted molecular mass is 66 kDa, presumably as a result of post-translational modification (Taguchi et al., 2011). To functionally assess the effect of E-cadherin on Nrf2 activity, we examined the expression of NAD(P)H quinone oxidoreductase 1 (NQO1) and hemeoxygenase-1 (HO-1) (Fig. 1B). As expected, overexpression of E-cadherin reduced the mRNA and protein levels of Nrf2 target genes. In addition, E-cadherin expression decreased the basal or tert-butylhydroquinone (t-BHQ)-inducible luciferase expression from the pGL-1651 reporter gene construct, which contains an antioxidant response element (ARE) in the promoter region (Fig. 1C, left). E-cadherin also prevented the Nrf2-induced pGL-1651 luciferase activity (Fig. 1C, right). The lack of inhibition of luciferase expression by E-cadherin from pGL-1651-ΔARE, a reporter construct with a deletion mutation in ARE, verified the role of Nrf2 in reporter gene expression. In HepG2 cells, a cell line that has a higher level of endogenous E-cadherin than HEK293 cells (supplementary material Fig. S1B), E-cadherin knockdown using siRNA significantly enhanced the basal or sulforaphane (5 μM, for 6 hours)-inducible nuclear accumulation of Nrf2 (Fig. 1D). Consistently, luciferase activity from the NQO1-ARE reporter construct was increased by a deficiency in E-cadherin, confirming the impact of endogenous E-cadherin on the activity of Nrf2. All of these results indicate that E-cadherin represses the activity of Nrf2 by inhibiting nuclear accumulation of Nrf2.

To explore the possibility of an interaction between E-cadherin and Nrf2, the ability of E-cadherin to bind Nrf2 was determined in HEK293 cells in which E-cadherin was overexpressed. Ectopically expressed E-cadherin bound with Nrf2 (Fig. 2A); as expected, Nrf2 interacted with Keap1. Of note, E-cadherin was also capable of interacting with Keap1, a protein that sequesters Nrf2 for its ubiquitylation and facilitates proteasomal degradation of ubiquitylated Nrf2 (Kobayashi et al., 2004). In HepG2 cells, the endogenous E-cadherin also interacted with Nrf2 and Keap1 (Fig. 2B). Moreover, in vitro GST pull-down assays confirmed the ability of in vitro translated [35S]E-cadherin to bind Nrf2, as did [35S]Keap1 (Fig. 2C, middle). In addition, we verified the ability of E-cadherin to directly bind with Keap1 (Fig. 2C, right). These results indicate that E-cadherin has the ability to bind Nrf2 and Keap1, presumably facilitating the interaction between Nrf2 and Keap1.

Immunocytochemical analysis showed that ectopically expressed Nrf2 was predominantly located in the nuclei but that simultaneously expressed E-cadherin prevented nuclear accumulation of Nrf2 (Fig. 2D), confirming the functional role of E-cadherin in inhibiting nuclear localization of Nrf2. As expected, endogenously expressed E-cadherin in the plasma membrane of HepG2 cells also interacted with Nrf2 (Fig. 2E). Collectively, our findings show that E-cadherin prevents nuclear accumulation of Nrf2 as a consequence of their interaction close to the plasma membrane.

E-cadherin binds to the C-terminus of Nrf2

The C-terminus of Nrf2 comprises Neh1 and Neh3 domains that are responsible for heterodimerization of Nrf2 and small Maf and DNA binding (Itoh et al., 2010). To locate the region of Nrf2 responsible for its interaction with E-cadherin, immunoprecipitation and immunoblot assays were performed in cells transfected with truncated mutants of Nrf2 in combination with E-cadherin. Our findings indicated that the C-terminus, but not the N-terminus, of Nrf2 was capable of binding E-cadherin (Fig. 3A). PKC∂ phosphorylates Nrf2 at Ser40 residue, which is located in the Neh2 domain of the N-terminus, and this activation process is reciprocally coupled with ubiquitylation and degradation of Nrf2 by Keap1 (Niture et al., 2009). We wondered whether the phosphorylation of Nrf2 is affected by E-cadherin. As expected, t-BHQ treatment promoted Nrf2 activation, as indicated by a decrease in the binding of Keap1 and Nrf2 (Fig. 3B). However, the intensity of E-cadherin and Nrf2 binding was not diminished. Moreover, Ser40 to alanine (S40A) mutation of Nrf2 failed to affect this effect, suggesting that E-cadherin and Nrf2 binding might not be coupled with the phosphorylation of Nrf2. This hypothesis was further supported by our result that E-cadherin inhibited luciferase expression from pGL-1651 elicited by an S40A mutant of Nrf2 (Fig. 3C). In addition, we observed that endogenous E-cadherin had the ability to bind Nrf2 in HepG2 cells treated with t-BHQ or sulforaphane (Fig. 3D), supporting our proposal that E-cadherin binds Nrf2 independently of Nrf2 phosphorylation.

Role of the β-catenin-binding domain in E-cadherin

E-cadherin contains domains that interact with p120-catenin and β-catenin. To further understand the molecular basis of E-cadherin and Nrf2 binding, we measured the abilities of several mutant constructs of E-cadherin to bind Nrf2 and inhibit luciferase expression from the NQO1-ARE reporter containing three copies of ARE. Either a C-terminal fragment of E-cadherin (ECAD-CT) or a mutant of E-cadherin in the p120-catenin-binding domain (ECAD-CT-Δp120-catenin) interacted with Nrf2 (Fig. 4A). However, E-cadherin deficient in the β-catenin-binding domain (ECAD-CT-Δβ-catenin) showed a decreased binding with Nrf2. Likewise, ECAD-CT or ECAD-CT-Δp120-catenin decreased the Nrf2 induction of luciferase from NQO1-ARE to a similar extent as did full-length E-cadherin (i.e. luciferase activity from pGL-1651) (Fig. 4B). The induction of luciferase by Nrf2 was significantly higher in the cells transfected with ECAD-CT-Δβ-catenin than those transfected with either ECAD-CT or ECAD-CT-Δp120-catenin. Similarly, the percentage of cells showing Nrf2 localization in nuclei or nuclei+cytoplasm was 16.8%, 18.0% and 40.6% after transfection of the cells with ECAD-CT, ECAD-CT-Δp120-catenin and ECAD-CT-Δβ-catenin, respectively (Fig. 4C). All of these results indicate that the β-catenin-binding domain is associated with binding between E-cadherin and Nrf2.

Fig. 1.

The effect of E-cadherin overexpression on the activity of Nrf2. (A) Nrf2 was immunoblotted on the subcellular fractions of HEK293 cells transfected for 24 hours with plasmid(s) encoding Nrf2 and/or E-cadherin. ECAD, E-cadherin; HSP70, heat shock protein 70. (B) Quantitative real-time PCR and immunoblotting for Nrf2 target gene products. The levels of NQO1 and HO-1 mRNA and protein were measured in HEK293 cells transfected as described above (significantly different compared with mock transfection, **P<0.01; significantly different compared with Nrf2 transfection, ##P<0.01; n=3). (C) The effect of enforced expression of E-cadherin on luciferase expression from pGL-1651 or pGL-1651-ΔARE. A scheme shows the C/EBP or Nrf2 binding site (ARE) in pGL-1651 that contains the promoter region of the GSTA2 gene. pGL-1651-ΔARE represents a mutant construct of pGL-1651 in which ARE was specifically deleted. Luciferase expression from pGL-1651 or pGL-1651-ΔARE was measured in the cells transfected with Nrf2 and/or E-cadherin followed by vehicle or t-BHQ treatment, and was calculated as a relative change to that of β-galactosidase. Values represent the mean ± s.e. (significantly different compared with control, **P<0.01, n=4). (D) The effect of E-cadherin knockdown on the activation of Nrf2 by sulforaphane. The levels of Nrf2 and E-cadherin or NQO1-ARE luciferase activities were measured in HepG2 cells treated with 5 μM sulforaphane for 6 hours after transfection with siRNA directed against E-cadherin for 48 hours. si-ECAD, siRNA directed against E-cadherin (significantly different compared with siRNA-transfected control, *P<0.05, **P<0.01; n=3).

