nuclearhemeoxygenase-1(ho-1)modulatessubcellular … · 2014-09-19 · matic action in heme...

14
Nuclear Heme Oxygenase-1 (HO-1) Modulates Subcellular Distribution and Activation of Nrf2, Impacting Metabolic and Anti-oxidant Defenses * Received for publication, March 25, 2014, and in revised form, August 5, 2014 Published, JBC Papers in Press, August 8, 2014, DOI 10.1074/jbc.M114.567685 Chhanda Biswas ‡§ , Nidhi Shah § , Manasa Muthu § , Ping La § , Amal P. Fernando § , Shaon Sengupta ‡§ , Guang Yang § , and Phyllis A. Dennery ‡§1 From the Department of Pediatrics, University of Pennsylvania Philadelphia, Pennsylvania 19104 and the § Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104 Background: A 28-kDa HO-1 isoform is induced by oxidative stress and cancer and accumulates in the nucleus. Results: Nuclear HO-1 interacts with Nrf2 and alters expression of its target genes. Conclusion: HO-1 modulates Nrf2 function. Significance: Exploiting the synergistic benefits of the HO-1Nrf2 protein complex is important for developing therapeutic strategies against oxidative stress or cancer. With oxidative injury as well as in some solid tumors and mye- loid leukemia cells, heme oxygenase-1 (HO-1), the anti-oxidant, anti-inflammatory, and anti-apoptotic microsomal stress pro- tein, migrates to the nucleus in a truncated and enzymatically inactive form. However, the function of HO-1 in the nucleus is not completely clear. Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor and master regulator of numerous antioxidants and anti-apoptotic proteins, including HO-1, also accumulates in the nucleus with oxidative injury and in various types of cancer. Here we demonstrate that in oxidative stress, nuclear HO-1 interacts with Nrf2 and stabilizes it from glycogen synthase kinase 3 (GSK3)-mediated phosphorylation cou- pled with ubiquitin-proteasomal degradation, thereby prolong- ing its accumulation in the nucleus. This regulation of Nrf2 post-induction by nuclear HO-1 is important for the preferen- tial transcription of phase II detoxification enzymes such as NQO1 as well as glucose-6-phosphate dehydrogenase (G6PDH), a regulator of the pentose phosphate pathway. Using Nrf2 knock-out cells, we further demonstrate that nuclear HO-1-as- sociated cytoprotection against oxidative stress depends on an HO-1/Nrf2 interaction. Although it is well known that Nrf2 induces HO-1 leading to mitigation of oxidant stress, we pro- pose a novel mechanism by which HO-1, by modulating the acti- vation of Nrf2, sets an adaptive reprogramming that enhances antioxidant defenses. The nuclear factor erythroid 2-related factor 2 or Nrf2, 2 a member of the cap’n’collar family of basic leucine zipper tran- scription factors, plays a crucial role in antioxidant defenses (1, 2). Nrf2 is constitutively expressed in the cytoplasm, and its accumulation and activation in the nucleus are favored in oxi- dative injury and also in cancer cells (3– 6). In the nucleus, Nrf2 in a heterodimer with small Maf proteins binds to the antioxi- dant response elements (ARE) in the promoter regions of Nrf2 target genes. This signaling facilitates transcription of numer- ous antioxidant enzymes and growth factors, thereby promot- ing tolerance to oxidative stress (2, 6, 7). In addition, Bach1, which competes with Nrf2 for binding to the ARE, suppresses ARE-mediated gene transcription (8). One important step in Nrf2 signaling is stabilization of the constitutive Nrf2 from Kelch-like ECH-associated protein 1 (KEAP1), which acts as an adaptor protein between Nrf2 via the Neh2 domain at the N terminus and Cul3 E3 ligase targeting Nrf2 for ubiquitin-pro- teasomal degradation. Many studies show that, with oxidative or electrophilic stress, both Nrf2 and Keap1 undergo post- translational modification, favoring release of Nrf2 by the deg- radation of Keap1 thereby facilitating stabilization and nuclear transport of Nrf2 (9 –13). However, the regulation of Nrf2 is complex and involves a network of intricate mechanisms beyond Keap1 (14 –19). More recently it was reported that GSK3-mediated phosphorylation of Ser 344 and Ser 347 within the phosphodegron motif of the Neh6 domain targets Nrf2 pro- tein for proteasomal degradation by another E3 ligase, -TrCP, in a Keap1-independent manner (20 –22). This process is thought to occur in the nucleus post-induction. It is well documented that heme oxygenase (HO)-1, a 32-kDa protein, is positively regulated by Nrf2 (1, 23). As a protective and adaptive response, most tissues exhibit robust activation of the highly inducible HO-1 (24 –28). The beneficial effects of HO-1 in cytoprotection have long been attributed to its enzy- * This work was supported, in whole or in part, by National Institutes of Health Grants HL-58752 and HL-11190 from the National Institutes of Health (to P. A. D.) 1 To whom correspondence should be addressed: Dept. of Pediatrics, Univer- sity of Pennsylvania and Division of Neonatology, Children’s Hospital of Philadelphia, 34th and Civic Center Blvd. Philadelphia, PA, 19104. Tel.: 215- 590-1653; Fax: 267-426-5693, E-mail: [email protected]. 2 The abbreviations used are: Nrf2, nuclear factor erythroid 2-related factor 2; ARE, antioxidant response element; HO-1, heme oxygenase-1; HO-1-TR, trun- cated HO-1, HO-1-FL, full-length HO-1; V, empty vector; MEF, mouse embry- onic fibroblast; IP, immunoprecipitation; NQO1, NAD(P)H dehydroge- nase, quinone 1; G6PDH, glucose-6-phosphate dehydrogenase; GSK3, glycogen synthase kinase 3; SOD2, superoxide dismutase 2; DHE, dihydroethidium; Keap1, Kelch-like ECH-associated protein 1; Bis-Tris, 2- (bis(2-hydroxyethyl)amino)-2-(hydroxymethyl)propane-1,3-diol; DMSO, dimethyl sulfoxide; XTT, 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tet- razolium-5-carboxanilide inner salt; TR, truncated; p, phosphorylated. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 39, pp. 26882–26894, September 26, 2014 © 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. 26882 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014 by guest on November 5, 2020 http://www.jbc.org/ Downloaded from

Upload: others

Post on 10-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

Nuclear Heme Oxygenase-1 (HO-1) Modulates SubcellularDistribution and Activation of Nrf2, Impacting Metabolic andAnti-oxidant Defenses*

Received for publication, March 25, 2014, and in revised form, August 5, 2014 Published, JBC Papers in Press, August 8, 2014, DOI 10.1074/jbc.M114.567685

Chhanda Biswas‡§, Nidhi Shah§, Manasa Muthu§, Ping La§, Amal P. Fernando§, Shaon Sengupta‡§, Guang Yang§,and Phyllis A. Dennery‡§1

From the ‡Department of Pediatrics, University of Pennsylvania Philadelphia, Pennsylvania 19104 and the §Division ofNeonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104

Background: A 28-kDa HO-1 isoform is induced by oxidative stress and cancer and accumulates in the nucleus.Results: Nuclear HO-1 interacts with Nrf2 and alters expression of its target genes.Conclusion: HO-1 modulates Nrf2 function.Significance: Exploiting the synergistic benefits of the HO-1�Nrf2 protein complex is important for developing therapeuticstrategies against oxidative stress or cancer.

With oxidative injury as well as in some solid tumors and mye-loid leukemia cells, heme oxygenase-1 (HO-1), the anti-oxidant,anti-inflammatory, and anti-apoptotic microsomal stress pro-tein, migrates to the nucleus in a truncated and enzymaticallyinactive form. However, the function of HO-1 in the nucleus isnot completely clear. Nuclear factor erythroid 2-related factor 2(Nrf2), a transcription factor and master regulator of numerousantioxidants and anti-apoptotic proteins, including HO-1, alsoaccumulates in the nucleus with oxidative injury and in varioustypes of cancer. Here we demonstrate that in oxidative stress,nuclear HO-1 interacts with Nrf2 and stabilizes it from glycogensynthase kinase 3� (GSK3�)-mediated phosphorylation cou-pled with ubiquitin-proteasomal degradation, thereby prolong-ing its accumulation in the nucleus. This regulation of Nrf2post-induction by nuclear HO-1 is important for the preferen-tial transcription of phase II detoxification enzymes such asNQO1 as well as glucose-6-phosphate dehydrogenase (G6PDH),a regulator of the pentose phosphate pathway. Using Nrf2knock-out cells, we further demonstrate that nuclear HO-1-as-sociated cytoprotection against oxidative stress depends on anHO-1/Nrf2 interaction. Although it is well known that Nrf2induces HO-1 leading to mitigation of oxidant stress, we pro-pose a novel mechanism by which HO-1, by modulating the acti-vation of Nrf2, sets an adaptive reprogramming that enhancesantioxidant defenses.

