13
Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing Radiosensitive Effects of Parthenolide in Normal and Cancer Cells Yong Xu 1 , Fang Fang 2 , Sumitra Miriyala 1 , Peter A. Crooks 3 , Terry D. Oberley 4 , Luksana Chaiswing 4 , Teresa Noel 1 , Aaron K. Holley 1 , Yanming Zhao 1 , Kelley K. Kiningham 5 , Daret K. St. Clair 1 , and William H. St. Clair 2 Abstract Elevated oxidative stress is observed more frequently in cancer cells than in normal cells. It is therefore expected that additional exposure to a low level of reactive oxygen species (ROS) will push cancer cells toward death, whereas normal cells might maintain redox homeostasis through adaptive antioxidant responses. We previously showed that parthenolide enhances ROS production in prostate cancer cells through activation of NADPH oxidase. The present study identies KEAP1 as the downstream redox target that contributes to parthenolide's radiosensitization effect in prostate cancer cells. In vivo, parthenolide increases radiosensitivity of mouse xenograft tumors but protects normal prostate and bladder tissues against radiation-induced injury. Mechanistically, parthenolide increases the level of cellular ROS and causes oxidation of thioredoxin (TrX) in prostate cancer cells, leading to a TrX-dependent increase in a reduced state of KEAP1, which in turn leads to KEAP1-mediated PGAM5 and Bcl-xL (BCL2L1) degradation. In contrast, parthenolide increases oxidation of KEAP1 in normal prostate epithelial cells, leading to increased Nrf2 (NFE2L2) levels and subsequent Nrf2- dependent expression of antioxidant enzymes. These results reveal a novel redox-mediated modication of KEAP1 in controlling the differential effect of parthenolide on tumor and normal cell radiosensitivity. Further- more, they show it is possible to develop a tumor-specic radiosensitizing agent with radioprotective properties in normal cells. Cancer Res; 73(14); 112. Ó2013 AACR. Introduction It is well documented that cancer cells are usually under more oxidative stress than normal cells are, in part, due to a hyperactive metabolism that fuels their rapid growth (1, 2). Thus, a therapy designed to increase reactive oxygen species (ROS) to a level above the threshold for cancer cell death, but to an adaptable level for normal cells, would be an attractive strategy to selectively kill cancer cells (3, 4). Redox homeostasis is thought to regulate many cellular processes that are essential for maintenance of normal physiologic conditions but is aberrantly modulated in cancers (5, 6). The functions of ROS are both benecial and deleterious due to their dual role in the prosurvival and antisurvival pathways. As a secondary messenger in cell signaling, ROS are required for normal development and can initiate adaptive responses in cellular defense (7, 8). On the other hand, ROS cause structural damage and functional decline in DNA, proteins, and lipids, and consequently act as an antitumorigenic factor by inducing cell senescence and apoptosis (9, 10). Indeed, ROS-mediated cell death is an important basis for radiotherapy and many chemotherapeutic treatments (11, 12). Currently, these ther- apeutic strategies are being used to kill cancer cells without benet of a rational design that exploits the intrinsic differ- ences in the cellular redox status of normal cells and cancer cells. Antioxidant defense systems are essential for the regula- tion of ROS levels, which have an important function in the maintenance of cellular redox hemostasis. Mounting evi- dence shows that a decline in antioxidant function may be involved in tumorigenesis due to prooxidant conditions that result from ROS accumulation. For example, manganese superoxide dismutase (MnSOD) is downregulated in many types of cancer and overexpression of MnSOD results in suppression of tumorigenesis (1315). High levels of antioxidants caused by therapy-mediated activation of prosurvival pathways, such as NF-kB and Nrf2, are thought to protect cancer cells against treatment (1618). Thus, inhibition of prosurvival pathways has been a traditional strategy to enhance therapeutic efcacy. Increasing evidence shows that certain mild prooxidant compounds derived from natural herbal medicines might enhance some anticancer treatments by modulating the Authors' Afliations: 1 Graduate Center for Toxicology, 2 Department of Radiation Medicine, University of Kentucky, Lexington, Kentucky; 3 Depart- ment of Pharmaceutical Sciences, UAMS College of Pharmacy, Little Rock, Arkansas; 4 Department of Pathology, University of Wisconsin, Madison, Wisconsin; and 5 Department of Pharmaceutical Sciences, Belmont Uni- versity School of Pharmacy, Nashville, Tennessee Corresponding Authors: William H. St. Clair, Department of Radiation Medicine, University of Kentucky, College of Medicine, Lexington, KY 40536. Phone: 859-257-4931; Fax: 859-323-6486; E-mail: [email protected]; and Daret K. St. Clair, E-mail: [email protected] doi: 10.1158/0008-5472.CAN-12-4297 Ó2013 American Association for Cancer Research. Cancer Research www.aacrjournals.org OF1 Research. on April 15, 2021. © 2013 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

Therapeutics, Targets, and Chemical Biology

KEAP1 Is a Redox Sensitive Target That Arbitrates theOpposing Radiosensitive Effects of Parthenolide in Normaland Cancer Cells

Yong Xu1, Fang Fang2, Sumitra Miriyala1, Peter A. Crooks3, Terry D. Oberley4, Luksana Chaiswing4, TeresaNoel1, Aaron K. Holley1, Yanming Zhao1, Kelley K. Kiningham5, Daret K. St. Clair1, and William H. St. Clair2

AbstractElevated oxidative stress is observed more frequently in cancer cells than in normal cells. It is therefore

expected that additional exposure to a low level of reactive oxygen species (ROS) will push cancer cells towarddeath, whereas normal cells might maintain redox homeostasis through adaptive antioxidant responses. Wepreviously showed that parthenolide enhances ROS production in prostate cancer cells through activation ofNADPH oxidase. The present study identifies KEAP1 as the downstream redox target that contributes toparthenolide's radiosensitization effect in prostate cancer cells. In vivo, parthenolide increases radiosensitivity ofmouse xenograft tumors but protects normal prostate and bladder tissues against radiation-induced injury.Mechanistically, parthenolide increases the level of cellular ROS and causes oxidation of thioredoxin (TrX) inprostate cancer cells, leading to a TrX-dependent increase in a reduced state of KEAP1, which in turn leads toKEAP1-mediated PGAM5 and Bcl-xL (BCL2L1) degradation. In contrast, parthenolide increases oxidation ofKEAP1 in normal prostate epithelial cells, leading to increased Nrf2 (NFE2L2) levels and subsequent Nrf2-dependent expression of antioxidant enzymes. These results reveal a novel redox-mediated modification ofKEAP1 in controlling the differential effect of parthenolide on tumor and normal cell radiosensitivity. Further-more, they show it is possible to develop a tumor-specific radiosensitizing agent with radioprotective propertiesin normal cells. Cancer Res; 73(14); 1–12. �2013 AACR.

IntroductionIt is well documented that cancer cells are usually under

more oxidative stress than normal cells are, in part, due to ahyperactive metabolism that fuels their rapid growth (1, 2).Thus, a therapy designed to increase reactive oxygen species(ROS) to a level above the threshold for cancer cell death, but toan adaptable level for normal cells, would be an attractivestrategy to selectively kill cancer cells (3, 4). Redox homeostasisis thought to regulatemany cellular processes that are essentialfor maintenance of normal physiologic conditions but isaberrantly modulated in cancers (5, 6). The functions ofROS are both beneficial and deleterious due to their dual rolein the prosurvival and antisurvival pathways. As a secondarymessenger in cell signaling, ROS are required for normaldevelopment and can initiate adaptive responses in cellular

defense (7, 8). On the other hand, ROS cause structural damageand functional decline in DNA, proteins, and lipids, andconsequently act as an antitumorigenic factor by inducingcell senescence and apoptosis (9, 10). Indeed, ROS-mediatedcell death is an important basis for radiotherapy and manychemotherapeutic treatments (11, 12). Currently, these ther-apeutic strategies are being used to kill cancer cells withoutbenefit of a rational design that exploits the intrinsic differ-ences in the cellular redox status of normal cells and cancercells.

