Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K...

7
Up-regulation of the ATPase Inhibitory Factor 1 (IF1) of the Mitochondrial H -ATP Synthase in Human Tumors Mediates the Metabolic Shift of Cancer Cells to a Warburg Phenotype * S Received for publication, May 19, 2010, and in revised form, June 7, 2010 Published, JBC Papers in Press, June 9, 2010, DOI 10.1074/jbc.M110.146480 Laura Sa ´ nchez-Cenizo, Laura Formentini, Marcos Aldea, A ´ lvaro D. Ortega, Paula García-Huerta, María Sa ´ nchez-Arago ´ , and Jose ´ M. Cuezva 1 From the Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Auto ´noma de Madrid, Centro de Investigacio ´n Biome ´dica en Red de Enfermedades Raras, ISCIII, Universidad Auto ´noma de Madrid, 28049 Madrid, Spain The H -ATP synthase is a reversible engine of mitochondria that synthesizes or hydrolyzes ATP upon changes in cell physi- ology. ATP synthase dysfunction is involved in the onset and progression of diverse human pathologies. During ischemia, the ATP hydrolytic activity of the enzyme is inhibited by the ATPase inhibitory factor 1 (IF1). The expression of IF1 in human tissues and its participation in the development of human pathology are unknown. Here, we have developed monoclonal antibodies against human IF1 and determined its expression in paired nor- mal and tumor biopsies of human carcinomas. We show that the relative mitochondrial content of IF1 increases significantly in carcinomas, suggesting the participation of IF1 in oncogenesis. The expression of IF1 varies significantly in cancer cell lines. To investigate the functional activity of IF1 in cancer, we have manipulated its cellular content. Overexpression of IF1 or of its pH-insensitive H49K mutant in cells that express low levels of IF1 triggers the up-regulation of aerobic glycolysis and the inhi- bition of oxidative phosphorylation with concurrent mitochon- drial hyperpolarization. Treatment of the cells with the H - ATP synthase inhibitor oligomycin mimicked the effects of IF1 overexpression. Conversely, small interfering RNA-mediated silencing of IF1 in cells that express high levels of IF1 promotes the down-regulation of aerobic glycolysis and the increase in oxidative phosphorylation. Overall, these findings support that the mitochondrial content of IF1 controls the activity of oxida- tive phosphorylation mediating the shift of cancer cells to an enhanced aerobic glycolysis, thus supporting an oncogenic role for the de-regulated expression of IF1 in cancer. In oxidative phosphorylation, ATP is synthesized by the mitochondrial ATP synthase, a H -driven rotatory engine of the inner membrane that utilizes as driving force for ATP syn- thesis the H electrochemical gradient generated by the respi- ratory chain (1– 4). The cellular expression level of -F1- ATPase, 2 which is the catalytic subunit of the H -ATP synthase, is diminished in diverse human pathologies (5), which include cancer (6 –9), affording a relevant marker of disease progression (6, 7, 10 –12) and of the response to chemotherapy (7, 13–15). Moreover, the down-regulation of -F1-ATPase in lung carcinomas (12) and colon cancer cells (15) also provides a mechanistic explanation to the increased glucose avidity of car- cinomas, i.e. to the enhanced aerobic glycolysis of cancer cells (16, 17). Interestingly, the quantitative determination of -F1- ATPase relative to the content of glyceraldehyde-3-phosphate dehydrogenase in human tumors has revealed that cancer abol- ishes the tissue-specific differences in the cellular complement of the bioenergetic -F1-ATPase protein (18). It is well established that when mitochondrial respiration is impaired, the H -ATP synthase can function in reverse acting as an ATP hydrolase for maintaining the proton motive force (1, 19). This process is regulated by an inhibitor peptide called ATPase inhibitory factor 1 or IF1 (19 –21), a highly conserved nuclearly encoded protein. When matrix pH drops, IF1 becomes activated and binds -F1-ATPase, blocking ATP hydrolysis and preventing a useless waste of energy (20). The substitution of histidine 49 in IF1 by a lysine residue renders a mutant form (H49K) that inhibits the ATP hydrolase activity in a pH-insensitive way (22). The structure and in vitro mecha- nism of action of IF1 has been studied in detail, and its role as an inhibitor of the hydrolase activity of the H -ATP synthase is well documented (19, 20, 23). However, the information on IF1 expression in human tissues and its putative contribution to the development of human pathology are unknown. In this study, we demonstrate that IF1 is overexpressed in human carcino- mas. Moreover, we document that IF1 plays a regulatory role in controlling cellular energetic metabolism, strongly supporting its participation as an additional molecular switch used by can- cer cells to trigger the induction of aerobic glycolysis, i.e. their Warburg phenotype. EXPERIMENTAL PROCEDURES Protein Extraction—Frozen tissue sections obtained from surgical specimens of untreated cancer patients with primary * This work was supported by Ministerio de Educacio ´ n y Ciencia Grant BFU2007-65253 and the Centro de Investigacio ´ n Biome ´dica en Red de Enfermedades Raras and Comunidad de Madrid Grant S-GEN-0269, Spain. S The on-line version of this article (available at http://www.jbc.org) contains supplemental Fig. S1. 1 To whom correspondence should be addressed: Centro de Biología Molec- ular Severo Ochoa, Universidad Auto ´ noma de Madrid, 28049 Madrid, Spain. Tel.: 34-91-196-4618; Fax: 34-91-196-4420; E-mail: jmcuezva@cbm. uam.es. 2 The abbreviations used are: -F1-ATPase, catalytic subunit of the H -ATP synthase; IF1, ATPase inhibitory factor 1; FCCP, carbonyl cyanide-p-trifluo- romethoxyphenylhydrazone; siRNA, small interfering RNA; NRK, normal rat kidney. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 33, pp. 25308 –25313, August 13, 2010 © 2010 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. 25308 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 33 • AUGUST 13, 2010 by guest on March 28, 2020 http://www.jbc.org/ Downloaded from