The nuclear factor erythroid 2-related factor 2 or Nrf2,2 amember of the cap’n’collar family of basic leucine zipper tran-

scription factors, plays a crucial role in antioxidant defenses (1,2). Nrf2 is constitutively expressed in the cytoplasm, and itsaccumulation and activation in the nucleus are favored in oxi-dative injury and also in cancer cells (3– 6). In the nucleus, Nrf2in a heterodimer with small Maf proteins binds to the antioxi-dant response elements (ARE) in the promoter regions of Nrf2target genes. This signaling facilitates transcription of numer-ous antioxidant enzymes and growth factors, thereby promot-ing tolerance to oxidative stress (2, 6, 7). In addition, Bach1,which competes with Nrf2 for binding to the ARE, suppressesARE-mediated gene transcription (8). One important step inNrf2 signaling is stabilization of the constitutive Nrf2 fromKelch-like ECH-associated protein 1 (KEAP1), which acts as anadaptor protein between Nrf2 via the Neh2 domain at the Nterminus and Cul3 E3 ligase targeting Nrf2 for ubiquitin-pro-teasomal degradation. Many studies show that, with oxidativeor electrophilic stress, both Nrf2 and Keap1 undergo post-translational modification, favoring release of Nrf2 by the deg-radation of Keap1 thereby facilitating stabilization and nucleartransport of Nrf2 (9 –13). However, the regulation of Nrf2 iscomplex and involves a network of intricate mechanismsbeyond Keap1 (14 –19). More recently it was reported thatGSK3�-mediated phosphorylation of Ser344 and Ser347 withinthe phosphodegron motif of the Neh6 domain targets Nrf2 pro-tein for proteasomal degradation by another E3 ligase, �-TrCP,in a Keap1-independent manner (20 –22). This process isthought to occur in the nucleus post-induction.

It is well documented that heme oxygenase (HO)-1, a 32-kDaprotein, is positively regulated by Nrf2 (1, 23). As a protectiveand adaptive response, most tissues exhibit robust activation ofthe highly inducible HO-1 (24 –28). The beneficial effects ofHO-1 in cytoprotection have long been attributed to its enzy-* This work was supported, in whole or in part, by National Institutes of Health

Grants HL-58752 and HL-11190 from the National Institutes of Health (toP. A. D.)

1 To whom correspondence should be addressed: Dept. of Pediatrics, Univer-sity of Pennsylvania and Division of Neonatology, Children’s Hospital ofPhiladelphia, 34th and Civic Center Blvd. Philadelphia, PA, 19104. Tel.: 215-590-1653; Fax: 267-426-5693, E-mail: [email protected].

2 The abbreviations used are: Nrf2, nuclear factor erythroid 2-related factor 2;ARE, antioxidant response element; HO-1, heme oxygenase-1; HO-1-TR, trun-cated HO-1, HO-1-FL, full-length HO-1; V, empty vector; MEF, mouse embry-

onic fibroblast; IP, immunoprecipitation; NQO1, NAD(P)H dehydroge-nase, quinone 1; G6PDH, glucose-6-phosphate dehydrogenase; GSK3�,glycogen synthase kinase 3�; SOD2, superoxide dismutase 2; DHE,dihydroethidium; Keap1, Kelch-like ECH-associated protein 1; Bis-Tris, 2-(bis(2-hydroxyethyl)amino)-2-(hydroxymethyl)propane-1,3-diol; DMSO,dimethyl sulfoxide; XTT, 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tet-razolium-5-carboxanilide inner salt; TR, truncated; p, phosphorylated.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 289, NO. 39, pp. 26882–26894, September 26, 2014© 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

26882 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

matic action in heme degradation and also to the catalyticbyproducts including carbon monoxide and biliverdin (29).More recently subcellular localization of HO-1 has been stud-ied in the context of oxidative stress and cancer (30). AlthoughHO-1 is an integral protein of the smooth endoplasmic reticu-lum (31, 32), it can localize to other compartments includingcaveolae (33), mitochondria (34), and the nucleus (35, 36),where it can mediate signaling functions (30).

We have previously described a more rapidly migrating iso-form of HO-1 in the nuclear extracts of hypoxia-exposed cellstransfected with N-terminally FLAG-tagged HO-1 (30). A neg-ative reactivity for an antibody directed to the C terminus iden-tified this isoform as a 28-kDa fragment of HO-1 missing 52amino acids from the C terminus (36). This nuclear HO-1 wasalso found to be enzymatically inactive; however, it was associ-ated with enhanced activation of transcription factor AP-1 (37).Earlier Hori et al. (38) showed that a mutated, enzymatically inac-tive form of HO-1 was still able to protect against oxidative injury.In addition, a HO-1 mutant with a deletion of the C-terminalamino acids was shown to bind to heme but could not degrade it tobiliverdin (39). We documented that the enzymatically inactiveHO-1 can alter its own transcription through activation of AP-1(36, 37). Because an AP-1 consensus sequence is found within theARE, and since both ARE and API sequences are found on thehuman NAD(P)H:quinone oxidoreductase 1 (NQO1), HMOX1,and sulfiredoxin genes responding to both AP-1 and Nrf2 activa-tion (40), we wondered whether the nuclear isoform of HO-1could also regulate Nrf2 activation and what would be the conse-quences of this activation.

To evaluate this, we used hyperoxia exposure, a clinicallyrelevant oxidative stress known to activate Nrf2 signaling (41–43) and induce HO-1, resulting in its migration to the nucleus(42). In the present study, mouse embryonic fibroblasts (MEFs)were used, as was a prostate cancer cell line (LnCap) becausethis cell line and human prostate cancer tissues have enhancednuclear localization of HO-1 when compared with the normalprostate tissue (35). The present study demonstrates thatnuclear HO-1 specifically interacts with Nrf2 in the nucleus tofacilitate its sustained stabilization from GSK3�-mediated pro-teolytic degradation. This leads to preferential activation ofcytoprotective pathways for sustained tolerance against oxida-tive injury, as well as cell survival.

EXPERIMENTAL PROCEDURES

Cell Lines—MEFs, HO-1-knock-out (HMOX1�/�, KO) andwild type (HMOX1�/�, WT), were generated from embryonicday 13.5 embryos of the same littermates. These cells wereimmortalized by transfection of plasmids pSV3-neo (ATCC37150) expressing the simian virus 40(SV40) large T-antigenand were maintained in 5% CO2, at 37 °C, in DMEM with 10%(v/v) FCS and 1% P/S (100 units/ml penicillin as well as 100�g/ml streptomycin). To express full-length HO-1 (FL),murine HO-1 cDNA was tagged with 3�FLAG epitope at the Cterminus and then cloned in a retroviral vector, pMX-puro,under the control of SV40 promoter. HO-1-KO cells werereconstituted with retrovirus containing empty vectors (con-trol) or FL HO-1. This plasmid was further used as a template togenerate and express truncated HO-1 (TR). In brief, the C-ter-

minal 53 amino acid residues of the FL HO-1 were deleted, andthree copies of a nuclear localization signal derived from SV40large T-antigen were inserted to express truncated HO-1(TR) specifically in the nucleus. High titer retroviruses werepackaged with the helper plasmid containing the packagingsequence (psi) in 293T cells. Immortalized HO-1-KO andNrf2-KO MEFs were infected with the retroviruses of emptyvector (V) or vector containing FL or TR HO-1 cDNA andfurther selected with 2 �g/ml puromycin. All retrovirus-com-plemented cells were maintained within 10 passages. In someexperiments, we also used prostate cancer cell lines (LnCap),gifts from Dr. William J Fredericks, University of Pennsylvania.The Nrf2 null mutant MEF cell lines were also transfected witha cDNA expressing Nrf2 fused with an HA tag at the N terminus(44), gifts from Drs. Melpo Christofidou-Solomidou and AlanDiehl, respectively, of the University of Pennsylvania.

Stable Knockdown of HO-1 with shRNA—To knock downendogenous HO-1 expression, lentiviruses carrying shRNA tar-geting HO-1 mRNA were produced as per the manufacturer’sinstructions (RMM4534-NM-010442, Thermo Fisher Scien-tific Open Biosystems products). Based on the highest knock-down efficiency, clone 6 (71758) was chosen for the knockdownexperiments, and clone 11, which did not reduce HO-1 expres-sion, was used as a control (nonspecific) shRNA.

Hyperoxia Exposure—Cells were seeded at 0.8 � 106/10-cmdish. After overnight culture, cells were transferred to freshmedium and exposed to hyperoxia (O2: 95% O2/5% CO2) ornormoxia (air: 21% O2/5% CO2) for 18 h (42). In some experi-ments, cells were exposed to air or O2 for 2, 6, and 18 h.

Preparation of Cell Lysates—Cells were harvested and homo-genized in the presence of protease and phosphatase inhibitorseither to extract whole cell homogenate using the M-PER mam-malian protein extraction reagent (78501) or to extract cyto-plasmic and nuclear fractions using NE-PER nuclear and cyto-plasmic extraction reagents (78835). Both extraction reagentswere obtained from Thermo Fisher Scientific.

Protein Content Estimation—The protein content in the wholecell lysate or nuclear and cytosolic fractions was measured by a96-well plate based Bradford assay.

Western Blotting—Proteins (15–30 �g) either from wholecell lysate or from the fractions of cytoplasm and nucleus wereresolved by SDS-PAGE on a precast 4 –12% Bis-Tris gels (Invit-rogen), electrotransferred to PVDF membrane (Millipore,IPVH00010), and assessed for the immunosignal of proteins,HO-1 (Enzo, ADI-SPA-896), FLAG (Sigma, F-7425), Myc(Santa Cruz Biotechnology, sc789), Nrf2 (Santa Cruz Biotech-nology, sc-722), pSer40-Nrf2 (Epitomics, 2073-1), HA (SantaCruz Biotechnology, sc805), polyubiquitin (Santa Cruz Bio-technology, sc8017), AKT (Cell Signaling, 9272), pSer473-AKT(Cell Signaling, 4058), GSK3� (Cell Signaling, 9832), pSer9-GSK3� (Cell Signaling, 9322), calnexin (Enzo, ADI-SPA-860),lamin B (Santa Cruz Biotechnology, sc-6216,), SOD2 (Stress-Gen, ADI-SOD-110), and HA (Santa Cruz Biotechnology,sc-805), using the respective antibodies as indicated in theparentheses. Protein immunosignals are shown with the load-ing controls. Simultaneously, the signals were plotted as bargraphs after normalization against the respective loadingcontrols.

