carbon monoxide expedites metabolic exhaustion to inhibit ... · bioenergetics, ultimately...

14
Therapeutics, Targets, and Chemical Biology Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit Tumor Growth Barbara Wegiel 1 , David Gallo 1 , Eva Csizmadia 1 , Clair Harris 1 , John Belcher 5 , Gregory M. Vercellotti 5 , Nuno Penacho 6 , Pankaj Seth 2 , Vikas Sukhatme 2 , Asif Ahmed 4 , Pier Paolo Pandol2 , Leszek Helczynski 3 , Anders Bjartell 3 , Jenny Liao Persson 3 , and Leo E. Otterbein 1 Abstract One classical feature of cancer cells is their metabolic acquisition of a highly glycolytic phenotype. Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. However, the functional contributions of CO and HO-1 to these processes are poorly dened. In human prostate cancers, we found that HO-1 was nuclear localized in malignant cells, with low enzymatic activity in moderately differentiated tumors correlating with relatively worse clinical outcomes. Exposure to CO sensitized prostate cancer cells but not normal cells to chemotherapy, with growth arrest and apoptosis induced in vivo in part through mitotic catastrophe. CO targeted mitochondria activity in cancer cells as evidenced by higher oxygen consumption, free radical generation, and mitochondrial collapse. Collectively, our ndings indicated that CO transiently induces an anti-Warburg effect by rapidly fueling cancer cell bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 700921. Ó2013 AACR. Introduction Epithelial cancers, including prostate, breast, and lung can- cer, are still leading causes of deaths in the United States and treatment for advanced disease is limited (1). A standard of rst-line care for advanced and metastatic cancers remains chemotherapy such as taxols, doxorubicin, and cisplatin (2). Rapid proliferation of primary tumor and cancer cell survival during spread to distant organs as well as resistance to treatment are possible in part due to the remarkable metabolic adaptation known as the Warburg effect (3). The Warburg effect is characterized by increased glucose uptake and ele- vated glycolysis with a limited oxygen consumption rate (OCR) resulting in lactic acid fermentation (4). High rates of energy consuming processes including protein, DNA, and fatty acid synthesis in cancer cells is often accompanied by an increased oxidative state of dysfunctional mitochondria (5). The promo- tion of tumor growth requires, in part, a selection of cancer cells with repressed mitochondrial activity and biogenesis (6). Defects in mitochondrial ROS metabolism from electron trans- port chains in cancer cells have been linked directly to increased cancer cell glucose metabolism. The free radical theory of cancer implicates ROS as a principal cause of early mutations as well as being involved in the response to treat- ment (711). Heme oxygenases (HO), which degrade heme to biliverdin, carbon monoxide (CO), and iron, are critical modulators of metabolism and mitochondrial activity. Expression of HO-1, the stress-inducible isoform, is strictly regulated, whereas HO- 2 is ubiquitously expressed primarily in brain and testes. Their functional role in cancer has not been clearly elucidated and remains controversial. HO-1 can impart potent antiprolifera- tive and proapoptotic effects via antioxidant mechanisms as shown in breast and lung cancer cell lines (12, 13). Better survival rates were observed in patients with colorectal cancer where HO-1 expression correlated with lower rates of lym- phatic tumor invasion. In contrast, overexpression of HO-1 has been shown to accelerate pancreatic cancer aggressiveness by increasing tumor growth, angiogenesis, and metastasis (14). Similar effects were observed in melanoma (15), gastric (16), and renal cancers (17). In patients with prostate cancer, HO-1 is localized in the nucleus and correlated with cancer progression (18). Nuclear HO-1 was also detected in head and neck squa- mous carcinomas and associated with tumor progression (19). Recently, nuclear HO-1 has been linked to resistance to ima- tinib in chronic myeloid leukemia (20). Further evidence for HO-1 in cancer incidence presides in the identication of a GT length polymorphism of the HO-1 promoter that is highly correlative with cancer severity (21). Individuals with long GT repeats in the HO-1 promoter and associated low expression of Authors' Afliations: 1 Department of Surgery, Transplant Institute, 2 Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; 3 Department of Clinical Sciences, Section of Urological Cancers, 4 School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, United Kingdom; 5 Department of Laboratory Medicine, University Hospital Malmo, Lund University, Malmo, Sweden; and 6 Department of Medicine and Vascular Biology Center, University of Minnesota, Minneapolis, Alfama Inc., Oeiras, Portugal Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Authors: Leo E. Otterbein, Harvard Medical School, Beth Israel Deaconess Medical Center, Transplant Institute, 3 Blackfan Circle, EC/CLS 603, Boston, MA 02215. Phone: 617-735-2851; Fax: 617-735- 2844; E-mail: [email protected]; and Barbara Wegiel. Phone: 617-735-2846; Email: [email protected] doi: 10.1158/0008-5472.CAN-13-1075 Ó2013 American Association for Cancer Research. Cancer Research www.aacrjournals.org 7009 on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Upload: others

Post on 23-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

Therapeutics, Targets, and Chemical Biology

Carbon Monoxide Expedites Metabolic Exhaustion to InhibitTumor Growth

Barbara Wegiel1, David Gallo1, Eva Csizmadia1, Clair Harris1, John Belcher5, Gregory M. Vercellotti5,Nuno Penacho6, Pankaj Seth2, Vikas Sukhatme2, Asif Ahmed4, Pier Paolo Pandolfi2, Leszek Helczynski3,Anders Bjartell3, Jenny Liao Persson3, and Leo E. Otterbein1

AbstractOne classical feature of cancer cells is their metabolic acquisition of a highly glycolytic phenotype. Carbon

monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, hasbeen implicated incarcinogenesis and therapeutic resistance.However, the functional contributions ofCOandHO-1to these processes are poorly defined. In human prostate cancers, we found that HO-1 was nuclear localized inmalignant cells, with low enzymatic activity in moderately differentiated tumors correlating with relatively worseclinical outcomes. Exposure to CO sensitized prostate cancer cells but not normal cells to chemotherapy, withgrowth arrest and apoptosis induced in vivo in part throughmitotic catastrophe. CO targetedmitochondria activityin cancer cells as evidenced by higher oxygen consumption, free radical generation, and mitochondrial collapse.Collectively, our findings indicated that CO transiently induces an anti-Warburg effect by rapidly fueling cancer cellbioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. �2013 AACR.

IntroductionEpithelial cancers, including prostate, breast, and lung can-

cer, are still leading causes of deaths in the United States andtreatment for advanced disease is limited (1). A standard offirst-line care for advanced and metastatic cancers remainschemotherapy such as taxols, doxorubicin, and cisplatin (2).Rapid proliferation of primary tumor and cancer cell survivalduring spread to distant organs as well as resistance totreatment are possible in part due to the remarkable metabolicadaptation known as the Warburg effect (3). The Warburgeffect is characterized by increased glucose uptake and ele-vated glycolysis with a limited oxygen consumption rate (OCR)resulting in lactic acid fermentation (4). High rates of energyconsuming processes including protein, DNA, and fatty acidsynthesis in cancer cells is often accompanied by an increasedoxidative state of dysfunctional mitochondria (5). The promo-

tion of tumor growth requires, in part, a selection of cancercells with repressed mitochondrial activity and biogenesis (6).Defects inmitochondrial ROSmetabolism from electron trans-port chains in cancer cells have been linked directly toincreased cancer cell glucose metabolism. The free radicaltheory of cancer implicates ROS as a principal cause of earlymutations as well as being involved in the response to treat-ment (7–11).

Heme oxygenases (HO), which degrade heme to biliverdin,carbon monoxide (CO), and iron, are critical modulators ofmetabolism and mitochondrial activity. Expression of HO-1,the stress-inducible isoform, is strictly regulated, whereas HO-2 is ubiquitously expressed primarily in brain and testes. Theirfunctional role in cancer has not been clearly elucidated andremains controversial. HO-1 can impart potent antiprolifera-tive and proapoptotic effects via antioxidant mechanisms asshown in breast and lung cancer cell lines (12, 13). Bettersurvival rates were observed in patients with colorectal cancerwhere HO-1 expression correlated with lower rates of lym-phatic tumor invasion. In contrast, overexpression of HO-1has been shown to accelerate pancreatic cancer aggressivenessby increasing tumor growth, angiogenesis, andmetastasis (14).Similar effects were observed in melanoma (15), gastric (16),and renal cancers (17). In patientswith prostate cancer, HO-1 islocalized in the nucleus and correlatedwith cancer progression(18). Nuclear HO-1 was also detected in head and neck squa-mous carcinomas and associated with tumor progression (19).Recently, nuclear HO-1 has been linked to resistance to ima-tinib in chronic myeloid leukemia (20). Further evidence forHO-1 in cancer incidence presides in the identification of a GTlength polymorphism of the HO-1 promoter that is highlycorrelative with cancer severity (21). Individuals with long GTrepeats in the HO-1 promoter and associated low expression of