Transcript of Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K...

Page 1: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

Up-regulation of the ATPase Inhibitory Factor 1 (IF1) of theMitochondrial H�-ATP Synthase in Human Tumors Mediatesthe Metabolic Shift of Cancer Cells to a Warburg Phenotype*□S

Received for publication, May 19, 2010, and in revised form, June 7, 2010 Published, JBC Papers in Press, June 9, 2010, DOI 10.1074/jbc.M110.146480

Laura Sanchez-Cenizo, Laura Formentini, Marcos Aldea, Alvaro D. Ortega, Paula García-Huerta,María Sanchez-Arago, and Jose M. Cuezva1

From the Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de InvestigacionesCientíficas-Universidad Autonoma de Madrid, Centro de Investigacion Biomedica en Red de Enfermedades Raras, ISCIII,Universidad Autonoma de Madrid, 28049 Madrid, Spain

The H�-ATP synthase is a reversible engine of mitochondriathat synthesizes or hydrolyzes ATP upon changes in cell physi-ology. ATP synthase dysfunction is involved in the onset andprogression of diverse human pathologies. During ischemia, theATPhydrolytic activity of the enzyme is inhibitedby theATPaseinhibitory factor 1 (IF1). The expression of IF1 in human tissuesand its participation in thedevelopment of humanpathology areunknown. Here, we have developed monoclonal antibodiesagainst human IF1 and determined its expression in paired nor-mal and tumor biopsies of human carcinomas.We show that therelative mitochondrial content of IF1 increases significantly incarcinomas, suggesting the participation of IF1 in oncogenesis.The expression of IF1 varies significantly in cancer cell lines. Toinvestigate the functional activity of IF1 in cancer, we havemanipulated its cellular content. Overexpression of IF1 or of itspH-insensitive H49K mutant in cells that express low levels ofIF1 triggers the up-regulation of aerobic glycolysis and the inhi-bition of oxidative phosphorylation with concurrent mitochon-drial hyperpolarization. Treatment of the cells with the H�-ATP synthase inhibitor oligomycin mimicked the effects of IF1overexpression. Conversely, small interfering RNA-mediatedsilencing of IF1 in cells that express high levels of IF1 promotesthe down-regulation of aerobic glycolysis and the increase inoxidative phosphorylation. Overall, these findings support thatthe mitochondrial content of IF1 controls the activity of oxida-tive phosphorylation mediating the shift of cancer cells to anenhanced aerobic glycolysis, thus supporting an oncogenic rolefor the de-regulated expression of IF1 in cancer.

In oxidative phosphorylation, ATP is synthesized by themitochondrial ATP synthase, a H�-driven rotatory engine ofthe inner membrane that utilizes as driving force for ATP syn-thesis the H� electrochemical gradient generated by the respi-ratory chain (1–4). The cellular expression level of �-F1-

ATPase,2 which is the catalytic subunit of the H�-ATPsynthase, is diminished in diverse humanpathologies (5), whichinclude cancer (6–9), affording a relevant marker of diseaseprogression (6, 7, 10–12) and of the response to chemotherapy(7, 13–15). Moreover, the down-regulation of �-F1-ATPase inlung carcinomas (12) and colon cancer cells (15) also provides amechanistic explanation to the increased glucose avidity of car-cinomas, i.e. to the enhanced aerobic glycolysis of cancer cells(16, 17). Interestingly, the quantitative determination of �-F1-ATPase relative to the content of glyceraldehyde-3-phosphatedehydrogenase in human tumors has revealed that cancer abol-ishes the tissue-specific differences in the cellular complementof the bioenergetic �-F1-ATPase protein (18).

It is well established that when mitochondrial respiration isimpaired, the H�-ATP synthase can function in reverse actingas an ATP hydrolase for maintaining the proton motive force(1, 19). This process is regulated by an inhibitor peptide calledATPase inhibitory factor 1 or IF1 (19–21), a highly conservednuclearly encoded protein. When matrix pH drops, IF1becomes activated and binds �-F1-ATPase, blocking ATPhydrolysis and preventing a useless waste of energy (20). Thesubstitution of histidine 49 in IF1 by a lysine residue renders amutant form (H49K) that inhibits the ATP hydrolase activity ina pH-insensitive way (22). The structure and in vitro mecha-nismof action of IF1 has been studied in detail, and its role as aninhibitor of the hydrolase activity of the H�-ATP synthase iswell documented (19, 20, 23). However, the information on IF1expression in human tissues and its putative contribution to thedevelopment of human pathology are unknown. In this study,we demonstrate that IF1 is overexpressed in human carcino-mas.Moreover, we document that IF1 plays a regulatory role incontrolling cellular energetic metabolism, strongly supportingits participation as an additional molecular switch used by can-cer cells to trigger the induction of aerobic glycolysis, i.e. theirWarburg phenotype.