Nuclear HO-1 Modulates Nrf2 Function

SEPTEMBER 26, 2014 • VOLUME 289 • NUMBER 39 JOURNAL OF BIOLOGICAL CHEMISTRY 26883

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

Immunohistochemistry—Cells (1 � 104/well in 2 ml) wereseeded on coverslips placed in 6-well plates and grown over-night. The cells on coverslips after fixation with 3% paraformal-dehyde were detected for compartmental localization of pro-teins as described previously (41, 42) and visualized byfluorescent microscopy.

Immunoprecipitation (IP)—IP of FLAG fusion HO-1-FL andHO-1-TR from O2 or air-exposed stably infected MEF cells wasperformed using the anti-FLAG M2 affinity gel (Sigma, A2220)following the manufacturer’s protocol. In brief 100 –200�g/200 –300 �l of total protein either from whole cell lysateor from cytoplasmic and nuclear fractions was incubated over-night with 20 �l of packed volume of M2 beads in a cold roomunder slow rotation. After washing repeatedly we used3�FLAG peptide (Sigma, F4799) at a final concentration of 150ng/�l in 50 �l of TBS to elute the proteins. IP of endogenousNrf2 was performed by overnight incubation with anti-Nrf2(Santa Cruz Biotechnology, sc-722) and anti-pSer40-Nrf2

(Epitomics, 2073-1) antibodies at a concentration of 2–3�g/100 �g of total protein. This was followed by further incu-bation with protein A-agarose beads (Invitrogen, 15918-014) atroom temperature for an hour. After washing the beads repeat-edly, the bound proteins from the beads were eluted with 0.1 M

glycine HCl, pH 3.5, and immediately restored in neutral pH byadding 0.5 M Tris-HCl, pH 7.4, containing 1.5 M NaCl. Similarly,IP of recombinant HA-tagged Nrf2 in the nuclear fractions wasperformed by incubation with anti-HA antibody (Santa CruzBiotechnology, sc-805) followed by protein-A-agarose beads,and the elution was performed by low pH glycine as describedabove.

Expression and Purification of Recombinant GST FusionHO-1-FL and HO-1-TR—GST fusion constructs of HO-1-FLand HO-1-TR were expressed in competent Escherichia colistrain BL21 (Invitrogen) as described previously (36). Bacteriawere grown to an optical density of 0.6 – 0.8 at 600 nm. Thefusion proteins thereafter were induced in the presence of 100

FIGURE 1. Oxidative stress induces nuclear enrichment of a 28-kDa HO-1 and a simultaneous enrichment of Nrf2 in the nucleus. A, the WT MEF cells wereevaluated for HO-1 and Nrf2 signal after exposure to air or O2. B and C, the cytoplasmic and nuclear fractions of the above cells were further evaluated for HO-1and Nrf2 signals (B), and normalization of these signals to calnexin and lamin B, respectively, is represented in the bar diagrams (C). D, the compartmentallocalization of HO-1 (green) and DAPI staining (red) with exposure to air (left column), O2 (middle column), or incubation with N-acetyl-L-cysteine (NAC) (60 nM)in O2 (right column) is shown in WT MEF cells; the yellow staining demonstrates co-localization in the overlay (bottom panel). E, after exposure to air or O2, theimmunosignal of HO-1, Nrf2, and calnexin is shown in whole cell lysates of WT MEF cells silenced for HO-1 and in control cells incubated with nonspecific shRNA.F, the signal of Nrf2 (from E) is quantitated after normalization to calnexin and represented in bar diagrams. G, after exposure to O2 for 2, 6, and 18 h, thesubcellular proportion of HO-1 and Nrf2 and loading controls in the cytoplasm (lowermost three panels) and in the nucleus (upper three panels) of WT MEF cellssilenced for HO-1 or control cells with nonspecific shRNA is shown in a representative Western blot. �, p � 0.05; values are the mean � S.E. of three separatedeterminations.

Nuclear HO-1 Modulates Nrf2 Function

26884 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

�M isopropyl 1-thio-�-D-galactopyranoside at 30 °C for 4 h.The fusion proteins in the bacterial lysate were purified using aGST purification module (GE Healthcare, catalog number18-1128-13AC). The eluted proteins were evaluated for proteincontent and stored in aliquots at �80 °C until used.

In Vitro Translation of Full-length Nrf2 and C-terminallyTruncated Nrf2 Fragments (�C1 to �C4)—Mice Nrf2 ORFcloned in Pmx-Puro vector were used as a template to generatedeletion mutants of Nrf2 (�C1 to �C4, see Fig. 4B) seriallytruncated from the C terminus. The specific forward primercontaining T7 promoter and the reverse primers are shown in atable (see Fig. 4D). DNA constructs were amplified by PCR. ThePCR products of Nrf2 and the Nrf2 fragments were verified onagarose gel and purified using the QIAquick PCR purificationkit (Qiagen, catalog number 28106) following the manufac-turer’s instructions. The pure PCR-generated constructs were

transcribed in vitro by using the T7 high yield RNA synthesis kit(New England Biolabs, catalog number E2050) in the presence ofT7 promoter enhancer according to the manufacturer’s instruc-tions, and the transcripts were detected on agarose gel. For in vitrotranslation, the transcripts were added to an aliquot of the TNT�T7 coupled reticulocyte lysate system, (Promega, catalog numberL4610) and incubated in a 50-�l reaction volume containing[35S]methionine instead of methionine for 60–90 min at 30 °C.After being resolved on SDS-PAGE, the radiolabeled Nrf2 andNrf2 fragments were detected on the dried gel by autoradiogram.An in vitro translation reaction in the absence of DNA templatewas also carried out as a specificity control.

In Vitro Binding of Nrf2 to HO-1 Captured on GST SpinTrapColumns—Equal amounts of the purified GST fusion HO-1-FLand HO-1-TR were captured on several GST SpinTrap col-umns (GE Healthcare, catalog number 18-1128-13AC). The

FIGURE 2. Nuclear 28-kDa HO-1 in a physical interaction with Nrf2 stabilizes its enrichment in the nucleus. A, schematic of the HO-1 clones: HO-1-FL,HO-1-TR, and empty vector (V) with 3�Myc motif at the N terminus and 3�FLAG motif at the C terminus. B, The HO-1-KO cells stably infected with the HO-1clones (FL, TR, and V) and exposed to air or O2 for 18 h were evaluated for compartment-specific expression of HO-1-FL and HO-1-TR (upper panel) and calnexinand lamin B (lower panel). C, representative immunostain of HO-1 (green, left panel), N-terminal Myc (red, middle panel), and C-terminal FLAG (green, right panel)in FL and TR cells is shown. D, HO-1 signal in FLAG IP of total cell lysate from V, FL, and TR exposed to air or O2 for 2, 6, and 18 h (upper panel) and Nrf2co-precipitation (lower panel) are shown. E, co-staining of Nrf2 in FLAG-IP from cytoplasmic and nuclear fractions of V, FL, and TR cells exposed to O2 is shownin the upper panel. A reverse IP of endogenous Nrf2 using anti-Nrf2 antibody (sc722) from the cytoplasmic and nuclear fractions was counterstained for HO-1(middle panel), and input of Nrf2 and HO-1 is shown in 10% of the nuclear and cytoplasmic fractions (bottom two panels). F, the transient expression ofHA-tagged Nrf2 (HA-N) in Nrf2-KO MEF whole cell lysate is shown. G, the HA-specific IP of HA-Nrf2 (HA-N) from these cells after O2 exposure was first stained forNrf2 (upper panel) followed by a counterstain for HO-1 (lower panel). The input of Nrf2 and HO-1 is also shown in 5% of the lysate.

Nuclear HO-1 Modulates Nrf2 Function

SEPTEMBER 26, 2014 • VOLUME 289 • NUMBER 39 JOURNAL OF BIOLOGICAL CHEMISTRY 26885

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

columns were washed repeatedly with phosphate buffer toremove any unbound protein. Each of these columns was thenincubated for 10 min individually with 20 �l of 35S-labeled Nrf2and fragments (�C1–�C4). The columns were then washedwith copious amounts of phosphate buffer. The Nrf2 and Nrf2fragments bound to trapped GST HO-1-FL or HO-1-TR werethen eluted using 30 �l of 10 mM reduced glutathione (suppliedwith the SpinTrap). Once the eluates were resolved on SDS-PAGE, the Nrf2 signal was evaluated by autoradiogram of driedgel. The nonspecific binding was verified by incubating radio-labeled Nrf2 on a GST SpinTrap without any HO-1 bound to it.

Quantitative Real Time PCR—Using the TaqMan geneexpression assay (Applied Biosystems), mRNA levels wereanalyzed and further normalized to 18s gene product. Theprimers used from Applied Biosystems were HO-1 (Mm00516004),Nrf2 (Mm00477784), NQO1 (Mm01253560), G6PDX(Mm00656735), SOD2 (Mm01313000), and 18s (HS99999901).

Use of Inhibitors—The cells were exposed to O2 for 18 h fol-lowed by an additional 30-min incubation with an inhibitorof AKT (MK2206, ChemieTek, catalog number CT-MK2206)alone at 1.0 �M or after co-incubation with an inhibitor ofGSK3� (SB 216763, Sigma-Aldrich, catalog number S3442) at10 �M. The cell pellets were evaluated for Nrf2 and HO-1 sig-nals by Western blot.