Fig. 1.

The effect of E-cadherin overexpression on the activity of Nrf2. (A) Nrf2 was immunoblotted on the subcellular fractions of HEK293 cells transfected for 24 hours with plasmid(s) encoding Nrf2 and/or E-cadherin. ECAD, E-cadherin; HSP70, heat shock protein 70. (B) Quantitative real-time PCR and immunoblotting for Nrf2 target gene products. The levels of NQO1 and HO-1 mRNA and protein were measured in HEK293 cells transfected as described above (significantly different compared with mock transfection, **P<0.01; significantly different compared with Nrf2 transfection, ##P<0.01; n=3). (C) The effect of enforced expression of E-cadherin on luciferase expression from pGL-1651 or pGL-1651-ΔARE. A scheme shows the C/EBP or Nrf2 binding site (ARE) in pGL-1651 that contains the promoter region of the GSTA2 gene. pGL-1651-ΔARE represents a mutant construct of pGL-1651 in which ARE was specifically deleted. Luciferase expression from pGL-1651 or pGL-1651-ΔARE was measured in the cells transfected with Nrf2 and/or E-cadherin followed by vehicle or t-BHQ treatment, and was calculated as a relative change to that of β-galactosidase. Values represent the mean ± s.e. (significantly different compared with control, **P<0.01, n=4). (D) The effect of E-cadherin knockdown on the activation of Nrf2 by sulforaphane. The levels of Nrf2 and E-cadherin or NQO1-ARE luciferase activities were measured in HepG2 cells treated with 5 μM sulforaphane for 6 hours after transfection with siRNA directed against E-cadherin for 48 hours. si-ECAD, siRNA directed against E-cadherin (significantly different compared with siRNA-transfected control, *P<0.05, **P<0.01; n=3).

Fig. 2.

Interaction of E-cadherin with Nrf2. (A) Immunoprecipitation and immunoblot assays. Nrf2 or E-cadherin immunoprecipitates were immunoblotted for the protein of interest in HEK293 cells transfected with E-cadherin or E-cadherin plus Nrf2. Overexpressed Nrf2 has a molecular mass of ∼110 kDa, whereas endogenous Nrf2 is smaller (i.e. predicted molecular mass of human Nrf2, 66 kDa). Asterisk indicates a nonspecific band. Keap1 immunoprecipitates were immunoblotted for E-cadherin in HEK293 cells transfected with E-cadherin or E-cadherin plus Keap1. IP, immunoprecipitation. (B) Interaction of endogenous E-cadherin with Nrf2 in HepG2 cells. E-cadherin was immunoblotted on IgG or Nrf2 immunoprecipitates, whereas either Nrf2 or Keap1 was blotted on IgG or E-cadherin immunoprecipitates. Input contained 10 μg of proteins. (C) In vitro binding of E-cadherin with Nrf2 or Keap1. GST pull-down assays were performed using in vitro translated [35S]E-cadherin, [35S]Keap1 or [35S]Nrf2, and bacterially expressed GST–Nrf2 or GST–Keap1. GST was used as a negative control. Radiolabeled proteins were synthesized from pcDNA-E-cadherin, pcDNA-Keap1 or pcDNA-Nrf2 using an in vitro translation method. Gels represent autoradiography. pcDNA, empty plasmid. (D) Immunocytochemistry for E-cadherin or Nrf2. HEK293 cells transfected with E-cadherin or E-cadherin plus Nrf2 were immunochemically stained for E-cadherin (green) or Nrf2 (red). To statistically assess subcellular distribution of Nrf2, 50 cells were counted in each arbitrary visual field on three samples per treatment (N, cells with Nrf2 predominantly concentrated in the nuclei; N+C, cells with Nrf2 located in the nuclei and cytoplasm equally; and C, cells with Nrf2 predominantly located in the cytoplasm). Values are means ± s.d. (significantly different compared with Nrf2 transfection, **P<0.01). (E) Immunoprecipitation and immunoblotting assays. Nrf2 immunoprecipitates were immunoblotted for E-cadherin or subcellular marker proteins in HepG2 cells.

Fig. 2.

Interaction of E-cadherin with Nrf2. (A) Immunoprecipitation and immunoblot assays. Nrf2 or E-cadherin immunoprecipitates were immunoblotted for the protein of interest in HEK293 cells transfected with E-cadherin or E-cadherin plus Nrf2. Overexpressed Nrf2 has a molecular mass of ∼110 kDa, whereas endogenous Nrf2 is smaller (i.e. predicted molecular mass of human Nrf2, 66 kDa). Asterisk indicates a nonspecific band. Keap1 immunoprecipitates were immunoblotted for E-cadherin in HEK293 cells transfected with E-cadherin or E-cadherin plus Keap1. IP, immunoprecipitation. (B) Interaction of endogenous E-cadherin with Nrf2 in HepG2 cells. E-cadherin was immunoblotted on IgG or Nrf2 immunoprecipitates, whereas either Nrf2 or Keap1 was blotted on IgG or E-cadherin immunoprecipitates. Input contained 10 μg of proteins. (C) In vitro binding of E-cadherin with Nrf2 or Keap1. GST pull-down assays were performed using in vitro translated [35S]E-cadherin, [35S]Keap1 or [35S]Nrf2, and bacterially expressed GST–Nrf2 or GST–Keap1. GST was used as a negative control. Radiolabeled proteins were synthesized from pcDNA-E-cadherin, pcDNA-Keap1 or pcDNA-Nrf2 using an in vitro translation method. Gels represent autoradiography. pcDNA, empty plasmid. (D) Immunocytochemistry for E-cadherin or Nrf2. HEK293 cells transfected with E-cadherin or E-cadherin plus Nrf2 were immunochemically stained for E-cadherin (green) or Nrf2 (red). To statistically assess subcellular distribution of Nrf2, 50 cells were counted in each arbitrary visual field on three samples per treatment (N, cells with Nrf2 predominantly concentrated in the nuclei; N+C, cells with Nrf2 located in the nuclei and cytoplasm equally; and C, cells with Nrf2 predominantly located in the cytoplasm). Values are means ± s.d. (significantly different compared with Nrf2 transfection, **P<0.01). (E) Immunoprecipitation and immunoblotting assays. Nrf2 immunoprecipitates were immunoblotted for E-cadherin or subcellular marker proteins in HepG2 cells.

Fig. 3.

E-cadherin binding to the C-terminus of Nrf2. (A) Immunoprecipitation-immunoblot assays. The scheme shows the N-terminal and C-terminal fragments of Nrf2 that contain Neh2, Neh4, Neh5 and Neh1, Neh3, Neh6, respectively (top). Each construct was tagged with GBD. Immunoprecipitates with either anti-GBD antibody (left) or anti-Nrf2 antibody (right) were immunoblotted for E-cadherin or Keap1 in HEK293 cells transfected with wild-type (WT), N-terminal or C-terminal fragment of Nrf2 for 24 hours, and treated with 10 μM MG132 for 4 hours. (B) Interaction of E-cadherin with a S40A mutant of Nrf2. HEK293 cells were treated with 30 μM t-BHQ and MG132 for 4 hours after transfection with E-cadherin and/or wild type or S40A mutant of Nrf2. Myc immunoprecipitates (Nrf2) were immunoblotted for E-cadherin or Keap1. (C) Luciferase reporter assays. pGL-1651 reporter activity was measured in HEK293 cells transfected with E-cadherin or E-cadherin plus Nrf2. The levels of ectopically expressed Nrf2 were immunoblotted (bottom). Data represented the mean ± s.e. (significantly different compared with mock transfection, **P<0.01, n=4). (D) The effects of Nrf2 activators on the interaction between E-cadherin and Nrf2. HepG2 cells were treated with 30 μM t-BHQ or 5 μM sulforaphane for 4 hours. Nrf2 activated by t-BHQ or sulforaphane had a molecular mass of ∼110 kDa. SFN, sulforaphane.

