essential role of mitochondria in apoptosis of cancer ... · essential role of mitochondria in...

12
Essential role of mitochondria in apoptosis of cancer cells induced by the marine alkaloid Lamellarin D Caroline Ballot, 1 Jérome Kluza, 1 Alain Martoriati, 1 Ulrika Nyman, 2 Pierre Formstecher, 1 Bertrand Joseph, 2 Christian Bailly, 1 and Philippe Marchetti 1,3 1 Inserm U837 and Faculté de Médecine, Université de Lille II 1 Place Verdun Cedex, France; 2 Department of Oncology- Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden; and 3 Plate-forme de Biothérapie, Centre de Bio-Pathologie, et Banque de Tissus, CHRU Lille, France Abstract Lamellarin D, a potent cytotoxic marine alkaloid, exerts its antitumor action through two complementary path- ways: a nuclear route via topoisomerase I inhibition and a mitochondrial targeting. The present study was designed to investigate the contribution of these two pathways for apoptosis in cancer cells. Lamellarin D pro- moted nuclear apoptosis in leukemia cells without prom- inent cell cycle arrest. Signals transmitted by lamellarin D initiated apoptosis via the intrinsic apoptotic pathway. The drug induced conformational activation of Bax and decreased the expression levels of antiapoptotic proteins Bcl-2 and cIAP2 in association with activation of cas- pase-9 and caspase-3. Upon lamellarin D exposure, Fas and Fas-L expression was not modified in leukemia cells. Moreover, leukemia cells deficient in caspase-8 or Fas-associated protein with death domain underwent ap- optosis through the typical mitochondrial apoptotic cas- cade, indicating that cell death induced by lamellarin D was independent of the extrinsic apoptotic pathway. La- mellarin D also exerted a topoisomerase I mediated DNA damage response resulting in H2AX phosphoryla- tion, and the upregulation of the DNA repair protein Rad51 and of p53, as well as the phosphorylation of p53 at serine 15. However, lamellarin D killed efficiently mutat- ed p53 or p53 null cancer cells, and sensitivity to lamellarin D was abrogated neither by cycloheximide nor in enu- cleated cells. Lamellarin Dinduced cytochrome c release occurs independently of nuclear factors in a cell-free sys- tem. These results suggest that lamellarin D exerts its cytotoxic effects primarily by inducing mitochondrial apo- ptosis independently of nuclear signaling. Thus, lamellarin D constitutes a new proapoptotic agent that may bypass certain forms of apoptosis resistance that occur in tumor cells. [Mol Cancer Ther 2009;8(12): 330717] Introduction Lamellarins represent a family of hexacyclic pyrrole alka- loids originally derived from marine invertebrates and have been shown to exert antineoplastic effects (1). Among lamel- larins, the natural compound lamellarin D (Fig. 1A) has been shown to inhibit a large panel of tumor cell types in vitro. At doses in the micromolar range, lamellarin D ex- hibits high apoptotic activities in leukemia cell lines (2, 3) characterized by the dissipation of the mitochondrial mem- brane potential (Δψm), the release of cytochrome c from mi- tochondria, and caspase-3 activation (2). The proapoptotic effects of lamellarin D can result from several mechanisms. Lamellarin D was first described to inhibit nuclear topoi- somerase I (4). In vitro, lamellarin D induced DNA cleavage with an efficiency just slightly lower than the prototype to- poisomerase I inhibitor campthotecin (5). Structure-activity studies indicate a good correlation between topoisomerase I inhibition and cytotoxicity of lamellarin D derivatives, lead- ing to the suggestion that one of the proapoptotic targets of lamellarin D is in the cell nucleus (5). Nevertheless, its role in apoptosis still needs to be shown. Interestingly, topoi- somerase I-mutated cancer cells are not totally resistant to- ward lamellarin Dinduced apoptosis, suggesting that induction of apoptosis could depend on other intracellular targets. We have previously reported strong evidence that lamellarin D and its amino synthetic derivative PM031379 can trigger apoptosis through a direct increase in mitochon- drial permeability (2). These results were obtained using a cell-free system of apoptosis in which supernatants of puri- fied mitochondria treated with lamellarin D were mixed with isolated nuclei. However, we could not conclude that apoptosis in intact cells results exclusively from this direct action on mitochondria. Thus, how lamellarin D triggers cell death still deserves further investigation. Most of the conventional anticancer treatments are thought to induce cell death through indirect activation of the mitochondria-dependent pathway of apoptosis, a path- way often found altered in drug-resistant cancer cells (6). Received 7/13/09; revised 10/12/09; accepted 10/23/09; published OnlineFirst 12/1/09. Grant support: INSERM, UNIVERSITÉ DE LILLE II and the Ligue Nationale contre le Cancer (Comité du Nord; to P. Marchetti). C. Ballot was recipient of grant from INSERM-Région Nord Pas de Calais. J. Kluza received a fellowship from Association pour la Recherche sur le Cancer (ARC). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Note: Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). The current address for C. Bailly is Institut de Recherche Pierre Fabre, 3 rue des satellites, BP94244, 31432 Toulouse, France. Requests for reprints: Philippe Marchetti, INSERM U 837 Faculté de médecine, 1, place Verdun F- 59045 Lille Cedex France. Phone: 33-3-20- 62-69-52; Fax: 33-3-20-62-68-84. E-mail: [email protected] Copyright © 2009 American Association for Cancer Research. doi:10.1158/1535-7163.MCT-09-0639 Mol Cancer Ther 2009;8(12). December 2009 3307 on May 15, 2020. © 2009 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