Evaluation of Oxidative Stress—After air or O2 exposure,cells were incubated with 10 �M dihydroethidium (DHE;Molecular Probes) for 30 min. The amount of oxyethidium gen-erated was measured by fluorescent microscopic analysis. Thenumber of fluorescence-positive cells was counted in an aver-age of 10 high power fields and expressed as a ratio of the totalnumber of cells.

Evaluation of Protein Carbonyls—The cell lysates were alsoanalyzed for the protein carbonyls using OxiSelect protein car-bonyl immunoblot kit (Cell Biolabs Inc., STA 309) as per themanufacturer’s instructions. In brief, the carbonyl groups werederivatized by dinitrophenylhydrazine followed by immuno-blotting with an anti-dinitrophenyl antibody.

G6PDH Enzyme Activity—Constitutive activity of glucose-6-phosphate dehydrogenase (G6PDH) in whole cell lysate wasdetected in a 96-well plate using a kit (Sigma, MAK015). Inbrief, 10 �l of lysate from 1 � 106 cell homogenate in 100 �l ofPBS was transferred in triplicates in 40 �l/well of master reac-tion mix. This assay detects oxidation of glucose 6-phosphategenerating NADH, which is measured every 5 min colorimetri-cally at 450 nm using a standard curve of NADH. After sub-tracting the values from the background, the nmol amounts ofNADH obtained between Tinitial and Tfinal were finallyexpressed in milliunits/ml by employing the following formula

G6PDH activity �B � sample dilution factor

�Reaction time � V(Eq. 1)

where B � amount (nmol) of NADH generated between Tinitialand Tfinal, Reaction time � Tfinal � Tinitial (minutes), and V �sample volume (ml) added to the well.

Measurement of Cell Viability by XTT Assay—The XTTassay (Biotium, 30007) was performed following the manufac-turer’s instructions to measure the proliferation rate/metabolic

activity of cells over a period of 6 h. Briefly the V, FL, and TRcells (20,000/well in a 96-microtiter plate) were grown inDMEM with high glucose (25 mM) or without any energy sub-strate in the presence of XTT reagent. XTT is a tetrazoliumanalog with a negatively charged inner salt that more readilyenters cells. In metabolically active cells XTT is rapidly reducedby NAD(P)H-dependent oxidoreductases and dehydrogenasesand produces solubilized formazan that is colorimetricallymeasured at 405 nm (45, 46).

Statistical Analysis—The values in figures were expressed asmeans � S.E. Unpaired t tests were used to compare twogroups. Values of p 0.05 and p 0.005 were considered asstatistically significant.

RESULTS

Hyperoxia Induces HO-1, Leading to Nuclear Localization ofa 28-kDa Isoform and Simultaneous Accumulation of Nrf2 inthe Nucleus—The WT MEF cells exposed to air or O2 wereanalyzed for HO-1 and Nrf2 proteins in Western blot (Fig. 1). Inair, HO-1 in the whole cell lysate appeared as a single band at 32kDa, whereas in O2, an additional faster migrating band alsoappeared at 28 kDa, which was accompanied with a simultane-ous increase of Nrf2 (Fig. 1A). The 32-kDa isoform constitu-

FIGURE 3. LnCap, the prostate cancer cells, predominantly express the28-kDa HO-1 localized to the nucleus as HO-1�Nrf2 complex. A, whole celllysate from LnCap cells was evaluated for HO-1 (upper panel), Nrf2 (middle panel),and calnexin (bottom panel). B, HO-1 signal from A normalized to calnexin is rep-resented by bar diagrams. C, the cytoplasmic and nuclear fractions of LnCapshow cytoplasm-specific 32-kDa and nucleus-specific 28-kDa HO-1 isoforms(upper panel), corresponding Nrf2 signal (middle panel), and calnexin and lamin B(lower two panels). D, the signal of HO-1 and Nrf2 from C is represented in bargraphs after normalization to calnexin and lamin B. E, Nrf2-IP from these celllysates using sc772 (middle lane) and an antibody against phosphorylated serine40 (Nrf2-pSer40, Epitomics, 2073-1, right lane) were evaluated for amount of Nrf2pulled down (upper panel) and HO-1 co-precipitation (bottom panel); the input ofNrf2 and HO-1 is shown in the left lane. �, p � 0.05; n � 3.Nrf2. Values are themean � S.E. of three separate determinations.

Nuclear HO-1 Modulates Nrf2 Function

26886 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

tively was predominant in the cytoplasm, whereas in O2, the28-kDa HO-1 was predominant and primarily localized to thenucleus. The latter was accompanied with a concomitantincrease of nuclear Nrf2 (Fig. 1, B and C). The immunohisto-chemistry further verified an O2-driven nuclear enrichment ofHO-1 (Fig. 1D). N-Acetyl-L-cysteine, an antioxidant, reversedthe O2-mediated nuclear shift of HO-1, suggesting that themigration of HO-1 to the nucleus is driven by oxidative stress.

Because we observed a coordinated nuclear enrichment ofNrf2 along with the enrichment of the 28-kDa HO-1, the latterwas silenced (Fig. 1E). This in turn significantly reduced bothbasal and O2-induced Nrf2 (Fig. 1, E and F). Furthermore,nuclear HO-1 increased in a time-dependent manner withexposure to O2 being maximal at 18 h (Fig. 1G). This subse-quently was accompanied with a nuclear accumulation of Nrf2,which was reversed when HO-1 was silenced (Fig. 1G). In thecytoplasm of the HO-1-silenced cells, Nrf2 was decreased aswell (Fig. 1G). These data indicate that HO-1 modulates stabi-lization and nuclear accumulation of Nrf2.

Nuclear HO-1 Physically Interacts with Nrf2—Nrf2 accumu-lation is predominantly regulated by post-translational mecha-nisms (17, 44, 47). To understand whether the nuclear accumu-lation of Nrf2 impacted by nuclear HO-1 was dictated by aphysical interaction between the two proteins, HO-1-KO-MEFcells reconstituted with HO-1-FL or HO-1-TR and the empty

vector (V) were used (Fig. 2A). The HO-1-TR was expressed inthe nucleus due to the presence of three nuclear localizationsequences, whereas the HO-1-FL expression was restricted tothe cytoplasm (Fig. 2B). The lack of migration of FL protein tothe nucleus could have been due to a resistance to proteolyticcleavage of the protein from the C terminus and release fromthe membrane. Alternatively, this may have resulted from astructural change due to the presence of the N-terminal Mycand C-terminal FLAG tags. The cytoplasmic and nuclear com-partment-specific expression of HO-1-FL and HO-1-TR,respectively, was further documented by immunostainingusing three different antibodies directed at HO-1, N-terminalMyc, and C-terminal FLAG tags (Fig. 2C).

The Nuclear HO-1�Nrf2 Complex Appears Late in OxidativeStress—After exposure of the FL, TR, and the control (V) cells toair or O2 for 2, 6, and 18 h, the FLAG fusion proteins wereimmunoprecipitated from whole cell lysate. In comparisonwith the air-exposed cells, the IP of the O2-exposed cellsshowed increased abundance of both HO-1-FL and HO-1-TRat all time points (Fig. 2D). Counterstaining indicated co-pre-cipitation of Nrf2, which in O2 appeared most abundantly withHO-1-TR at 18 h, whereas a comparatively weak signal wasobserved with HO-1-FL. Regardless of exposure, Nrf2 signalwas absent in the IP from control cells lacking HO-1 (Fig. 2D).These data suggested that a preferential interaction exists

FIGURE 4. Nuclear HO-1 interacts with Nrf2 via transactivation domain, Neh4. A, the GST fusion HO-1-FL and HO-1-TR purified from bacterial lysate and apurification from uninduced bacterial culture (Control) are shown. B, schematic presents clones of Nrf2 and C-terminal deleted Nrf2 fragments (�1 to �4). C andD, primer sequences (C) and the amplified PCR products of the Nrf2 and the Nrf2 fragments on a 1% agarose gel (D) are shown. E, the transcripts obtained byreverse transcription of the PCR products are shown. F, the [35S]methionine-radiolabeled Nrf2 signal from in vitro translation and a translation reaction in theabsence of any transcript representing specificity control (far right lane) are shown. G, the autoradiogram signals of Nrf2 and the Nrf2 fragments bound toGST-HO-1 (TR: lanes 1–5 and FL: lanes 7–11) on GST SpinTrap columns are represented; lane 6 shows the eluate from a GST SpinTrap without any HO-1 and servesas a control for the specificity.

Nuclear HO-1 Modulates Nrf2 Function

SEPTEMBER 26, 2014 • VOLUME 289 • NUMBER 39 JOURNAL OF BIOLOGICAL CHEMISTRY 26887

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

between nuclear HO-1 and Nrf2. The absence of the Nrf2 signalat early time points (2 and 6 h) further indicated that this inter-action occurs late during oxidative stress.

The HO-1�Nrf2 Complex Localizes to the Nucleus—To fur-ther assess compartment-specific localization of the nuclearHO-1�Nrf2 complex, FLAG IP from cytoplasmic and nuclearfractions of cells exposed to O2 for 18 h was performed. Theco-precipitation of Nrf2 was strongest in the nucleus withHO-1-TR, indicating localization of HO-1-TR�Nrf2 complexpredominantly in the nucleus (Fig. 2E). The IP of HO-1-FL,abundant in the cytoplasm, weakly counterstained with Nrf2,and the control (V) cells were completely devoid of Nrf2 signal.This further supported the preferential interaction between28-kDa nuclear HO-1 with Nrf2 and predominant localizationof the complex in the nucleus. A reverse IP of the endogenousNrf2 using anti-Nrf2 antibody (sc772) further confirmed thispreferential interaction between Nrf2 and nuclear HO-1 (Fig.2E). We did observe a considerable amount of HO-1-TR in thecytoplasm, as shown by its co-precipitation with Nrf2 and in theinput lane as well. Although this may have resulted fromcontamination of cytoplasm with the nuclear portion duringfractionation, this could also suggest that Nfr2 interacts withHO-1 in the cytoplasm prior to migration to the nucleus as acomplex. An IP of HA-tagged Nrf2 (HA-N) ectopicallyexpressed in Nrf2-KO MEF cells (Fig. 2F) further confirmedthe interaction of endogenous 28-kDa HO-1 with Nrf2 (Fig.2G). As expected, this interaction was absent in Nrf2-KO-MEF cells.

