Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 45, Issue of November 7, pp.

44675–44682, 2003
© 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

Phosphorylation of Nrf2 at Ser40 by Protein Kinase C in Response to


Antioxidants Leads to the Release of Nrf2 from INrf2, but Is
Not Required for Nrf2 Stabilization/Accumulation in the
Nucleus and Transcriptional Activation of Antioxidant
Response Element-mediated NAD(P)H:Quinone
Oxidoreductase-1 Gene Expression*
Received for publication, July 15, 2003, and in revised form, August 20, 2003
Published, JBC Papers in Press, August 28, 2003, DOI 10.1074/jbc.M307633200

David A. Bloom and Anil K. Jaiswal‡


From the Department of Pharmacology, Baylor College of Medicine, Houston, Texas 77030

The antioxidant response element (ARE) and tran- ing NAD(P)H:quinone oxidoreductase-1 (NQO1), glutathione
scription factor Nrf2 regulate basal expression and an- S-transferase Ya, ␥-glutamylcysteinyl synthetase, and heme
tioxidant induction of NAD(P)H:quinone oxidoreduc- oxygenase-1, contribute significantly to cellular protection
tase-1 (NQO1) and other detoxifying genes. Under against redox cycling, oxidative stress, and neoplasia (reviewed
normal conditions, Nrf2 is targeted for proteasomal deg- in Refs. 1 and 2). Specific nuclear protein complexes bind to the
radation by INrf2. Oxidative stress causes release of AREs of various genes (reviewed in Refs. 1 and 2). Among these
Nrf2 from INrf2. Nrf2 translocates to the nucleus, binds proteins, the role of Nrf2 (NF-E2-related factor-2) is the most
to the ARE, and activates gene expression. In this study,
clearly established (reviewed in Refs. 1 and 2). Nrf2 is a basic
we demonstrate that protein kinase C (PKC) plays a
leucine zipper protein that does not homodimerize, but that
significant role in the regulation of ARE-mediated NQO1
gene expression and induction in response to t-butylhy- requires heterodimerization with another leucine zipper pro-
droquinone. Treatment of HepG2 cells with the PKC tein to be active (reviewed in Refs. 1 and 2). c-Jun and small
inhibitors staurosporine and calphostin C repressed Maf proteins have been shown to heterodimerize with Nrf2;
ARE-mediated induction of a luciferase reporter as well these complexes bind to the ARE and alter ARE-mediated gene
as that of the endogenous NQO1 gene. Similar experi- expression (reviewed in Refs. 1 and 2).
ments with inhibitors of MEK/ERK, p38, phosphatidyli- A cytosolic inhibitor of Nrf2, Keap1/INrf2, was identified (3,
nositol 3-kinase, and tyrosine kinases failed to repress 4). Under normal conditions, it is believed that INrf2 retains
ARE-mediated gene expression. The PKC inhibitor stau- Nrf2 in the cytoplasm (3, 4). When cells are challenged by
rosporine blocked the nuclear translocation of Nrf2, chemically induced oxidative stress, Nrf2 is released from
suggesting that Nrf2 might be the target for PKC regu- INrf2. Nrf2 then translocates to the nucleus, resulting in the
lation. A Prosite search revealed the presence of seven activation of ARE-mediated gene expression. Recently, reports
putative PKC sites in mouse Nrf2. The PKC site at Ser40 have shown that the interaction of Nrf2 with INrf2 targets
is conserved among species and lies in the Neh2 domain,
Nrf2 for proteasomal degradation (5–7). Nrf2 accumulates in
which interacts with INrf2. We demonstrate that phos-
the nucleus of cells from INrf2-deficient mice, in a manner
phorylation of Ser40 is necessary for Nrf2 release from
INrf2, but is not required for Nrf2 stabilization/accumu- similar to challenge with chemicals that induce oxidative stress
lation in the nucleus and transcriptional activation of (7).
ARE-mediated NQO1 gene expression. A peptide that In vitro studies have demonstrated that several kinases,
competes with endogenous Nrf2 for INrf2 binding was including JNK1, MAPK, p38, and MEK, are involved in regu-
able to induce ARE activity more effectively than t-bu- lating ARE-mediated gene expression (8 –11), yet other studies
tylhydroquinone, and Nrf2 that accumulated in the nu- have implicated phosphatidylinositol 3-kinase and tyrosine ki-
cleus as a result was not phosphorylated. nases as the kinases responsible (12, 13). However, another
report has disputed the involvement of p38 and MEK kinases
and instead has indicated that protein kinase C (PKC) is the
Antioxidant response element (ARE)1-mediated expression major kinase involved in antioxidant induction of ARE-medi-
and coordinated induction of detoxifying enzyme genes, includ- ated gene expression (14). In vitro studies from the same group
showed preferential phosphorylation of a serine residue in the
Neh2 domain of Nrf2 by PKC (15).
* This work was supported by National Institutes of Health Grant Our laboratory has long-term interest in Nrf2 regulation of
GM47466. The costs of publication of this article were defrayed in part
by the payment of page charges. This article must therefore be hereby ARE-mediated gene expression with special emphasis on
marked “advertisement” in accordance with 18 U.S.C. Section 1734 NQO1 gene expression. Because of the conflicting reports on
solely to indicate this fact. the kinases involved, we felt further studies were necessary. In
‡ To whom correspondence should be addressed: Dept. of Pharmacol- addition, there is no information available on the kinase(s)
ogy, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030.
Tel.: 713-798-7691; Fax: 713-798-3145; E-mail: ajaiswal@bcm.tmc.edu. involved in the induction of ARE-mediated human NQO1 gene
1
The abbreviations used are: ARE, antioxidant response element; expression in response to antioxidants. Furthermore, it is un-
NQO1, NAD(P)H:quinone oxidoreductase-1; JNK1, c-Jun N-terminal known if phosphorylation of Nrf2 is required only for its release
kinase-1; MAPK, mitogen-activated protein kinase; MEK, mitogen-ac-
from INrf2 or is required also for stabilization/accumulation of
tivated protein kinase/extracellular signal-regulated kinase kinase;
PKC, protein kinase C; ERK, extracellular signal-regulated kinase; Nrf2 in the nucleus and transcriptional activation of ARE-
t-BHQ, t-butylhydroquinone. mediated gene expression.