Fig. 3.

E-cadherin binding to the C-terminus of Nrf2. (A) Immunoprecipitation-immunoblot assays. The scheme shows the N-terminal and C-terminal fragments of Nrf2 that contain Neh2, Neh4, Neh5 and Neh1, Neh3, Neh6, respectively (top). Each construct was tagged with GBD. Immunoprecipitates with either anti-GBD antibody (left) or anti-Nrf2 antibody (right) were immunoblotted for E-cadherin or Keap1 in HEK293 cells transfected with wild-type (WT), N-terminal or C-terminal fragment of Nrf2 for 24 hours, and treated with 10 μM MG132 for 4 hours. (B) Interaction of E-cadherin with a S40A mutant of Nrf2. HEK293 cells were treated with 30 μM t-BHQ and MG132 for 4 hours after transfection with E-cadherin and/or wild type or S40A mutant of Nrf2. Myc immunoprecipitates (Nrf2) were immunoblotted for E-cadherin or Keap1. (C) Luciferase reporter assays. pGL-1651 reporter activity was measured in HEK293 cells transfected with E-cadherin or E-cadherin plus Nrf2. The levels of ectopically expressed Nrf2 were immunoblotted (bottom). Data represented the mean ± s.e. (significantly different compared with mock transfection, **P<0.01, n=4). (D) The effects of Nrf2 activators on the interaction between E-cadherin and Nrf2. HepG2 cells were treated with 30 μM t-BHQ or 5 μM sulforaphane for 4 hours. Nrf2 activated by t-BHQ or sulforaphane had a molecular mass of ∼110 kDa. SFN, sulforaphane.

Fig. 4.

Interaction between E-cadherin mutants and Nrf2. (A) The domain structures of E-cadherin mutant constructs. E-cadherin has two catenin binding domains at its intracellular region; the region of 762-EED-764 was replaced with AAA to eliminate the p120-catenin-binding domain, whereas the last 35 amino acids were deleted to remove the β-catenin-binding domain. E-cadherin immunoprecipitates were immunoblotted for Nrf2 in HEK293 cells transfected with Nrf2 or Nrf2 plus each E-cadherin mutant. Asterisk indicates light chain of IgG. (B) The effects of E-cadherin mutants on luciferase expression from NQO1-ARE. Luciferase activity was measured on the lysates of HEK293 cells transfected as indicated above for 24 hours, and was normalized to that of β-galactosidase. Data indicate the mean ± s.e. (significantly different compared with mock-transfected control, **P<0.01, n=4). (C) Immunocytochemical assay performed on HEK293 cells transfected with the plasmid encoding Nrf2 or Nrf2 plus a mutant of E-cadherin (E-cadherin, green; and Nrf2, red). To statistically assess subcellular distribution of Nrf2, 50 cells were counted in each arbitrary visual field on three samples per treatment (N, cells with Nrf2 predominantly concentrated in the nuclei; N+C, cells with Nrf2 located in the nuclei and cytoplasm equally; and C, cells with Nrf2 predominantly located in the cytoplasm) (table). Values are means ± s.d. significantly different compared with Nrf2 transfection, **P<0.01; significantly different compared with ECAD-CT or ECAD-CT-Δp120ctn transfection, #P<0.05). Histograms show intracellular distribution of Nrf2 after transfection with each mutant construct (right). Two hundred cells were counted in each visual field. ctn, catenin.

Fig. 4.

Interaction between E-cadherin mutants and Nrf2. (A) The domain structures of E-cadherin mutant constructs. E-cadherin has two catenin binding domains at its intracellular region; the region of 762-EED-764 was replaced with AAA to eliminate the p120-catenin-binding domain, whereas the last 35 amino acids were deleted to remove the β-catenin-binding domain. E-cadherin immunoprecipitates were immunoblotted for Nrf2 in HEK293 cells transfected with Nrf2 or Nrf2 plus each E-cadherin mutant. Asterisk indicates light chain of IgG. (B) The effects of E-cadherin mutants on luciferase expression from NQO1-ARE. Luciferase activity was measured on the lysates of HEK293 cells transfected as indicated above for 24 hours, and was normalized to that of β-galactosidase. Data indicate the mean ± s.e. (significantly different compared with mock-transfected control, **P<0.01, n=4). (C) Immunocytochemical assay performed on HEK293 cells transfected with the plasmid encoding Nrf2 or Nrf2 plus a mutant of E-cadherin (E-cadherin, green; and Nrf2, red). To statistically assess subcellular distribution of Nrf2, 50 cells were counted in each arbitrary visual field on three samples per treatment (N, cells with Nrf2 predominantly concentrated in the nuclei; N+C, cells with Nrf2 located in the nuclei and cytoplasm equally; and C, cells with Nrf2 predominantly located in the cytoplasm) (table). Values are means ± s.d. significantly different compared with Nrf2 transfection, **P<0.01; significantly different compared with ECAD-CT or ECAD-CT-Δp120ctn transfection, #P<0.05). Histograms show intracellular distribution of Nrf2 after transfection with each mutant construct (right). Two hundred cells were counted in each visual field. ctn, catenin.

Role of β-catenin in the E-cadherin and Nrf2 interaction, and in Nrf2 ubiquitylation

We next examined whether β-catenin affects the binding of E-cadherin with Nrf2. siRNA knockdown of β-catenin decreased the intensity of E-cadherin and Nrf2 binding (Fig. 5A). Consistently, a deficiency in β-catenin relieved Nrf2 from repression by E-cadherin, as shown by the nuclear accumulation of Nrf2 as well as the induction of its target gene (Fig. 5B,C). Nrf2 is degraded by the 26S proteasome system after multiple ubiquitylation (Nguyen et al., 2003). In this process, Keap1 is required for formation of the proteasome complex that degrades Nrf2. In HepG2 cells, β-catenin bound endogenous E-cadherin, Nrf2 and Keap1 (Fig. 5D), supporting the notion that these molecules form a quaternary complex. To precisely understand the molecular interactions, we tested whether β-catenin mediates the interaction of E-cadherin and Nrf2 in conjunction with Keap1 ubiquitylation of Nrf2. Interestingly, knockdown of β-catenin markedly decreased the ubiquitylation of Nrf2 (Fig. 5E, left). In the absence of E-cadherin, knockdown of β-catenin had no effect on Nrf2 ubiquitylation (Fig. 5E, middle). As expected, knockdown of Keap1 decreased Nrf2 ubiquitylation (Fig. 5E, right). In HepG2 cells, a deficiency in E-cadherin caused a decrease in Nrf2 ubiquitylation (Fig. 5F, top), and diminished β-catenin binding to either Nrf2 or Keap1 (Fig. 5F, bottom). Our results indicate that E-cadherin interacts with Nrf2 through its β-catenin binding, which might facilitate the ubiquitylation of Nrf2 by Keap1.