EXPERIMENTAL PROCEDURES

Protein Extraction—Frozen tissue sections obtained fromsurgical specimens of untreated cancer patients with primary

* This work was supported by Ministerio de Educacion y Ciencia GrantBFU2007-65253 and the Centro de Investigacion Biomedica en Red deEnfermedades Raras and Comunidad de Madrid Grant S-GEN-0269, Spain.

□S The on-line version of this article (available at http://www.jbc.org) containssupplemental Fig. S1.

1 To whom correspondence should be addressed: Centro de Biología Molec-ular Severo Ochoa, Universidad Autonoma de Madrid, 28049 Madrid,Spain. Tel.: 34-91-196-4618; Fax: 34-91-196-4420; E-mail: [email protected].

2 The abbreviations used are: �-F1-ATPase, � catalytic subunit of the H�-ATPsynthase; IF1, ATPase inhibitory factor 1; FCCP, carbonyl cyanide-p-trifluo-romethoxyphenylhydrazone; siRNA, small interfering RNA; NRK, normal ratkidney.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 285, NO. 33, pp. 25308 –25313, August 13, 2010© 2010 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

25308 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 33 • AUGUST 13, 2010

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

breast (ductal invasive), lung, and colorectal adenocarcinomasas well as squamous lung carcinomas were obtained from theBanco de Tejidos y Tumores, Instituto de Investigaciones Bio-medicas Pi y Suner, Hospital Clinic, Barcelona, Spain. Routinehistopathological study of all cases had been previously per-formed by an experienced pathologist, and the histologicaltype, grade, and size of the tumor aswell as regional lymphnodeinvolvement were recorded (24). Coded samples were receivedto protect patient confidentiality after approval of the projectby the Institutional Review Board. Tissue sections of pairednormal and tumor tissue derived from each patient were pro-cessed (25). Details of the clinicopathological features of thepatients have been recently provided (see Table 1 in Ref. 24).Protein concentration in the extracts was determined with theBradford reagent (Bio-Rad) using bovine serum albumin asstandard.Cloning, Expression, and Purification of Recombinant IF1—

The cDNA (BC009677) encoding human IF1 (AAH09677) wasamplified by PCR using the IMAGE 3877506 clone obtainedfrom the ATCC collection (Manassas, VA) and primers 5�-cgc-gagctcatggcagtgacggc-3� and 5�-atagtttagcggccgcatcatcatgttt-tagc-3�, which add SacI and NotI restriction sites, respectively.The resulting product was purified and first cloned into pGEM-Teasy vector (Promega) and after into pQE-Trisystem (18). Theresulting plasmid, pQE-IF1 that encodes IF1 with C-terminalHis6 and streptavidin tags, was used to transform Escherichiacoli BL-21 cells. Protein expression was induced by addition of1 mM isopropyl 1-thio-�-D-galactopyranoside. After overnightinduction, the cells were collected, and the expressed proteinwas purified using nickel-nitrilotriacetic acid superflow resin(Qiagen) (18).Monoclonal Antibody Production—To produce antibodies

against human IF1, we proceed as described recently (18). Inbrief, BALB/c mice were immunized with various doses of thepurified protein (20 �g) and the hybridomas produced by fus-ing spleen cells with the SP2 myelomas (18). Supernatants ofthe hybridomas were screened by indirect enzyme-linkedimmunosorbent assay on IF1-coated polystyrene plates. Boundantibodies were detected using horseradish peroxidase-labeledgoat anti-mouse antibodies (1:1,000 DAKO, Carpinteria, CA)(18). The positive colonies were cloned by limiting dilution.Mouse monoclonal antibodies were purified with Montageantibody purification kit (Millipore, Billerica; MA).Plasmid Constructs—The pCMV-SPORT6-IF1 plasmid con-

taining human IF1 cDNA was used to generate the IF1-H49Kmutant by standard techniques. In brief, two overlapping frag-ments, including the mutation, were amplified using pCMV-SPORT6-IF1 as template and the following primers: SP6 (att-taggtgacactatag) and H49K reverse (gaacgatttcttcttccttgtgtt),T7 (taatacgactcactataggg) and H49K forward (aacacaaggaagaa-gaaatcgttc). The final product was amplified, digested withEcoRI andNotI, and re-cloned into the pCMV-SPORT6 vector.The pTRE2hyg-IF1 and pTRE2hyg-H49K plasmids were gen-erated frompCMV-SPORT6 constructs using BamHI andNotIrestriction sites and pTRE2hyg (Clontech). The plasmidpTRE2hyg-Luc containing a luciferase gene (luc) was used as acontrol (Clontech). The pBI-L-IF1 and pBI-L-H49K plasmidswere generated following standard cloning techniques from