This paper is available on line at http://www.jbc.org 44675

This is an Open Access article under the CC BY license.


44676 PKC Regulation of ARE-mediated NQO1 Gene Expression
EXPERIMENTAL PROCEDURES
Construction of Plasmids—The construction of pGL2-NQO1-ARE,
pcDNA-Nrf2, and pcDNA-INrf2 was as described previously (3). The
plasmid pcDNA-Nrf2 was used as the template for all mutations of
Nrf2. The seven putative PKC sites in Nrf2 were mutated using the
QuikChange® II site-directed mutagenesis kit (Stratagene, La Jolla,
CA). Briefly, two complementary PAGE-purified primers that con-
tained the desired mutation were produced (Integrated DNA Technol-
ogies, Inc., Coralville, IA). Fifteen cycles of PCR (94 °C for 1 min, 48 °C
for 0.5 min, and 68 °C for 8 min) were performed using pcDNA-Nrf2 as
the template, the primer pair containing the mutation, and PfuUltra
HF DNA polymerase. The parental plasmid was then digested with
DpnI, and the remaining nicked, circular, mutated plasmid was trans-
formed into maximum efficiency DH5␣ chemically competent cells (In-
vitrogen). The forward primers for each mutation are as follows:
Nrf2⌬PKC1, 5⬘-GTG TTT GAC TTT GCT CAG CGA CAG AAG GAC-3⬘;
FIG. 1. Effect of PKC inhibitors on t-BHQ induction of ARE-
Nrf2⌬PKC2, 5⬘-GTG CCC CTG GAG CTG TCA AAC AGA ACG GCC-3⬘; mediated gene expression. HepG2 cells were seeded in 6-well dishes.
Nrf2⌬PKC3, 5⬘-CCA ATG TGA AAA TGC AGC AAA AAA AGA AGT Twenty-four h later, they were cotransfected with a reporter construct
TCC-3⬘; Nrf2⌬PKC4, 5⬘-GTG AAA AGA CAA ACA TGC AGC CCG CTT expressing the luciferase gene under the control of the NQO1 gene ARE
AGA GGC-3⬘; Nrf2⌬PKC5, 5⬘-CCT TGT TCC CAA AGC TAA GAA GCC and a control reporter expressing Renilla luciferase. Thirty-six h after
AGA TAC-3⬘; and Nrf2⌬PKC6, 5⬘-CAA GAA GCC AGA TGC TAA GAA transfection, the cells were pretreated with staurosporine, calphostin C,
AAA CTA GG-3⬘. The reverse primers are the exact complements of the or Me2SO (DMSO; control) for 2 h and then treated with Me2SO, with
forward primers. The pcDNA-Neh2 and pcDNA-Neh2⌬S plasmids ex- t-BHQ, or with t-BHQ plus inhibitor for 16 h. The cells were lysed, and
pressing Neh2 and Neh2⌬S domains, respectively, were created by the lysates were analyzed by dual luciferase assay. The results repre-
PCR, followed by TA cloning into the pcDNA3.1 vector. pcDNA-Nrf2 sent the mean ⫾ S.E. of three independent experiments, with each
was used as the template for Neh2, and pcDNA-Nrf2⌬PKC1 was used experiment done in triplicate.
as the template for the Neh2⌬S domain. The same primer set was used
to PCR both constructs: 5⬘-CCC TCA GCA TGA TGG ACT TGG-3⬘ trifugation. Cytosolic and nuclear extracts were prepared as follows.
(forward) and 5⬘-GGC CGG CTG AAT TTG GGG AGG-3⬘ (reverse). The cell pellet was suspended in 10 volumes of isotonic buffer (0.15 M
Kinase Inhibitor/Luciferase Reporter Assay—HepG2 cells were NaCl, 1.5 mM MgCl2, 10 mM Tris-HCl (pH 7.4), 0.5% (v/v) Nonidet P-40,
grown in monolayer cultures by procedures described previously (3). 1 mM dithiothreitol, 1 mM phenylmethylsulfonyl fluoride, 50 ␮M NaF, 1
The cells were cotransfected with a reporter construct containing the mM Na3VO4, and 1 ␮M okadaic acid) and homogenized in a Dounce
human NQO1 gene ARE driving luciferase expression and a Renilla homogenizer 10 times with a loose pestle. The nuclei were pelleted at
3500 rpm at 4 °C. The cytosolic extract was removed, and the pelleted
luciferase reporter as an internal control. Thirty-six h after transfec-
nuclei were lysed using radioimmune precipitation assay buffer (50 mM
tion, the cells were pretreated with inhibitors to various kinases
Tris (pH 8.0), 150 mM NaCl, 1% Nonidet P-40, 0.5% deoxycholate, 0.1%
(PD98059 for MEK/ERK, SB23580 for p38, wortmannin for phosphati-
SDS, 1 mM phenylmethylsulfonyl fluoride, 50 ␮M NaF, 1 mM Na3VO4,
dylinositol 3-kinase, genistein for tyrosine kinase, and calphostin C and
and 1 ␮M okadaic acid). Both extracts were cleared by centrifugation at
staurosporine for PKC) for 2 h. All the inhibitors were purchased from
12,000 rpm for 10 min. Nrf2 was immunoprecipitated from the nuclear
Calbiochem and were the highest purity available. The cells were
extracts using anti-Nrf2 antibody (H-300, Santa Cruz Biotechnology,
treated with a combination of kinase inhibitor and 50 ␮M t-butylhydro-
Inc., Santa Cruz, CA). Equal amounts of precipitate were run on an
quinone (t-BHQ) for 16 h. The cells were harvested, washed, homoge- SDS-polyacrylamide gel, and Western blotting was performed with
nized, and analyzed for luciferase activity. The dual luciferase reporter antibody specific to Nrf2 or phosphoserine (Sigma), phosphothreonine
assay from Promega (Madison, WI) and a Packard Topcount NXT (Cell Signaling, Salem, MA), or phosphotyrosine (Cell Signaling). The