Effects of loss of E-cadherin on Nrf2 activity and cancer cell resistance to doxorubicin

Treatment with transforming growth factor β1 (TGFβ1) inhibits E-cadherin expression through a process of cadherin switch (Zavadil and Böttinger, 2005). To functionally assess the effect of E-cadherin loss on Nrf2 activity, we determined whether a decrease in E-cadherin has an impact on the transcriptional activity of Nrf2. In HepG2 cells, TGFβ1 treatment (5 μg/μl, for 48 hours), which caused a loss of E-cadherin, increased the content of Nrf2 in the nuclear fraction, and significantly enhanced NQO1-ARE reporter gene activity (Fig. 6A). Moreover, sulforaphane treatment (5 μM, for 6 hours) further increased the activity of Nrf2 in cells treated with TGFβ1, compared with untreated cells.

Finally, we compared expression of E-cadherin and the nuclear accumulation of Nrf2 in a set of hepatocarcinoma cell lines. In these cells, E-cadherin levels were reciprocal to the levels of nuclear Nrf2 (Fig. 6B, left). It is now accepted that Nrf2 activation in cancer cells contributes to their resistance to chemotherapeutic agents (e.g. doxorubicin and cisplatin) (Lau et al., 2008; Shim et al., 2009; Wang et al., 2008). As expected, the SNU449 cell line, which had the lowest level of E-cadherin was the most resistant to doxorubicin treatment (0.3–3 μM, for 12 hours) (Fig. 6B, right). Levels of nuclear Nrf2 were the highest in SNU449 cells. Overall, resistance of the cell lines to doxorubicin paralleled not only E-cadherin repression, but also the level of Nrf2 expression. Functionally, E-cadherin knockdown decreased the sensitivity of HepG2 cells to doxorubicin and induced Nrf2 target genes (Fig. 6C).

EMT is a fundamental biological process that involves not only physiological conditions such as embryonic development, but also pathological changes including cancer progression and organ fibrosis (Arumugam et al., 2009; Huber et al., 2005; Wheelock et al., 2008). Cells that have passed through EMT exhibit a higher level of ROS production (Felton et al., 2009). In this process, persistent oxidative stress might induce adaptive responses within the tumor cell, which might increase resistance to apoptosis. Nrf2 is a key regulator determining cell survival or death in stressful situations, and might help cancer cells to survive in a deleterious environment. In cancer biology, the upstream regulators of Nrf2 remain to be established, particularly in association with tumor malignancy because the degree of Nrf2 activation might vary depending on the transdifferentiation status of the cell (Ikeda et al., 2004; Lau et al., 2008; Wang et al., 2008). Here, we demonstrate for the first time that E-cadherin has an inhibitory effect on Nrf2 by making it inactive, which might result from the interaction of E-cadherin with Nrf2.

The cadherins consist of five superfamily members: classical cadherins, desmosomal cadherins, atypical cadherins, proto-cadherins and cadherin-related signaling proteins (Gumbiner, 2005). Classical cadherins comprising E-(epithelial), N-(neuronal), P-(placental) and VE-(vascular-endothelial) types have an extracellular domain for cell–cell adhesion, a transmembrane domain, and a cytoplasmic tail that binds intracellular catenins (Angst et al., 2001). In the EMT process, cadherin switch from E-cadherin to N-cadherin is a key feature. Our results from GST pull-down and pGL-1651 reporter assays showed that E-cadherin interacts with Nrf2, repressing the activity of Nrf2 by inhibiting its nuclear translocation. Moreover, E-cadherin bound the C-terminus of Nrf2, which contains cap’n’collar, DNA-binding domain, leucine zipper and Neh3 domain (Itoh et al., 2004). Although the functional effect of E-cadherin on the nuclear localization signal of Nrf2 remains to be elucidated, the ability of E-cadherin to interact with the C-terminus is likely to prevent nuclear accumulation of Nrf2 because Nrf2 has a putative nuclear localization signal and a nuclear export signal at its C-terminus (Jain et al., 2005). In the present study, N-cadherin had only a minor effect. N-cadherin transfection decreased the expression of luciferase from pGL-1651, but the degree of Nrf2 inhibition by N-cadherin was much lower than that by E-cadherin (supplementary material Fig. S2A). It also diminished luciferase expression from pGL-1651-ΔARE (supplementary material Fig. S2A), suggesting that the minor inhibitory effect of N-cadherin is due to another molecule and/or component rather than Nrf2.

In the present study, the ability of t-BHQ to activate Nrf2 was confirmed by measuring the release of Myc–Nrf2 from Keap1. The lack of change in overexpressed Nrf2 by t-BHQ (Fig. 3B) might be due to an experimental condition using exogenous Myc–Nrf2 and MG132, a proteasomal inhibitor that causes accumulation of the Keap1–Nrf2 complex and Keap1-dependent Nrf2 ubiquitylation. The functional impact of endogenous E-cadherin on Nrf2 ubiquitylation was strengthened by decreased Nrf2 ubiquitylation in HepG2 cells transfected with siRNA against E-cadherin; the lack of a detectable change in Keap1-dependent Nrf2 ubiquitylation by E-cadherin overexpression might result from saturation of ubiquitylated Nrf2 caused by MG132. Overall, our data showing endogenous protein–protein interactions support the functional role of E-cadherin in Nrf2 activity and physiological relevance.

β-Catenin is a protein whose core armadillo repeat structure allows it to bind many partners and conveys structural and functional information (Daugherty and Gottardi, 2007; Hoogeboom and Burgering, 2009). It functions as a latent signaling molecule responsible for transcription factor activation and gene induction through recruitment of co-activator of the Tcf/Lef DNA-binding protein family (Daugherty and Gottardi, 2007).

Fig. 5.

Role of β-catenin on the binding between E-cadherin and Nrf2. (A) Interaction between E-cadherin and Nrf2 after knockdown of β-catenin. E-cadherin and Nrf2 binding was determined on the lysates of HEK293 cells transfected with the plasmids encoding E-cadherin and Nrf2 for 24 hours, followed by transfection of siRNA directed against β-catenin (si-βctn), or on those of HepG2 cells transfected with si-βctn. Nrf2 immunoprecipitates were immunoblotted for E-cadherin. (B) The effect of β-catenin knockdown on the nuclear accumulation of Nrf2. Data indicate the mean ± s.e. (significantly different compared with Nrf2-transfected control, **P<0.01, n=3). (C) The effect of si-βctn on NQO1-ARE luciferase activity and Nrf2 target gene expression. Luciferase activity was measured on the lysates of HEK293 cells transfected as described in A. Data indicate the mean ± s.e. (significant compared with the respective control, **P<0.01, n=3). The levels of NQO1 and HO-1 were measured in HEK293 cells transfected as indicated. (D) Immunoprecipitation and immunoblotting assays. The protein of interest in HepG2 cells was immunoblotted on IgG or β-catenin immunoprecipitates. Input contained 10 μg of proteins. (E) The effect of β-catenin knockdown on Nrf2 ubiquitylation. Keap1 immunoprecipitates were immunoblotted for Nrf2 (top). Ubiquitylated Nrf2 in lysates was immunoprecipitated with anti-Nrf2 antibody, and the immunoprecipitate was subjected to immunoblotting for ubiquitin (middle). Ubi, ubiquitin. (F) The effect of E-cadherin knockdown on Nrf2 ubiquitylation and interaction of β-catenin with Nrf2 or Keap1. Nrf2 immunoprecipitates were immunoblotted for Keap1 (top). Ubiquitylated Nrf2 in lysates was immunoprecipitated with anti-Nrf2 antibody, and the immunoprecipitate was subjected to immunoblotting for ubiquitin (middle). β-Catenin immunoprecipitates were immunoblotted for the protein of interest (bottom).

Fig. 5.