The LnCap Prostate Cancer Cells Constitutively Are Enrichedwith the 28-kDa HO-1 That Resides in the Nucleus and Forms aComplex with Nrf2—To further evaluate whether the HO-1/Nrf2 interaction is physiological, we used LnCap prostate ade-nocarcinoma cell lines. The whole cell lysate exhibited predom-inantly a 28-kDa HO-1 isoform (Fig. 3, A and B), which uponfractionation of cytoplasm and nucleus exclusively localized tothe nucleus (Fig. 3, C and D). The Nrf2 signal as well was signif-icantly increased in the nucleus when compared with cyto-plasm (Fig. 3, C and D). The Nrf2 IP from LnCap cells showedco-precipitation of HO-1 particularly of the 28 kDa (Fig. 3E).Because phosphorylation of Nrf2 at Ser40 is known to modulateNrf2 activity (17), an antibody specific to pSer40 (Epitomics,2073-1) was used to immunoprecipitate Nrf2. Although the IPshowed abundant Nrf2, HO-1 co-precipitation was absent (Fig.3E). These data indicated that Nrf2, if phosphorylated at Ser40,does not interact with nuclear HO-1 and hence does not par-ticipate in Ser40-mediated Nrf2 signaling.

Nuclear HO-1 Interacts with Nrf2 via the Neh4 Transactiva-tion Domain—The purified GST fusion HO-1-FL and HO-1-TRproteins expressed in bacteria are presented in Fig. 4A. Nrf2and Nrf2 fragments (�C1 to �C4) generated with an in vitrotranslation system are shown stepwise in Fig. 4, B–F. The bindingof Nrf2 and Nrf2 fragments with HO-1 was verified with a bind-ing assay using purified GST fusion HO-1-FL and HO-1-TR pro-teins trapped on GST columns (Fig. 4G). The binding of the[35S]methionine-radiolabeled Nrf2 fragments to GST-HO-1revealed that Nrf2 could bind to HO-1-TR and that this binding

FIGURE 5. A, diagram of Nrf2 depicting domains and possible phosphorylation (P) and ubiquitination (Ub) sites (adapted from Refs. 20 and 21). ab sc772,antibody sc772. B, the FL and TR cells were exposed to O2 followed by an additional 30 min of incubation with MK2206 (5 �M). The immunosignal of p-AKT(pSer473, Cell Signaling, 9272), p-GSK3� (pSer9, Cell Signaling, 9832), and corresponding total Nrf2 levels (sc722) are shown in whole cell lysates. C, The wholecell lysates of FL and TR cells exposed to O2 followed by an additional 30 min incubation with DMSO or MK2206 (5 �M) or SB216763 (10 �M) alone or inco-incubation were assessed for levels of Nrf2 and calnexin. D, the Nrf2 signal quantitated after normalization to calnexin expressed in bar diagrams. MK,MK2206; SB, SB216763. E, cell lysates of V, FL, and TR exposed to air or O2 were assessed for HO-1 pulled down by FLAG-IP (upper panel), Nrf2 co-precipitation(middle panel), and polyubiquitination signal (lower panel). On the left, 5% input shows HO-1, polyubiquitinated proteins (Poly-Ub), Nrf2, and calnexin. �, p �0.05; n � 3. Values are the mean � S.E. of three separate measurements.

Nuclear HO-1 Modulates Nrf2 Function

26888 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

did not occur with HO-1-FL. Furthermore, the presence of bind-ing signal for �C1 to �C3 and the absence of this signal with �C4indicated that the transactivation domain Neh4 is crucial for theinteraction between Nuclear HO-1 and Nrf2.

Nuclear HO-1 Stabilizes Nrf2 from GSK3�-mediated Degra-dation—Active GSK3� phosphorylates serine moieties (343and 347) in the Neh6 domain of Nrf2 (Fig. 5A) and targets Nrf2for ubiquitination and degradation (20). To understandwhether HO-1-TR protects Nrf2 from GSK3�-mediated pro-teolytic degradation, AKT, the negative regulator of GSK3�,was inhibited by MK2206. This in turn results in the activa-tion of GSK3� by preventing phosphorylation at Ser-9 (48,49). The phosphorylation signals of AKT and GSK3� wereverified with specific antibodies (pSer473-AKT, Cell Signal-ing, 9272; and pSer9-GSK3�, Cell Signaling, 9832) in FL and

TR cells after exposure to O2 followed by an additional30-min incubation with MK2206 (Fig. 5B). In the presence ofMK2206, when GSK3� activity is increased, Nrf2 signal wasreduced in FL, whereas it remained unaltered in TR. Thisobservation was further verified in the next experimentwhere FL and TR cells exposed to O2 were treated similarlywith MK2206 alone or in co-incubation with SB16763, aninhibitor of GSK3�. When compared with the Nrf2 level inFL cells incubated only with DMSO (Fig. 5, C and D), the FLcells incubated with MK2206 showed significant loss of Nrf2,indicating that activation of GSK3� results in degradation ofNrf2. Restoration of this loss by co-incubation with SB16763verifies that this effect is dependent on GSK3�. On the con-trary, in TR cells, Nrf2 level was relatively higher andretained regardless of inhibitor treatment (Fig. 5, C and D).

FIGURE 6. Cells enriched with nuclear HO1 exhibit a significant increase in transcription and activation of Nrf2 target genes. A, HO-1-silenced andcontrol MEF cells with scrambled shRNA were exposed to air or O2 and analyzed for steady state mRNA levels of HO-1, Nrf2, NQO1, and G6PDH by quantitativereal time PCR. B, similarly, Nrf2, NQO1, and G6PDH mRNA levels were evaluated in V and TR cells. C and D, constitutive G6PDH enzyme activity in control(scrambled shRNA) and HO-1-silenced-MEF cells (C) as well as in V, FL, and TR cells (D) is shown. E and F, cell viability or rate of metabolic activity of V, FL, andTR as evaluated using XTT reagent for indicated times in medium devoid of energy substrates (E) or in glucose-enriched medium (25 mM) is shown (F). �, p �0.05; ��, p � 0.005. Values are the mean � S.E. of three separate measurements.

Nuclear HO-1 Modulates Nrf2 Function

SEPTEMBER 26, 2014 • VOLUME 289 • NUMBER 39 JOURNAL OF BIOLOGICAL CHEMISTRY 26889

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

This validates the role of HO-1-TR in rescuing Nrf2 fromGSK3�-mediated degradation.

Because ubiquitination is a general mechanism to target pro-teins for proteasomal degradation, this was evaluated in FL, TR,and control cells (Fig. 5E). Interestingly, maximum Nrf2 co-precipitation signal with FLAG-HO-1-TR IP was associatedwith minimal polyubiquitination signal, and the FLAG IP fromFL and control cells showed less Nrf2 co-precipitation yet max-imum polyubiquitination signal. The input samples of total celllysates from TR also exhibited less polyubiquitin signal whencompared with FL and control cells (Fig. 5E). These data couldreflect sustenance of protein homeostasis and limited ubiquiti-nation in TR. Collectively, these data support the hypothesisthat HO-1-TR prevents Nrf2 ubiquitination by physically inter-acting with the protein and thus preventing it from GSK3�-mediated degradation, thereby facilitating Nrf2 accumulationin the nucleus.

Nuclear HO-1-mediated Stabilization of Nrf2 RegulatesTranscription of Specific Downstream Antioxidants and Meta-bolic Genes—To understand the physiologic importance ofHO-1-mediated nuclear accumulation of Nrf2, we evaluatedsteady state mRNA levels of HO-1, Nrf2, and a group of Nrf2downstream target genes including NQO1 and G6PDH in WT-MEF and HO-1-silenced MEF cells (Fig. 6A). As expected, afterexposure to air or O2, HO-1 mRNA was suppressed in theHO-1-silenced cells. In O2-exposed WT cells, levels of NQO1mRNA and G6PDH mRNA increased concomitantly with theincrease in HO-1. Disruption of this signal in HO-1-silencedcells indicated that regulation of the Nrf2 downstream genes is

dependent on HO-1. Of note, Nrf2 mRNA induction was alsoincreased in O2 exposure, indicating that nuclear HO-1 mayinfluence Nrf2 transcription as well (Fig. 6A). In addition, inHO-1-TR cells, there was significant induction of NQO1 andG6PDH mRNA levels both at baseline and after O2 exposurewhen compared with control cells (Fig. 6B).

Cells Enriched with Nuclear HO-1 Exhibit Increased G6PDHActivity—Because HO-1-TR was associated with enhancedtranscription of G6PDH in TR cells and disruption of HO-1 inWT cells diminished it, constitutive G6PDH activity wasassessed in these cells. In fact, the basal G6PDH activityobserved was indeed decreased significantly with the disrup-tion of HO-1 by shRNA (Fig. 6C). Additionally, G6PDH activitywas significantly higher in TR when compared with FL andcontrol cells (Fig. 6D). These data further indicate that modu-lation of G6PDH activity by Nrf2 is particularly influenced bynuclear HO-1.