luminometer (PerkinElmer Life Sciences) were used to measure lucif- Neh2 and Neh2⌬S domains were pulled down using Ni2⫹-nitrilotriace-
erase activity according to the manufacturers’ protocols. tic acid-Sepharose beads (QIAGEN Inc.) according to the manufactur-
NQO1 Activity—HepG2 cells were pretreated with calphostin C for er’s protocol.
2 h, and then fresh medium was added with Me2SO, with 50 ␮M t-BHQ, Similar assays were also performed with Hepa-1 cells transfected
or with 50 ␮M t-BHQ plus increasing concentrations of calphostin C. with the pcDNA-Neh2-V5 and pcDNA-Neh2⌬S-V5 plasmids in separate
Two h after treatment, the cells were harvested; cytosolic extracts were experiments. The cytosolic fractions from the transfected cells were
prepared; and NQO1 activity was assayed by a previously described prepared by the procedures described above. Cytosolic proteins (1 mg)
method (16). The cytosolic extracts from HepG2 cells treated with were immunoprecipitated with anti-V5 antibodies. Equal amounts of
t-BHQ and with t-BHQ plus calphostin C were also resolved by SDS- immunoprecipitated proteins from the transfected cells were separated
PAGE and Western-blotted, and the NQO1 protein level was visualized by 12% SDS-PAGE, Western-blotted, and probed with anti-V5 and
using antibodies to NQO1 and actin. anti-phosphoserine antibodies.
Bacterial Expression and Purification of Nrf2—Wild-type Nrf2 and Nrf2 Localization and Immunofluorescence—Hepa-1 cells were
Nrf2⌬PKC1 through Nrf2⌬PKC6 mutants were subcloned into the grown on Lab-Tek II chamber slides. The cells were pretreated with 10
pProExHTb vector from Invitrogen. These clones were transformed into nM staurosporine for 1 h and then treated with 50 ␮M t-BHQ alone or
subcloning efficiency DH5␣ competent cells (Invitrogen). The bacteria with 50 ␮M t-BHQ plus 10 nM staurosporine for 2 h. The cells were fixed
were grown at 37 °C and induced with 0.5 mM isopropyl-␤-D-thiogalac- using formalin and permeabilized with cold acetone. Immunofluores-
topyranoside when they reached A590 ⫽ 0.5. The bacteria were grown cence was then performed using anti-Nrf2 antibody H-300 and fluores-
for another 2 h at 37 °C and then harvested. His6-tagged Nrf2 was cein isothiocyanate-labeled secondary antibody (Rockland, Philadel-
purified on a nickel column (QIAGEN Inc.) under denaturing conditions phia, PA). The cells were stained with Hoechst stain to visualize the
according to the manufacturer’s protocol. Before Nrf2 was eluted, the nuclei. A Nikon Eclipse TE2000-U fluorescent microscope fitted with a
column was washed with diminishing amounts of urea. Nrf2 was then Photometrics CoolSnap CF camera and the appropriate filters was used
eluted under native conditions with 500 mM imidazole. to capture the fluorescent images.
In Vitro Kinase Assay—Equal amounts of purified Nrf2 or Nrf2⌬PKC
RESULTS
mutants were used as the substrate for purified PKC consisting pri-
marily of the ␣- and ␤-isozymes (Promega). The purified proteins were To determine which kinase pathway(s) is involved in ARE-
incubated with the PKC enzyme and [␥-32P]ATP in PKC buffer (20 mM mediated induction, HepG2 cells were transfected with a lucif-
Hepes (pH 7.4), 1 mM dithiothreitol, 10 mM MgCl2, 1.7 mM CaCl2, and erase reporter plasmid driven by the NQO1 gene ARE. Thirty-
0.1 mg/ml phosphatidylserine) for 1 h at 30 °C. The proteins were then
six h after transfection, the cells were treated with 50 ␮M
resolved by SDS-PAGE and visualized by autoradiography.
Immunoprecipitation and Phospho-specific Antibodies—Hepa-1 cells
t-BHQ for 16 h, resulting in ⬃3.5-fold induction over basal
were grown in 150-mm dishes. The cells were treated with Me2SO or 50 activity. This induction was inhibited by cotreatment with the
␮M t-BHQ for 2 h, washed twice with phosphate-buffered saline, PKC inhibitor staurosporine (Fig. 1). Similar results were also
scraped into 10 ml of phosphate-buffered saline, and pelleted by cen- observed with a second PKC inhibitor, calphostin C (Fig. 1).
PKC Regulation of ARE-mediated NQO1 Gene Expression 44677

FIG. 2. Effect of inhibitors of MEK/ERK, p38, phosphatidylinositol 3-kinase, and tyrosine kinases on t-BHQ induction of ARE-
mediated gene expression. HepG2 cells were grown in monolayers and cotransfected with ARE-luciferase and Renilla luciferase reporter
plasmids. Thirty-six h after transfection, the cells were pretreated with inhibitors for 2 h and then treated with Me2SO, with t-BHQ, or with t-BHQ
plus inhibitors for 16 h. The cells were harvested and lysed, and the lysates were analyzed for luciferase activity. The results are presented as the
mean ⫾ S.E. of three independent experiments, with each experiment done in triplicate. DMSO, Me2SO; PI3-K, phosphatidylinositol 3-kinase.