Role of β-catenin on the binding between E-cadherin and Nrf2. (A) Interaction between E-cadherin and Nrf2 after knockdown of β-catenin. E-cadherin and Nrf2 binding was determined on the lysates of HEK293 cells transfected with the plasmids encoding E-cadherin and Nrf2 for 24 hours, followed by transfection of siRNA directed against β-catenin (si-βctn), or on those of HepG2 cells transfected with si-βctn. Nrf2 immunoprecipitates were immunoblotted for E-cadherin. (B) The effect of β-catenin knockdown on the nuclear accumulation of Nrf2. Data indicate the mean ± s.e. (significantly different compared with Nrf2-transfected control, **P<0.01, n=3). (C) The effect of si-βctn on NQO1-ARE luciferase activity and Nrf2 target gene expression. Luciferase activity was measured on the lysates of HEK293 cells transfected as described in A. Data indicate the mean ± s.e. (significant compared with the respective control, **P<0.01, n=3). The levels of NQO1 and HO-1 were measured in HEK293 cells transfected as indicated. (D) Immunoprecipitation and immunoblotting assays. The protein of interest in HepG2 cells was immunoblotted on IgG or β-catenin immunoprecipitates. Input contained 10 μg of proteins. (E) The effect of β-catenin knockdown on Nrf2 ubiquitylation. Keap1 immunoprecipitates were immunoblotted for Nrf2 (top). Ubiquitylated Nrf2 in lysates was immunoprecipitated with anti-Nrf2 antibody, and the immunoprecipitate was subjected to immunoblotting for ubiquitin (middle). Ubi, ubiquitin. (F) The effect of E-cadherin knockdown on Nrf2 ubiquitylation and interaction of β-catenin with Nrf2 or Keap1. Nrf2 immunoprecipitates were immunoblotted for Keap1 (top). Ubiquitylated Nrf2 in lysates was immunoprecipitated with anti-Nrf2 antibody, and the immunoprecipitate was subjected to immunoblotting for ubiquitin (middle). β-Catenin immunoprecipitates were immunoblotted for the protein of interest (bottom).

Under normal conditions, β-catenin enhances cell proliferation and differentiation through Wnt signaling (Daugherty and Gottardi, 2007; Hoogeboom and Burgering, 2009; Thévenod and Chakraborty, 2010). It also serves as a mediator that promotes cell survival under conditions of ROS challenge, which would increase stress resistance and ROS clearance. β-Catenin interacts with some transcription factors activated by ROS challenge, such as HIF-1, c-Jun, TCF and Foxo (Thévenod and Chakraborty, 2010). Interactions of β-catenin with these molecules enhance the transcriptional activity of target genes (Thévenod and Chakraborty, 2010). Our results identified the role of β-catenin in the binding of E-cadherin with Nrf2 or Keap1. They also support the notion that the complex of E-cadherin and β-catenin is required for E-cadherin-mediated repression of Nrf2 activity. Thus, β-catenin might have the ability to reprogram transcriptional activity in response to stimuli, which is consistent with a previous report (Hoogeboom and Burgering, 2009). In our study, E-cadherin interacted with β-catenin for Nrf2 binding, supporting the idea that E-cadherin forms a complex with β-catenin in association with its binding to Nrf2 and Keap1. Nonetheless, the finding that a deficiency in E-cadherin diminished the interaction of β-catenin and Nrf2 does not exclude the possibility of direct binding of β-catenin with Nrf2. Collectively, E-cadherin apparently provides a docking site for the molecules. Our observations showed that E-cadherin deficient in the β-catenin-binding domain (ECAD-CT-Δβ-catenin) still weakly binds Nrf2, suggesting that another region of E-cadherin (e.g. the intervening region) also provides a direct binding site for Nrf2. Our finding showing a decrease in Keap1 activity by β-catenin knockdown corroborated the concept that β-catenin enhances the interaction between E-cadherin and Nrf2 and might contribute to Keap1-mediated ubiquitylation.

Fig. 6.

The effect of loss of E-cadherin on the activity of Nrf2. (A) The effect of TGF-β1 treatment on Nrf2 activity. Nrf2 was immunoblotted on the nuclear fraction of HepG2 cells treated with TGF-β1 for 48 hours and continuously exposed to 5 μM sulforaphane for 6 hours. E-cadherin was immunoblotted on the lysates (significantly different compared with control, *P<0.05, **P<0.01, n=3). (B) Cytotoxicity of hepatoma cell lines to doxorubicin. E-cadherin or Nrf2 was immunoblotted on total lysates or nuclear fractions. Each lane contained 20 μg of protein. Data indicate the mean ± s.e. (significant compared with the level of E-cadherin or nuclear Nrf2 in HepG2 cells, *P<0.05, **P<0.01, n=3) (left). The cells were treated with 0.3–3 μM doxorubicin for 12 hours following 12 hours of serum starvation, and the viability was assessed using MTT assays. Data indicate the mean ± s.e. (significant compared with the viability of HepG2 cells at respective time point, #P<0.05, ##P<0.01, n=5) (right). For data points where error bars cannot be seen, the standard error is covered by the data point. (C) The effects of E-cadherin knockdown on Nrf2 target gene expression and chemoresistance to doxorubicin. NQO1 and HO-1 were immunoblotted on the lysates of HepG2 cells transfected with a nontargeting control siRNA or si-ECAD (top). Cell viability was assessed using MTT assays in HepG2 cells treated with 3 μM doxorubicin for 12 hours following 12 hours of serum starvation. Data indicate the mean ± s.e. (significant compared with mock-transfected control, **P<0.01, n=4) (bottom).

Fig. 6.

The effect of loss of E-cadherin on the activity of Nrf2. (A) The effect of TGF-β1 treatment on Nrf2 activity. Nrf2 was immunoblotted on the nuclear fraction of HepG2 cells treated with TGF-β1 for 48 hours and continuously exposed to 5 μM sulforaphane for 6 hours. E-cadherin was immunoblotted on the lysates (significantly different compared with control, *P<0.05, **P<0.01, n=3). (B) Cytotoxicity of hepatoma cell lines to doxorubicin. E-cadherin or Nrf2 was immunoblotted on total lysates or nuclear fractions. Each lane contained 20 μg of protein. Data indicate the mean ± s.e. (significant compared with the level of E-cadherin or nuclear Nrf2 in HepG2 cells, *P<0.05, **P<0.01, n=3) (left). The cells were treated with 0.3–3 μM doxorubicin for 12 hours following 12 hours of serum starvation, and the viability was assessed using MTT assays. Data indicate the mean ± s.e. (significant compared with the viability of HepG2 cells at respective time point, #P<0.05, ##P<0.01, n=5) (right). For data points where error bars cannot be seen, the standard error is covered by the data point. (C) The effects of E-cadherin knockdown on Nrf2 target gene expression and chemoresistance to doxorubicin. NQO1 and HO-1 were immunoblotted on the lysates of HepG2 cells transfected with a nontargeting control siRNA or si-ECAD (top). Cell viability was assessed using MTT assays in HepG2 cells treated with 3 μM doxorubicin for 12 hours following 12 hours of serum starvation. Data indicate the mean ± s.e. (significant compared with mock-transfected control, **P<0.01, n=4) (bottom).

Nrf2 binds depolymerized actin after a challenge of oxidative stress, and then the complex of Nrf2–actin can translocate into the nucleus for ARE activation (Kang et al., 2002). Thus, treatment with phalloidin, an actin-stabilizing agent, prevented the activation of Nrf2 (Kang et al., 2002). In our experiment, phalloidin treatment did not alter the interaction between E-cadherin and Nrf2 (supplementary material Fig. S2B). However, this interaction was weakened by a deficiency in β-catenin. Keap1 can also bind with actin filament and this Keap1–actin complex functions as a scaffold to retain Nrf2 in the cytoplasm under unstressed conditions (Kang et al., 2004). Our result raises the possibility that E-cadherin provides a docking site for Nrf2 and Keap1 in association with actin and β-catenin interaction. Therefore, it is likely that β-catenin contributes to the regulatory effect of E-cadherin for the activity of Nrf2, presumably facilitating Keap1-mediated ubiquitylation.