Authors' Affiliations: 1Department of Surgery, Transplant Institute,2Department of Medicine, Beth Israel Deaconess Medical Center, HarvardMedical School, Boston, Massachusetts; 3Department of ClinicalSciences, Section of Urological Cancers, 4School of Life and HealthSciences, Aston University, Aston Triangle, Birmingham, United Kingdom;5Department of Laboratory Medicine, University Hospital Malm€o, LundUniversity, Malm€o, Sweden; and 6Department of Medicine and VascularBiology Center, University of Minnesota, Minneapolis, Alfama Inc., Oeiras,Portugal

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

Corresponding Authors: Leo E. Otterbein, Harvard Medical School, BethIsrael Deaconess Medical Center, Transplant Institute, 3 Blackfan Circle,EC/CLS 603, Boston, MA 02215. Phone: 617-735-2851; Fax: 617-735-2844; E-mail: [email protected]; and Barbara Wegiel. Phone:617-735-2846; Email: [email protected]

doi: 10.1158/0008-5472.CAN-13-1075

�2013 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 7009

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 2: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

HO-1 showed a higher frequency of gastric or lung adenocar-cinoma and oral squamous cancer versus those with short GTrepeats and higher HO-1 expression (22). CO, biliverdin, bili-rubin as well as iron and ferritin serve as potential modulatorsof tumorigenesis however all have been minimally studied incancer (23).

In the present studies, we first conducted a detailed analysisof a large cohort of patients with prostate cancer and con-firmed HO-1 nuclear localization in moderately advancedtumors where it is enzymatically inactive and therefore maybe a critical regulator of cancer progression. We tested thehypothesis that HO-1, through its ability to generate CO,modulates cancer cell growth in vitro and in vivo using humanand murine prostate and lung cancer models. Paradoxically,CO rapidly enhancedmitochondria activity of cancer cells thatresults in metabolic exhaustion and cellular collapse causingtumor regression. Furthermore, CO increased cancer cell sen-sitivity to chemotherapeutics 1,000-fold while simultaneouslyprotecting normal cell growth and viability.

Materials and MethodsProstate cancer samples and tissue microarray

Benign and malignant samples of 482 patients undergoingradical prostatectomy for localized prostate cancer weresubjected in duplicate to tissue microarray (TMA) constructsof 1.0 mm in diameter and scored for immunohistochemicalstaining intensity as previously described (24). The majority ofsamples were successfully prepared (�95%), and Gleasongrades were evaluated by a National Board–certified pathol-ogist (L. Helczynski) in the prostate cancer specimens from351 before preparation of TMA. The group of samples con-sisted of 246 samples with Gleason grade 3 and 105 sampleswith Gleason grades 4 and 5. The study was approved by theEthics Committee, Lund University and the Helsinki Decla-ration of Human Rights was strictly observed.

ImmunohistochemistryImmunohistochemical staining of paraffin-embedded sec-

tions from human TMA blocks was conducted as previouslydescribed (25).Mouse antibody againstHO-1 (Stressgen, #OSA-110) was used at 1:200 dilution. Secondary antibody was usedas a negative control. Immunohistochemistry, immunofluo-rescence, and immunostaining on the cell lines and mousetumor samples were conducted as previously described (26).Briefly, formalin-fixed or Zn-fixed paraffin-embedded tumortissues were processed for antigen retrieval with high-pressurecooking in citrate buffer for 1 hour. Tissues sections were thenblocked with 7% horse serum for 30 minutes, followed byincubation with primary antibody overnight. Biotin-labeledsecondary antibody (Vector Laboratories) was applied for 1hour at room temperature followed byVectastain Elite ABCKitand detection with ImmPact DAB (Vector Laboratories).Immunofluorescence staining was conducted on frozen tumorsections or cultured cells on glass coverslips. Tissues or cellswere fixed with 2% paraformaldehyde for 10 minutes followedby permeabilization with 0.05% Triton X-100. After blockingwith 7% horse serum, sections were probed with primary

antibody and secondaryfluorescently labeled antibody (Molec-ular Probes). Hoechst-33258 (Molecular Probes, Invitrogen)was used to stain the nuclei.

The following antibodies were applied: polyclonal rabbitanti-HO-1 (Stressgen) was used at 1:1,000. Prohibitin antibodywas purchased from Epitomics Inc. Cytochrome c antibodywas obtained from Cell Signaling. mtTFA and vimentin anti-bodies were purchased from Santa Cruz Biotechnology. Anti-mouseKi-67 antibodywas fromDako. Anti-mouse Thy-1, CD31antibody was from BD Biosciences, Pharmingen and anti-P(Ser10)-histone H3 was from Cell Signaling. Terminal deox-ynucleotidyl transferase–mediated dUTP nick end labeling(TUNEL) staining was conducted using ApopTag PeroxidaseIn Situ Apoptosis Detection Kit (Chemicon, Millipore) follow-ing manufacturer's protocol.

Animal tumor modelsThe tumor models were approved by the BIDMC IACUC.

Nu/nu mice were purchased from Taconic at 7 weeks of age.Mice were provided food and water ad libitum as needed. After1-week acclimatization, 1� 106 PC3 cells were injected into theright flank of mice. Tumors were established for 2 weeks, atwhich point treatmentwithCO�doxorubicinwas begun.Micewere exposed to CO (250 ppm, 1 h/d) as previously described(27). Doxorubicin (8 mg/kg) was given i.v. twice per week justbefore CO/air exposure. Tumor volume (0.52�width� length)was measured and calculated every day for 14 days. After 14days, mice were sacrificed and tumors were harvested. In theorthotopic model of PC3 xenografts, 1 � 106 PC3 cells wereinjected to the right lobe of prostate under anesthesia. Tumors(n ¼ 4 per group) were established for 2 weeks followed byCO/air treatment for 4 weeks. CO was administrated every dayfor 1 hour at 250 ppm. TRAMP mice (Jackson Laboratories)were treated with CO (250 ppm/d, 5 d/wk) starting at 5 weeksof age. Treatment was continued for 20 weeks. Tumors wereharvested and processed for immunohistochemical analysisthereafter. Kras4bG12D transgenic mice were kindly providedby Dr. Varmus and were on doxycycline chow at 5 weeks ofage and COwas started after 8 weeks of doxycycline treatment.CO (250 ppm, 5 d/wk) was continued for 5 weeks and thelungs were harvested for histology.

Cell culture and treatmentThe human prostatic cell line, PC3, human breast cancer

cells (MDA-468 andT47D), hepatocarcinoma (HepG2), cervicalcancer cells (HeLa), skin fibroblasts (NIH3T3), and lung ade-nocarcinoma cell line (A549) were purchased from AmericanType Culture Collection or provided by Dr. Balk and Dr. Chen(BIDMC) and were cultured in RPMI or Dulbecco's ModifiedEagle Medium (DMEM) supplemented with 10% FBS andantibiotics. Human bronchial epithelial cells were purchasedfrom American Type Culture Collection (ATCC) and culturedfollowing manufacturer's protocol. PNT1A were kindly giftedby Dr. Nishtman Dizeyi (Lund University, Malmo, Sweden) andwere cultured in RPMI medium supplemented with 10% FBSand antibiotics. A549 rho(o) cells that are deprived of mito-chondria were generated after 3 weeks of culture in mediumcontaining ethidium bromide (20 mg/mL). Mouse embryonic

Wegiel et al.

Cancer Res; 73(23) December 1, 2013 Cancer Research7010

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 3: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

fibroblasts from p53þ/þ and p53�/� mice were kindly giftedby Dr. Tyler Jacks (MIT, Boston, MA) and were cultured inDMEM with 10% FBS and antibiotics. Cells were treatedwith camptothecin (0.001–10 mg/mL, Sigma) or doxorubicin(10 mg/mL, Sigma) and vehicle control (dimethyl sulfoxide) for1 to 24 hours. Cotreatment with CO (250 ppm) was conductedas previously described (27). Pegylated catalase (3000 U/mL)and SOD (10 U/mL) were from Sigma and were used 1 hourbefore CO treatment.

Transient transfectionPC3 cells were transfected using Amaxa nucleofection or

Lipofectamine 2000 as previously described (24). pCMV con-trol vector or human HO-1 cDNA encoding vector were usedfor transfection. Transfection efficiency was between 40% and50% as estimated with overexpression of GFP by fluorescencemicroscopy.

Comet assayPC3 cells were treated with CO or air for 24 hours and

harvested for COMET SCGE Assay (Assay Designs). The man-ufacturer's protocol was followed. Cells were viewed by ZeissAxiovert Apotome Fluorescent microscope at �40. At least100 cells were counted, and the amount of comets was calcu-lated and expressed per 100 cells.