pCMV-SPORT6-IF1 and pCMV-SPORT6-H49K and the bidi-rectional plasmid pBI-L (Clontech). The pBI-GL plasmid con-tains the lacZ gene, which was used as a control (Clontech). Allthe constructs were checked by sequencing.Cell Cultures, Transfections, and siRNA Silencing—Mouse

hepatoma (Hepa 1–6), normal rat kidney (NRK), human em-bryonic kidney (HEK293T),human hepatocarcinoma (HepG2),breast (HS578T and T47D Tet-Off), colon (HCT116), lung(A549), and cervix (HeLa Tet-Off) carcinoma cells were cul-tured following the suppliers’ indications (ATCC and Clon-tech) up to�70–80% confluence. Transfection of NRK, T47D,HeLa, and Hepa 1–6 cells was performed using Lipofectamineand Plus Reagent (Invitrogen). Transfections of HepG2 cellswere carried out with JetPEI (Polyplus transfections). Optimaltransfection conditions were previously determined for eachcell line. A plasmid encoding a mitochondrial version of greenfluorescent protein (26) was co-transfected with the plasmid ofinterest at a 1:10 ratio to assess transfection efficiencies and forselection of cells in flow cytometry experiments. Harvestedcells were lysed and the lysates cleared by centrifugation (26).Luciferase activity was determined in protein extracts with theluciferase assay system kit (Promega). Luminescence was regis-tered using a FLUOstar OPTIMA (BMG Labtech) plate lumi-nometer. siRNA (Qiagen S100908075) was used to suppressthe expression of IF1 (23). An inefficient siRNA sequence,Silencer� Select Negative Control number 1 plasmid (Ambion/Applied Biosystems), was used as a control.Determination of the Rates of Glycolysis and of Oxygen

Consumption—Twenty four h after seeding the cells, the cul-ture medium was replaced with fresh medium supplementedwith 0.5% fetal bovine serum and with or without 6 �M oligo-mycin. Aliquots of the medium were collected at various times(up to 2 h), and the lactate content was determined enzymati-cally to verify a linear production rate of the metabolite (12).Cellular oxygen consumption rates were determined in anXF24 Extracellular Flux Analyzer (Seahorse Bioscience). Cellswere seeded in XF24-well cell culture microplates (SeahorseBioscience) at �70% confluence and incubated at 37 °C and 7%CO2 for 24 h. The final concentration and order of injectedsubstances was 6 �M oligomycin, 1 �M FCCP, 1 �M rotenone,and 1 �M antimycin.Determination of the Mitochondrial Membrane Potential

(��m)—Transfectedcellsweretreatedwith0.5�Mtetramethyl-rhodamine methyl ester (Molecular Probes) and processed forflow cytometry (26). In some experiments, cells were treatedwith or without 6 �M oligomycin (2 h) or 5 �M FCCP (30 min)before assessing tetramethylrhodamine methyl ester retention.The fluorescence intensity of at least 10,000 events was deter-mined in a FACScan cytometer (BD Biosciences) usingCellQuest (BD Biosciences) acquisition software. Data of thegreen population of transfected cells was analyzed with FlowJosoftware (TreeStar). To estimate��m, the fluorescent signal ofthe cells obtained with FCCP treatment was subtracted fromthe signals obtained without FCCP treatment.Protein Electrophoresis and Western Blot Analysis—Proteins

extracts were fractionated on SDS-PAGE, and the fractionatedproteins were transferred onto polyvinylidene difluoride mem-branes. The primary antibodies used were as follows: rabbit

Inhibition of the H�-ATP Synthase in Cancer

AUGUST 13, 2010 • VOLUME 285 • NUMBER 33 JOURNAL OF BIOLOGICAL CHEMISTRY 25309

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

anti-�-F1-ATPase (1:20,000) andmousemonoclonal anti-glyc-eraldehyde-3-phosphate dehydrogenase (Abcam, 1:20,000),anti-�-tubulin (Sigma, 1:2,500) and anti-IF1 (1:200) antibodies.Peroxidase-conjugated anti-mouse or anti-rabbit IgGs (NordicImmunology, 1:3,000) were used as secondary antibodies. Themitochondrial expression of IF1 in carcinomas, cell lines, andsilencing experiments was assessed relative to the mitochon-drial �-F1-ATPase. In overexpression experiments, we used asloading controls the cellular markers glyceraldehyde-3-phos-phate dehydrogenase and/or tubulin. The ECL systemwas usedto visualize the bands. The intensity of the bandswas quantifiedusing aKodakDC120digital camera and theKodak 1DAnalysissoftware.Statistical Analysis—Statistical analyses were performed

using a two-tailed Student’s t test. Analysis of variancewith posthoc Dunnett’s test was used for multiple comparisons to thecontrol, using the SPSS 17.0 software package. The resultsshown are means � S.E. The number of experiments (n) isindicated. A p � 0.05 was considered statistically significant.

RESULTS

The recombinant human IF1 protein was expressed in bac-teria, purified from inclusion bodies (Fig. 1A), and used toimmunizemice for the development of monoclonal antibodies.