Inhibitors of MEK/ERK (PD98059), p38 (SB23580), phosphati-


dylinositol 3-kinase (wortmannin), and tyrosine kinases (genis-
tein) failed to block t-BHQ induction of the ARE (Fig. 2). The
small amount of inhibition observed with phosphatidylinositol
3-kinase and p38 inhibitors was not significant (p ⬎ 0.1). In-
terestingly, the inhibitors of MEK/ERK and tyrosine kinases
stimulated NQO1 ARE-mediated gene expression.
Calphostin C was also able to inhibit the induction of NQO1
activity in vivo (Fig. 3). Untransfected HepG2 cells were
treated with calphostin C or with calphostin C plus t-BHQ. The
lysates were then assayed for NQO1 activity, and the amount
of NQO1 was determined by Western blot analysis. Both NQO1
activity and the amount of NQO1 protein decreased in a dose-
dependent manner in response to treatment with the PKC
inhibitor.
To determine whether PKC inhibitors affect the localization
of Nrf2, immunofluorescence was used to follow the distribu-
tion of endogenous Nrf2 before and after treatment. Hepa-1
cells were grown on chamber slides; treated with Me2SO, with
t-BHQ, with staurosporine, or with t-BHQ and staurosporine;
fixed; and probed with anti-Nrf2 antibody (Fig. 4A). In control
cells, Nrf2 was found localized in the cytoplasm and nucleus,
although the fluorescence was weak. After treatment with t-
BHQ, Nrf2 accumulated in the nucleus, resulting in a much
stronger signal. The PKC inhibitor staurosporine was able to
FIG. 3. In vivo effect of calphostin C on NQO1 activity and
block the t-BHQ-induced nuclear accumulation of Nrf2. These protein. HepG2 cells were treated with Me2SO, with t-BHQ, or with
results were also confirmed by Western blot analysis. Cytosolic t-BHQ plus increasing concentrations of calphostin C for 16 h. Cytosolic
and nuclear extracts were prepared from treated and untreated extracts were prepared and analyzed for NQO1 activity (upper panel).
Hepa-1 cells. The extracts were resolved by SDS-PAGE and The cytosolic extracts from HepG2 cells treated with t-BHQ and with
t-BHQ plus calphostin C were also analyzed for NQO1 protein by
probed with anti-Nrf2 antibody (Fig. 4B). Nrf2 was hardly SDS-PAGE and Western blotting (lower panel). Western blots were
visible in any of the cytosolic extracts or in the nuclear extracts probed with antibodies to NQO1 and actin. Anti-actin antibody was
from control cells or from cells treated with staurosporine plus used to show equal loading of proteins in the lanes.
44678 PKC Regulation of ARE-mediated NQO1 Gene Expression

FIG. 4. Effect of staurosporine on t-BHQ-induced nuclear accumulation of Nrf2. A, Hepa-1 cells were grown and treated on chamber
slides. They were fixed and probed with anti-Nrf2 antibody, followed by fluorescein isothiocyanate (FITC)-labeled secondary antibody. The cells
were also stained with Hoechst stain to visualize the nuclei. B, Hepa-1 cells were treated, and cytosolic and nuclear extracts were prepared. The
extracts were resolved by SDS-PAGE and probed with anti-Nrf2 antibody. The membrane was then stripped and probed with anti-actin antibody
to show equal loading. DMSO, Me2SO.