ImmunoblottingCells were lysed by brief sonication in ice-cold lysis buffer

[150 mmol/L NaCl, 50 mmol/L Tris-HCl, pH 7.5, 1% NP-40, 10mmol/L NaF, 1% SDS, 1 mmol/L EDTA, pH 8.0, 10 mmol/Lphenylmethylsulfonyl fluoride (PMSF)] and the protease inhib-itor cocktail CompleteMini (Roche). Samples were centrifugedfor 20minutes at 14,000� g at 4�C and clear supernatants werecollected. Protein concentrations were measured using a BCAProtein Assay Kit (Pierce). Twenty to 30 micrograms of eachprotein sample was electrophoresed on a NuPAGE 4%–12%Bis-Tris Gel (Invitrogen) in NuPAGE MES SDS Running Buffer(Invitrogen) followed by transfer to polyvinylidene difluoride(PVDF) membrane (Ready Gel Blotting Sandwiches; BioRad).Themembranes were blocked with 5% nonfat drymilk, probedwith appropriate primary antibodies at concentration of1:1,000 followed by horseradish peroxidase-conjugated sec-ondary antibodies at a dilution of 1:5,000, and visualized usingSuper SignalWest Pico chemiluminescent substrate (Pierce) orFemto Maximum Sensitivity substrate (Pierce), followed byexposure to the autoradiography film (ISC BioExpress). Thefollowing antibodieswere applied: rabbit anti-caspase-3, rabbitanti-cleaved caspase-3 (Cell Signaling Technologies); goat anti-Gadd45 (Santa Cruz Biotechnology); rabbit anti-heme oxyge-nase-1 (RDI-Fitzgerald Industries Int); mouse anti-b-actin(Sigma-Aldrich); mouse anti-GAPDH (Calbiochem); and rabbitanti-cyclin A2 (Santa Cruz Biotechnology).

HO-1 activity and CO levels in the tissuesPC3 cells were treated with 10 mg/mL doxorubicin or camp-

tothecin for 4 hours, and nuclear/cytoplasmic fractions wereisolated. HO-1 activity was measured as previously described(28).We assume that onlyHO-1was contributing to the changes

inHOactivity assay asHO-2 remainedunchanged in response todoxorubicin and camptothecin treatment for 4 hours (data notshown). Measurements of CO concentrations were conductedon tissue sections as described by Verma and colleagues (29).Tumor and normal tissues were harvested immediately afterthe last exposure of mice to CO and tissues were processed forgas chromatography as described. Briefly, CO was liberated asa gas in a sealed glass vial by adding 25 mL of water and 5 mLof sulfosalicylic acid [SSA; 30% (wt/vol)] to 30 mL of dilutedtissue homogenate. The vials were incubated on ice for 10minutes before being analyzed. The gas in the headspace ofthe vials was then analyzed quantitatively with a gas chromato-graph equipped with a reducing-compound photometry detec-tor (Peak Laboratories), which allowed the CO in gas to bequantified to concentrations as low as 1 to 2 parts per billion.The amount of COwas then calculated using a calibration curveprepared from CO standards.

RNA isolation and real-time PCRTotal RNAwas isolated fromPC3 cells after treatmentwith 10

mg/mL camptothecin with or without CO for 6 or 24 hours usingRNeasy Kit (Qiagen, Inc). Two micrograms of RNA was used forcDNA synthesis (Invitrogen). The region of Gadd45awas ampli-fiedwith specific primers: F-50 TGCTCAGCAAAGCCCTGAGT30;R-50GCAGGCACAACACCACGTTA30. The following primers tob-actinwereused: F- 50CGCGAGAGAAGATGACCCAGATC30; R-50TCACCGGAGTCCATCACGA30. Real-time PCR reactions wereconducted using 1�SYBR Green PCR Master Mix (BioRad).Primers for MMP2, urokinase plasminogen activator (uPA), andVEGF were previously described (24).

Apoptosis, viability, and clonogenic assaysPC3 cells were seeded at 1 � 105 concentration in 6-well

plates 24 hours before experiment. Cells were treated withcamptothecin (0.001–10 mg/mL) or doxorubicin (10 mg/mL)with CO/air for 24 hours. Cells were harvested and apoptosisrate was measured using Annexin V-FITC staining accordingto the manufacturer's protocol (BD Biosciences). To test cellviability, PC3 cells were stained with crystal violet solution(Sigma-Aldrich) for 10 minutes, followed by extensive washwith double distilled water. Cells were dried and dissolved in10% acetic acid, followed bymeasurement of absorbance at 560nm on the ELISA reader.

Clonogenic assay. Cells were treated in the presence orabsence of CO � chemotherapeutics as above for 24 hours.After 24 hours, cells were trypsinized and seeded as a subcul-ture with dilution of 1:10 to 1:100. Colony counts were con-ducted 2 weeks after seeding.

Cell fractionationNuclear and cytoplasmic fraction of PC3 cells were con-

ducted using the Subcellular Fractionation Kit following themanufacturer's protocol (Pierce) or BioVision Nuclear/Cyto-plasmic Kit extraction (BioVision). Briefly, cells were treated asdescribed above, washed twice with PBS, and trypsinized.Pellets were collected and resuspended in ice-cold cytoplasmicextraction reagent (CERI) buffer and vortexed. Lysates wereincubated on ice for 10 minutes, and CERII buffer was added

Carbon Monoxide Regulates Cancer Cell Metabolism

www.aacrjournals.org Cancer Res; 73(23) December 1, 2013 7011

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 4: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

and lysates were incubated for additional 1minute on ice. Aftervortexing, lysates were centrifuge at 16,000 � g and super-natants were collected for the cytoplasmic fraction. Pelletswere resuspended in ice-cold nuclear extraction reagent (NER)buffer and incubated on ice for 30 minutes. Nuclear andcytoplasmic fractions were analyzed by immunoblotting asdescribed above.

Mitochondrial activity and metabolic profilingSPECT. We used technetium-99m methoxyisobutyl isoni-

trile (99mTc-sestamibi) single-photon emission computedtomography (SPECT) in PC3 cells treated with CO (250 ppm)or CORM-A1 (20 or 100 mmol/L) for 24 hours in vitro. Tech-netium labeling was conducted at BIDM Imaging Facility, andthe radiation intensity was evaluated that corresponded to themitochondrial activity.

Seahorse experimentsCells were seeded in 24-well plate (3 � 104) 24 hours before

experiment and treated with CO (250 ppm) or air for 30minutes or CO was bubbled into the medium and transferredto the reader immediately prior measurment. OCR and extra-cellular acidification rate (ECAR) were measured over time.

LDH and glucose measurementsPC3 cells were treated for 6 hours with CO (250 ppm) and

harvested in the manufacturer's lysis buffers to measure glu-cose or lactate (Biovision, Glucose or Lactate Kits) and processfor colorimetric analysis of metabolites.

MitoTracker RedCMCRos was purchased from Invitrogen, Molecular Probes

and was used at 0.2 mmol/L concentration. Cells were treatedfor 3.5 hours with doxorubicin (10 ng/mL) � CO and Mito-Tracker Red CMCRos was added for the last 30 minutes oftreatment. Cells were washed and fixed with 2% paraformal-dehyde followed by staining with Hoechst.

Statistical analysesStatistical analyses were conducted using SPSS version 13.0

(SPSS Inc.). The differences in the expression of different mar-kers between benign and cancer specimens were evaluated bypairedWilcoxon rank-sum test or Kruskal–Wallis test. Distribu-tions of overall survival and disease-free survival were estimatedby the method of Kaplan–Meier, with 95% confidence intervals.Differences between survival curves were calculated using thelog-rank test. Data are presented as the mean � SD and arerepresentative for at least 3 independent experiments. Student t,ANOVA, and Wilcoxon tests were used for estimation of statis-tical significance for the experiments (P < 0.05).

ResultsNuclear HO-1 with low activity in prostate cancercorrelates with poorer prognosis

HO-1 has been shown to be expressed and function in thenucleus as a regulator of oxidative stress response transcrip-tion factor at the expense, however, of losing enzymatic activity(30). On the basis of these data, we hypothesized that nucleartranslocation of HO-1 influences the response of cancer cells to

therapy. We evaluated 482 tumor specimens from patientswith prostate cancer with corresponding benign tissues bytissue microarray to assess the correlation between nuclearHO-1 expression and clinical tumor characteristics. Themajor-ity of benign samples showed low overall expression of HO-1(Fig. 1A–B and Supplementary Fig. S1A). In contrast, Gleasongrade 3 specimens showed significantly higher expression ofHO-1 in the nucleus than that of corresponding benign tissue(Fig. 1C and D; �, P ¼ 0.047). The intensity of nuclear HO-1staining declined inGleason grade 4–5 specimens as comparedwithGleason grade 3but did not achieve statistical significance

Figure 1. Nuclear expression of HO-1 in 482 biopsy samples of humanprostate cancer. A and B, representative pictures of HO-1 staining inspecimens from benign tissues adjacent to prostate cancer cells ofGleason grade 3 (C and D) and Gleason grades 4–5 (E and F).Magnification, �40 (insets at 100�). G, statistical analysis of nuclearHO-1 expression in different stages of prostate cancer (Gleason 3 and 4–5) versus adjacent benign tissues (BPH). �, P < 0.05, prostate cancerversus BPH. H, survival curves of time to relapse against low or highnuclear HO-1 expression in samples from all biopsies. Low nuclearstaining intensity corresponds to a score of 1 and high nuclear stainingintensity corresponds to a score of 3. �, P ¼ 0.046. Images arerepresentative of all biopsies. Scale bar, 50 mm.