It is well recognized that TGF-β1 represses E-cadherin. Moreover, E-cadherin deficiency facilitates TGF-β1 induction because E-cadherin inhibits RhoA-dependent phosphorylation of Smad3 (Cho et al., 2010). Our findings corroborated the notion that an increase in Nrf2 activity plays a role in the EMT process of HepG2 cells treated with TGF-β1. Consistently, TGF-β1 enhanced Nrf2-mediated ARE activity and HO-1 induction in smooth muscle cells (Churchman et al., 2009). Presumably, this phenomenon is associated with enhanced superoxide production and the expression of NAD(P)H oxidase subunit p22(phox). Evidence is accumulating that the activating mutations of Nrf2 frequently occurred in human cancers and such mutations might help to increase the constitutive expression of pro-survival factors (Hayes and McMahon, 2009). It should also be pointed out that the expression levels of Nrf2 and its target genes were elevated in several aggressive cancer cells (i.e. LK2, EBC1 and HO1-U1) or in lung and colon cancer tissues (Kim et al., 2011; Shibata et al., 2008). All of these findings are in agreement with our observation that cancer cells deficient in E-cadherin exhibited higher levels of Nrf2 and were more resistant to the cytotoxicity caused by doxorubicin.

In summary, the results of this study showed that (1) E-cadherin inhibits nuclear accumulation of Nrf2 and Nrf2-mediated gene transcription by its interaction with Nrf2; (2) E-cadherin binds the C-terminus of Nrf2, thus it does not affect phosphorylation of Ser40; (3) E-cadherin interacts with Keap1; (4) E-cadherin forms a quaternary complex with Nrf2, Keap1 and β-catenin, which might contribute to Keap1-dependent ubiquitylation of Nrf2. Collectively, E-cadherin binding with Nrf2 might restrain nuclear localization of Nrf2 and its transcriptional activity, implying that the loss of E-cadherin promotes tumor proliferation and metastasis by increasing the activity of Nrf2.

Materials

L-[35S]methionine (>1000 Ci/mmol) was purchased from PerkinElmer (Waltham, MA). Anti-E-cadherin antibody was obtained from BD Bioscience (San Jose, CA). Antibodies directed against Nrf2, lamin A/C, heat shock protein 70, NQO-1, Keap1, Gal4 DNA-binding domain (GBD) and β-catenin were supplied from Santa Cruz Biotechnology (Santa Cruz, CA). Anti-HO-1 antibody was purchased from Stressgen (San Diego, CA). Sulforaphane, anti-ubiquitin antibody, and anti-β-actin antibody were purchased from Sigma (St Louis, MO). Horseradish-peroxidase-conjugated goat anti-rabbit and goat anti-mouse IgG were obtained from Zymed Laboratories (San Francisco, CA). MG132 was provided by Calbiochem (Darmstadt, Germany).

Plasmids

pGL-1651 reporter construct containing the promoter region of the GSTA2 gene was generated according to method described previously (Park et al., 2004). Briefly, the region containing –1651 to +66 bp of the GSTA2 gene was amplified by PCR using pGTB-1.65 as a template. The DNA product was cloned into pGEM-T EasyVector (Promega, Madison, WI) and subcloned into the pGL3 luciferase reporter. The deletion mutants of GSTA2 promoter luciferase plasmid, pGL-1651-ΔC/EBP and pGL-1651-ΔARE, in which the C/EBP-binding site and ARE were deleted, respectively, were constructed previously (Park et al., 2004). Mammalian expression plasmids for GBD-Nrf2 functional domain fusion proteins were provided by Masayuki Yamamoto (Tohoku University, Sendai, Japan); GBD-NT contains the N-terminal 1–317 amino acid region of Nrf2, whereas GBD-CT contains the C-terminal 317–597 amino acid region of Nrf2 (Katoh et al., 2001). A Ser40 to alanine mutant of Nrf2 was generated by oligonucleotide-mediated mutagenesis (Cho et al., 2007). The constructs encoding deletion mutants of catenin-binding domains in E-cadherin were gifts from Richard A. Anderson (University of Wisconsin-Madison, Madison, WI).

Cell culture

HEK293 and HepG2 cells were supplied by the American Type Culture Collection (Manassas, VA), and SNU886 and SNU449 cells were obtained from the Korean Cell Line Bank (Seoul, Korea). The cells were maintained in Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum (FBS), 50 U/ml penicillin, and 50 μg/ml streptomycin at 37°C in a humidified atmosphere with 5% CO2. Cells that had undergone fewer than 20 passages were used. They were incubated in the presence or absence of t-BHQ (30 μM), sulforaphane (5 μM) or recombinant human TGF-β1 (5 μg/μl) for the indicated time periods.

Transient transfection and luciferase reporter assays

Transient transfection was done with Lipofectamine 2000 reagent (Invitrogen, Carlsbad, CA). Briefly, cells were re-plated 24 hours before transfection at a density of 7×105 cells in six-well plates. The cells were incubated with the plasmids of interest and Lipofectamine reagent for 3 hours for transfection, and continuously incubated for additional 24 hours in minimal essential medium containing 1% FBS. Control cells were transfected with an equal amount of the respective empty plasmid (i.e. mock transfection). In luciferase reporter assays, the cells were transiently transfected with pGL-1651 or pGL-NQO1-ARE plasmid that contains three copies of ARE derived from the promoter of the NQO1. The activity of luciferase was measured by adding Luciferase Assay Reagent (Promega, Madison, WI).

Immunoblot analysis

Cell lysates and subcellular fractions were prepared according to previously published methods (Kang et al., 2002). Membrane fractions were prepared using ProteoExtract subcellular proteome extraction kit (Calbiochem, Darmstadt, Germany). SDS-polyacrylamide gel electrophoresis and immunoblot analyses were performed as previously described (Ki et al., 2005). Immunoreactive proteins were visualized using an ECL chemiluminescence detection kit (Amersham Biosciences, Amersham, UK). At least three separate experiments were performed with different samples to measure changes in protein levels.

Real-time PCR assays

Total RNA was extracted using Trizol (Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions. Total RNA was reverse-transcribed using an oligo(dT)16 primer to obtain cDNA. The cDNA was amplified by polymerase chain reaction (PCR). PCR was performed with a Light Cycler 1.5 apparatus (Roche, Mannheim, Germany) using a Light Cycler DNA master SYBR green-I kit according to the manufacturer’s instructions. The following primer sequences were used: human NQO1, 5′-AGGCTGGTTTGAGCGAGT-3′ (sense) and 5′-TTGAATTCGGGCGTCTGCTG-3′ (antisense); and human HO-1, 5′-CAGGCAGAGAATGCTGAGTTC-3′ (sense) and 5′-CATCACCAGCTTAAAGCCTT-3′ (antisense).

Immunoprecipitation and immunoblotting assays

To determine protein–protein interaction, a fraction of cell lysates (500 μg protein in 1 ml) was incubated with antibody directed against the protein of interest overnight at 4°C. The antigen–antibody complex was immunoprecipitated following incubation for 2 hours at 4°C with Protein-G–agarose. Immune complexes were solubilized in 2× Laemmli buffer and boiled for 5 minutes. The samples were immunoblotted with the antibody of interest.