Antioxidant defense systems are essential for the regula-tion of ROS levels, which have an important function in themaintenance of cellular redox hemostasis. Mounting evi-dence shows that a decline in antioxidant function may beinvolved in tumorigenesis due to prooxidant conditions thatresult from ROS accumulation. For example, manganesesuperoxide dismutase (MnSOD) is downregulated in manytypes of cancer and overexpression of MnSOD resultsin suppression of tumorigenesis (13–15). High levels ofantioxidants caused by therapy-mediated activation ofprosurvival pathways, such as NF-kB and Nrf2, are thoughtto protect cancer cells against treatment (16–18). Thus,inhibition of prosurvival pathways has been a traditionalstrategy to enhance therapeutic efficacy.

Increasing evidence shows that certain mild prooxidantcompounds derived from natural herbal medicines mightenhance some anticancer treatments by modulating the

Authors' Affiliations: 1Graduate Center for Toxicology, 2Department ofRadiationMedicine, University of Kentucky, Lexington, Kentucky; 3Depart-ment of Pharmaceutical Sciences, UAMSCollege of Pharmacy, Little Rock,Arkansas; 4Department of Pathology, University of Wisconsin, Madison,Wisconsin; and 5Department of Pharmaceutical Sciences, Belmont Uni-versity School of Pharmacy, Nashville, Tennessee

Corresponding Authors: William H. St. Clair, Department of RadiationMedicine, University of Kentucky, College of Medicine, Lexington, KY40536. Phone: 859-257-4931; Fax: 859-323-6486; E-mail:[email protected]; and Daret K. St. Clair, E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-12-4297

�2013 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org OF1

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 2: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

redox state of cancer cells to high prooxidant levels (19, 20).Parthenolide, an active ingredient derived from the tradi-tional anti-inflammatory medical plant feverfew (Tanacetumparthenium), belongs to the family of sesquiterpene lactonescontaining an a-methylene-g-lactone moiety and an epoxidegroup, which is able to conjugate thiols of proteins through aMichael addition reaction (21). In addition to its anti-inflammatory effect, parthenolide seems to be toxic to avariety of cancer cells (22–25). Importantly, parthenolide hasno cytotoxic effect on normal cells (25). Mechanistically,parthenolide has been shown to increase apoptosis in cancercells through inhibition of multiple prosurvival pathways,such as NF-kB and PI3K-AKT (19, 26). However, thesefindings do not explain why parthenolide is not toxic tonormal cells.

Posttranslational modification is a keymechanism by whichproteins dramatically increase their functional diversity.Reversible redox modification of protein cysteine residuesplays an important role in vital cell signaling pathways relatedto many physiologic and pathogenic processes (27, 28). TheKeap1-Nrf2 pathway is one of the main cellular defensemechanisms against oxidative stress (29, 30). The present studyexamines the role of cysteine modifications in modulatingradiation responses in prostate cancer cells versus normalprostate epithelial cells. It elucidates the functional linkbetween redox modulation and cell signaling transductionpathways, and it provides evidence for the differential effectof parthenolide on cellular redox status in normal and cancercells. The results reveal that parthenolide-mediated redoxmodification of Keap1 serves as a central regulator of differ-ential responses to radiotherapy in normal and tumor cells.The present study provides a proof-of-concept for using anintrinsic difference in cellular redox conditions to kill tumorcells while protecting normal cells from the unwanted sideeffects of radiation.

Materials and MethodsCell culture, cell transfection, treatment, and cellsurvival analysis

Human prostate carcinoma/adenocarcinoma LNCaP,PC3, and DU145 cell lines as well as human prostate epi-thelial viral transformed PZ-HPV-7 (PZ) and RWPE-1 celllines were obtained from American Type Culture Collection.Normal prostate epithelial PrEC cells were purchased fromLonza. All cell lines were cultured and maintained in themedia recommended in the manufacturer's protocols.Plasmid-cloned Keap1 cDNA and Bcl-xL cDNA (OriGene)and siRNAs for knocking down Keap1, Nrf2, thioredoxin(TrX), PGAM5, and Bcl-xL (Dharmacon) were transfectedinto cultured cells before treatment. Parthenolide andits water-soluble prodrug, dimethylamino-parthenolide(DMAPT), were synthesized as previously described (31).The cells were treated with 0 to 5 mmol/L parthenolidefollowed by irradiation by a 250 kV X-ray machine (FaxitronX-ray Corp.) with peak energy of 130 kV, 0.05 mm Al filter, ata dose of 0 to 6 Gy. Cell survival rates were quantified bycolony survival fraction, Trypan blue exclusion assay, andMTT assay, as previously described (25, 32).

AnimalsFour- to five-week-old male NCRNU (nu/nu athymic nude)

mice were obtained from Taconic (Hudson). For formation ofxenograft tumors, 1.8 � 106 cells mixed in Matrigel (BDBiosciences) were subcutaneously injected into the right flankof themice. Tumor volumeswere routinelymeasured and theirsizes calculated on the basis of a protocol described elsewhere(33). Animals with an average tumor size of 500 mm3 wererandomized into several groups for DMAPT and radiationtreatments. The tumors were treated 5 times with 10 mg/kgDMAPT and 3 Gy IR. The tumor tissues were collected, and 100mg of tissue was lysed to quantify amounts of oxidized orreduced Keap1 and levels of downstream proteins. To deter-mine the protective effect of parthenolide against radiationdamage, mice were pretreated with DMAPT followed by radi-ation treatment (5 � 3Gy). Prostate and bladder tissues werefixed, embedded, and processed for routine electron micros-copy. The embedded blocks were sectioned and transferred tocopper grids and counterstained with uranyl acetate, followedby lead citrate. Grids were observed in an electron microscope(Hitachi H-600) operated at 75 kV. Mitochondrial damage wasquantified by a pathologist (T.D. Oberley) using identifiedultrastructural changes including swelling, vacuolization,myelination, disorganization, and loss of cristae, lysosomaldegradation, and membrane disruption. Animal experimentalprocedures were approved by the Institutional Animal Careand Use Committee of the University of Kentucky (Lexington,KY), Approval Protocol No. 01077M2006.

Quantification of intracellular superoxide andprooxidant

Dihydroethidium (DHE, Invitrogen), which exhibits blue fluo-rescence in the cytosol until oxidized, was used to estimate thelevels of superoxide after parthenolide treatment. To confirm thelevel of superoxide induced by parthenolide, the cells were pre-treated with 50 mg PEG-SOD (Sigma) for 1 hour followed byparthenolide treatment. Antimycin (Sigma) was used as a posi-tive control because it has been shown to increase superoxide inall tested cell lines. A dichlorofluorescein (DCF) assay was usedto quantify the levels of intracellular ROS after parthenolidetreatment. The cells were labeled by both carboxy-H2DCFDA(sensitive to oxidation, Invitrogen) and carboxy-DCFDA (insen-sitive to oxidation, Invitrogen). The H2DCFDA:DCFDA ratio wasused to optimize the controls of cell number, dry uptake, andester cleavage. The procedures for both DHE and DCF wereconducted by theUniversity of Kentucky FlowCytometer Facilityusing a FACScan protocol provided by Dr. Douglas R. Spitz(Professor of Radiation Oncology at the University of Iowa; 34).

Measurement of oxygen consumption ratesTo determine how parthenolide changes mitochondrial

function in cancer and normal cells, a Seahorse BioscienceXF24 Extracellular Flux Analyzer was used to measure oxygenconsumption rates (OCR) after parthenolide treatment. TheXF24 creates a transient, 7 mL chamber in specialized micro-plates, which allows determination of oxygen and protonconcentrations in real time. To allow comparison betweenexperiments, data are expressed as the OCR in pmoL/min or

Xu et al.

Cancer Res; 73(14) July 15, 2013 Cancer ResearchOF2

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 3: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

the rate of extracellular acidification in mpH/min. Reservecapacity, an important index that indicates capacity of mito-chondrial respiration, is calculated by subtracting baselineOCR from maximal OCR.