t-BHQ. However, there was an abundance of Nrf2 in the nu- were made and are referred to as Nrf2⌬PKC1 through
cleus of t-BHQ-treated cells. These results coincide with the Nrf2⌬PKC6 (Fig. 6A). All seven PKC sites were mutated from
proposed model that Nrf2 is rapidly degraded in the cytosol as serine/threonine to alanine. The two threonines located next to
a result of the Nrf2/INrf2 interaction and is stabilized/accumu- each other at positions 417 and 418 were mutated in the same
lated in nuclei after chemically induced oxidative stress (5–7). construct, Nrf2⌬PKC3 (Fig. 6A).
Next, we studied the ability of PKC to phosphorylate Nrf2 in Mutating the putative PKC sites individually did not have a
vitro. Histidine-tagged Nrf2 was overexpressed in a bacterial significant effect on the activity of any of the mutants (Fig. 6B,
system and purified using nickel-coated Sepharose beads. This data shown only for Nrf2⌬PKC1). However, mutating the PKC
Nrf2 was used as the substrate in an in vitro kinase assay site in the Neh2 domain resulted in INrf2 inhibiting
utilizing purified PKC as the enzyme (Fig. 5A). Nrf2 was phos- Nrf2⌬PKC1 more efficiently than wild-type Nrf2 (Fig. 6C). The
phorylated by PKC in vitro. Staurosporine was able to inhibit inhibition of the other mutants by INrf2 was not affected (data
the phosphorylation, whereas wortmannin, a phosphatidyli- not shown). In vitro kinase results revealed that Nrf2⌬PKC1
nositol 3-kinase inhibitor, was not. was not phosphorylated by PKC (Fig. 6D).
To determine whether Nrf2 is phosphorylated in vivo, we To further study the effect of this mutation in vivo, con-
utilized antibodies specific to phosphoserine, phosphothreo- structs were created that would express the Neh2 domain or
nine, and phosphotyrosine. Hepa-1 cells were treated with the Neh2 domain with the PKC1 serine mutation, Neh2⌬S
Me2SO or t-BHQ. Nrf2 was immunoprecipitated from the cy- (Fig. 7A). Both domains were tagged with V5 and His6. HepG2
tosolic and nuclear extracts, and Western analysis was per- cells were transfected with increasing amounts of these con-
formed (Fig. 5B). This revealed that Nrf2 that accumulated in structs as well as the ARE-luciferase reporter. As expected, the
the nucleus in response to t-BHQ treatment was phosphoryl- wild-type Neh2 domain was able to compete with endogenous
ated at a serine residue(s). Phosphorylation of Nrf2 in the Nrf2 for INrf2. This resulted in a concentration-dependent
untreated nuclear extracts could not be detected. It is possible
increase in ARE-luciferase activity (Fig. 7B). At the highest
that it is not phosphorylated or that the amount of protein was
dose, the Neh2 domain induced ARE activity to the same extent
not sufficient to detect the phosphorylation. Likewise, we could
as t-BHQ in cells that were not transfected with the Neh2
not detect phosphorylation with anti-phosphothreonine or anti-
domain. However, the Neh2⌬S domain was much more effi-
phosphotyrosine antibody. The question arises whether PKC
cient in inducing ARE activity. At the highest dose, it induced
inhibitors can block the phosphorylation. However, there was
ARE activity almost twice as much as t-BHQ alone or the
an insufficient amount of Nrf2 in the staurosporine-treated
wild-type Neh2 domain.
nuclear extracts to determine phosphorylation.
To demonstrate binding of the Neh2 and Neh2⌬S domains to
A Prosite search revealed seven putative PKC sites in Nrf2.
INrf2 and that the domains can cause the nuclear accumula-
Of these, four are serine residues.2 PKC site 1, Ser40, is located
tion of Nrf2, Hepa-1 cells were transfected with an empty
in the N-terminal Neh2 domain of Nrf2, the domain that in-
vector or with vector encoding the Neh2 or Neh2⌬S domain.
teracts with INrf2 (2, 3). This PKC site is conserved across the
species (human, rat, mouse, and chicken).2 Six Nrf2 mutants The cells were treated with Me2SO or t-BHQ, and cytosolic and
nuclear extracts were made. Western analysis revealed that
the Neh2 and Neh2⌬S domains were able to cause the nuclear
2
D. A. Bloom and A. K. Jaiswal, unpublished data. accumulation of Nrf2 (Fig. 7C, upper panel). Treatment with
PKC Regulation of ARE-mediated NQO1 Gene Expression 44679

FIG. 5. In vitro and in vivo phospho-


rylation of Nrf2 by PKC. A, Nrf2 was
expressed in an isopropyl-␤-D-thiogalacto-
pyranoside-inducible bacterial system.
Purified Nrf2 was phosphorylated in vitro
using a mixture of PKC isoforms. B, en-
dogenous Nrf2 was immunoprecipitated
from cytosolic (C) and nuclear (N) extracts
prepared from Hepa-1 cells treated with
Me2SO or t-BHQ. The immunoprecipi-
tated proteins were resolved by SDS-
PAGE and probed with anti-Nrf2, anti-
phosphoserine, anti-phosphothreonine, or
anti-phosphotyrosine antibody.

FIG. 6. PKC phosphorylation of wild-type and mutant Nrf2. A, shown is a graphical representation of Nrf2 showing the locations of several
important domains and the PKC phosphorylation sites. Nrf2⌬PKC3 contains mutations of two overlapping PKC sites in Nrf2. b-Zip, basic leucine
zipper. B, HepG2 cells were transfected with the NQO1 ARE-luciferase reporter along with wild-type Nrf2 or one of the Nrf2⌬PKC mutants.
Renilla luciferase was included in each transfection as transfection efficiency control. Thirty-six h later, the cells were treated with Me2SO (DMSO)
or t-BHQ for 16 h. The cells were harvested, and luciferase activity was determined. C, HepG2 cells were cotransfected with the NQO1
ARE-luciferase reporter plasmid, Nrf2, or Nrf2⌬PKC1 with or without INrf2. The cells were harvested and analyzed for luciferase activity. D,
bacterially expressed and purified Nrf2 or Nrf2⌬PKC1 was used as the substrate in an in vitro kinase assay following the procedures described
under “Experimental Procedures.” The proteins were analyzed by SDS-PAGE and autoradiographed (upper panel) or stained with Coomassie Blue
(lower panel).

t-BHQ or expression of the Neh2 domain caused nearly an The Neh2 and Neh2⌬S domains tagged with histidines were
equal amount of Nrf2 accumulation in the nucleus. However, precipitated from the cytosolic extract using nickel-coated
expression of the Neh2⌬S domain caused a greater amount of Sepharose beads. Western analysis using anti-INrf2 antibody
Nrf2 to accumulate in the nucleus, consistent with the ARE revealed that both the Neh2 and Neh2⌬S domains bound to
activity data. The Neh2 and Neh2⌬S domains were equally INrf2 in uninduced cells (Fig. 7D). However, after t-BHQ treat-
expressed (Fig. 7C, middle panel). ment, the wild-type Neh2 domain no longer associated with
44680 PKC Regulation of ARE-mediated NQO1 Gene Expression