The expression of IF1 in normal human tissues was assessedrelative to the expression of the mitochondrial �-F1-ATPaseprotein (Fig. 1B). Human tissues derived from the digestivetrack had the highest relative expression level of IF1 (Fig. 1B),whereas breast and lung expressed nondetectable levels of IF1(Fig. 1B). The relative expression of IF1 in cell lines indicated avery large variability in the mitochondrial content of theATPase inhibitor (Fig. 1C). Immunofluorescence microscopyrevealed the mitochondrial localization of IF1 (Fig. 1D). Next,we analyzed the relative expression of IF1 (IF1/�-F1-ATPaseratio) in paired normal and tumor biopsies derived from breast,colon, and lung cancer patients (Fig. 1, E–H). Remarkably, in allhuman tissues analyzed carcinogenesis promoted a significantincrease in the relative tumor content of IF1 (Fig. 1, E–H).The augmented expression of IF1 in tumors (Fig. 1, E–H)

suggested that the protein might regulate the energetic metab-olism of aerobic cells bymodulating the activity of theH�-ATPsynthase. Therefore, cell lines displaying low basal levels of IF1(Fig. 1C) were transiently transfected with control, IF1, or themutant H49K plasmid, and the glycolytic flux was determined(Fig. 2). The NRK (Fig. 2A) and Hepa 1–6 (Fig. 2B) cell linesshowed a significant increase in aerobic glycolysis when IF1 orthe mutant H49K was overexpressed. Similar findings were

FIGURE 1. Expression of IF1 in human tumors. A, purification of recombinant IF1 (r-IF1). The gel shows protein extracts from noninduced (�) and induced (�)bacterial extracts and the purified recombinant IF1. B and C, IF1 and �-F1-ATPase (�F1) expression in different human tissues (B) and cell lines (C). The migrationof the native IF 12-kDa isoform (n-IF1) is indicated. In the right panel, two different exposures of the IF1 film are presented. Cells with high and low IF1 contentcould be distinguished. D, immunofluorescence microscopy of HeLa cells stained with 200 nM MitoTracker (red) and with the IF1 monoclonal antibody (green)revealing the co-localization (Merge) of IF1 in mitochondria. Images are shown at 63 magnification. Bar, 20 �m. E–H, Western blots of IF1 and �-F1-ATPase(�F1) in paired normal (N) and tumor (T) biopsies derived from three representative patients are shown. The histograms represent the fold of control of theIF1/�F1 ratio in ductal invasive breast (E, n 9), colon (F, n 12), and lung (G, n 15) adenocarcinomas and squamous lung carcinomas (H, n 7) relative topaired normal samples. *, p � 0.05 when compared with normal by Student’s t test.

Inhibition of the H�-ATP Synthase in Cancer

25310 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 33 • AUGUST 13, 2010

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

obtained when the cells were treated with the H�-ATP syn-thase inhibitor oligomycin (Fig. 2,A and B). In HepG2 cells, theeffect of IF1 or H49K was less marked than that triggered byoligomycin (Fig. 2C), but it reached statistical significancewhenthemutantH49Kwas overexpressed (Fig. 2C), perhaps becauseof the low transfection efficiencies attained in this cell line. Sim-ilar experiments in cells that naturally overexpress high levels ofIF1 (supplemental Fig. S1) revealed no significant effect of IF1or H49K in the flux of aerobic glycolysis.

Determination of oligomycin-sensitive respiration in NRK cellstransfected with IF1 showed thatthe oxygen consumption rates weresignificantly diminished when com-pared with controls (Fig. 3A), indi-cating that IF1 is interfering withthe activity of the H�-ATP syn-thase. A more stringent inhibitionof oxidative phosphorylation wasnoted when the cells expressed thepH-insensitive H49K mutant (Fig.3A), consistent with the nonregu-lated binding activity of the mutantto �-F1-ATPase (22).In normal aerobic conditions, the

H�-ATP synthase utilized the H�

gradient for the synthesis of ATP. Ifthe enzyme is inhibited, mitochon-

drial hyperpolarization ensues due to the interruption of thebackflow of H� into the matrix. Consistently, when any of thecell lines under study were treated with oligomycin, the mito-chondrial membrane potential (��m) increased (Fig. 3, B–D).Similarly, NRK (Fig. 3B) and Hepa 1–6 (Fig. 3C) cells trans-fected with IF1 or H49K showed significant increases in ��mwhen compared with control cells. The same studies in HepG2cells using a bidirectional plasmid with a doxycycline-regulatedpromoter that expresses luciferase and any of the two trans-genes confirmed that IF1 or H49K promoted an increase in��m (Fig. 3D).The siRNA-mediated silencing of IF1 in HeLa cells that

naturally overexpress the inhibitor (Fig. 1C) promoted alarge and significant reduction in the rates of aerobic glycol-ysis (Fig. 4A). Remarkably, oligomycin treatment of IF1-si-lenced cells restored the original rates of aerobic glycolysis(Fig. 4A), strongly supporting the link between the bioener-getic activity of mitochondria and glucose consumptionrates (12, 15). Consistently, the determination of the activityof oxidative phosphorylation indicated that IF1-silencedcells had a higher oligomycin sensitive respiratory rate thancontrols (Fig. 4B).