Detection of oxidized and reduced forms of Keap1protein3-N-maleimido-propionyl biocytin was used to selectively

label sulfhydryl (SH) and then was detected by biotin–strep-tavidin integration on the blots, as previously described (35).To quantify disulfide (S-S) bonds, the SH formwas stabilized bytreating with N-ethylmaleimide and then the S-S bonds werereduced by treating with 2-mercaptoethanol. To identify SHand S-S moieties of Keap1 protein, the labeled proteins wereimmunoprecipitated by Keap1 antibody (Abcam) and sub-jected to SDS-PAGE, followed by detection with horseradishperoxidase-conjugated streptavidin (Sigma).

Immunoblots and immunoprecipitationHomogenized cells and tumor tissues were electrophoresed

on an 8% (w/v) SDS-PAGE gel, transferred onto a nitrocellulosemembrane, and subsequently incubated with primary antibo-dies against Keap1 (Abcam), Nrf2 (Abcam), MnSOD (UpstateBiotech), CuZnSOD (eBiosci), Gpx (Abcam), catalase (Milli-pore), TrX (BD Sciences), PGAM5 (Santa Cruz Biotech.), Bcl-xL(Santa Cruz Biotech.), LC3B (Cell Signaling), b-actin (Sigma),and pCNA (Santa Cruz Biotech.). All secondary antibodieswereobtained from Santa Cruz Biotech. Immunoblots were visual-ized using an enhanced chemiluminescence detection system(Amersham Pharmacia Biotech.). For immunoprecipitation,cell extracts were incubated overnight with one primary anti-body at 4�C and integrated with a protein A/G agarose (SantaCruz Biotech). Immunocomplexes were precipitated and frac-tionated on an SDS-PAGE gel. Interacting proteins weredetected by immunoblotting with their primary antibodies.

SOD enzymatic assayMnSOD activities were measured by the nitroblue tetrazo-

lium-bathocuproin sulfonate reduction inhibition method.Sodium cyanide (2 mmol/L) was used to inhibit CuZnSODactivity (36).

Statistical data analysesMultiple independent experiments were conducted for each

set of data presented. Images in Immunoblots were quantifiedusing Carestream Molecular Imaging software (CarestreamHealth Inc.). Statistical significance was analyzed using one-way ANOVA and Tukey multiple comparison test, followed bydata analysis with GraphPad Prism.

ResultsParthenolide enhances radiosensitivity of prostatetumors but protects normal tissues from radiationOur previous studies showed that parthenolide is able to

selectively enhance the radiosensitivity of prostate cancer cellswithout injury to normal prostate epithelial cells (24, 25). Todetermine whether parthenolide enhances radiotherapy invivo, human prostate cancer PC3 cells were subcutaneously

implanted into the right flank of nudemale mice. When tumorsize reached a volume of 500 mm3, animals were randomizedinto groups according to the treatment consisting of saline or10 mg/kg DMAPT. One hour after saline or DMAPT wasadministered, the tumors were treated with fractionated radi-ation of 1 Gy or 2 Gy per day for 5 days followed by routinemeasurement of tumor volume. The tumor growth curves areshown in Fig. 1A. Mice were humanely killed when a tumorreached the maximum size of 2,000 mm3. Tumor growth wasclearly delayed in the treatment groups, particularly when thedrug and radiation were combined, compared with growth inthe untreated group. The tumor growth rates after treatmentwere compared according to the days needed for tumorvolume to reach 2,000 mm3. DMAPT significantly enhancedradiotherapeutic efficiency compared with the effects of radi-ation treatment alone (Fig. 1B). A separate group of nude malemice that had no cancer cell implantation was treated withDMAPT and radiation to determine the toxicity of DMAPT toorgans that can be affected by radiotherapy of prostate cancer.Prostates and bladders of the animals were examined by lightand electron microscopy. At 60 days after irradiation, no grosspathology was observed (data not shown). However, ultra-structural damage was clearly observed by electron micros-copy (Fig. 1C). Mitochondrial damage was most pronouncedand this was morphometrically analyzed. The number ofdamaged mitochondria in prostate and bladder was propor-tional to radiation exposure. Pretreatment with DMAPT sig-nificantly reduced the number of damaged mitochondria inboth organs compared with the group without DMAPT treat-ment (Fig. 1C and D). These results indicate that DMAPT, thewater-soluble prodrug of parthenolide, is a promising agent forselectively enhancing the sensitivity of prostate cancer cells toradiation while protecting normal tissues from damage causedby radiation.

Parthenolide differentiallymodulates cellularROS levelsin cancer and normal cells

To determine the effect of parthenolide on redox homeo-stasis in cancer and normal cells, the levels of superoxide andtotal ROS after parthenolide treatment were measured usingflow cytometry. The mean fluorescence intensity of DHE andtheH2DCFDA:DCFDA ratiowere higher in prostate cancer PC3cells than in normal prostate PZ and PrEC cells, indicatinghigher basal levels of superoxide and total ROS, respectively(Fig. 2A and B). Following parthenolide treatment, DHE andDCF levels increased further in PC3 cells but declined slightly inboth types of noncancerous cells. Combining PEG-SOD withparthenolide treatment restored the basal level of DHE fluo-rescence. As positive controls, addition of equivalent concen-trations of the ROS-stimulating agents antimycin (Fig. 2A) andphorbol-12-myristate-13-acetate (PMA; Fig. 2B) caused highlevels of ROS in all the tested cell lines.

The levels of antioxidant proteins were also quantified(Fig. 2C). Parthenolide altered the protein level of the antiox-idant enzymes, in particular mitochondria-localized antioxi-dant enzymes. MnSOD and glutathione peroxidase (GpX)were significantly reduced in all 3 parthenolide-treated pros-tate cancer cell lines. Intriguingly, parthenolide had the

Keap1, a Central Regulator of Cellular Redox Signaling

www.aacrjournals.org Cancer Res; 73(14) July 15, 2013 OF3

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 4: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

opposite effect in the 3 normal prostate cell lines. However,neither cancer nor normal cells showed any obvious changesafter parthenolide treatment for the major cytosolic superox-ide removal protein, copper, and zinc-containing SOD (CuZn-SOD). This observation was confirmed by quantification of thecorresponding enzyme activity (Fig. 2D). These results suggestthat parthenolide-mediated alteration of cellular redox statusis mediated, at least in part, by changing the activities ofantioxidant enzymes in mitochondria.

To probe whether altering cellular redox status is associatedwith a change in mitochondrial respiration, the OCR in theparthenolide-treated cells was measured using a SeahorseBioscience FX OxygenFlux Analyzer. The basal and maximalOCR in normal cells was higher than in cancer cells (Fig. 2E).Importantly, parthenolide was able to increase the OCR andreserve capacity in PZ cells, whereas parthenolide had no effecton PC3 OCR. Finally, the cytotoxicity of parthenolide was

tested in all the cell lines using an MTT assay, which requiresactive mitochondria. As shown in Fig. 2F, parthenolide wastoxic to all the cancer cells but not to the normal cell lines.Taken together, these results suggest that changes in cellularredox status andmitochondrial functionmay be a cause for thedifferential biologic effects of parthenolide on cancer andnormal cells.