Cells Enriched with Nuclear HO-1 Are Metabolically MoreViable in Glucose-enriched Medium—Increased activity ofG6PDH should improve growth and proliferation of cells inglucose medium. This was tested in V, FL, and TR cells. Cellsgrown in medium without any energy substrate as expected didnot grow (Fig. 6E). In glucose-enriched (25 mM) medium, TRcells showed proliferation significantly at a higher rate whencompared with FL and control cells (Fig. 6F).

Nrf2 Requires Participation of Nuclear HO-1 to Induce Anti-oxidant Defenses—Because NQO1 by utilizing NAD(P)H andNADH plays a major role in scavenging toxic oxidized sub-strates, we measured the oxidation burden in both air-exposed

FIGURE 7. Nuclear HO-1 coordinates with Nrf2 to mediate enhanced antioxidant defense. A, the amount of fluorescence generated in V, FL, and TR cells afterincubation with DHE was assessed by fluorescent microscopy. B, the percentage of DHE-positive cells/high power field based on an average of 10 fields is alsorepresented by bar diagram. C, Nrf2 immunosignal in the whole lysates of Nrf2-WT and Nrf2-KO cells infected with V, FL, and TR (top panel), HO-1-FL and HO-1-TR(middle panel), and calnexin (bottom panel) is shown. D, the quantity of expression level in a ratio to calnexin is shown. E, after exposure to air or O2, protein carbonylsin the whole lysates of these cells were assessed by derivation with dinitrophenylhydrazine and immunoblotting with anti-dinitrophenyl (Anti-DNP) antibody.

Nuclear HO-1 Modulates Nrf2 Function

26890 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 10: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

and O2-exposed V, FL, and TR cells by measuring the intensityof 2-hydroxyethidium fluorescence in cells after incubationwith DHE and also assessed protein carbonyls as indices of pro-tein oxidation. All cell groups showed low DHE fluorescence inair, whereas in O2, only TR showed low levels of DHE fluores-cence when compared with V and FL cells (Fig. 7, A and B).

To understand whether the enhanced anti-oxidant protec-tion in the TR cells was by virtue of nuclear HO-1 interactionwith Nrf2, we used Nrf2-WT and KO cells as such and alsoinfected with cDNA constructs of V, FL, and TR. Fig. 7, C and D,indicate the levels of HO-1-TR and HO-1-FL expression inboth Nrf2 WT and KO cells. We then evaluated levels of car-bonylated proteins in these cells after exposure to air or O2 (Fig.7E). The protein carbonyls did not differ much between thetypes of cells in air. However, in O2, Nrf2-WT cells withHO-1-TR showed reduced levels of carbonylated proteinswhen compared with those having HO-1-FL and empty vector,whereas the Nrf2-KO cells exhibited enhanced protein carbon-yls, which were not reversed even by HO-1-TR or HO-1-FL.

Furthermore, Nrf2-WT cells containing HO-1-TR showedsignificantly increased transcription of NQO1 (Fig. 8A) andG6PDH (Fig. 8C) when compared with those with HO-1-FLand empty vector. The fact that NQO1 and G6PDH transcrip-tion was lower in Nrf2-KO background even in the presence of

HO-1-TR (Fig. 8, B and D) further supports our hypothesis thatnuclear HO-1 partners with Nrf2 or vice versa to enhance anti-oxidant defenses. Nevertheless, steady state levels of SOD2were not modulated by the interaction of nuclear HO-1 andNrf2 (Fig. 8, E and F).

DISCUSSION

We demonstrate in various ways that oxidative stress medi-ates induction of HO-1 along with nuclear localization of a28-kDa isoform of HO-1, which physically interacts with Nrf2.This interaction results in the stabilization of Nrf2 in thenucleus. We further established that under physiologic condi-tions, Nrf2 interacts with the 28-kDa endogenous nuclearHO-1. Many have described how Nrf2 results in the inductionof HO-1 via binding at the ARE (25, 50), but this is the firstreport of the converse, that is, the modulation of Nrf2 functionby HO-1.

Participation of various kinase signaling pathways has beendocumented in ARE-mediated transcription (52). In particular,phosphorylation of Ser40 by protein kinase C has been impliedfor the modulation of Nrf2 in a Keap1-dependent manner (17,18). In the present study, neither isoform of HO-1 interactedwith Nrf2-pSer40. Nevertheless, activation of PI3K/Akt is welldocumented to provide protection from oxidative stress-medi-

FIGURE 8. Nrf2 requires participation of nuclear HO-1 to induce antioxidant defenses. A, C, and E, the Nrf2-WT cells infected with V, FL, and TR exposed toair or O2 were evaluated for steady state mRNA levels of NQO1 (A), G6PDX (C), and SOD2 (E). B, D, and F, similarly exposed Nrf2 KO cells infected with V, FL, andTR were evaluated for NQO1 (B), G6PDX (D), and SOD2 (F). �, p � 0.05; Values are the mean � S.E. of three separate determinations.

Nuclear HO-1 Modulates Nrf2 Function

SEPTEMBER 26, 2014 • VOLUME 289 • NUMBER 39 JOURNAL OF BIOLOGICAL CHEMISTRY 26891

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 11: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

ated injury via Nrf2 activation (1). The PI3K-Akt-GSK3� axismay regulate Nrf2 activation in several ways. Active GSK3� candirectly phosphorylate the phosphodegron motif in the Neh6domain of Nrf2 protein, leading to Keap1-independent and�-TrCP-dependent proteasomal degradation of Nrf2 protein(20–22). Alternatively, GSK3� can phosphorylate and activateFyn kinase, leading to phosphorylation and nuclear exclusion ofNrf2 protein (53). Therefore, phosphorylation of Nrf2 might dic-tate its accumulation and nucleocytoplasmic trafficking (52).

A cellular model where the truncated form of HO-1 is con-stitutively overexpressed in the nucleus of cells devoid ofendogenous HO-1 allowed for documentation of a distinctfunction of nuclear HO-1, specifically that it preferentiallybinds to Nrf2 and influences its nuclear abundance. Accumu-lation of nuclear HO-1 and consequent interaction with Nrf2may take up to 18 h, indicating a specific role for nuclear HO-1in the regulation of Nrf2 post-induction. Nuclear enrichment ofNrf2 was observed in TR cells even with GSK3� activation,whereas in FL cells lacking nuclear HO-1, this resulted in loss ofNrf2. These data document specifically that by interacting withNrf2, nuclear HO-1 stabilizes it from GSK3�-mediated proteo-lytic degradation. The observed decreased polyubiquitin signalin the lysates of O2-exposed TR cells could reflect sustainedprotein homeostasis by limited ubiquitination, although thiswould need to be systematically evaluated.

HO-1 has long been identified as proto-oncogenic due to itsanti-apoptotic and pro-angiogenic properties (54, 55), andthere is a positive relationship between high nuclear HO-1 con-tent and severity of prostate cancer (35), Nonetheless, the pre-cise role of HO-1 in cancer biology is far from being completelyunderstood. Cancer cells display altered metabolic circuitryand nutrient uptake to increase proliferation preferentially,leading to glucose-dependent ATP production (Warburg the-ory). Increased expression and activity of G6PDH are fre-quently observed in various cancers (56 –58). It is the first keyrate-limiting enzyme in the pentose phosphate pathway, whichprovides a readily available supply of pentose sugars for RNAand DNA synthesis (59, 60). Also, by regulating the pentosephosphate pathway, G6PDH generates NAD(P)H, which pro-vides reducing equivalents for the maintenance of a pool ofreduced glutathione to balance redox state (61). Additionally,by utilizing NAD(P)H, NQO1 scavenges a wide variety of oxi-dized and toxic substrates such as quinones (62, 63). Thisshould provide protection against oxidative stress. In accor-dance, the cells enriched with nuclear HO-1 show increasedNQO1 transcripts and decreased oxidative damage of proteinsand an increased G6PDH activity as well, and this indicates thatthese effects are attained by the interaction of nuclear HO-1with Nrf2.

Interestingly, nuclear HO-1-mediated activation of Nrf2 ledto the transcription of specific downstream genes includingNrf2 itself. This is explained by the fact that the pattern ofresultant downstream genes depends on the alignment of theARE and AP1 primary core sequences that Nrf2 binds to (51,64). How the binding of Nrf2 to HO-1 mediates preferentialtranscription of NQO1 and G6PDH is still not clear. In addi-tion, the fact that nuclear HO-1 may also regulate the transcrip-tion of Nrf2 needs further verification.

In summary, we demonstrate that with oxidative stress, thereis increased expression of nuclear HO-1. This regulates subcel-lular distribution and activation of Nrf2 post-induction, result-ing in transcriptional regulation of late phase II antioxidantenzymes and enabling a feed forward adaptive reprogrammingfor recovery and a survival advantage in oxidative stress.

Acknowledgments—We thank Drs. Melpo Christofidou-Solomidouand Dr. William J Fredericks, University of Pennsylvania for supply-ing us with the Nrf2-KO cells, and LnCap cell lines, respectively. Wealso thank Dr. Alan Diehl, University of Pennsylvania, for valuablesuggestions and providing us with the HA-tagged Nrf2 cDNA. Weacknowledge Prof. Yair Argon, University of Pennsylvania for valua-ble comments.