Wegiel et al.

Cancer Res; 73(23) December 1, 2013 Cancer Research7012

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 5: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

(Fig. 1A–H). Finally, there was no significant difference incytoplasmic staining of HO-1 in prostate cancer versus adja-cent benign samples (data not shown). We reasoned that highnuclear HO-1 expression in moderately differentiated tumorsmay correspond to the level of oxidative stress or hypoxia thatdrives progression of differentiated tumors to more advanceddisease.We next evaluated the clinical significance of nuclear HO-1

expression by correlating HO-1 staining with biochemicalrecurrence (BCR) of prostate cancer as measured by pros-tate-specific antigen in the circulation (31) as well as overallpatient survival (Fig. 1H). High nuclear HO-1 (arbitrary inten-sity¼ 3) expression in the cancer biopsies in comparison to low(arbitrary intensity ¼ 1) nuclear HO-1 predicts a shorter timeto BCR (Fig. 1F; �, P¼ 0.007). After a 20- to 30-month follow-upperiod, a 20% greater number of patientswith high nuclearHO-1 relapsed in comparison to patients with low nuclear HO-1(P ¼ 0.046), indicating a shorter time to recurrent disease inpatients with prostate cancer with a lack of enzymaticallyactive nuclear HO-1.

HO-1 activity and CO influence the cancer cell responseto genotoxinsNuclear localized HO-1 is a hallmark of cancer progression

in prostate cancer (18). Comparing the HO-1 localizationprofiles and activity of noncancerous cells (epithelial cells andfibroblasts) to cancerous cells (PC3, A549, HepG2, T47D, MDA-468, HeLa), we found that cancer cell lines showed high nuclearexpression and low cytoplasmic expression, whereas noncan-cerous cells showed high cytosolic expression and low nuclearexpression (Fig. 2A and B). We show that when HO-1 islocalized to the nucleus it loses its activity in response tocommon chemotherapeutics (Fig. 2C; ref. 30). On the basis ofnuclear localization of HO-1 with low activity (30), we nextasked whether HO-1 is important in mediating the effective-ness of chemotherapeutic treatment in vitro. DNA damagecausesmutations and carcinogenesis as well as induces cancercell death. HO-1 expression in normal cells is required forappropriate DNA repair in response to chemotherapy orirradiation (32). Using camptothecin and doxorubicin toinduce DNA damage, we tested the role of HO-1 and CO oncancer and normal cell death. We observed significant celldeath with camptothecin or doxorubicin in vector control–treated PC3 cells as expected. Overexpression of HO-1 inducedfurther death of PC3 cells in response to camptothecin ordoxorubicin (Fig. 2D). A similar effect was observed in cellsafter exposure to a low concentration of CO (Fig. 2 andSupplementary Fig. S1B and S1C; ref. 33). Dose escalationexperiments to induce cell death with camptothecin showedthat in the presence of CO, the sensitivity of cells to camp-tothecin increased synergistically to approximately 1,000-foldover chemotherapy alone–treated cells (Fig. 2E and Supple-mentary Fig. S1B and S1C). The effect was specific to DNAdamage–inducing agents as apoptotic responses induced byTNF and cycloheximide were not altered after CO treatment(data not shown). Further evidence of the effects of CO on PC3cells in the presence of camptothecin showed that comparedwith air controls, CO induced significantly stronger cleavage of

caspase-3, augmented DNA damage as measured by cometassay, and induced Gadd45a expression, an important regu-lator of apoptosis and DNA repair that is induced in responseto chemotherapy (Supplementary Figs. S1D–S1F and S2).

Many reports detail the protective effects afforded by COand HO-1 in response to oxidative stress in primary cells (34–36), which is in contrast to the above effects in prostate cancercells. Treatment of normal prostate epithelial cells (PNT1A)with doxorubicin showed that unlike prostate cancer cells, COprevented doxorubicin-induced death of normal prostate cells(Fig. 2F and Supplementary Fig. S1G) supporting the remark-able selectivity ofHO-1 andCO to enhance killing of tumor cellswhile protecting primary cells. We confirmed these findingsusing primary mouse embryonic fibroblasts (MEF) treatedwith camptothecin or doxorubicin at the highest doses usedin cancer cells and evaluated the effects of CO. As expected, weobserved 75% to 90% cell death in response to both chemother-apeutics in air-treated cells. Similar to PNT1A, CO protectedprimary MEFs against doxorubicin-induced cell death versusair-treated cells (Supplementary Fig. S1H). The protection ofprimary cells afforded by CO is independent of p53, a well-characterized modulator of cell death in response to DNAdamage. CO was also able to protect p53�/� MEF (Supple-mentary Fig. S1H).

CO inhibits growth of human prostate cancer xenograftsWe next evaluated the role of CO alone and CO in com-

bination with doxorubicin on the growth of tumors in vivo,using PC3 human prostate cancer xenografts implantedsubcutaneously in nude mice. Mice with established tumorswere exposed to a regimen of CO (250 ppm for 1 hour once aday at the same time of day) � doxorubicin (doxorubicin wasinjected immediately after CO treatment) and growth wasmonitored over 2 weeks, at which point volumes of tumorswere measured and harvested for histologic analyses. Micetreated with CO or doxorubicin alone showed significanttumor growth arrest versus controls as measured by tumorvolume and Ki-67 expression and had no effect on bodyweight (Fig. 3A and Supplementary Fig. S3A and S3B). Com-bination treatment with doxorubicin plus CO showed furtheradditive effects with regard to tumor size, proliferative index,and neovascularization (Fig. 3B–F). We did, however, observea significant effect of CO alone or in combination withdoxorubicin on cell death that was not observed with doxo-rubicin alone, suggesting a distinct cellular target for CO (Fig.3G and H). Mitotic catastrophe as assessed by occurrence ofaberrant nuclei after cell division was present among the p-Histone H3–positive cells in tumors treated with CO ordoxorubicin (insets in Fig. 3E). Validation of the effects ofCO with immunostaining showed that CO inhibited the cell-cycle marker cyclin A2 (densitometric analyses: air þ doxo-rubicin: 1.15 � 0.28, CO þ doxorubicin: 0.68 � 0.09; P < 0.05;Supplementary Fig. S4A and S4B). The number of Thy-positivefibroblasts was markedly decreased in animals receiving COversus air treatment. It was not determined whether this wasdue to less infiltration or increased cell death (SupplementaryFig. S4C). Depletion of Thy-1–positive stromal cells maybe an additional early molecular target of CO leading to

Carbon Monoxide Regulates Cancer Cell Metabolism

www.aacrjournals.org Cancer Res; 73(23) December 1, 2013 7013

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 6: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

accelerated cell death and inhibition of angiogenesis. Thecontribution of endothelial cell proliferation and recruitmentof stroma cells in addition to tumor cell growth needs to befurther evaluated.

CO induces apoptosis and blocks mitosis andangiogenesis in a model of orthotopic prostatecancer in mice

In a more clinically relevant model of orthotopic PC3prostate cancer tumors, CO suppressed the mitotic index asmeasured by p-Histone H3 and CD31 staining in establishedtumors (8% and 6%, respectively; Fig. 4A–C). Importantly, COsignificantly suppressed a key marker of invasion and earlymetastasis known as uPA (Fig. 4D). Furthermore, CO showed

the trend toward decreased expression of the tumor markersMMP2 and VEGF; however, it did not reach significance (Fig.4D). Similar to the subcutaneous tumor xenograft model, COinduced greater cell death in orthotopic tumors as comparedwith tumors from air-treated mice (Fig. 4E and F; 35% in COversus 15% in air), suggesting that in addition to arrestinggrowth, CO reduced the invasive and aggressive phenotypewith a signature of mitochondrial collapse and tissue necrosis.This was different than that observed in vitro where CO alonedid not affect cancer cell death yet inhibited cell cycle. In bothtumor models, we observed a significant increase in theamount of CO present within the tumor tissue as well as inthe liver and lung of CO-exposed mice (Fig. 4G and data notshown).