GST pull-down assays

E-cadherin and Nrf2 binding was analyzed using agarose-immobilized GST fusion proteins according to methods described previously (Ki et al., 2005). Briefly, Escherichia coli BL21(DE3) cells harboring the plasmid encoding GST-Nrf2 plasmid was cultured at 37°C with shaking. GST fusion protein was produced by incubating the cells with 1 mM isopropyl-β-D-thiogalactopyranoside for 6 hours. Bacteria expressing GST–Nrf2 or GST were precipitated and resuspended with one-tenth volume of PBS containing 0.1% Tween-20, 2 mM EDTA, 0.1% β-mercaptoethanol and 0.2 mM PMSF. Bacterial cell lysates was mixed with GSH agarose beads (100 μl, 50% slurry) pre-swollen with PBS containing 0.1% Tween-20. GST–Keap1 was purchased from Abnova (Taipei City, Taiwan). GST fusion protein was immobilized to GSH agarose by incubation at 4°C for 20 minutes and washed three times with PBS containing 0.1% Tween-20 and 0.2 mM PMSF and once with 20 mM HEPES buffer (pH 7.8, buffer A) containing 50 mM KCl, 5 mM MgCl2, 5 mM EDTA, 6% glycerol, 0.05% Triton X-100, 0.1 mM PMSF, 1 mM DTT and 0.05 mg/ml BSA.

Radiolabeled protein was produced with the TNT system (Promega, Madison, WI) using pcDNA-E-cadherin or pcDNA-Keap1 or pcDNA-Nrf2 (1 μg). For binding reactions, GSH agarose immobilized to GST–Nrf2 (or GST as control) (25 μl) was mixed with [35S]E-cadherin, [35S]Keap1 or [35S]Nrf2 (10 μl) in 500 μl buffer A. The protein-bound agarose was washed three times with 20 mM HEPES buffer (pH 7.8) containing 50 mM KCl, 5 mM MgCl2, 5 mM EDTA, 6% glycerol, 0.05% Triton X-100 and 0.1 mM PMSF. Bound proteins were eluted with 20 μl of 50 mM Tris-HCl (pH 8.0) containing 10 mM reduced GSH and 10% glycerol. The eluate was resolved by SDS-PAGE. Labeled proteins were visualized by autoradiography.

Immunocytochemistry

HEK293 cells were grown on Lab-TEK chamber slides (Nalge Nunc International, Rochester, NY). Standard immunocytochemical methods were used for immunostaining of Nrf2 or E-cadherin, as previously described (Kang et al., 2002). For immunostaining, the cells were fixed in 100% methanol for 30 minutes. After blocking in 5% bovine serum albumin in PBS for 1 hour, the cells were incubated for 1 hour with anti-Nrf2 antibody and monoclonal mouse anti-E-cadherin antibody. The cells were incubated with Alexa-Fluor-488-conjugated goat anti-rabbit IgG antibody and Alexa-Fluor-555-conjugated rabbit anti-mouse IgG antibody (Invitrogen, Carlsbad, CA). Stained cells were examined using a laser-scanning confocal microscope (Leica TCS NT, Leica Microsystems, Wetzlar, Germany).

siRNA knockdown

To knockdown the protein of interest, cells were transfected with siRNA directed against β-catenin, E-cadherin or Keap1, or a nontargeting control siRNA (100 pmol/ml) using Dharmafect (Dharmacon, Lafayette, CO). All siRNAs were purchased from Dharmacon. The knockdown effect was confirmed by immunoblotting for target protein.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assays

HepG2, SNU886 or SNU449 cells were plated in a 48-well dish (1×105 cells/well), and incubated in Eagle’s minimum essential medium without 10% FBS for 12 hours. The cells were then incubated with doxorubicin dissolved in dimethylsulfoxide at indicated concentrations for 12 hours. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assays were performed to assess cell viability as previously described (Kim et al., 2008).

Statistical analysis

One-way analysis of variance was used to assess significant differences among treatment groups. For each significant effect of treatment, the Newman–Keuls test was used for comparisons of multiple group means. The criterion for statistical significance was set at P<0.05 or P<0.01.

Funding

This work was supported by the World Class University project (Ministry of Education, Science and Technology Development) [grant number R32-2010-000-10098-0]; and a National Research Foundation of Korea grant [MEST No. 2011-0001204] funded by the Government of Korea.