FIG. 7. In vivo effect of t-BHQ and the Neh2 and mutant Neh2 domains on ARE-mediated gene expression, stabilization/nuclear
accumulation, and phosphorylation of Nrf2. A, Nrf2 is shown with functional protein domains including Neh2. The Neh2 and mutant Neh2
domains tagged with V5 are also shown. Ser40 in the Neh2 domain was mutated to alanine to generate the mutant Neh2 domain. The construction
of recombinant plasmids expressing Neh2 and mutant Neh2 domains was described under “Experimental Procedures.” Constructs expressing the
wild-type Neh2 domain or the Neh2⌬S domain tagged with V5 and His6 were created. b-Zip, basic leucine zipper. B, the plasmids expressing the
Neh2 or Neh2⌬S domain were transfected into HepG2 cells along with the ARE-luciferase reporter. Thirty-six h after transfection, the cells were
treated with Me2SO (DMSO) or t-BHQ. Forty-eight h after transfection, the cells were harvested, and the extracts were analyzed by dual luciferase
assay. C, Hepa-1 cells were either treated with t-BHQ or transfected with either the Neh2 or Neh2⌬S domain. Cytosolic and nuclear extracts were
prepared, and the lysates were resolved by SDS-PAGE. The proteins were transferred to nylon membranes. The membranes were then probed with
anti-Nrf2 antibody, stripped, probed with anti-V5 antibody, stripped again, and probed with anti-actin antibody. D, the cytosolic extracts were
made from untreated or t-BHQ-treated Hepa-1 cells that had been transfected with either the Neh2 or Neh2⌬S domain of Nrf2 tagged with V5 and
histidines. The Neh2 and mutant Neh2 domains were affinity-purified on nickel-coated Sepharose beads. After washing, the proteins were eluted
and resolved by SDS-PAGE and transferred to nylon membranes. The membranes were probed with anti-INrf2 antibody, stripped, and probed with
anti-V5 antibody. E, the Hepa-1 cells were either treated with t-BHQ or transfected with the Neh2 or Neh2⌬S domain. The t-BHQ-treated and
transfected Hepa-1 cells were homogenized; nuclei were isolated; and nuclear extracts were prepared. Nrf2 was immunoprecipitated from these
nuclear extracts with anti-Nrf2 antibody. The eluted proteins were analyzed by SDS-PAGE and Western blotting. Western blots were probed with
either anti-Nrf2 or anti-phosphoserine antibody. Equal amounts of the immunoprecipitated proteins were loaded in the lanes. F, the Hepa-1 cells
were transfected with plasmid pcDNA-Neh2-V5 or pcDNA-Neh2⌬S-V5, and cytosolic fractions were prepared by standard procedures. Cytosolic
proteins (1 mg) from the transfected cells were immunoprecipitated with anti-V5 antibody. Equal amounts of immunoprecipitated proteins from
each transfection were separated by 12% SDS-PAGE, Western-blotted, and probed with anti-V5 and anti-phosphoserine antibodies.

INrf2. The interaction of the Neh2⌬S domain with INrf2 was for Nrf2 to accumulate in the nucleus. Furthermore, the un-
not affected by t-BHQ treatment. phosphorylated form of Nrf2 is able to activate transcription of
Next, Nrf2 was immunoprecipitated from the nuclear ex- the ARE.
tracts from the previous experiment (Fig. 7E). Western analy-
sis demonstrated that only Nrf2 from cells induced with t-BHQ DISCUSSION
was phosphorylated at serine residues. Nrf2 that accumulated NQO1 is a phase II detoxifying enzyme that competes with
in the nucleus of cells transfected with either the Neh2 or the one-electron reducing enzyme cytochrome P450 reductase
Neh2⌬S domain was not phosphorylated. It may be noteworthy and catalyzes two-electron reductive metabolism and detoxifi-
that each lane contains an equal amount of immunoprecipi- cation of quinones (1). NQO1-null mice have been generated,
tated Nrf2 in Fig. 7E. It is this reason that the increase in Nrf2 and studies have shown that NQO1 plays an important role in
in the presence of t-BHQ in Fig. 7C is not visible in Fig. 7E. regulating the intracellular redox status of cells and protects
Further studies with Hepa-1 cells transfected with Neh2-V5 against myelogenous hyperplasia and quinone-induced redox
and Neh2⌬S-V5 and anti-phosphoserine antibody demon- cycling, oxidative stress, and cytotoxicity (17–19). Studies have
strated that Neh2-V5 and not Neh2⌬S-V5 was phosphorylated also shown that NQO1 protects the skin against benzo-
(Fig. 7F). [a]pyrene- and 9,10-dimethyl-1,2-benzanthracene-induced car-
Taken together, these results indicate that the phosphoryl- cinogenesis (20, 21).
ation of Nrf2 at Ser40 by PKC is required for Nrf2 to evade Antioxidants and oxidative stress induce a battery of more
INrf2-mediated degradation. Phosphorylation is not required than two dozen defensive genes, including NQO1, by an ARE-
PKC Regulation of ARE-mediated NQO1 Gene Expression 44681

FIG. 8. Role of PKC in transcriptional activation of ARE-mediated gene expression. Two alternative hypotheses are shown. Both require
PKC-mediated phosphorylation of Nrf2, leading to either release of Nrf2 from INrf2 (Hypothesis I) or escape of Nrf2 from INrf2 (Hypothesis II).
The Nrf2 heterodimeric partner is shown as Unknown. This is because Jun and small Maf proteins have been shown to form heterodimers with
Nrf2. However, none of these proteins have been clearly established as the activating heterodimeric partner of Nrf2.