Figure 2. Nuclear HO-1 is enzymatically inactive. A and B, immunoblot of HO-1 in the nuclear (A) and cytoplasmic (B) fractions of various normal (NIH3T3fibroblasts, NIH; primary human bronchial epithelial cells (E); primary lung fibroblasts (F), as well as cancer cells lines (PC3-PC, A549-A5, HepG2-Hep, HeLa-Hel, T47D-T47,MDA-468-MD). Data are representative of 3 independent experiments. C,HO-1 activity in the nuclear (white bars) and cytoplasmic (black bars)fractions of PC3 cells treated with camptothecin (1 mg/mL) or doxorubicin (10 mg/mL) for 4 hours. �, P < 0.05. D, crystal violet staining of PC3 cellsoverexpressing HO-1 (black bars) or control vector (white bars) treated for 24 hours with camptothecin (1–10 mg/mL) or doxorubicin (1–10 mg/mL). #,P < 0.05, doxorubicin/camptothecin treated versus untreated; ��, P < 0.01, HO-1 versus control vector. E, dose-dependent treatment of PC3 cells withcamptothecin (0.001–10 mg/mL) � CO for 24 hours. ��, P < 0.01; �, P < 0.05, CO � camptothecin (black bars) versus air � camptothecin (white bars).F, cell viabilitywasmeasured by crystal violet staining of PNT1A normal primary prostate epithelial cells treatedwith doxorubicin (10mg/mL)�CO (black bars)or air (white bars) for 48 hours. �, CO þ doxorubicin versus air þ doxorubicin, P < 0.05.

Wegiel et al.

Cancer Res; 73(23) December 1, 2013 Cancer Research7014

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 7: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

CO prevents development of PIN/carcinoma lesions inTRAMP mice and inhibits lung carcinoma growth inKras4bG12D transgenic miceCO was tested in 2 additional models of spontaneously

developed tumors using the prostate tumor TRAMP andthe lung tumor Kras mouse models (26, 37). We started COtreatment in mice 8 to 20 weeks of age, which is aftertumors are established (26, 37). Untreated mice showed ahigh proliferative index as measured by Ki-67 staining inthe tumors showing multiple PIN foci and adenocarcinoma(Fig. 5A and B). In contrast, CO-treated mice had fewer PINfoci and significantly less Ki-67–positive cells and some ofthe CO-treated TRAMP mice were completely lesion-free(Fig. 5A and B). Importantly, during the progression of thedisease and in contrast to the normal prostate where HO-1is localized to the cytoplasm, HO-1 was expressed in thenucleus in the PIN lesions (Fig. 5C). Staining for themitochondrial stress response marker mtTFA showed asignificant increase in the number of mitochondria in thelesions of CO-treated TRAMP mice as compared with theneoplastic lesions in air-treated TRAMP mice at the sameage (Fig. 5D).

A similar effect on tumor growth was observed in the Kraslung tumormodelwhereweobserved a significant inhibition inboth the frequency (average, n ¼ 24 in air vs. n ¼ 15 in CO) aswell as the size of the tumor nodules in the lung in animalstreatedwith CO versus air (Fig. 5E). Proliferation of tumorswasalso significantly inhibited as evidenced by decreased Ki-67staining (Fig. 5F). Longer treatment with CO resulted in furtherinhibition of tumor growth and size of the nodules (data notshown).

CO targets mitochondria to induce death of cancer cellsWe next explored potential molecular mechanisms by

which CO exerted its effects using prostate cancer cells asour model. Of note, HO-1 and CO have been shown to inducegrowth arrest of human cancer cells (38) as well as murineAC29 mesothelioma cells (55% less cells at day 2 and 80% atday 5 vs. air controls), which support a more global effectof CO in a different type of cancer cells. Given that CO isknown to target mitochondria, and the data presentedin Fig. 3, we next assessed the activity and number ofmitochondria in cancer cells in the presence and absenceof CO. CO increased OCR within minutes after exposure,

Figure 3. CO blocks tumor growth of established PC3 xenografts and sensitizes tumor cells to doxorubicin treatment. A and B, growth curves (% oforiginal tumor volume) of PC3 cells inoculated into nude mice for 2 weeks before initiation of treatment with air or CO (250 ppm, 1 hour) � doxorubicin,which was continued for an additional 2 weeks before harvest. Results are mean � SD of n ¼ 4–5 per group. Immunostaining of CD31 (C)and p-Histone H3 (E) in PC3 tumors. Quantitation of CD31 (D) or PH3-positive (F) cells was counted as the percentage of positive cells/field of view(magnification, �20). Results represent mean � SD of 8 to 10 sections from each harvested tumor. E and F, TUNEL staining quantitated as percentageof positive cells per field of view (magnification, �20). Results represent mean � SD of 8 to 10 sections from each harvested tumor. ��, P < 0.01 versusair; �, P < 0.05 versus air.

Carbon Monoxide Regulates Cancer Cell Metabolism

www.aacrjournals.org Cancer Res; 73(23) December 1, 2013 7015

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 8: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

which was accompanied by ROS generation in PC3 cellsthat correlated with decreased ECAR and lactate levels(Fig. 6A–E). Use of the mitochondrial complex I poisonrotenone to compare CO with another mitochondrial target-ing compound and whether it would have similar effects inthese cells as CO showed complete blockage of the OCR aswas expected with rotenone and this effect occurred inboth cancer and noncancer cells (Fig. 6B). We also testedmitochondrial potential using the JC-1 dye. As previouslyreported in other cell types (39), CO increased mitochondriamembrane potential in PC3 cells (Supplementary Fig. S5).CO did inhibit OCR in normal cells, again suggesting aselective difference in the response of cancer cells andnormal PNT1A cells to CO (Fig. 6B). The activation of

mitochondrial activity by CO is likely contributes to a lesscancerous phenotype. As the treatment time with CO wasextended, PC3 cells showed clear G1 arrest (Fig. 6F), whichcorresponded to significant inhibition of metabolic activityof cancer cells as measured by SPECT (Fig. 6G). Further-more, exposure to CO induced mitochondrial stress asevidenced by increased mtTFA, cytochrome c–positivestaining, and an inhibition in the oxidized state of mito-chondria (Fig. 6H–K). Metabolic screening showed thatexposure of PC3 cells to CO for 6 hours resulted in a decreasein glucose metabolism and influenced synthetic pathwaysinvolved in nucleotide and amino acid synthesis, all of whichcorroborate the effects of CO on cancer cell growth (Table 1;Supplementary Table S1 and Supplementary Fig. S6).

Figure 4. CO induces apoptosis in an orthotopic prostate cancer model in mice. A–C, coimmunostaining of p-Histone H3 (green) and CD31 (red) of orthotopictumors in mice treated for 4 weeks with CO or air. Representative pictures are shown in A, and quantification of staining is shown in B and C (n¼ 4 per group).Scale bar, 50 mm. D, real-time PCR with primers for MMP2, VEGF, and uPA in mice treated with CO or air. Results represent mean � SD of n ¼ 4 tumorsper group. �, P < 0.05 versus control. E and F, TUNEL staining for apoptosis and the percentage of positive cells per field of view (magnification, �20)quantitated as above. Images are representative of 8 to 10 sections from each harvested tumor. Results represent mean � SD of 8 to 10 fields of view fromeach tumor. ��,P <0.01 versus air. G,CO levels in the tumors frommicewith subcutaneously or orthotopically inoculated tumors. n¼3–6 per group. Scale bar,100 mm.

Wegiel et al.

Cancer Res; 73(23) December 1, 2013 Cancer Research7016

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 9: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

Figure 5. CO arrests growth of established prostate and lung cancer. A and B, immunohistochemistry analysis with antibody against Ki-67 andhistological analysis (H&E) was conducted on the tissues from wild-type (wt) or TRAMP mice that were treated with CO (250 ppm/daily/5 d/wk) oruntreated (air). Quantitation of Ki-67 staining is shown in A (n ¼ 6–7 mice per group). C, immunohistochemistry with antibodies to HO-1 was applied inthe normal (wt) and PIN lesion containing prostates (TRAMP) as well as in the lung tumors of FVB/N-Tg(teto-Kras2)12Hev (provided by Dr. H.Varmus, National Cancer Institute, Bethesda, MD). Scale bar, 200 mm. D, immunostaining of prostates from TRAMP treated as above with antibodiesto mitochondrial transcription factor A. Two representative pictures from air and CO-treated mice are shown. Scale bar, 200 mm. E and F, H&Estaining in the lungs of FVB/N-Tg(teto-Kras2)12Hev mice that were treated with doxycycline for 8 weeks and continued with CO treatment forthe following 5 weeks. The number of nodules in the lung cross-sections was evaluated in at least n ¼ 5 to 6 per animal. The representative sectionsand immunohistochemistry with antibody against Ki-67 are shown in F. Data are representative for air (n ¼ 8) and CO (n ¼ 9 animals). P < 0.0002.Scale bar, 200 mm.