Angst
B. D.
,
Marcozzi
C.
,
Magee
A. I.
(
2001
).
The cadherin superfamily: diversity in form and function
.
J. Cell Sci.
114
,
629
641
.
Arumugam
T.
,
Ramachandran
V.
,
Fournier
K. F.
,
Wang
H.
,
Marquis
L.
,
Abbruzzese
J. L.
,
Gallick
G. E.
,
Logsdon
C. D.
,
McConkey
D. J.
,
Choi
W.
(
2009
).
Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer
.
Cancer Res.
69
,
5820
5828
.
Cho
I. J.
,
Kim
Y. W.
,
Han
C. Y.
,
Kim
E. H.
,
Anderson
R. A.
,
Lee
Y. S.
,
Lee
C. H.
,
Hwang
S. J.
,
Kim
S. G.
(
2010
).
E-cadherin antagonizes transforming growth factor β1 gene induction in hepatic stellate cells by inhibiting RhoA-dependent Smad3 phosphorylation
.
Hepatology
52
,
2053
2064
.
Cho
M. K.
,
Kim
W. D.
,
Ki
S. H.
,
Hwang
J. I.
,
Choi
S.
,
Lee
C. H.
,
Kim
S. G.
(
2007
).
Role of Galpha12 and Galpha13 as novel switches for the activity of Nrf2, a key antioxidative transcription factor
.
Mol. Cell. Biol.
27
,
6195
6208
.
Churchman
A. T.
,
Anwar
A. A.
,
Li
F. Y.
,
Sato
H.
,
Ishii
T.
,
Mann
G. E.
,
Siow
R. C.
(
2009
).
Transforming growth factor-beta1 elicits Nrf2-mediated antioxidant responses in aortic smooth muscle cells
.
J. Cell Mol. Med.
13
,
2282
2292
.
Daugherty
R. L.
,
Gottardi
C. J.
(
2007
).
Phospho-regulation of Beta-catenin adhesion and signaling functions
.
Physiology (Bethesda)
22
,
303
309
.
Drees
F.
,
Pokutta
S.
,
Yamada
S.
,
Nelson
W. J.
,
Weis
W. I.
(
2005
).
Alpha-catenin is a molecular switch that binds E-cadherin-beta-catenin and regulates actin-filament assembly
.
Cell
123
,
903
915
.
Felton
V. M.
,
Borok
Z.
,
Willis
B. C.
(
2009
).
N-acetylcysteine inhibits alveolar epithelial-mesenchymal transition
.
Am. J. Physiol. Lung Cell Mol. Physiol.
297
,
L805
L812
.
Gumbiner
B. M.
(
2005
).
Regulation of cadherin-mediated adhesion in morphogenesis
.
Nat. Rev. Mol. Cell Biol.
6
,
622
634
.
Hayes
J. D.
,
McMahon
M.
(
2009
).
Nrf2 and Keap1 mutations: permanent activation of an adaptive response in cancer
.
Trends Biochem. Sci.
34
,
176
188
.
Hoogeboom
D.
,
Burgering
B. M.
(
2009
).
Should I stay or should I go: beta-catenin decides under stress
.
Biochim. Biophys. Acta.
1796
,
63
74
.
Huber
M. A.
,
Kraut
N.
,
Beug
H.
(
2005
).
Molecular requirements for epithelial-mesenchymal transition during tumor progression
.
Curr. Opin. Cell Biol.
17
,
548
558
.
Ikeda
H.
,
Nishi
S
,
Sakai
M.
(
2004
).
Transcription factor Nrf2/MafK regulates rat placental glutathione S-transferase gene during hepatocarcinogenesis
.
Biochem. J.
380
,
515
521
.
Itoh
K.
,
Tong
K. I.
,
Yamamoto
M.
(
2004
).
Molecular mechanism activating Nrf2-Keap1 pathway in regulation of adaptive response to electrophiles
.
Free Radic. Biol. Med.
36
,
1208
1213
.
Itoh
K.
,
Mimura
J.
,
Yamamoto
M.
(
2010
).
Discovery of the negative regulator of Nrf2, Keap1: a historical overview
.
Antioxid. Redox Signal.
13
,
1665
1678
.
Jain
A. K.
,
Bloom
D. A.
,
Jaiswal
A. K.
(
2005
).
Nuclear import and export signals in control of Nrf2
.
J. Biol. Chem.
280
,
29158
29168
.
Jeong
W. S.
,
Jun
M.
,
Kong
A. N.
(
2006
).
Nrf2: a potential molecular target for cancer chemoprevention by natural compounds
.
Antioxid. Redox Signal.
8
,
99
106
.
Kang
K. W.
,
Lee
S. J.
,
Park
J. W.
,
Kim
S. G.
(
2002
).
Phosphatidylinositol 3-kinase regulates nuclear translocation of NF-E2-related factor 2 through actin rearrangement in response to oxidative stress
.
Mol. Pharmacol.
62
,
1001
1010
.
Kang
M. I.
,
Kobayashi
A.
,
Wakabayashi
N.
,
Kim
S. G.
,
Yamamoto
M.
(
2004
).
Scaffolding of Keap1 to the actin cytoskeleton controls the function of Nrf2 as key regulator of cytoprotective phase 2 genes
.
Proc. Natl. Acad. Sci. USA
101
,
2046
2051
.
Katoh
Y.
,
Itoh
K.
,
Yoshida
E.
,
Miyagishi
M.
,
Fukamizu
A.
,
Yamamoto
M.
(
2001
).
Two domains of Nrf2 cooperatively bind CBP, a CREB binding protein, and synergistically activate transcription
.
Genes Cells
6
,
857
868
.
Ki
S. H.
,
Cho
I. J.
,
Choi
D. W.
,
Kim
S. G.
(
2005
).
Glucocorticoid receptor (GR)-associated SMRT binding to C/EBPbeta TAD and Nrf2 Neh4/5: role of SMRT recruited to GR in GSTA2 gene repression
.
Mol. Cell. Biol.
25
,
4150
4165
.
Kim
T. H.
,
Hur
E. G.
,
Kang
S. J.
,
Kim
J. A.
,
Thapa
D.
,
Lee
Y. M.
,
Ku
S. K.
,
Jung
Y.
,
Kwak
M. K.
(
2011
).
NRF2 blockade suppresses colon tumor angiogenesis by inhibiting hypoxia-induced activation of HIF-1α
.
Cancer Res.
71
,
2260
2275
.
Kim
Y. W.
,
Zhao
R. J.
,
Park
S. J.
,
Lee
J. R.
,
Cho
I. J.
,
Yang
C. H.
,
Kim
S. G.
,
Kim
S. C.
(
2008
).
Anti-inflammatory effects of liquiritigenin as a consequence of the inhibition of NF-kappaB-dependent iNOS and proinflammatory cytokines production
.
Br. J. Pharmacol.
154
,
165
173
.
Kobayashi
A.
,
Kang
M. I.
,
Okawa
H.
,
Ohtsuji
M.
,
Zenke
Y.
,
Chiba
T.
,
Igarashi
K.
,
Yamamoto
M.
(
2004
).
Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2
.
Mol. Cell. Biol.
24
,
7130
7139
.
Lau
A.
,
Villeneuve
N. F.
,
Sun
Z.
,
Wong
P. K.
,
Zhang
D. D.
(
2008
).
Dual roles of Nrf2 in cancer
.
Pharmacol. Res.
58
,
262
270
.
Nguyen
T.
,
Sherratt
P. J.
,
Huang
H. C.
,
Yang
C. S.
,
Pickett
C. B.
(
2003
).
Increased protein stability as a mechanism that enhances Nrf2-mediated transcriptional activation of the antioxidant response element. Degradation of Nrf2 by the 26 S proteasome
.
J. Biol. Chem.
278
,
4536
4541
.
Niture
S. K.
,
Jain
A. K.
,
Jaiswal
A. K.
(
2009
).
Antioxidant-induced modification of INrf2 cysteine 151 and PKC-delta-mediated phosphorylation of Nrf2 serine 40 are both required for stabilization and nuclear translocation of Nrf2 and increased drug resistance
.
J. Cell Sci.
15
,
4452
4464
.
Park
E. Y.
,
Cho
I. J.
,
Kim
S. G.
(
2004
).
Transactivation of the PPAR-responsive enhancer module in chemopreventive glutathione S-transferase gene by the peroxisome proliferator-activated receptor-gamma and retinoid X receptor heterodimer
.
Cancer Res.
64
,
3701
3713
.
Parrish
A. R.
,
Catania
J. M.
,
Orozco
J.
,
Gandolfi
A. J.
(
1999
).
Chemically induced oxidative stress disrupts the E-cadherin/catenin cell adhesion complex
.
Toxicol. Sci.
51
,
80
86
.
Rao
R. K.
,
Basuroy
S.
,
Rao
V. U.
,
Karnaky
K. J.
Jr
,
Gupta
A.
(
2002
).
Tyrosine phosphorylation and dissociation of occludin-ZO-1 and E-cadherin-beta-catenin complexes from the cytoskeleton by oxidative stress
.
Biochem. J.
368
,
471
481
.
Satoh
H.
,
Moriguchi
T.
,
Taguchi
K.
,
Takai
J.
,
Maher
J. M.
,
Suzuki
T.
,
Winnard
P. T.
Jr
,
Raman
V.
,
Ebina
M.
,
Nukiwa
T.
, et al. 
. (
2010
).
Nrf2-deficiency creates a responsive microenvironment for metastasis to the lung
.
Carcinogenesis
31
,
1833
1843
.
Shibata
T.
,
Ohta
T.
,
Tong
K. I.
,
Kokubu
A.
,
Odogawa
R.
,
Tsuta
K.
,
Asamura
H.
,
Yamamoto
M.
,
Hirohashi
S.
(
2008
).
Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy
.
Proc. Natl. Acad. Sci. USA
105
,
13568
13573
.
Shim
G. S.
,
Manandhar
S.
,
Shin
D. H.
,
Kim
T. H.
,
Kwak
M. K.
(
2009
).
Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the Nrf2 pathway
.
Free Radic. Biol. Med.
47
,
1619
1631
.
Taguchi
K.
,
Motohashi
H.
,
Yamamoto
M.
(
2011
).
Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution
.
Genes Cells
16
,
123
140
.
Thévenod
F.
,
Chakraborty
P. K.
(
2010
).
The role of Wnt/beta-catenin signaling in renal carcinogenesis: lessons from cadmium toxicity studies
.
Curr. Mol. Med.
10
,
387
404
.
Wang
X. J.
,
Sun
Z.
,
Villeneuve
N. F.
,
Zhang
S.
,
Zhao
F.
,
Li
Y.
,
Chen
W.
,
Yi
X.
,
Zheng
W.
,
Wondrak
G. T.
, et al. 
. (
2008
).
Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2
.
Carcinogenesis
29
,
1235
1243
.
Wheelock
M. J.
,
Johnson
K. R.
(
2003
).
Cadherin-mediated cellular signaling
.
Curr. Opin. Cell Biol.
15
,
509
514
.
Wheelock
M. J.
,
Shintani
Y.
,
Maeda
M.
,
Fukumoto
Y.
,
Johnson
K. R.
(
2008
).
Cadherin switching
.
J. Cell Sci.
15
,
727
735
.
Zavadil
J.
,
Böttinger
E. P.
(
2005
).
TGF-beta and epithelial-to-mesenchymal transitions
.
Oncogene
24
,
5764
5774
.

Supplementary information