Keap1 is susceptible to parthenolide-mediated redoxmodification

Keap1, a redox-sensitive protein, has been reported to playan important role in cell survival under oxidative stress (29). Toinvestigate whether parthenolide modifies Keap1 function, aKeap1 antibody linked to biotin was used to immunoprecip-itate redox-modified Keap1 protein and the presence of oxi-dized (-S-S-) and reduced (-SH) cysteine residues was detectedusing a secondary antibody linked to streptavidin. In the 3

A

B

5 x 3 Gy DMAPT + 5 x 3 Gy

Prostate

Bladder

0

20

40

60

80

100

Prostate Bladder

5 x 3Gy DMAPT/5 x 3Gy

*

*

% D

amag

ed m

ito

cho

nd

ria

/ to

tal

mit

och

on

dri

a

* P < 0.05

C

D

a b

c d

12,000X 12,000X

8,000X 8,000X

0

10

20

30

40

50

60

70

P < 0.05

P < 0.05

P < 0.01

Day

s w

hen

tum

or

volu

me

reac

hes

to

2,0

00 (

mm

3 )

Days after treatment

Tu

mo

r vo

lum

e (m

m3 )

0

500

1,000

1,500

2,000

2,500

3,000

0 2 4 6 8 11 14 17 20 24 28 32 36

Untreated

DMAPT

1 Gy

1 Gy + DMAPT

2 Gy

2 Gy + DMAPT

Untre

ated

DMAPT1 G

y

1 Gy +

DMAPT

2 Gy

2 Gy +

DMAPT

Figure 1. The effect of parthenolide on radiosensitivities of prostate cancer and normal cells. A, prostate cancer PC3 cells were injected into the flanks of nudemale mice. The resulting tumors were treated with DMAPT and IR. Tumor volume was measured and tumor growth was calculated. B, time needed fortumor growth to reach 2,000mm3 volume after treatment was calculated and plotted. C, mice without cancer were treated with radiation alone (5� 3 Gy) andDMAPT (10 mg/kg) with radiation. Prostate and bladder tissues were removed for pathologic analysis using electron microscopy. Arrows indicate normalmitochondria and asterisks indicate mitochondria with myelin figures. M, normal mitochondria; Ly, lysosome; and V, mitochondria with vacuoles. D,quantification of mitochondrial damage in mice prostate and bladder tissues.

Xu et al.

Cancer Res; 73(14) July 15, 2013 Cancer ResearchOF4

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 5: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

normal cell lines, parthenolide increased the oxidized form ofKeap1 but decreased the reduced form of Keap1 (Fig. 3A).Interestingly, the results from the 3 cancer cell lines seemed tobe completely opposite to results observed in normal cellstreated with parthenolide: the level of the oxidized form wasdecreased, but the level of the reduced form was increased

(Fig. 3B). To verify that the observed increase in reduced Keap1also occurred in vivo, mouse xenograft tumor tissues with andwithoutDMAPT treatmentwere also used for determination ofKeap1 redox status. Consistent with data obtained from cul-tured tumor cells treated with parthenolide, the in vivo resultsshow that parthenolide decreased the oxidized form of Keap1

A

B

Mea

n flu

ores

cenc

e in

tens

ity

MnSOD

β-Actin

- + - + - + - + - + - +

GpX

CuZnSOD

Catalase

Cancer cells Normal cells0

50

100

150

200

250

300

Basal ATP-linked Maximal OCR Reservecapacity

OC

R (

pMol

es/m

in/m

gpro

tein

)

PZPZ+PNPC3PC3+PN

NS

NS

NS

NS

P < 0.01

P < 0.05

P < 0.01

P < 0.01

DHE assay

0

500

1,000

1,500

2,000

DMSO PN (5 μmol/L) PN+PEG-SOD(50 μ/mL)

Antimycin(5 μmol/L)

PC3

PZ

PrEC

*

*#

$

DCF assay

0

20

40

60

80

100

120

DMSO PN (5 μmol/L)

PN (5 μmol/L)

PN (μmol/L)

PMA (5 μmol/L)

PC3

PZ

PrEC* *# $

C

E

F

0

20

40

60

80

100

120

140

160

180

200

MnSOD CuZnSOD Catalase GpX

PC3/DMSO

PC3/PN

PZ/DMSO

PZ/PN

PrEC/DMSO

PrEC/PN

*

*

*

*

**

*P < 0.05

Enz

yme

activ

ity (

units

/min

/mg)

Cel

l sur

viva

l (%

)

0

20

40

60

80

100

120

0 1 2 3 4 5

PC3

DU-145

LNCaP

PZ

RWPE-1

PrEC

PC3DU-1

45

LNCaP

PZ RWPE-1

PrEC

D

H2D

CF

DA

/DC

FD

A

Figure 2. The effect of parthenolide on redox homeostasis in prostate cancer and normal cells. A–B, cells were treated with parthenolide and then labeledwith DHE or DCF. Themean fluorescence intensity of DHE (A) and the ratio of H2DCFDA to DCFDA (B) were determined using flow cytometry. Concentrationsof cellular superoxide and total ROS were estimated by quantification of fluorescence intensity. Antimycin and PMA were used as positive controls forgeneration of ROS. PEG-SOD was used as a control to remove superoxide generated by DHE. PN, parthenolide. �, #, $ indicate the significances oftreatments compared to DMSO untreated control in PC3 (�), PZ (#), and PrEC ($) cells. C, the levels of antioxidant proteins were quantified by Western blots.D, the activities of the corresponding enzymes were measured. E, quantification of the basal oxygen consumption, ATP-linked oxygen consumption, themaximal OCR after the addition of FCCP, and the reserve capacity of the cells. NS, not significant. F, three cancer cell lines and 3 noncancer cell lines weretreated with parthenolide (PN) at the indicated concentrations. Cell survival fraction was determined by MTT assay.

Keap1, a Central Regulator of Cellular Redox Signaling

www.aacrjournals.org Cancer Res; 73(14) July 15, 2013 OF5

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 6: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

but increased the reduced form of Keap1 in the tumors (Fig.3C). Changes in antioxidant proteins inmouse xenograft tumortissues treated with DMAPT are also consistent with the resultobtained from in vitro studies (Fig. 3D), indicating that parthe-nolide decreases the level of mitochondrial antioxidant pro-teins in prostate tumors.

Oxidization of Keap1 leads to activation of the Nrf2prosurvival pathway in normal cells

Activation of the Nrf2 signaling pathway through dissocia-tion with Keap1 resulting in Nrf2 nuclear translocation isconsidered to be a primary prosurvival pathway in responseto oxidative stress (30, 37). To examine whether parthenolidechanges Nrf2 nuclear translocation, the levels of Nrf2 in nucleiwere measured. As shown in Fig. 4A, the nuclear levels of Nrf2were increased in the 3 normal cell lines treated with parthe-nolide, but no changes were observed in the 3 cancer cell lines.To examine whether activation of the Nrf2 pathway is a majormechanism by which parthenolide protects normal cellsagainst radiation injury, Keap1 and Nrf2 were silenced in PZcells by transfecting their siRNA (Fig. 4B, left). Cell survivaldecreased when Nrf2 was silent. IR significantly reduced cellsurvival but the cell survival was restored when Keap1 wassilenced (Fig. 4B, right panel). These results suggest that

oxidation of Keap1 and subsequent activation of Nrf2 byparthenolide is essential for normal cell survival after radiationtreatment.

Thioredoxin is necessary for parthenolide-mediatedreduction of Keap1 in cancer cells

TrX is highly expressed in cancer cells and stimulates cellgrowth. We previously reported that parthenolide decreasesthe reduced form of TrX but increases the oxidized form of TrXin prostate cancer cells (25). In the present study, we verify thatTrX was expressed at a high level in all 3 cancer cell lines,whereas a low level was observed in the 3 noncancer cell lines(Fig. 5A). Immunoprecipitation of Keap1 protein from PC3 cellextracts using a TrX antibody suggests an interaction betweenKeap1 and TrX that is increased by parthenolide (Fig. 5B). Todetect whether the parthenolide-influenced reduction ofKeap1 in cancer cells is dependent on TrX, we selectivelysilenced TrX by transfecting its siRNA before parthenolidetreatment (Fig. 5C, left). As expected, the reduced form ofKeap1 was decreased, but the oxidized form of Keap1 wasincreased when TrX was silent (Fig. 5C, middle and right). Theresults suggest that TrX is interacting with Keap1 to keepKeap1 in a reduced state in parthenolide-treated cells. Tofurther confirm that the function of Keap1 leads to cell death

A

B

IgG

PN − + − +

Keap1

IgG

PN − + − +

Keap1

IgG

PN − + − +

Keap1

PrECPZ-HPV-7 RWPE-1

-SH-S-S- -SH-S-S- -SH-S-S-

-SH

IgG

PN − + − +

Keap1

-S-S-

PC-3

-SH

IgG

PN − + − +

Keap1

-S-S-

DU-145

-SH

IgG

PN − + − +

Keap1

-S-S-

LNCaP

-SH -S-S-

DMAPT − + − +

Keap1

IgG

C

Fol

d ch

ange

(%

)

*P < 0.05

0

20

40

60

80

100

120

140

MnSOD GpX CuZnSOD Catalase

DMSO

DMAPT

DMSO

DMAPT

* *

Catalase

MnSOD

GPX

CuZnSOD

β-Actin

D

Figure 3. Keap1 redox modification by parthenolide. A and B, 3 normal cell lines (A) and 3 prostate cancer cell lines (B) were treated with saline orparthenolide (PN). Keap1was immunoprecipitated with its antibody and SH- and S-Smoieties of Keap1were shown by ECL detection. C, tumor tissueswerehomogenized and incubated with MPB. Reduced (SH-) and oxidized (S-S) forms of Keap1 in the tissues were detected. D, tumor tissues were homogenizedand antioxidant proteins were quantified by Western blots. Right, representative blots. Left, the average of multiple blots.