dependent mechanism (reviewed in Ref. 1). This coordinated A recent report had demonstrated that Nrf2 is phosphoryl-
induction of detoxifying genes is of critical importance for an- ated in vitro by PKC and that this affects the interaction of
tioxidant action and chemoprevention (reviewed in Ref. 1). The Nrf2 with INrf2 in vitro (14). However, the involvement of PKC
transcription factor Nrf2 is known to bind to the NQO1 ARE as had been contradicted by other reports (8 –13). We confirm that
well as to the AREs of several other detoxifying enzyme genes PKC activity is critical for the induction of ARE-mediated gene
and to activate their transcription in response to antioxidants expression by t-BHQ. Furthermore, we show for the first time
and xenobiotics (reviewed in Ref. 1). Under normal conditions, in vivo evidence that phosphorylation of Ser40 is necessary for
Nrf2 is believed to be retained in the cytoplasm by a cytosolic Nrf2 to dissociate from INrf2. Mutation of this residue in the
inhibitor, INrf2 (3, 4). Recently, it has also been shown that Neh2 domain resulted in the Neh2⌬S domain/INrf2 interaction
INrf2 targets Nrf2 for proteasomal degradation (5–7). Treat- being unresponsive to t-BHQ treatment.
ment of cells with antioxidants disrupts the Nrf2/INrf2 inter- Importantly, inducing Nrf2 nuclear accumulation by overex-
action (3, 4). When Nrf2 is no longer associated with INrf2, it is pressing the Neh2 domain resulted in an increase in ARE
stabilized and accumulates in the nucleus. It forms het- activity similar to that caused by t-BHQ induction. The mutant
erodimers with other leucine zipper proteins, leading to the Neh2⌬S domain induced ARE expression and Nrf2 accumula-
coordinated activation of NQO1 and other detoxifying enzyme tion much more efficiently than the wild-type Neh2 domain or
genes, including glutathione S-transferase Ya, ␥-glutamylcys- t-BHQ. These data point to the fact that phosphorylation of
teinyl synthetase, and heme oxygenase-1 (22–25). c-Jun and Nrf2 is not required for it to activate transcription. We dem-
small Maf proteins have been shown to be heterodimeric part- onstrated this by showing that Nrf2 that accumulated in the
ners of Nrf2, leading to the activation of detoxifying genes (26, nucleus in response to t-BHQ treatment was phosphorylated at
27). However, the role of small Maf proteins is controversial. a serine residue(s), but that Nrf2 that accumulated in the
Other studies have shown that Nrf2-small Maf heterodimers nucleus as a result of Neh2 overexpression was not. Clearly, it
repress ARE-mediated gene expression (23, 25). t-BHQ treat- is the association of Nrf2 with INrf2 that is regulated by PKC,
ment does not affect the levels of Nrf2 mRNA (2). Therefore, we not Nrf2 stability or activity.
suspected that t-BHQ treatment leads to the modification of The role of PKC phosphorylation of Nrf2 in ARE-mediated
Nrf2 and that these modifications are critical for ARE-medi- gene expression is shown in Fig. 8. Two alternative hypotheses
ated induction of detoxifying enzyme genes. are depicted. Briefly, in Hypothesis I, Nrf2 is retained in the
It is interesting that inhibitors of MEK/ERK and tyrosine cytoplasm by INrf2. Antioxidants induce the expression of or
kinases activate ARE-mediated gene expression. It is possible activate PKC. PKC then phosphorylates Nrf2 that is bound to
that phosphorylation of Nrf2 by members of these kinase fam- INrf2. This phosphorylation leads to the release of Nrf2 from
ilies facilitates the binding of Nrf2 to INrf2 and therefore INrf2 and the stabilization/nuclear accumulation of Nrf2.
degradation of Nrf2. Tyrosine kinases are involved in regulat- Phosphorylated Nrf2 binds to the ARE and activates ARE-
ing cell growth, so it would not be surprising if this pathway is mediated gene expression. This hypothesis is based on cur-
linked to defensive measures. Inhibition of cell growth could rently accepted results on Nrf2 interaction with INrf2, antiox-
serve as a warning to the cell that the environmental condi- idant-induced release of Nrf2 from INrf2, and nuclear
tions are unfavorable. The cell would then activate defensive localization of Nrf2 (3, 4). However, an alternative hypothesis
genes, through the ARE, to limit cellular damage. that can not be ruled out is possible. In Hypothesis II, INrf2 is
44682 PKC Regulation of ARE-mediated NQO1 Gene Expression
an inhibitor of unphosphorylated Nrf2. INrf2 binds to unphos- 2. Jaiswal, A. K. (2000) Free Radic. Biol. Med. 29, 254 –262
3. Dhakshinamoorthy, S., and Jaiswal, A. K. (2001) Oncogene 20, 3906 –3917
phorylated Nrf2 and stimulates its degradation (5–7). Antioxi- 4. Itoh, K., Wakabayashi, N., Katoh, Y., Ishii, T., Igarashi, K., Engel, J. D., and
dants/oxidants/other stresses induce/activate PKC, which Yamamoto, M. (1999) Genes Dev. 13, 76 – 86