REFERENCES1. Chen, H. H., Chen, Y. T., Huang, Y. W., Tsai, H. J., and Kuo, C. C. (2012)

4-Ketopinoresinol, a novel naturally occurring ARE activator, induces theNrf2/HO-1 axis and protects against oxidative stress-induced cell injuryvia activation of PI3K/AKT signaling. Free Radic. Biol. Med. 52,1054 –1066

2. Moi, P., Chan, K., Asunis, I., Cao, A., and Kan, Y. W. (1994) Isolation ofNF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcrip-tional activator that binds to the tandem NF-E2/AP1 repeat of the �-glo-bin locus control region. Proc. Natl. Acad. Sci. U.S.A. 91, 9926 –9930

3. Shelton, P., and Jaiswal, A. K. (2013) The transcription factor NF-E2-related factor 2 (Nrf2): a protooncogene? FASEB J. 27, 414 – 423

4. Jaramillo, M. C., and Zhang, D. D. (2013) The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 27, 2179 –2191

5. Sporn, M. B., and Liby, K. T. (2012) NRF2 and cancer: the good, the badand the importance of context. Nat. Rev. Cancer 12, 564 –571

6. Nguyen, T., Nioi, P., and Pickett, C. B. (2009) The Nrf2-antioxidant re-sponse element signaling pathway and its activation by oxidative stress.J. Biol. Chem. 284, 13291–13295

7. Malhotra, D., Portales-Casamar, E., Singh, A., Srivastava, S., Arenillas, D.,Happel, C., Shyr, C., Wakabayashi, N., Kensler, T. W., Wasserman, W. W.,and Biswal, S. (2010) Global mapping of binding sites for Nrf2 identifiesnovel targets in cell survival response through ChIP-Seq profiling andnetwork analysis. Nucleic Acids Res. 38, 5718 –5734

8. Dhakshinamoorthy, S., Jain, A. K., Bloom, D. A., and Jaiswal, A. K. (2005)Bach1 competes with Nrf2 leading to negative regulation of the antioxi-dant response element (ARE)-mediated NAD(P)H:quinone oxidoreduc-tase 1 gene expression and induction in response to antioxidants. J. Biol.Chem. 280, 16891–16900

9. Villeneuve, N. F., Lau, A., and Zhang, D. D. (2010) Regulation of the Nrf2-Keap1 antioxidant response by the ubiquitin proteasome system: an in-sight into Cullin-ring ubiquitin ligases. Antioxid. Redox Signal 13,1699 –1712

10. Kobayashi, A., Kang, M. I., Watai, Y., Tong, K. I., Shibata, T., Uchida, K.,and Yamamoto, M. (2006) Oxidative and electrophilic stresses activateNrf2 through inhibition of ubiquitination activity of Keap1. Mol. Cell. Biol.26, 221–229

11. Velichkova, M., and Hasson, T. (2005) Keap1 regulates the oxidation-sensitive shuttling of Nrf2 into and out of the nucleus via a Crm1-depen-dent nuclear export mechanism. Mol. Cell. Biol. 25, 4501– 4513

12. Cullinan, S. B., Gordan, J. D., Jin, J., Harper, J. W., and Diehl, J. A. (2004)The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3ligase: oxidative stress sensing by a Cul3-Keap1 ligase. Mol. Cell. Biol. 24,8477– 8486

13. Ichimura, Y., Waguri, S., Sou, Y. S., Kageyama, S., Hasegawa, J., Ishimura,R., Saito, T., Yang, Y., Kouno, T., Fukutomi, T., Hoshii, T., Hirao, A.,Takagi, K., Mizushima, T., Motohashi, H., Lee, M. S., Yoshimori, T.,Tanaka, K., Yamamoto, M., and Komatsu, M. (2013) Phosphorylation ofp62 activates the Keap1-Nrf2 pathway during selective autophagy. Mol.Cell 51, 618 – 631

Nuclear HO-1 Modulates Nrf2 Function

26892 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 12: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

14. Lau, A., Tian, W., Whitman, S. A., and Zhang, D. D. (2013) The predictedmolecular weight of Nrf2: it is what it is not. Antioxid Redox. Signal 18,91–93

15. He, X., and Ma, Q. (2009) NRF2 cysteine residues are critical for oxidant/electrophile-sensing, Kelch-like ECH-associated protein-1-dependentubiquitination-proteasomal degradation, and transcription activation.Mol. Pharmacol. 76, 1265–1278

16. Kawai, Y., Garduno, L., Theodore, M., Yang, J., and Arinze, I. J. (2011)Acetylation-deacetylation of the transcription factor Nrf2 (nuclear factorerythroid 2-related factor 2) regulates its transcriptional activity and nu-cleocytoplasmic localization. J. Biol. Chem. 286, 7629 –7640

17. Huang, H. C., Nguyen, T., and Pickett, C. B. (2002) Phosphorylation ofNrf2 at Ser-40 by protein kinase C regulates antioxidant response ele-ment-mediated transcription. J. Biol. Chem. 277, 42769 – 42774

18. Sun, Z., Huang, Z., and Zhang, D. D. (2009) Phosphorylation of Nrf2 atmultiple sites by MAP kinases has a limited contribution in modulatingthe Nrf2-dependent antioxidant response. PLoS One 4, e6588

19. Watai, Y., Kobayashi, A., Nagase, H., Mizukami, M., McEvoy, J., Singer,J. D., Itoh, K., and Yamamoto, M. (2007) Subcellular localization and cy-toplasmic complex status of endogenous Keap1. Genes Cells 12,1163–1178

20. Chowdhry, S., Zhang, Y., McMahon, M., Sutherland, C., Cuadrado, A.,and Hayes, J. D. (2013) Nrf2 is controlled by two distinct �-TrCP recog-nition motifs in its Neh6 domain, one of which can be modulated byGSK-3 activity. Oncogene 32, 3765–3781

21. Rada, P., Rojo, A. I., Evrard-Todeschi, N., Innamorato, N. G., Cotte, A.,Jaworski, T., Tobon-Velasco, J. C., Devijver, H., García-Mayoral, M. F.,Van Leuven, F., Hayes, J. D., Bertho, G., and Cuadrado, A. (2012) Struc-tural and functional characterization of Nrf2 degradation by the glycogensynthase kinase 3/�-TrCP axis. Mol. Cell. Biol. 32, 3486 –3499

22. Rada, P., Rojo, A. I., Chowdhry, S., McMahon, M., Hayes, J. D., andCuadrado, A. (2011) SCF/�-TrCP promotes glycogen synthase kinase3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol. Cell. Biol. 31, 1121–1133

23. Kilic, U., Kilic, E., Tuzcu, Z., Tuzcu, M., Ozercan, I. H., Yilmaz, O., Sahin,F., and Sahin, K. (2012) Melatonin suppresses cisplatin-induced nephro-toxicity via activation of Nrf-2/HO-1 pathway. Nutr. Metab. (Lond.) 10, 7

24. Wagener, F. A., Eggert, A., Boerman, O. C., Oyen, W. J., Verhofstad, A.,Abraham, N. G., Adema, G., van Kooyk, Y., de Witte, T., and Figdor, C. G.(2001) Heme is a potent inducer of inflammation in mice and is counter-acted by heme oxygenase. Blood 98, 1802–1811

25. Alam, J., Shibahara, S., and Smith, A. (1989) Transcriptional activation ofthe heme oxygenase gene by heme and cadmium in mouse hepatoma cells.J. Biol. Chem. 264, 6371– 6375

26. Applegate, L. A., Luscher, P., and Tyrrell, R. M. (1991) Induction of hemeoxygenase: a general response to oxidant stress in cultured mammaliancells. Cancer Res. 51, 974 –978

27. Keyse, S. M., and Tyrrell, R. M. (1989) Heme oxygenase is the major32-kDa stress protein induced in human skin fibroblasts by UVA radia-tion, hydrogen peroxide, and sodium arsenite. Proc. Natl. Acad. Sci. U.S.A.86, 99 –103

28. Dennery, P. A. (2000) Regulation and role of heme oxygenase in oxidativeinjury. Curr Top. Cell Regul. 36, 181–199

29. Agarwal, A., and Bolisetty, S. (2013) Adaptive responses to tissue injury:role of heme oxygenase-1. Trans Am. Clin. Climatol. Assoc. 124, 111–122

30. Dennery, P. A. (2014) Signaling function of heme oxygenase proteins.Antioxid. Redox Signal. 20, 1743–1753

31. Yoshida, T., and Sato, M. (1989) Posttranslational and direct integration ofheme oxygenase into microsomes. Biochem. Biophys. Res. Commun. 163,1086 –1092

32. Yoshida, T., Ishikawa, K., and Sato, M. (1991) Degradation of heme by asoluble peptide of heme oxygenase obtained from rat liver microsomes bymild trypsinization. Eur. J. Biochem. 199, 729 –733

33. Jung, N. H., Kim, H. P., Kim, B. R., Cha, S. H., Kim, G. A., Ha, H., Na, Y. E.,and Cha, Y. N. (2003) Evidence for heme oxygenase-1 association withcaveolin-1 and -2 in mouse mesangial cells. IUBMB Life 55, 525–532

34. Slebos, D. J., Ryter, S. W., van der Toorn, M., Liu, F., Guo, F., Baty, C. J.,Karlsson, J. M., Watkins, S. C., Kim, H. P., Wang, X., Lee, J. S., Postma,

D. S., Kauffman, H. F., and Choi, A. M. (2007) Mitochondrial localizationand function of heme oxygenase-1 in cigarette smoke-induced cell death.Am. J. Respir. Cell Mol. Biol. 36, 409 – 417

35. Sacca, P., Meiss, R., Casas, G., Mazza, O., Calvo, J. C., Navone, N., andVazquez, E. (2007) Nuclear translocation of haeme oxygenase-1 is associ-ated to prostate cancer. Br. J. Cancer 97, 1683–1689