Carbon Monoxide Regulates Cancer Cell Metabolism

www.aacrjournals.org Cancer Res; 73(23) December 1, 2013 7017

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 10: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

In efforts to further investigate the role of mitochondria inthe effects observed with CO, we depleted mitochondria fromcancer cells (Rhoo cells), which resulted in epithelial-to-mes-enchymal transition as evident by the emergence of fibroblast-

like–shaped cells and enhanced expression of vimentin in theRhoo cells comparedwith control, whichwas unaffected by CO,further suggestive of mitochondrial targeting by CO (Fig. 6L).Rhoo cells are more prone to doxorubicin-induced cell death

Figure6. Effects ofCOonmitochondria in prostate cancer cells. A,OCRandECARweremeasured inPC3cells treatedwithmediumbubbledwith250ppmCO.Relative mitochondria activity was measured over time and the levels are shown at 30 minutes. B, OCR was measured in PC3 or PNT1A � Rotenone(20 nmol/L) for 10 minutes followed by 30minutes�CO (250 ppm). Data are representative for 2 independent experiments in duplicates. C, levels of glucoseand lactate were measured in the lysates of PC3 cells � CO (250 ppm) for 6 hours. D and E, mitochondrial superoxide production was measuredby flowcytometry usingMitoSox. Data are representative for 3 independent experiments in triplicates. ��,P<0.01. F, cell-cycle analysiswas conducted inPC3and PNT1A cells by propidium iodide staining. Cells were synchronized for 48 hours via serum starvation and then treated for 48 hours with FBS-containingmedium � CO. Data are representative of 3 independent experiments in triplicates. ��, P < 0.01. G, SPECT analysis in PC3 cells � CO (250 ppm) orCORM-A1 (20 or 100 mmol/L) for 24 hours. Intensity of radiation is presented as amean�SDof 2 independent experiments carried out in triplicate. �,P < 0.05versus control; ��, P < 0.01 versus control. H, immunohistochemical staining for cytochrome c in subcutaneous prostate cancer xenografts as an indicator ofcell death. Representative sections from n ¼ 3 to 4 animals are shown. Scale bar, 150 mm. I and J, MitoTracker Red CMXRos staining of PC3 cellstreated with doxorubicin (Dox) � CO for 4 hours. Hoechst was used to stain nuclei. Representative pictures are shown in C, and densitometric intensityanalyses of 3 fields are shown in D. Results represent mean � SD of 3 independent experiments. �, P < 0.001 versus control. Scale bar, 10 mm. K,immunoblotting with antibodies against mtTFA in the lysates of CO-treated PC3 and A549 cells (4 hours, 250 ppm). L, immunofluorescence staining withantibody against vimentin in lung adenocarcinoma cell line A549, control, andRhoo (EtBr)�CO (250 ppm, 48 hours). Data are representative of 2 independentexperiments in duplicate. Scale bar, 30 mm.

Wegiel et al.

Cancer Res; 73(23) December 1, 2013 Cancer Research7018

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 11: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

then normal cells; however, CO did not amplify the effects ofdoxorubicin in these cells in contrast to normal cells (% ofsurviving colonies: A549: air þ doxorubicin: 29.8% � 1.2%; COþ doxorubicin: 13.4% � 0.7%; A549Rhoo: air þ doxorubicin:17.1%� 1%; COþ doxorubicin: 17.6%� 0.39%). On the basis ofchanges in mitochondrial function described above and thespecifically the increase in ROS generation, we next depletedROS using a cocktail of pegylated catalase and superoxidedismutase. Addition of the antioxidant cocktail before treat-ment with CO resulted in reversal of the effects of CO ondoxorubicin-induced apoptosis, further supporting ROS andthe mitochondria as the cellular target for CO (SupplementaryFig. S7).

DiscussionIn the present study, we elucidate a role for CO in

modulation of prostate and lung tumor growth and survival.CO not only mimics the effects of chemotherapy alone byblocking proliferation but also amplifies tumor cell deathwhen treated in combination with either doxorubicin orcamptothecin. Furthermore, we provide evidence that COreduces the tumor burden in xenograft and transgenicmouse models of prostate and lung cancers and effectivelyblocks progression of neoplasia and adenocarcinoma. Wededuce that CO effectively switches the metabolic state ofthe cancer cell to fuel oxidative metabolism and decreasesin nucleotide and amino acid synthetic pathways. Collec-tively, this causes cell-cycle arrest and collapse underintense mitochondrial-dependent oxidative stress. In vitro,this elicits a 1,000-fold increase in sensitivity to genotoxinsfostered, in part, by a mitochondria-dependent increase inROS generation. In vivo, the role of CO and ROS on tumor

growth is less clear and likely involves other cellularmechanisms including hypoxia and perhaps a heightenedimmune response. Importantly, CO protects normal cellsfrom DNA damage by cytotoxic agents, in part, by reducingoxygen consumption and eliciting a hibernation-like statethat has been observed in other cell types such as T cellsand smooth muscle cells with the induction of DNA repaircomplexes (40).

HO-1 is an accepted cytoprotective gene in most stress-related pathologies acting to reestablish homeostasis (41, 42).Others and we have shown that nuclear-localized HO-1 nolonger possesses enzymatic activity and as such becomeshighly associated with cancer and tumor growth (30, 43). Weshow thatCOcanmimic the effects of enzymatically activeHO-1 and that exposure toCOor reinstatingHO-1 activity results inenhanced sensitization and acceleration of apoptosis of cancercells and tumors while protecting normal cells against che-motherapeutic toxicity. This is consistent with prior observa-tions that HO-1–derived CO is important in DNA repair innormal cells (32). We posit that the subsequent decrease inendogenous CO generation whenHO-1 is in the nucleus resultsin accelerated DNA stress and damage and thus serves as ahallmark of early carcinogenesis.

In our cohort of patients, nuclear HO-1 is increased in asubset of patients with prostate cancer with a shorter time toBCR. We did not observe a significant association of HO-1expression with Gleason grades due to the relatively lownumber of cohort cases with the most advanced disease(Gleason grade 5). We would anticipate however statisticallysignificant correlations in a larger cohort of patients asreported by Sacca and colleagues who showed a clear associ-ation between HO-1 and prostate cancer incidence. Inhibitionof HO-1 pharmacologically using zinc-protoporphyrin (Zn-PP)induced apoptosis and suppressed growth of sarcomas in rats(44), but Zn-PP treatment failed to potentiate the antitumoreffects of 5-fluorouracil, cisplatin, and doxorubicin in 3 differ-ent tumor models (45). HO-1 overexpression or CO exposurereduced TPA-induced invasion of breast cancer cells (13) aswell as lung adenocarcinoma cells (38). The effects of HO-1 ontumor growth and progression are complex and likely dependon cancer types, the cell-cycle status, the model system, as wellas the methods of pharmacologic or genetic manipulation ofHO-1 activity.

In our model of human tumor xenografts in vivo, weobserved strong inhibition of angiogenic markers, mitosis, andaccelerated apoptosis in tumors treatedwithCO. Furthermore,CO sensitized cancer cells and solid tumors to apoptosis withapparent mitotic catastrophe likely influenced by cellularexhaustion as evidenced by our biochemical data (Fig. 6). Theeffects of CO onmitochondria have beenwell described andwespeculate that the high metabolic requirements of cancer cellslend itself to greater effects of CO on survival given that CO isknown to target mitochondrial respiratory complexes (33).This is supported by the effects of CO on mitochondrialmembrane potential (46), biogenesis (36), and decreased pro-hibitin (data not shown), which regulate cancer cell survivaland growth. PC-3 xenografts are partially resistant to doxoru-bicin-induced apoptosis, which delays tumor growth through

Table 1. Metabolic analysis was conductedusing MetaboAnalyst 2.0

Peaks (mz/rt) Fold change

S-Adenosyl-L-homocysteine-positive 7.9552S-Adenosyl-L-homocysteine-negative 7.82181-Methyladenosine 6.7669NADH-nega 6.28492-Deoxyglucose-6-phosphate 6.0176Pyridoxine 5.4215Pyridoxamine 5.4073NG-dimethyl-L-arginine 5.3147Pyrophosphate 5.1344Ribose phosphate 5.0937NADH 5.0912Thymine 5.04943-Phospho-serine 5.0256

NOTE: Top 13 of 290 metabolites were inhibited by CO (250ppm, 6 hours) treatment of PC3 cells by more than 5-fold.n ¼ 3 in duplicates. Univariate fold change analysis wasconducted by MetaboAnalyst 2.0.