Xu et al.

Cancer Res; 73(14) July 15, 2013 Cancer ResearchOF6

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 7: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

in parthenolide-treated cancer cells, a Keap1 expression con-struct was transfected into PC3 cells, followed by parthenolideand IR treatments. Overexpression of Keap1 resulted inincreases in cell death in both treated and untreated cells (Fig.5D, top). The levels ofmitochondrial phosphoglyceratemutase5 (PGAM5), a protein serine/threonine phosphatase that inter-acts with Bcl-xL in the mitochondrial membrane (38), and Bcl-xLwere clearly decreased in the Keap1-transfected cells, but nochanges were observed in Nrf2, Ikka, and IkBa (Fig. 5D,bottom). These results suggest that the parthenolide-increasedreduced form of Keap1 facilitates Keap1-mediated ubiquitin/proteasome-dependent degradation of PGAM5 and Bcl-xL,which is an established mechanism for parthenolide-mediatedcell death in cancer cells.

Keap1 triggers PGAM5-mediated Bcl-xL ubiquitindegradation in parthenolide-treated cancer cellsTo further investigate themechanismbywhich parthenolide

enhances the radiosensitivity of prostate cancer cells, wedetermined the interactions between Keap1, PGAM5, andBcl-xL. The results show that a reduced form of Keap1, whichis increased in parthenolide-treated PC3 cells, enhanced inter-action between Keap1 and PGAM5, as detected by immuno-precipitation using a PGAM5 antibody (Fig. 6A). Bcl-xL, aprosurvival mitochondrial protein, was also increased in thepulled down complex (Fig. 6A). Interestingly, the proteins thatare associated with Keap1 were decreased in whole-cellextracts (Fig. 6B). A time course of parthenolide treatmentshows that PGAM5 and Bcl-xL proteins were slightly increasedat 12 hours but decreased at 24 and 48 hours after treatment(Fig. 6C). Proteins in different cellular fractions were alsoquantified (Fig. 6D). The mitochondria-associated proteinsPGAM5 and Bcl-xL were reduced by the parthenolide treat-ment, but no change was observed in Hsp75, a control formitochondrial protein. Parthenolide had nomajor effect on the

levels of Nrf2 and Ikka in treated cells. These results suggestthat parthenolide enhances Keap1-mediated ubiquitin/pro-teasome-dependent degradation of PGAM5 and Bcl-xL (39).In addition, parthenolide increased the level of mitochondria-associated autophagic protein LC3B, suggesting that parthe-nolidemay enhance the radiation sensitivity of prostate cancercells partially through triggering the autophagy pathway.

Because Keap1 interacts with PGAM5/Bcl-xL/Nrf2, wedecided to determine the effect of PGAM5/Bcl-xL/Nrf2 inmediating parthenolide's effect on cancer cells. PGAM5, Bcl-xL, and Nrf2 were silenced using their siRNAs, followed byparthenolide treatment (Fig. 6E, bottom). The cell survivalfraction was decreased when PGAM5 or Bcl-xL was silent,which is similar to the effect of parthenolide (Fig. 6E, top). Nosignificant additive effects were observed when parthenolidewas combined with PGAM5 or Bcl-xL siRNA. In contrast, asignificant effect was observed when Nrf2 was silent. Theseresults suggest that Keap1-mediated PGAM5/Bcl-xL degrada-tion, but not Nrf2 degradation, is important for parthenolide-induced cancer cell death.

To further determine whether the function of Bcl-xL plays amajor role in protecting cancer cells against parthenolide-induced cell death, a plasmid carrying Bcl-xL cDNA wastransfected into PC3 cells followed by parthenolide treatment(Fig. 6F, bottom). The results show that expression of Bcl-xLefficiently protects cells from cytotoxicity caused by parthe-nolide (Fig. 6F, top). Taken together, these results suggest thatparthenolide enhances the radiosensitivity of prostate cancercells, in part, by triggering ubiquitin/proteasome-based deg-radation of Bcl-xL.

In summary, parthenolide provides radiosensitization inprostate cancer cells but radioprotection in normal cells, andthe observed differential effects are mediated, in part, by redoxmodification of Keap1, i.e., reducing Keap1 in cancer cellsbut oxidizing Keap1 in normal cells. The distinct redox

A

B

PCNA

− + − + − + − + − + − +

PC3 DU-145 LNCaP PZ PrEC RWPE-1

Nrf2

Keap1

Nrf2

β-Actin

PN (5 μmol/L)

PN (5 μmol/L)

− + − + − +

P < 0.01

P < 0.01

Cel

l su

rviv

al (

%)

0

20

40

60

80

100

120

140

Untreated 6 Gy PN 5 μmol/L PN + 6Gy

Ctrl siRNA

Keap1 siRNA

Nrf2 siRNACtrl

siRNA

Keap1

siRNA

Nrf2 si

RNA

Figure 4. Activation of Keap1-Nrf2pathway by parthenolide in normalcells. A, parthenolide (PN)increases nuclear levels of Nrf2 innormal cells but not in cancer cells.Nuclear proteins extracted fromthe parthenolide-treated cell lineswere immunoblotted to quantifynuclear levels of Nrf2 using PCNAas loading control. B, the effect ofKeap1 and Nrf2 in normal cells. PZcells were transfected with siRNAsto knock down Keap1 or Nrf2,respectively. After treatment withparthenolide and IR, the cellsurvival fraction was quantifiedusing Trypan blue exclusion assay(right) and the knocked downKeap1 and Nrf2 were confirmed byWestern blots (left).

Keap1, a Central Regulator of Cellular Redox Signaling

www.aacrjournals.org Cancer Res; 73(14) July 15, 2013 OF7

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 8: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

modification of Keap1 initiates different signaling pathwaysthat affect mitochondrial function, leading to cell survival orcell injury in response to radiation, as illustrated in Fig. 7.

DiscussionThe majority of anticancer therapies fail because cancers

develop phenotypes that are treatment resistant and becausetreatments cause unwanted and/or detrimental side effects tonormal cells or to untargeted tissues. While conventionaladjuvant therapies improve tumor response to radiotherapy,they generally cause additional damage to normal tissues.Thus, the focus of the present study is to identify adjuvanttherapeutics that can reduce the side effects of radiotherapy.Our study provides a proof-of-concept for improving theefficacy of radiotherapy while protecting against injury to

normal tissues. It has been shown that parthenolide, theanti-inflammatory phytochemical, is able to suppress tumorgrowth in many organs (22–25). In addition, parthenolideseems to synergically enhance chemotherapeutic efficiencywhen it is combined with taxol or cisplatin to treat lung andgastric cancer cells (23, 40). Parthenolide also sensitizes radio-resistant osteosarcoma cells to radiotherapy (41). Here, weshow that DMAPT, a parthenolide prodrug, sensitized prostatecancer cells to radiotherapy in vivo and protected normalprostate and bladder against radiation-induced tissue injury.These results extend our previous survival studies in prostatecancer cell lines and normal prostate epithelial cells.