DISCUSSION

We show for the first time that human carcinomas have anincreased expression of IF1. We support that the expressionlevel of IF1 participates in the regulation of tumor energeticmetabolism by controlling the synthase activity of the H�-ATPsynthase. Indeed, cells that overexpress IF1 or H49K show aremarkable increase in their rates of aerobic glycolysis and asignificant decrease in the activity of oxidative phosphorylationconcurrent with an increase in��m. All these findings are con-sistent with IF1 interfering with the activity of the H�-ATPsynthase as illustrated by the results obtained when using theH�-ATP synthase inhibitor oligomycin. Moreover, the silenc-ing of IF1 resulted in a sharp reduction in the rates of aerobicglycolysis and the stimulation of oxidative phosphorylation.These findings are of utmost importance for understanding themetabolism of cancer cells (9, 27).

FIGURE 2. Overexpression of IF1 or H49K triggers the induction of aerobic glycolysis. A–C, histogramsshow the changes in the rates of aerobic glycolysis (Ctr, open bars) mediated by the overexpression of IF1(dotted bars) or H49K (hatched bars) in NRK (A), Hepa 1– 6 (B), and HepG2 (C) cells, as representative examples ofcell lines with negligible or low content of IF1 (Fig. 1C). For comparison, the effect of 6 �M oligomycin (OL.)treatment (closed bars) on the rates of aerobic glycolysis in the same cellular types (A–C) is shown. *, p � 0.05when compared with control by Student’s t test. Multiple comparisons by analysis of variance and post hocDunnett’s tests confirmed the statistical significance reported except in HepG2 cells. GAPDH, glyceraldehyde-3-phosphate dehydrogenase; ns, nonsignificant.

FIGURE 3. Overexpression of IF1 or H49K triggers the inhibition of oxida-tive phosphorylation (A) and mitochondrial hyperpolarization (B–D).A, NRK cells were transfected with green fluorescent (control (Ctr), open bar),IF1 (dotted bar), or H49K (hatched bar) plasmids, and the effect on oligomycin-sensitive respiration (OSR) was determined. *, #, p � 0.05 when comparedwith control or IF1 by Student’s t test, respectively. B–D, histograms show therelative changes in the mitochondrial membrane potential (��m) (open bars)mediated by the overexpression of IF1 (dotted bars) or H49K (hatched bars) inNRK (B), Hepa 1– 6 (C), and HepG2 (D) cells. The effect of 6 �M oligomycin (OL)(closed bars) in ��m (B–D) is shown. D, HepG2 Tet-On advanced cell line wastransfected with a bidirectional luciferase-galactosidase (control), a lucifer-ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity or in ��m in induced (�Dox)and noninduced (�Dox) cells are shown. *, #, p � 0.05 when compared withcontrol or noninduced cells by Student’s t test, respectively. In all panels,multiple comparisons by analysis of variance and post hoc Dunnett’s testsconfirmed the statistical significance reported.

Inhibition of the H�-ATP Synthase in Cancer

AUGUST 13, 2010 • VOLUME 285 • NUMBER 33 JOURNAL OF BIOLOGICAL CHEMISTRY 25311

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

The functional role of IF1 as a natural inhibitor of the activityof the ATP synthase is debated (21, 23, 28). In this regard, arecent study claims that the overexpression of IF1 in HeLa cellstriggers the down-regulation of��m (23). This result contrastswith the findings reported in our study. We suggest that thehigh basal expression of IF1 in some cell lines (Fig. 1C) hampersthe detection of reliable physiological responses in overexpres-sion experiments. In fact, cells lines with high basal expressionof the protein (HeLa and T47D, see Fig. 1C) revealed nochanges in glycolysis and in ��m (supplemental Fig. S1). Thebinding of IF1 to �-F1-ATPase is regulated by the energeticstate of mitochondria (19, 20). However, it is reasonable to sug-gest that IF1 binding is also dependent on the mass action ratioand, in situations of increased IF1 expression such as in certaintumors (Fig. 1, E–H) and cancer cells (Fig. 1C), the protein willinhibit both the synthetic and hydrolytic activities. In agree-ment with our proposal, recent findings indicate that deletionof IEX-1, a stress-inducible gene that apparently targets IF1 fordegradation, results in the inhibition of theATP synthase activ-ity in vivo (29).A hallmark of the cancer phenotype is the reprogramming of

energetic metabolism toward glycolysis (9, 16, 27, 30). Severalmechanisms directly promoting glycolysis, the alteration of thebioenergetic function of mitochondria or both have been pro-posed to explain theWarburg phenotype (Refs. 9, 27, 30, 31 andreferences therein).More recently, deregulation of the E3 ubiq-uitin ligaseAPC/C-Cdh1 (32) and c-myc (33) has been shown topromote the glycolytic shift of cancer cells by controlling theturnover (32) and the splicing (33) of glycolysis-promotingenzymes. We propose that the increased expression of IF1 intumors promotes the metabolic switch in cancer cells acting atthe level of mitochondrial oxidative phosphorylation. Three