Carbon Monoxide Regulates Cancer Cell Metabolism

www.aacrjournals.org Cancer Res; 73(23) December 1, 2013 7019

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 12: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

mitotic blockade (47). This effectwas amplified by the presenceof CO in vitro and in vivo. The increase in ROS by CO in cancercells, likely augments protein and mitochondrial DNA (48).Indeed, inhibition of HO-1 activity in prostate cancer cellscorrelated with a reduction in protein carbonylation and ROSformation (49).

The finding that CO protects normal cells from genotoxintoxicity offers an added benefit and may permit chemosparingtherapy, thus decreasing negative side effects. We have nottested the response to irradiation, however considering thatCO protects normal cells against cell death via amplification ofDNA repair (32), we expect that CO may have similar sparingeffects on normal cells in response to irradiation while ampli-fying cancer cell death in response to irradiation. CO-elicitedprotection of normal cells suggests that normal cells toleratealterations in metabolic demands more efficiently and aremore capable of repairing damaged DNA. The effect of CO toenhance killing is not specific to prostate cancer cells, as weobserved accelerated cell death of breast, lung, and otherprostate cancer cell lines treated with doxorubicin and CO aswell as dysregulated smooth muscle cells that lead to vascularstenosis in models of angioplasty, transplantation, and pul-monary hypertension. CO at these doses has no untowardeffects on the animals and in fact also protects against doxo-rubicin-induced cardiomyopathy (36).

In summary, we show that HO-1 expression in cancerspecimens is targeted to the nucleus in moderately differ-entiated tumor cells for reasons that remain to be elucidat-ed. Collectively, CO influences cellular bioenergetics that wefind differs between cancer cells and normal cells. COaccelerates oxidative metabolism and ROS generation,unlike in noncancer cells where CO inhibits respiration andprotects against cell death. Insufficient respiration due todysfunctional mitochondria drives what is known as theWarburg effect. Exposure to CO uses Warburg physiology asan advantage, compelling the cancer cell to consume moreoxygen that in turn drives metabolic demand, leading togrowth inhibition, cellular exhaustion, and death. These dataprovide the first evidence showing the potential for safe, lowamounts of CO to be used as an adjuvant therapeutic optionfor the treatment of cancer. One might envision a specific

inhalational therapy regimen being implemented such asthat used here to treat patients with cancer with inhaled COusing a specific metered dosing delivery device or a COreleasing molecule (CO-RM; ref. 50).

Disclosure of Potential Conflicts of InterestJ. Belcher has a commercial research grant from Sangart Inc. and Seattle

Genetics. No potential conflicts of interest were disclosed by the otherauthors.

Authors' ContributionsConception and design: B. Wegiel, V. Sukhatme, A. Ahmed, J.L. Persson, L.E.OtterbeinDevelopment of methodology: J. Belcher, N. Penacho, L.E. OtterbeinAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): B. Wegiel, D. Gallo, C. Harris, J. Belcher, G.M.Vercellotti, P. Seth, P.P. Pandolfi, A. Bjartell, J.L. Persson, L.E. OtterbeinAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): B. Wegiel, C. Harris, P. Seth, L. Helczynski, L.E.OtterbeinWriting, review, and/or revision of the manuscript: B. Wegiel, J. Belcher,G.M. Vercellotti, A. Ahmed, A. Bjartell, J.L. Persson, L.E. OtterbeinAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): D. Gallo, J.L. Persson, L.E. OtterbeinStudy supervision: B. Wegiel, V. Sukhatme, L.E. OtterbeinReference pathologist: L. Helczynski

AcknowledgmentsThe authors thankDr. Nardella for helpful discussions and for providing KRas

and TRAMPmice; Drs. Balk andChen for the PC3 cells; Dr. Dizeyi for PNT1A cells;and Dr. Jacks for MEF cells. They also thank the Swedish Cancer Society, TheSwedish National Research Council, MAS Cancer Foundation, Swedish RoyalPhysiographic Society in Lund, Gunnar Nilsson Cancer Foundation, CrafoordFoundation andMAS Foundation (J. Belcher and A. Bjartell) and The Foundationfor Urology Research in Malm€o (A. Bjartell). They are grateful to Elise Nilsson forthe excellent technical skills.

Grant SupportThisworkwas supported byNIHgrantHL-071797&HL-076167 (LEO). BW is a

recipient of AHA (10SDG2640091) grant. The authors thank the Julie Henry Fundat the Transplant Center of the BIDMC for their support. This study was partiallyfunded by grants from the British Heart Foundation (PG/06/114) and MedicalResearch Council (G0601295 and G0700288).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received April 17, 2013; revised September 16, 2013; accepted September 18,2013; published OnlineFirst October 11, 2013.

References1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics,

2009. CA Cancer J Clin 2009;59:225–49.2. Winquist E, Waldron T, Berry S, Ernst DS, Hotte S, Lukka H. Non-

hormonal systemic therapy in men with hormone-refractory prostatecancer and metastases: a systematic review from the Cancer CareOntario Program in Evidence-based Care's Genitourinary CancerDisease Site Group. BMC Cancer 2006;6:112.

3. Levine AJ, Puzio-Kuter AM. The control of the metabolic switch incancers by oncogenes and tumor suppressor genes. Science2010;330:1340–4.

4. Warburg O. Iron, the oxygen-carrier of respiration-ferment. Science1925;61:575–82.

5. He Y, Wu J, Dressman DC, Iacobuzio-Donahue C, Markowitz SD,Velculescu VE, et al. Heteroplasmic mitochondrial DNA mutations innormal and tumour cells. Nature 2010;464:610–4.

6. Sanchez-AragoM, Chamorro M, Cuezva JM. Selection of cancer cellswith repressed mitochondria triggers colon cancer progression.Carcinogenesis 2010;31:567–76.

7. Oberley LW, Buettner GR. Role of superoxide dismutase in cancer: areview. Cancer Res 1979;39:1141–9.

8. Bize IB,Oberley LW,MorrisHP. Superoxide dismutase andsuperoxideradical in Morris hepatomas. Cancer Res 1980;40:3686–93.

9. Spitz DR, Sim JE, Ridnour LA, Galoforo SS, Lee YJ. Glucose depri-vation-induced oxidative stress in human tumor cells. A fundamentaldefect in metabolism? Ann N Y Acad Sci 2000;899:349–62.

10. Ahmad IM, Aykin-BurnsN, Sim JE,Walsh SA,HigashikuboR, BuettnerGR, et al. Mitochondrial O2�- and H2O2 mediate glucose deprivation-induced stress in human cancer cells. J Biol Chem 2005;280:4254–63.

11. Aykin-Burns N, Ahmad IM, Zhu Y, Oberley LW, Spitz DR. Increasedlevels of superoxide andH2O2mediate the differential susceptibility of

Wegiel et al.

Cancer Res; 73(23) December 1, 2013 Cancer Research7020

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 13: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

cancer cells versus normal cells to glucose deprivation. Biochem J2009;418:29–37.

12. Hill M, Pereira V, ChauveauC, Zagani R, RemyS, Tesson L, et al. Hemeoxygenase-1 inhibits rat and human breast cancer cell proliferation:mutual cross inhibition with indoleamine 2,3-dioxygenase. FASEB J2005;19:1957–68.

13. Lin CW, Shen SC, Hou WC, Yang LY, Chen YC. Heme oxygenase-1inhibits breast cancer invasion via suppressing the expression ofmatrix metalloproteinase-9. Mol Cancer Ther 2008;7:1195–206.

14. Sunamura M, Duda DG, Ghattas MH, Lozonschi L, Motoi F, YamauchiJ, et al. Heme oxygenase-1 accelerates tumor angiogenesis of humanpancreatic cancer. Angiogenesis 2003;6:15–24.

15. Was H, Cichon T, Smolarczyk R, Rudnicka D, Stopa M, Chevalier C,et al. Overexpression of heme oxygenase-1 in murine melanoma:increased proliferation and viability of tumor cells, decreased survivalof mice. Am J Pathol 2006;169:2181–98.

16. YinY, LiuQ,WangB,ChenG, Xu L, ZhouH. Expression and function ofheme oxygenase-1 in human gastric cancer. Exp Biol Med 2012;237:362–71.

17. Banerjee P, Basu A, Wegiel B, Otterbein LE, Mizumura K, Gasser M,et al. Heme Oxygenase-1 Promotes Survival of Renal Cancer Cellsthrough Modulation of Apoptosis- and Autophagy-regulating Mole-cules. J Biol Chem 2012;287:32113–23.

18. Sacca P, Meiss R, Casas G, Mazza O, Calvo JC, Navone N, et al.Nuclear translocation of haeme oxygenase-1 is associated to prostatecancer. Br J Cancer 2007;97:1683–9.

19. Gandini NA, FermentoME, SalomonDG,Blasco J, Patel V,Gutkind JS,et al. Nuclear localization of heme oxygenase-1 is associated withtumor progression of head and neck squamous cell carcinomas. ExpMol Pathol 2012;93:237–45.