ROS, as products of cell metabolism, play a dual role intumorigenesis and tumor suppression. The "two-faced" char-acter of ROS has emerged as a potential source for discoveringanticancer drugs. Redox homeostasis is frequently deregulated

β-Actin

β-Actin

β-Actin

TrX

− + − + − + − + − + − +

PC3 DU145 LNCaP PZ PrEC RWPE-1

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L) − + +

Keap1

TrX

IP TrX IgGA B

C

− + − +

siRNA ctrl siRNA TrX

-SH

Keap1

IgG

− + − +

-S-S-

siRNA ctrl siRNA TrX

Keap1

TrX

− + − +

siRNA ctrl siRNA TrX

PGAM5

Nrf2

Keap1

− + − +

Vector Keap1

Bcl-xL

Ikkα

IkBα

0

20

40

60

80

100

120

Untreated 6Gy PN+6Gy

VectorKeap1

*

*

**

*P < 0.05

Cel

l sur

viva

l (%

)

D

Figure 5. TrX-dependent Keap1 reduction by parthenolide in prostate cancer cells. A, the levels of TrX in normal and cancer cells before and after treatmentwith parthenolide (PN) were detected by Western blots. B, after the indicated treatment, Keap1 was immunoprecipitated using a TrX antibody. C, PC3cells were transfectedwith TrX siRNA before the indicated treatment (top). SH- and S-S bands in Keap1 protein were detected as described in Fig. 3 (bottom).D, a Keap1 cDNA construct was transfected into PC3 cells. The levels of related proteins were detected by Western blots (bottom). The effect of Keap1 onradiosensitivity was analyzed using a Trypan blue exclusion assay (top).

Xu et al.

Cancer Res; 73(14) July 15, 2013 Cancer ResearchOF8

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 9: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

in cancers, as it is constantly exposed to high levels of ROScompared with normal counterparts. Our data show thatconstitutively elevated levels of oxidative stress in cancer cellsrepresent a specific vulnerability that can be selectively tar-geted by direct- or indirect-acting prooxidants and antioxi-dants or redoxmodulators. Theoretically, the differential redoxstatus of cancer cells compared with normal cells shouldprovide a therapeutic window for selective redox interventionvia additional increases in ROS. In this context, normal andcancer cells should respond differently to the same level ofprooxidant action generated either by direct production ofoxidative species or by modulation of specific cellular targetsinvolved in redox regulation. In this study, we show thatparthenolide serves as a prooxidant and displays a selectiveredox modification capability that differentially modulatescellular redox signals and targets. The Michael acceptor reactswith a thiol group of target proteins through covalent adduc-

tion (21). Parthenolide contains electrophilic a-methylene-g-lactone, a bisfunctional Michael acceptor, and displays apotential for bifunctional target alkylation and crosslinking.The present study shows the inverse effects of parthenolide onredoxmodification in cancer cells comparedwith normal cells.Remarkably, observations of the cytotoxic and cytoprotectiveeffects of parthenolide are consistent with its action in themodulation of ROS levels in both cancer and normal cells.Alteration of cellular ROS by parthenolide is attributed tofunctionally up- or downregulating antioxidant enzymes inmitochondria, which consequently regulates mitochondrialrespiration. Parthenolide is able to selectively reduce theactivity of several enzymes involved in oxidative stress removalin cancer cells, which in turn can cause ROS levels to rise abovethe threshold for cell death. This finding predicts that antiox-idant proteins and mitochondria are feasible therapeutictargets.

A

Keap1

Bcl-xL

Nrf2

Keap1

PGAM5

− +

LC3B1

LC3B2

IP: PGAM5

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PN (5 μmol/L)

PGAM5

Bcl-xL

IgG

− ++

IgG

B

− + − + − +

12 h 24 h 48 h

Keap1

Vecto

r

Bcl-xL

PGAM5

BcL-xL

β-Actin

β-Actin β-Actin

β-Actin

CC

ell s

urvi

val (

%)

P < 0.01

P < 0.01

0

20

40

60

80

100

120

Ctrl siRNA PGAM5 siRNA Bcl-xL siRNA Nrf2 siRNA

Ctrl si

RNA

Who

le

Cytoso

l

Mito

chon

dria

PGAM5

siRNA

Bcl-xL

siRNA

Nrf2 si

RNA

DMSOPN

P < 0.01

P < 0.01

Nrf2

PGAM5

Bcl-xL

− + − + − + − +

− + − + − +

D

E

− + − +

Bcl-xL

0

20

40

60

80

100

120

Vector Bcl-xL

DMSO

PN P < 0.01

Cel

l sur

viva

l (%

)

Fβ-Actin

LC3B1LC3B2

PGAM5

Keap1

Nrf2

Ikkα

Hsp75

Bcl-xL

Figure 6. Degradation of PGAM5-Bcl-xL caused by parthenolide-mediated reduction of Keap1 in prostate cancer cells. A, PC3 cells were treated withparthenolide (PN). Keap1 and Bcl-xL were immunoprecipitated using a PGAM5 antibody. B, the total levels of Nrf2, PGAM5, and Bcl-xL werequantified by Western blots. C, the levels of mitochondria- associated proteins after parthenolide treatments. D, proteins in various cellular fractionswere identified with antibody specific for each protein. E, PC3 cells were transfected with siRNA to knockdown PGAM5, Bcl-xL, and Nrf2, respectively(bottom). Cell survival fraction was quantified by Trypan blue exclusion assay (top). F, a Bcl-xL expression construct was transfected into PC3 cells and theexpression of Bcl-xL was monitored by Western blots (bottom). Cell survival fraction was quantified by Trypan blue exclusion assay (top).

Keap1, a Central Regulator of Cellular Redox Signaling

www.aacrjournals.org Cancer Res; 73(14) July 15, 2013 OF9

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 10: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

It has been reported that parthenolide is a potent inhibitor ofNF-kB, which is a ROS-responsive transcriptional factorinvolved in both tumor progression and tumor resistance totreatment through upregulation of antiapoptotic genes, suchas Bcl-2, Bcl-xL, survivin, and XIAP (42). We previously showedthat NADPH oxidase-mediated inactivation of the Foxo 3signaling pathway is involved in the parthenolide-enhancedradiosensitivity of prostate cancer (25). However, previousstudies did not explain how parthenolide exerts such anopposing effect in tumor and normal cells. The present studyidentifies Keap1 as a redox signaling sensor that plays a pivotalrole in the differential regulation of the downstream signalingtargets in response to radiation-mediated cytotoxicity in pros-tate cancer and normal cells. Keap1, an adaptor protein forubiquitin-based processing by the CUL3/RBX1-dependent E3ubiquitin ligase complex, functions as a sensor for thiol-reac-tive redox modification (43). The present study shows thatstabilization of Nrf2 by oxidation of Keap1 serves as a majormechanism by which parthenolide protects normal tissuesagainst radiotoxicity through upregulation of antioxidantenzymes in mitochondria. However, Nrf2 transcriptional acti-vation did not play a major role in parthenolide-treatedprostate cancer cells. Thus, it is interesting to note that unliketraditional chemotherapeutic agents, parthenolide is unable toenhance resistance of prostate cancer to radiation treatmentby stimulating Nrf2 target genes.

In addition to regulating the Nrf2 signaling pathway, Keap1is able to bind other proteins such as p62 and PGAM5 (44).Interaction between Keap1 and p62 facilitates release of Nrf2from the complex, which is considered to be a noncanonicalcysteine-independent mechanism for the autophagy deficien-

cy–activated Nrf2 pathway (45). The N-terminus of PGAM5interacts with the Kelch domain of Keap1 and its C-terminusbinds to Bcl-xL. Keap1-dependent ubiquitination results inproteasome-dependent degradation of PGAM5 and Bcl-xL(38). Bcl-xL, an important member of the Bcl-2 family, is apotent antiapoptotic factor that plays a crucial role in cellsurvival by maintaining the electrochemical and osmotichomeostasis of mitochondria (46). The present study showsthat parthenolide increases the level of reduced Keap1 andconsequently induces Keap1-dependent degradation ofPGAM5 and Bcl-xL in cancer cells, suggesting that formationof the Keap1-PGAM5-Bcl-xL complex is a mechanism under-lying the effect of parthenolide on radiosensitization of pros-tate cancer cells.