independent mechanisms affect the overall bioenergetic activ-ity of the H�-ATP synthase in cancer. Two of them limit thetumor content of the catalytic �-F1-ATPase (7, 9, 12) either bymasking the translation of the transcript (34, 35), as recentlyshown in breast, colon, and lung tumors (24), or by limiting theamount of �-F1-ATPase mRNA after hypermethylation of thepromoter of theATP5B gene in leukemia (36). The thirdmech-anism limits the activity of the complex and is triggered byincreasing themitochondrial content of IF1 as described in thisstudy.It is worth mentioning that IF1 not only plays a role in limit-

ing oxidative phosphorylation and thus in promoting glycolysis.Glycolysis is known to provide an advantageous phenotype thatfavors cellular proliferation and invasion (15, 37). Moreover,limiting the activity of theH�-ATP synthase also contributes totumor growth because oxidative phosphorylation is requiredfor the efficient execution of cell death (38–40). We suggestthat studies aimed at characterizing the mechanisms that reg-ulate IF1 expression in cancer as well as the basic cell biology ofthe protein will provide promising targets to halt diseaseprogression.

Acknowledgments—We are indebted to J. Satrustegui for helpful dis-cussions and critical reading of the manuscript. M. Chamorro and C.Nunez de Arenas are acknowledged for expert technical assistance.The Xarxa de Bancs de Tumors de Catalunya is acknowledged. TheCentro de Biología Molecular Severo Ochoa is recipient of an institu-tional grant from Fundacion Ramon Areces.

REFERENCES1. Boyer, P. D. (1997) Annu. Rev. Biochem. 66, 717–7492. Mitchell, P. (1961) Nature 191, 144–1483. Abrahams, J. P., Leslie, A. G., Lutter, R., and Walker, J. E. (1994) Nature

370, 621–6284. Yoshida,M.,Muneyuki, E., andHisabori, T. (2001)Nat. Rev.Mol. Cell Biol.

2, 669–6775. Kucharczyk, R., Zick, M., Bietenhader, M., Rak, M., Couplan, E., Blondel,

M., Caubet, S. D., and di Rago, J. P. (2009) Biochim. Biophys. Acta 1793,186–199

6. Cuezva, J. M., Krajewska, M., de Heredia, M. L., Krajewski, S., Santamaría,G., Kim, H., Zapata, J. M., Marusawa, H., Chamorro, M., and Reed, J. C.(2002) Cancer Res. 62, 6674–6681

7. Lin, P. C., Lin, J. K., Yang, S. H., Wang, H. S., Li, A. F., and Chang, S. C.(2008) Int. J. Colorectal. Dis. 23, 1223–1232

8. Unwin, R. D., Craven, R. A., Harnden, P., Hanrahan, S., Totty,N., Knowles,M., Eardley, I., Selby, P. J., andBanks, R. E. (2003)Proteomics 3, 1620–1632

9. Cuezva, J.M., Ortega, A. D.,Willers, I., Sanchez-Cenizo, L., Aldea,M., andSanchez-Arago, M. (2009) Biochim. Biophys. Acta 1792, 1145–1158

10. Isidoro, A., Casado, E., Redondo, A., Acebo, P., Espinosa, E., Alonso, A.M.,Cejas, P., Hardisson, D., Fresno Vara, J. A., Belda-Iniesta, C., Gonzalez-Baron, M., and Cuezva, J. M. (2005) Carcinogenesis 26, 2095–2104

11. Cuezva, J. M., Chen, G., Alonso, A. M., Isidoro, A., Misek, D. E., Hanash,S. M., and Beer, D. G. (2004) Carcinogenesis 25, 1157–1163

12. Lopez-Ríos, F., Sanchez-Arago, M., García-García, E., Ortega, A. D., Ber-rendero, J. R., Pozo-Rodríguez, F., Lopez-Encuentra, A., Ballestín, C., andCuezva, J. M. (2007) Cancer Res. 67, 9013–9017

13. Shin, Y. K., Yoo, B. C., Chang, H. J., Jeon, E., Hong, S. H., Jung, M. S., Lim,S. J., and Park, J. G. (2005) Cancer Res. 65, 3162–3170

14. Hernlund, E., Hjerpe, E., Avall-Lundqvist, E., and Shoshan,M. (2009)Mol.Cancer Ther. 8, 1916–1923

15. Sanchez-Arago, M., Chamorro, M., and Cuezva, J. M. (2010) Carcinogen-esis 31, 567–576

FIGURE 4. Silencing of IF1 triggers the inhibition of aerobic glycolysis andactivates oxidative phosphorylation. The rates of aerobic glycolysis (A) andoxidative phosphorylation (B) were determined in HeLa cells expressing aninefficient (control: Ctr, open bars) or IF1 siRNA to promote the silencing of IF1(closed bars). A, effect of 6 �M oligomycin treatment (OL. �) is shown in controland IF1-silenced cells. *, and #, p � 0.05 when compared with control oroligomycin-treated cells by Student’s t test, respectively.