20. Tibullo D, Barbagallo I, Giallongo C, La Cava P, Parrinello N, Vanella L,et al. Nuclear translocation of heme oxygenase-1 confers resistance toImatinib in chronic myeloid leukemia cells. Curr Pharm Des 2013;19:2765–70.

21. Kikuchi A, YamayaM, Suzuki S, YasudaH, KuboH, NakayamaK, et al.Association of susceptibility to the development of lung adenocarci-nomawith the heme oxygenase-1 gene promoter polymorphism. HumGenet 2005;116:354–60.

22. Lo SS, Lin SC, Wu CW, Chen JH, Yeh WI, Chung MY, et al. Hemeoxygenase-1 gene promoter polymorphism is associated with risk ofgastric adenocarcinoma and lymphovascular tumor invasion. AnnSurg Oncol 2007;14:2250–6.

23. Ollinger R, Kogler P, Troppmair J, Hermann M, Wurm M, Drasche A,et al. Bilirubin inhibits tumor cell growth via activation of ERK. CellCycle 2007;6:3078–85.

24. Wegiel B, Bjartell A, Tuomela J, Dizeyi N, Tinzl M, Helczynski L, et al.Multiple cellular mechanisms related to cyclin A1 in prostate cancerinvasion and metastasis. J Natl Cancer Inst 2008;100:1022–36.

25. WegielB,BjartellA,EkbergJ,GadaleanuV,BrunhoffC,PerssonJL.AroleforcyclinA1 inmediatingtheautocrineexpressionofvascularendothelialgrowth factor in prostate cancer. Oncogene 2005;24:6385–93.

26. Fisher GH,Wellen SL, Klimstra D, Lenczowski JM, Tichelaar JW, LizakMJ, et al. Induction and apoptotic regression of lung adenocarcinomasby regulation of a K-Ras transgene in the presence and absence oftumor suppressor genes. Genes Dev 2001;15:3249–62.

27. Otterbein LE, Zuckerbraun BS, Haga M, Liu F, Song R, Usheva A,et al. Carbon monoxide suppresses arteriosclerotic lesions asso-ciated with chronic graft rejection and with balloon injury. Nat Med2003;9:183–90.

28. Maines MD, Trakshel GM, Kutty RK. Characterization of two con-stitutive forms of rat liver microsomal heme oxygenase. Only onemolecular species of the enzyme is inducible. J Biol Chem 1986;261:411–9.

29. Verma K, Penney DG, Helfman CC, Sutariya BB. Carboxyhemoglobinin the rat: improvements in the spectrophotometric measurement andcomparison to other studies. J Appl Toxicol 1989;9:323–30.

30. Lin Q, Weis S, Yang G, Weng YH, Helston R, Rish K, et al. Hemeoxygenase-1 protein localizes to the nucleus and activates transcrip-tion factors important in oxidative stress. J Biol Chem 2007;282:20621–33.

31. Bjartell AS, Al-Ahmadie H, Serio AM, Eastham JA, Eggener SE, FineSW, et al. Association of cysteine-rich secretory protein 3 and beta-microseminoprotein with outcome after radical prostatectomy. ClinCancer Res 2007;13:4130–8.

32. Otterbein LE, Hedblom A, Harris C, Csizmadia E, Gallo D, Wegiel B.Heme oxygenase-1 and carbon monoxide modulate DNA repairthrough ataxia-telangiectasia mutated (ATM) protein. Proc Natl AcadSci U S A 2011;108:14491–6.

33. D'Amico G, Lam F, Hagen T, Moncada S. Inhibition of cellular respi-ration by endogenously produced carbon monoxide. J Cell Sci 2006;119:2291–8.

34. Brouard S, Otterbein LE, Anrather J, Tobiasch E, Bach FH, Choi AM,et al. Carbon monoxide generated by heme oxygenase 1 suppressesendothelial cell apoptosis. J Exp Med 2000;192:1015–26.

35. G�eunther L, Berberat PO, Haga M, Brouard S, Smith RN, Soares MP,et al. Carbon monoxide protects pancreatic beta-cells from apoptosisand improves islet function/survival after transplantation. Diabetes2002;51:994–9.

36. Suliman HB, Carraway MS, Ali AS, Reynolds CM, Welty-Wolf KE,PiantadosiCA. TheCO/HOsystem reverses inhibition ofmitochondrialbiogenesis and prevents murine doxorubicin cardiomyopathy. J ClinInvest 2007;117:3730–41.

37. Foster BA, Gingrich JR, Kwon ED, Madias C, Greenberg NM. Char-acterization of prostatic epithelial cell lines derived from transgenicadenocarcinoma of the mouse prostate (TRAMP) model. Cancer Res1997;57:3325–30.

38. Lee PJ, Alam J, Wiegand GW, Choi AM. Overexpression of hemeoxygenase-1 in human pulmonary epithelial cells results in cell growtharrest and increased resistance to hyperoxia. Proc Natl Acad Sci U SA1996;93:10393–8.

39. Zuckerbraun BS, Chin BY, Bilban M, d'Avila JC, Rao J, Billiar TR, et al.Carbon monoxide signals via inhibition of cytochrome c oxidase andgeneration of mitochondrial reactive oxygen species. FASEB J 2007;21:1099–106.

40. SongR,MahidharaRS, ZhouZ,HoffmanRA,Seol DW, Flavell RA, et al.Carbon monoxide inhibits T lymphocyte proliferation via caspase-dependent pathway. J Immunol 2004;172:1220–6.

41. Otterbein LE, Soares MP, Yamashita K, Bach FH. Heme oxygenase-1:unleashing the protective properties of heme. Trends Immunol 2003;24:449–55.

42. WasH, Dulak J, Jozkowicz A. Heme oxygenase-1 in tumor biology andtherapy. Curr Drug Targets 2010;11:1551–70.

43. FerrandoM,GueronG,ElgueroB,Giudice J, SallesA, LeskowFC, et al.Heme oxygenase 1 (HO-1) challenges the angiogenic switch in pros-tate cancer. Angiogenesis 2011;14:467–79.

44. Fang J, Sawa T, Akaike T, Akuta T, Sahoo SK, Khaled G, et al. In vivoantitumor activity of pegylated zinc protoporphyrin: targeted inhi-bition of heme oxygenase in solid tumor. Cancer Res 2003;63:3567–74.

45. Nowis D, Bugajski M, Winiarska M, Bil J, Szokalska A, Salwa P, et al.Zinc protoporphyrin IX, a heme oxygenase-1 inhibitor, demon-strates potent antitumor effects but is unable to potentiate antitu-mor effects of chemotherapeutics in mice. BMC Cancer 2008;8:197.

46. Piantadosi CA, Carraway MS, Suliman HB. Carbon monoxide, oxida-tive stress, and mitochondrial permeability pore transition. Free RadicBiol Med 2006;40:1332–9.

47. Grunwald V, DeGraffenried L, Russel D, Friedrichs WE, Ray RB,Hidalgo M. Inhibitors of mTOR reverse doxorubicin resistance con-ferred by PTEN status in prostate cancer cells. Cancer Res 2002;62:6141–5.

48. Song W, Su H, Song S, Paudel HK, Schipper HM. Over-expression ofheme oxygenase-1 promotes oxidative mitochondrial damage in ratastroglia. J Cell Physiol 2006;206:655–63.

49. Alaoui-Jamali MA, Bismar TA, Gupta A, Szarek WA, Su J, Song W,et al. A novel experimental heme oxygenase-1-targeted therapyfor hormone-refractory prostate cancer. Cancer Res 2009;69:8017–24.

50. Motterlini R, Otterbein LE. The therapeutic potential of carbon mon-oxide. Nat Revi Drug Discov 2010;9:728–43.

Carbon Monoxide Regulates Cancer Cell Metabolism

www.aacrjournals.org Cancer Res; 73(23) December 1, 2013 7021

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075

Page 14: Carbon Monoxide Expedites Metabolic Exhaustion to Inhibit ... · bioenergetics, ultimately resulting in metabolic exhaustion. Cancer Res; 73(23); 7009–21. 2013 AACR. Introduction

2013;73:7009-7021. Published OnlineFirst October 11, 2013.Cancer Res   Barbara Wegiel, David Gallo, Eva Csizmadia, et al.   GrowthCarbon Monoxide Expedites Metabolic Exhaustion to Inhibit Tumor

  Updated version

  10.1158/0008-5472.CAN-13-1075doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2013/10/11/0008-5472.CAN-13-1075.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/73/23/7009.full#ref-list-1

This article cites 48 articles, 21 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/73/23/7009.full#related-urls

This article has been cited by 7 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/73/23/7009To request permission to re-use all or part of this article, use this link

on December 9, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst October 11, 2013; DOI: 10.1158/0008-5472.CAN-13-1075