Although a high rate of aerobic glycolysis in tumors, knownas the Warburg effect, has been observed in various types ofcancer, cancers have functional mitochondria, and mitochon-drial respiration is necessary for cancer cell proliferation (47).Cancer cells depend on a hyperactive metabolism to fuel theirrapid growth and also on antioxidative enzymes to quenchpotentially toxic ROS generated by such a high metabolicdemand (48). Our results show that parthenolide not onlysuppresses MnSOD and GpX, 2 major antioxidant enzymes inmitochondria, but also activates Bcl-xL degradation in cancercells, which suggests thatmitochondria are a feasible target foranticancer treatment. The present study also shows thatparthenolidemaymaintain normal cell survival through induc-tion ofMnSODandGpXactivity. Thus, amore efficient and safetherapymay involvemodification of cellular redox signaling byalteration of the antioxidant response coupled to selectivedegradation of prosurvival members of the Bcl2 family in

Cell defenseProtection

Cytosol

Parthenolide

Normal cells

Radiation

Cancer cells

Nucleus

Antioxidants

Degradation

Cell death

red

red

ox

Figure 7. A proposed mechanisticmodel for parthenolide-mediatedinverse therapeutic effects onradiosensitivity of prostate cancerand radioresistance of normalcells. Parthenolide sensitizescancer cells to radiation, in part, bymaintaining Keap1 in a reducedstate and enhancing its interactionwith PGAM5 and Bcl-xL, resultingin degradation of Bcl-xL inmitochondria. In contrast,parthenolide protects normal cellsagainst radiation via oxidation ofKeap1 and release of the Nrf2transcription factor for activationofmitochondrial antioxidantenzymes.

Xu et al.

Cancer Res; 73(14) July 15, 2013 Cancer ResearchOF10

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 11: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

cancer cells, as conventional anticancer therapy mainly causescell growth arrest or cell death by raising cellular ROS, whichoxidizes and damages DNA, proteins, and lipids. Optimizingprototype redox chemotherapeutics from natural sources pro-vides an exciting opportunity to further develop even bettercandidates to enhance therapeutic efficacy with less off-targettoxicity.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: Y. Xu, P.A. Crooks, D.K. St. Clair, W.H. St. ClairDevelopment of methodology: Y. Xu, F. Fang, S. MiriyalaAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): Y. Xu, F. Fang, L. Chaiswing, A.K. Holley, Y. Zhao, K.K.Kiningham, D.K. St. Clair, W.H. St. ClairAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y. Xu, F. Fang, S. Miriyala, P.A. Crooks, T.D. Oberley,D.K. St. Clair, W.H. St. Clair

Writing, review, and/or revision of the manuscript: Y. Xu, S. Miriyala, P.A.Crooks, D.K. St. Clair, W.H. St. ClairAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): Y. Xu, T. NoelStudy supervision: Y. Xu, D.K. St. Clair, W.H. St. Clair

AcknowledgmentsThe authors thank Yulan Sun, a previous graduate student, who initiated the

work.

Grant SupportThis work was supported by NIH grants CA49797, CA115801, and CA143428

(to D.K. St. Clair and William St. Clair). Additional support was provided by theEdward P. Evans Foundation and by the resources and facilities of the WilliamS. Middleton Veterans Administration Hospital (Madison, WI).

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

Received November 27, 2012; revised March 18, 2013; accepted April 5, 2013;published OnlineFirst May 14, 2013.

References1. Irani K, Xia Y, Zweier JL, Sollott SJ, Der CJ, Fearon ER, et al. Mitogenic

signaling mediated by oxidants in Ras-transformed fibroblasts. Sci-ence 1997;275:1649–52.

2. Rhee SG. Cell signaling. H2O2, a necessary evil for cell signaling.Science 2006;312:1882–83.

3. Pelicano H, Carney D, Huang P. ROS stress in cancer cells andtherapeutic implications. Drug Resist Updat 2004;7:97–110.

4. TewKD, TownsendDM.Redoxplatforms in cancer drugdiscovery anddevelopment. Curr Opin Chem Biol 2011;15:156–61.

5. D'Autreaux B, Toledano MB. ROS as signalling molecules: Mechan-isms that generate specificity in ROS homeostasis. Nat Rev Mol CellBiol 2007;8:813–24.

6. Xia C, Meng Q, Liu LZ, Rojanasakul Y, Rojanasakul Y, Wang XR, et al.Reactive oxygen species regulate angiogenesis and tumor growththrough vascular endothelial growth factor. Cancer Res 2007;67:10823–30.

7. Dr€oge W. Free radicals in the physiological control of cell function.Physiol Rev 2002;82:47–95.

8. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Freeradicals and antioxidants in normal physiological functions and humandisease. Int J Biochem Cell Biol 2007;39:44–84.

9. Cabello CM, Bair WB III, Wondrak GT. Experimental therapeutics:targeting the redox Achilles heel of cancer. Curr Opin Investig Drugs2007;8:1022–37.

10. Trachootham D, Lu W, Ogasawara MA, Nilsa RD, Huang P. Redoxregulation of cell survival. Antioxid Redox Signal 2008;10:1343–74.

11. Wang J, Yi J. Cancer cell killing via ROS to increase or decrease, that isthe question. Cancer Biol Ther 2008;7:1875–84.

12. Spyratou E, Makropoulou M, Mourelatou EA, Demetzos C. Biopho-tonic techniques for manipulation and characterization of drugdelivery nanosystems in cancer therapy. Cancer Lett 2012;327:111–22.

13. Oberley L, Oberley TD. Role of antioxidant enzymes in cell immor-talization and transformation. Mol Cell Biochem 1988;84:147–53.

14. ZhaoY, Chaiswing L, Oberley TD, Batinic-Haberle I, St ClairW, EpsteinCJ, et al. A mechanism-based antioxidant approach for the reductionof skin carcinogenesis. Cancer Res 2005;65:1401–5.

15. Dhar SK, Tangpong J, Chaiswing L, Oberley TD, St Clair DK. Manga-nese superoxide dismutase is a p53-regulated gene that switchescancers between early and advanced stages. Cancer Res 2011;71:6684–95.

16. Josson S, Xu Y, Fang F, Dhar SK, St Clair DK, St Clair WH. RelBregulates manganese superoxide dismutase gene and resistance toionizing radiation of prostate cancer cells. Oncogene 2006;25:1554–59.

17. Fan M, Ahmed KM, Coleman MC, Spitz DR, Li JJ. Nuclear factor-kappaB and manganese superoxide dismutase mediate adaptiveradioresistance in low-dose irradiated mouse skin epithelial cells.Cancer Res 2007;67:3220–28.

18. Homma S, Ishii Y, Morishima Y, Yamadori T, Matsuno Y, Haraguchi N,et al. Nrf2 enhances cell proliferation and resistance to anticancerdrugs in human lung cancer. Clin Cancer Res 2009;15:3423–32.

19. Hassane DC, Sen S, Minhajuddin M, Rossi RM, Corbett CA, Balys M,et al. Chemical genomic screening reveals synergism between parthe-nolide and inhibitors of the PI-3 kinase and mTOR pathways. Blood2010;116:5983–90.

20. Raj L, Ide T, Gurkar AU, Foley M, Schenone M, Li X, et al. Selectivekilling of cancer cells by a small molecule targeting the stress responseto ROS. Nature 2011;475:231–34.

21. Hwang DR, Wu YS, Chang CW, Chang CW, Lien TW, Chen WC, et al.Synthesis and anti-viral activity of a series of sesquiterpene lactonesand analogues in the subgenomic HCV replicon system. Bioorg MedChem 2006;14:83–91.