36. Lin, Q., Weis, S., Yang, G., Weng, Y. H., Helston, R., Rish, K., Smith, A.,Bordner, J., Polte, T., Gaunitz, F., and Dennery, P. A. (2007) Heme oxyge-nase-1 protein localizes to the nucleus and activates transcription factorsimportant in oxidative stress. J. Biol. Chem. 282, 20621–20633

37. Lin, Q. S., Weis, S., Yang, G., Zhuang, T., Abate, A., and Dennery, P. A.(2008) Catalytic inactive heme oxygenase-1 protein regulates its own ex-pression in oxidative stress. Free Radic. Biol. Med. 44, 847– 855

38. Hori, R., Kashiba, M., Toma, T., Yachie, A., Goda, N., Makino, N., Soejima,A., Nagasawa, T., Nakabayashi, K., and Suematsu, M. (2002) Gene trans-fection of H25A mutant heme oxygenase-1 protects cells against hy-droperoxide-induced cytotoxicity. J. Biol. Chem. 277, 10712–10718

39. Wilks, A., and Ortiz de Montellano, P. R. (1993) Rat liver heme oxygenase.High level expression of a truncated soluble form and nature of the meso-hydroxylating species. J. Biol. Chem. 268, 22357–22362

40. Soriano, F. X., Baxter, P., Murray, L. M., Sporn, M. B., Gillingwater, T. H.,and Hardingham, G. E. (2009) Transcriptional regulation of the AP-1 andNrf2 target gene sulfiredoxin. Mol. Cells 27, 279 –282

41. Wright, C. J., and Dennery, P. A. (2009) Manipulation of gene expressionby oxygen: a primer from bedside to bench. Pediatr. Res. 66, 3–10

42. Yang, G., Biswasa, C., Lin, Q. S., La, P., Namba, F., Zhuang, T., Muthu, M.,and Dennery, P. A. (2013) Heme oxygenase-1 regulates postnatal lungrepair after hyperoxia: Role of �-catenin/hnRNPK signaling. Redox. Biol.1, 234 –243

43. Cho, H. Y., Jedlicka, A. E., Reddy, S. P., Kensler, T. W., Yamamoto, M.,Zhang, L. Y., and Kleeberger, S. R. (2002) Role of NRF2 in protectionagainst hyperoxic lung injury in mice. Am. J. Respir. Cell Mol. Biol. 26,175–182

44. Cullinan, S. B., and Diehl, J. A. (2004) PERK-dependent activation of Nrf2contributes to redox homeostasis and cell survival following endoplasmicreticulum stress. J. Biol. Chem. 279, 20108 –20117

45. Berridge, M. V., Herst, P. M., and Tan, A. S. (2005) Tetrazolium dyes astools in cell biology: new insights into their cellular reduction. Biotechnol.Annu. Rev. 11, 127–152

46. Marshall, N. J., Goodwin, C. J., and Holt, S. J. (1995) A critical assessmentof the use of microculture tetrazolium assays to measure cell growth andfunction. Growth Regul. 5, 69 – 84

47. Kimura, M., Yamamoto, T., Zhang, J., Itoh, K., Kyo, M., Kamiya, T., Abu-ratani, H., Katsuoka, F., Kurokawa, H., Tanaka, T., Motohashi, H., andYamamoto, M. (2007) Molecular basis distinguishing the DNA bindingprofile of Nrf2-Maf heterodimer from that of Maf homodimer. J. Biol.Chem. 282, 33681–33690

48. De Sarno, P., Li, X., and Jope, R. S. (2002) Regulation of Akt and glycogensynthase kinase-3 � phosphorylation by sodium valproate and lithium.Neuropharmacology 43, 1158 –1164

49. Sangai, T., Akcakanat, A., Chen, H., Tarco, E., Wu, Y., Do, K. A., Miller,T. W., Arteaga, C. L., Mills, G. B., Gonzalez-Angulo, A. M., and Meric-Bernstam, F. (2012) Biomarkers of response to Akt inhibitor MK-2206 inbreast cancer. Clin. Cancer Res. 18, 5816 –5828

50. Heiss, E. H., Schachner, D., Zimmermann, K., and Dirsch, V. M. (2013)Glucose availability is a decisive factor for Nrf2-mediated gene expression.Redox. Biol. 1, 359 –365

51. Wasserman, W. W., and Fahl, W. E. (1997) Functional antioxidant re-sponsive elements. Proc. Natl. Acad. Sci. U.S.A. 94, 5361–5366

52. Das, B. N., Kim, Y. W., and Keum, Y. S. (2013) Mechanisms of Nrf2/Keap1-dependent phase II cytoprotective and detoxifying gene expressionand potential cellular targets of chemopreventive isothiocyanates. Oxid.Med. Cell Longev. 2013, 839409

53. Jain, A. K., and Jaiswal, A. K. (2007) GSK-3� acts upstream of Fyn kinase inregulation of nuclear export and degradation of NF-E2 related factor 2.J. Biol. Chem. 282, 16502–16510

54. Tanaka, S., Akaike, T., Fang, J., Beppu, T., Ogawa, M., Tamura, F., Miy-amoto, Y., and Maeda, H. (2003) Antiapoptotic effect of haem oxyge-

Nuclear HO-1 Modulates Nrf2 Function

SEPTEMBER 26, 2014 • VOLUME 289 • NUMBER 39 JOURNAL OF BIOLOGICAL CHEMISTRY 26893

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 13: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

nase-1 induced by nitric oxide in experimental solid tumour. Br. J. Cancer88, 902–909

55. Sunamura, M., Duda, D. G., Ghattas, M. H., Lozonschi, L., Motoi, F.,Yamauchi, J., Matsuno, S., Shibahara, S., and Abraham, N. G. (2003) Hemeoxygenase-1 accelerates tumor angiogenesis of human pancreatic cancer.Angiogenesis 6, 15–24

56. Koudstaal, J., Makkink, B., and Overdiep, S. H. (1975) Enzyme histochem-ical pattern in human tumours. II. Oxidoreductases in carcinoma of thecolon and the breast. Eur. J. Cancer 11, 111–115

57. Baba, M., Yamamoto, R., Iishi, H., Tatsuta, M., and Wada, A. (1989) Role ofglucose-6-phosphate dehydrogenase on enhanced proliferation of pre-neoplastic and neoplastic cells in rat liver induced by N-nitrosomorpho-line. Int. J. Cancer 43, 892– 895

58. Pretlow, T. G., 2nd, Harris, B. E., Bradley, E. L., Jr., Bueschen, A. J., Lloyd,K. L., and Pretlow, T. P. (1985) Enzyme activities in prostatic carcinomarelated to Gleason grades. Cancer Res. 45, 442– 446

59. Pretlow, T. G., 2nd, Whitehurst, G. B., Pretlow, T. P., Hunt, R. S.,Jacobs, J. M., McKenzie, D. R., McDaniel, H. G., Hall, L. M., and Brad-ley, E. L., Jr. (1982) Decrease in creatine kinase in human prostaticcarcinoma compared to benign prostatic hyperplasia. Cancer Res. 42,

4842– 484860. Boros, L. G., Puigjaner, J., Cascante, M., Lee, W. N., Brandes, J. L., Bassilian,

S., Yusuf, F. I., Williams, R. D., Muscarella, P., Melvin, W. S., and Schirmer,W. J. (1997) Oxythiamine and dehydroepiandrosterone inhibit the non-oxidative synthesis of ribose and tumor cell proliferation. Cancer Res. 57,4242– 4248

61. Kuo, W. Y., and Tang, T. K. (1998) Effects of G6PD overexpression inNIH3T3 cells treated with tert-butyl hydroperoxide or paraquat. FreeRadic. Biol. Med. 24, 1130 –1138

62. Ross, D., Kepa, J. K., Winski, S. L., Beall, H. D., Anwar, A., and Siegel, D.(2000) NAD(P)H:quinone oxidoreductase 1 (NQO1): chemoprotection,bioactivation, gene regulation and genetic polymorphisms. Chem. Biol.Interact. 129, 77–97

63. Cadenas, E., and Ernster, L. (1990) Quinoid compounds: high-perfor-mance liquid chromatography with electrochemical detection. MethodsEnzymol. 186, 180 –196

64. Wasserman, W. W., and Fahl, W. E. (1997) Comprehensive analysis ofproteins which interact with the antioxidant responsive element: correla-tion of ARE-BP-1 with the chemoprotective induction response. Arch.Biochem. Biophys. 344, 387–396

Nuclear HO-1 Modulates Nrf2 Function

26894 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 289 • NUMBER 39 • SEPTEMBER 26, 2014

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 14: NuclearHemeOxygenase-1(HO-1)ModulatesSubcellular … · 2014-09-19 · matic action in heme degradation and also to the catalytic byproducts including carbon monoxide and biliverdin

Sengupta, Guang Yang and Phyllis A. DenneryChhanda Biswas, Nidhi Shah, Manasa Muthu, Ping La, Amal P. Fernando, Shaon

Activation of Nrf2, Impacting Metabolic and Anti-oxidant DefensesNuclear Heme Oxygenase-1 (HO-1) Modulates Subcellular Distribution and

doi: 10.1074/jbc.M114.567685 originally published online August 8, 20142014, 289:26882-26894.J. Biol. Chem. 

  10.1074/jbc.M114.567685Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/289/39/26882.full.html#ref-list-1

This article cites 64 references, 27 of which can be accessed free at

by guest on Novem

ber 5, 2020http://w

ww

.jbc.org/D

ownloaded from