5. Itoh, K., Wakabayashi, N., Katoh, Y., Tetsuro, I., O’Connor, T., and Yamamoto,
phosphorylates free Nrf2. Phosphorylated Nrf2 escapes INrf2- M. (2003) Genes Cells 8, 379 –391
mediated degradation and accumulates in the nucleus, where it 6. Nguyen, T., Sherratt, P. J., Huang, H. C., Yang, C. S., and Pickett, C. B. (2003)
activates ARE-mediated gene expression. Hypothesis II is J. Biol. Chem. 278, 4536 – 4541
7. McMahon, M., Itoh, K., Yamamoto, M., and Hayes, J. D. (2003) J. Biol. Chem.
based on several observations. First, INrf2 targets Nrf2 for 278, 21569 –21600
proteasomal degradation (5–7). Second, PKC phosphorylates 8. Yu, R., Jiao, J. J., Duh, J. L., Tan, T.-H., and Tony Kong, A.-N. (1996) Cancer
Res. 56, 2954 –2959
Nrf2 in the Neh2 domain, which binds to INrf2 (this study and 9. Yu, R., Mandlekar, S., Weber, M. J., Der, C. J., Wu, J., and Tony Kong, A.-N.
Ref. 4). The binding of INrf2 to the Neh2 domain might inter- (1999) J. Biol. Chem. 274, 27545–27552
fere with the availability of Ser40 for phosphorylation by PKC. 10. Yu, R., Mandlekar, S., Lei, W., Fahl, W. E., Tan, T.-H., and Tony Kong, A.-N.
(2000) J. Biol. Chem. 275, 2322–2327
Third, Nrf2 does not require phosphorylation to accumulate in 11. Yu, R., Chen, C., Mo, Y. Y., Hebbar, V., Owuor, E. D., Tan, T.-H., and Tony
the nucleus and to activate ARE-mediated gene expression Kong, A.-N. (2000) J. Biol. Chem. 275, 39907–39913
12. Kang, K. W., Ryu, H., and Kim, S. G. (2000) Mol. Pharmacol. 58, 1017–1025
(this study). Future experiments should select one hypothesis 13. Dieter, M. Z., Freshwater, S. L., Solis, W. A., Nebert, D. W., and Dalton, T. P.
and rule out the other. (2001) Biochem. Pharmacol. 61, 215–225
The data suggest that any mechanism that modifies Nrf2, 14. Huang, H. C., Nguyen, T., and Pickett, C. B. (2000) Proc. Natl. Acad. Sci.
U. S. A. 97, 12475–12480
allowing it to disrupt the Nrf2-INrf2 complex and/or to escape 15. Huang, H. C., Nguyen, T., and Pickett, C. (2002) J. Biol. Chem. 277,
Nrf2 binding to INrf2, will result in the up-regulation of ARE- 42769 – 42774
16. Shaw, P. M., Reiss, A., Adesnik, M., Nebert, D. W., Schembri, J., and Jaiswal,
mediated gene expression. Recently, four sulfhydryl groups in A. K. (1991) Eur. J. Biochem. 195, 171–176
INrf2 that are sensitive to the redox state of the cell were 17. Radjendirane, V., Joseph, P., Lee, Y.-H., Kimura, S., Klein-Sazanto, A. J. P.,
reported (29). Alteration of these sulfhydryl groups leads to the Gonzalez, F. J., and Jaiswal, A. K. (1998) J. Biol. Chem. 273, 7382–7389
18. Gaikwad, A., Long, D. J., II, Stringer, J. L., and Jaiswal, A. K. (2001) J. Biol.
loss of the association of Nrf2 with INrf2, leading to the acti- Chem. 276, 22559 –22564
vation of ARE-mediated gene expression (28). It is possible that 19. Long, D. J., II, Gaikwad, A., Multani, A., Pathak, S., Montgomery, C. A.,
Gonzalez, F. J., and Jaiswal, A. K. (2002) Cancer Res. 62, 3030 –3036
this mechanism is redundant to the phosphorylation of Nrf2 by 20. Long, D. J., II, Waikel, R. L., Wang, X., Perlaky, L., Roop, D. R., and Jaiswal,
PKC or that the two mechanisms work in concert. It is also A. K. (2000) Cancer Res. 60, 5913–5915
possible that these two mechanisms are activated in a stimu- 21. Long, D. J., II, Waikel, R. L., Wang, X. J., Roop, D. R., and Jaiswal, A. K. (2001)
J. Natl. Cancer Inst. 93, 1166 –1170
lus- and/or cell type-specific manner. Likewise, there could be 22. Venugopal, R., and Jaiswal, A. K. (1996) Proc. Natl. Acad. Sci. U. S. A. 93,
other cell type-specific factors that disrupt the Nrf2-INrf2 com- 14960 –14965
23. Wild, A. C., Moinova, H. R., and Mulcahy, R. T. (1999) J. Biol. Chem. 274,
plex or modify Nrf2, preventing its interaction with INrf2 in 33627–33636
response to various stimuli. 24. Gong, P., Hu, B., Stewart, D., Ellerbe, M., Figueroa, Y. G., Blank, V., Beckman,
B. S., and Alam, J. (2001) J. Biol. Chem. 276, 27018 –27025
Acknowledgments—We are grateful to Drs. Jefferson Y. Chan and 25. Nguyen, T., Huang, H. C., and Pickett, C. B. (2000) J. Biol. Chem. 275,
Yuet W. Kan (both from University of California, San Francisco) for 15466 –15473
26. Itoh, K., Chiba, T., Takahashi, S., Ishii, T., Igarashi, K., Katoh, K., Oyake, T.,
providing cDNA encoding Nrf2. We thank Dr. S. Dhakshinamoorthy for
Hayashi, N., Satoh, K., Hatayama, I., Yamamoto, M., and Nabeshima, Y.
helpful suggestions. (1997) Biochem. Biophys. Res. Commun. 236, 313–322
27. Venugopal, R., and Jaiswal, A. K. (1998) Oncogene 17, 3145–3156
REFERENCES
28. Dinkova-Kostova, A. T., Holtzclaw, W. D., Cole, R. N., Itoh, K., Wakabayashi,
1. Dhakshinamoorthy, S., Long, D. J., II, and Jaiswal, A. K. (2000) Curr. Top. N., Katoh, Y., Yamamoto, M., and Talalay, P. (2002) Proc. Natl. Acad. Sci.
Cell. Regul. 36, 201–216 U. S. A. 99, 11908 –11913

You might also like