22. Pajak B, Gajkowska B, Orzechowski A. Molecular basis of partheno-lide-dependent proapoptotic activity in cancer cells. Folia HistochemCytobiol 2008;46:129–35.

23. Gill KK, Kaddoumi A, Nazzal S. Mixed micelles of PEG(2000)-DSPEand vitamin-E TPGS for concurrent delivery of paclitaxel and parthe-nolide: enhanced chemosensitization and antitumor efficacy againstnon-small cell lung cancer (NSCLC) cell lines. Eur J Pharm Sci2012;46:64–71.

24. Sun Y, St Clair DK, Fang F, Warren GW, Rangnekar VM, Crooks PA,et al. The radiosensitization effect of parthenolide in prostate cancercells is mediated by nuclear factor-kappaB inhibition and enhanced bythe presence of PTEN. Mol Cancer Ther 2007;9:2477–86.

25. Sun Y, St Clair DK, Xu Y, Crooks PA, St Clair WH. A NADPH oxidase-dependent redox signaling pathway mediates the selective radiosen-sitization effect of parthenolide in prostate cancer cells. Cancer Res2010;70:2880–90.

26. Kishida Y, Yoshikawa H, Myoui A. Parthenolide, a natural inhibitor ofNuclear Factor-kappaB, inhibits lung colonization of murine osteosar-coma cells. Clin Cancer Res 2007;13:59–67.

27. Salmeen A, Andersen JN, Myers MP, Meng TC, Hinks JA, Tonks NK,et al. Redox regulation of protein tyrosine phosphatase 1B involves asulphenyl-amide intermediate. Nature 2003;423:769–73.

28. Rasmussen HH, Hamilton EJ, Liu CC, Figtree GA. Reversible oxidativemodification: implications for cardiovascular physiology and patho-physiology. Trends Cardiovasc Med 2010;20:85–90.

29. Wakabayashi N, Dinkova-Kostova AT, Holtzclaw WD, Kang MI,Kobayashi A, Yamamoto M, et al. Protection against electrophile and

Keap1, a Central Regulator of Cellular Redox Signaling

www.aacrjournals.org Cancer Res; 73(14) July 15, 2013 OF11

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 12: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

oxidant stress by induction of the phase 2 response: fate of cysteinesof the Keap1 sensor modified by inducers. Proc Natl Acad Sci USA2004;101:2040–45.

30. Adam J, Hatipoglu E, O'Flaherty L, Ternette N, Sahgal N, Lockstone H,et al. Renal cyst formation in Fh1-deficient mice is independent of theHif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2signaling. Cancer Cell 2011;20:524–37.

31. Neelakantan S, Nasim S, Guzman ML, Jordan CT, Crooks PA. Ami-noparthenolides as novel anti-leukemic agents: discovery of the NF-kappaB inhibitor, DMAPT (LC-1). Bioorg Med Chem Lett 2009;19:4346–9.

32. Xu Y, Fang F, St Clair DK, Josson S, Sompol P, Spasojevic I, et al.Suppression of RelB-mediated manganese superoxide dismutaseexpression reveals a primary mechanism for radiosensitization effectof 1alpha, 25-dihydroxyvitamin D(3) in prostate cancer cells. MolCancer Ther 2007;6:2048–56.

33. Xu Y, Josson S, Fang F, St Clair DK, Wan XS, Sun Y, et al. RelBenhances prostate cancer growth: implications for the role of the NF-kB alternative pathway in tumorigenicity. Cancer Res 2009;69:3267–71.

34. Slane BG, Aykin-Burns N, Smith BJ, Kalen AL, Goswami PC, DomannFE, et al. Mutation of succinate dehydrogenase subunit C results inincreased O2

.�, oxidative stress, and genomic instability. Cancer Res2006;66:7615–20.

35. Bayer EA, Safars M, Wilchek M. Selective labeling of sulfhydryls anddisulfides on blot transfers using avidin-biotin technology: studies onpurified proteins and erythrocyte membranes. Anal Biochem1987;161:262–71.

36. Keller JN, Kindy MS, Holtsberg FW, St Clair DK, Yen HC, Germeyer A,et al., Mitochondrial manganese superoxide dismutase prevents neu-ral apoptosis and reduces ischemic brain injury: suppression of per-oxynitrite production, lipid peroxidation, and mitochondrial dysfunc-tion. J Neurosci 1998;18:687–97.

37. Wu RP, Hayashi T, Cottam HB, Jin G, Yao S, Wu CC, et al. Nrf2responses and the therapeutic selectivity of electrophilic compounds

in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2010;107:7479–84.

38. Lo SC, Hannink M. PGAM5, a Bcl-xL-interacting protein, is a novelsubstrate for the redox-regulated Keap1-dependent ubiquitin ligasecomplex. J Biol Chem 2006;281:37893–903.

39. Niture SK, Jaiswal AK. Inhibitor of Nrf2 (INrf2 or Keap1) proteindegrades Bcl-xL via phosphoglycerate mutase 5 and controls cellularapoptosis. J Biol Chem 2011;286:44542–56.

40. Sohma I, Fujiwara Y, Sugita Y, Yoshioka A, Shirakawa M, Moon JH,et al. Parthenolide, an NF-kB inhibitor, suppresses tumor growth andenhances response to chemotherapy in gastric cancer. Cancer Geno-mics Proteomics 2011;8:39–47.

41. Zuch D, Giang AH, Shapovalov Y, Schwarz E, Rosier R, O'Keefe R,et al. Targeting radioresistant osteosarcoma cells with parthenolide.J Cell Biochem 2012;113:1282–91.

42. Montero AJ, Jassem J.Cellular redox pathways as a therapeutic targetin the treatment of cancer. Drugs 2011;71:1385–96.

43. SekharKR,RachakondaG,FreemanML.Cysteine-based regulationofthe CUL3 adaptor protein Keap1. Toxicol Appl Pharmacol 2010;244:21–26.

44. Lau A, Wang XJ, Zhao F, Villeneuve NF, Wu T, Jiang T, et al. Anoncanonical mechanism of Nrf2 activation by autophagy deficiency:direct interaction between Keap1 and p62. Mol Cell Biol 2010;30:3275–85.

45. Rusten TE, Stenmark H. p62, an autophagy hero or culprit? Nat CellBiol 2010;12:207–9.

46. Vander Heiden MG, Chandel NS, Williamson EK, Schumacker PT,Thompson CB. Bcl-xL regulates the membrane potential and volumehomeostasis of mitochondria. Cell 1997;91:627–37.

47. Weinberg F, Chandel NS. Mitochondrial metabolism and cancer. AnnNY Acad Sci 2009;1177:66–73.

48. Pavlides S, Vera I, Gandara R, Sneddon S, Pestell RG, Mercier I, et al.Warburg meets autophagy: cancer-associated fibroblasts acceleratetumor growth and metastasis via oxidative stress, mitophagy, andaerobic glycolysis. Antioxid Redox Signal 2012;16:1264–84.

Xu et al.

Cancer Res; 73(14) July 15, 2013 Cancer ResearchOF12

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297

Page 13: KEAP1 Is a Redox Sensitive Target That Arbitrates the ......2013/07/05  · Therapeutics, Targets, and Chemical Biology KEAP1 Is a Redox Sensitive Target That Arbitrates the Opposing

Published OnlineFirst May 14, 2013.Cancer Res   Yong Xu, Fang Fang, Sumitra Miriyala, et al.   and Cancer CellsOpposing Radiosensitive Effects of Parthenolide in Normal KEAP1 Is a Redox Sensitive Target That Arbitrates the

  Updated version

  10.1158/0008-5472.CAN-12-4297doi:

Access the most recent version of this article at:

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. (CCC)Click on "Request Permissions" which will take you to the Copyright Clearance Center's

.http://cancerres.aacrjournals.org/content/early/2013/07/05/0008-5472.CAN-12-4297To request permission to re-use all or part of this article, use this link

Research. on April 15, 2021. © 2013 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 14, 2013; DOI: 10.1158/0008-5472.CAN-12-4297