Inhibition of the H�-ATP Synthase in Cancer

25312 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 285 • NUMBER 33 • AUGUST 13, 2010

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

16. Warburg, O. (1956) Science 123, 309–31417. Warburg, O. (1956) Science 124, 269–27018. Acebo, P., Giner, D., Calvo, P., Blanco-Rivero, A., Ortega, A. D., Fernan-

dez, P. L., Roncador, G., Fernandez-Malave, E., Chamorro, M., and Cu-ezva, J. M. (2009) Transl. Oncol. 2, 138–145

19. Gledhill, J. R., Montgomery, M. G., Leslie, A. G., and Walker, J. E. (2007)Proc. Natl. Acad. Sci. U.S.A. 104, 15671–15676

20. Cabezon, E., Montgomery, M. G., Leslie, A. G., and Walker, J. E. (2003)Nat. Struct. Biol. 10, 744–750

21. Pullman, M. E., and Monroy, G. C. (1963) J. Biol. Chem. 238, 3762–376922. Cabezon, E., Butler, P. J., Runswick, M. J., and Walker, J. E. (2000) J. Biol.

Chem. 275, 25460–2546423. Campanella, M., Casswell, E., Chong, S., Farah, Z., Wieckowski, M. R.,

Abramov,A. Y., Tinker, A., andDuchen,M. R. (2008)CellMetab.8, 13–2524. Willers, I.M., Isidoro, A., Ortega, A. D., Fernandez, P. L., andCuezva, J.M.

(2010) Biochem. J. 426, 319–32625. Isidoro, A., Martínez, M., Fernandez, P. L., Ortega, A. D., Santamaría, G.,

Chamorro,M., Reed, J. C., andCuezva, J.M. (2004)Biochem. J. 378, 17–2026. Martínez-Diez,M., Santamaría, G., Ortega, A. D., andCuezva, J.M. (2006)

PLoS ONE 1, e10727. Vander Heiden,M. G., Cantley, L. C., and Thompson, C. B. (2009) Science

324, 1029–103328. Walker, J. E. (1994) Curr. Opin. Struct. Biol. 4, 912–91829. Shen, L., Zhi, L., Hu, W., and Wu, M. X. (2009) Cell Death Differ. 16,

603–61230. Kroemer, G., and Pouyssegur, J. (2008) Cancer Cell 13, 472–48231. Semenza, G. L., Artemov, D., Bedi, A., Bhujwalla, Z., Chiles, K., Feldser, D.,

Laughner, E., Ravi, R., Simons, J., Taghavi, P., and Zhong, H. (2001) No-vartis Found. Symp. 240, 251–264

32. Almeida, A., Bolanos, J. P., and Moncada, S. (2010) Proc. Natl. Acad. Sci.U.S.A. 107, 738–741

33. David, C. J., Chen, M., Assanah, M., Canoll, P., and Manley, J. L. (2010)Nature 463, 364–368

34. de Heredia, M. L., Izquierdo, J. M., and Cuezva, J. M. (2000) J. Biol. Chem.275, 7430–7437

35. Ortega, A. D.,Willers, I.M., Sala, S., and Cuezva, J.M. (2010) J. Cell. Sci., inpress

36. Li, R. J., Zhang,G. S., Chen, Y.H., Zhu, J. F., Lu,Q. J., Gong, F. J., andKuang,W. Y. (2010) Ann. Oncol. 21, 1506–1514

37. Fang, J. S., Gillies, R. D., and Gatenby, R. A. (2008) Semin. Cancer. Biol. 18,330–337

38. Matsuyama, S., Xu, Q., Velours, J., and Reed, J. C. (1998) Mol. Cell 1,327–336

39. Santamaría, G., Martínez-Diez, M., Fabregat, I., and Cuezva, J. M. (2006)Carcinogenesis 27, 925–935

40. Tomiyama, A., Serizawa, S., Tachibana, K., Sakurada, K., Samejima, H.,Kuchino, Y., and Kitanaka, C. (2006) J. Natl. Cancer Inst. 98, 1462–1473

Inhibition of the H�-ATP Synthase in Cancer

AUGUST 13, 2010 • VOLUME 285 • NUMBER 33 JOURNAL OF BIOLOGICAL CHEMISTRY 25313

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: Up-regulationoftheATPaseInhibitoryFactor1(IF1)ofthe ... · ase-IF1 (IF1), or a luciferase-H49K (H49K) doxycycline (Dox.)-responsive plas-mid. The relative changes in luciferase activity

García-Huerta, María Sánchez-Aragó and José M. CuezvaLaura Sánchez-Cenizo, Laura Formentini, Marcos Aldea, Álvaro D. Ortega, Paula

a Warburg Phenotype-ATP Synthase in Human Tumors Mediates the Metabolic Shift of Cancer Cells to

+Up-regulation of the ATPase Inhibitory Factor 1 (IF1) of the Mitochondrial H

doi: 10.1074/jbc.M110.146480 originally published online June 9, 20102010, 285:25308-25313.J. Biol. Chem. 

  10.1074/jbc.M110.146480Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

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

Supplemental material:

  http://www.jbc.org/content/suppl/2010/06/09/M110.146480.DC1

  http://www.jbc.org/content/285/33/25308.full.html#ref-list-1

This article cites 39 references, 12 of which can be accessed free at

by guest on March 28, 2020

http://ww

w.jbc.org/

Dow

nloaded from