Upload: others

Post on 16-May-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

3307

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

Essential role of mitochondria in apoptosis of cancer cellsinduced by the marine alkaloid Lamellarin D

Caroline Ballot,1 Jérome Kluza,1 Alain Martoriati,1

Ulrika Nyman,2 Pierre Formstecher,1

Bertrand Joseph,2 Christian Bailly,1

and Philippe Marchetti1,3

1Inserm U837 and Faculté de Médecine, Université de Lille II 1Place Verdun Cedex, France; 2Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), KarolinskaInstitutet, Stockholm, Sweden; and 3Plate-formede Biothérapie, Centre de Bio-Pathologie, et Banque deTissus, CHRU Lille, France

AbstractLamellarin D, a potent cytotoxic marine alkaloid, exertsits antitumor action through two complementary path-ways: a nuclear route via topoisomerase I inhibitionand a mitochondrial targeting. The present study wasdesigned to investigate the contribution of these twopathways for apoptosis in cancer cells. Lamellarin D pro-moted nuclear apoptosis in leukemia cells without prom-inent cell cycle arrest. Signals transmitted by lamellarinD initiated apoptosis via the intrinsic apoptotic pathway.The drug induced conformational activation of Bax anddecreased the expression levels of antiapoptotic proteinsBcl-2 and cIAP2 in association with activation of cas-pase-9 and caspase-3. Upon lamellarin D exposure,Fas and Fas-L expression was not modified in leukemiacells. Moreover, leukemia cells deficient in caspase-8 orFas-associated protein with death domain underwent ap-optosis through the typical mitochondrial apoptotic cas-cade, indicating that cell death induced by lamellarin Dwas independent of the extrinsic apoptotic pathway. La-mellarin D also exerted a topoisomerase I–mediatedDNA damage response resulting in H2AX phosphoryla-

Received7/13/09; revised10/12/09; accepted10/23/09;publishedOnlineFirst12/1/09.

Grant support: INSERM, UNIVERSITÉ DE LILLE II and the Ligue Nationalecontre le Cancer (Comité du Nord; to P. Marchetti). C. Ballot was recipientof grant from INSERM-Région Nord Pas de Calais. J. Kluza received afellowship from Association pour la Recherche sur le Cancer (ARC).

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 toindicate this fact.

Note: Supplementary material for this article is available at MolecularCancer Therapeutics Online (http://mct.aacrjournals.org/).

The current address for C. Bailly is Institut de Recherche Pierre Fabre,3 rue des satellites, BP94244, 31432 Toulouse, France.

Requests for reprints: Philippe Marchetti, INSERM U 837 Faculté demédecine, 1, place Verdun F- 59045 Lille Cedex France. Phone: 33-3-20-62-69-52; Fax: 33-3-20-62-68-84. E-mail: [email protected]

Copyright © 2009 American Association for Cancer Research.

doi:10.1158/1535-7163.MCT-09-0639

Mol Cancer Ther 2009;8(12). December 2009

on May 15, 2020.mct.aacrjournals.org Downloaded from

tion, and the upregulation of the DNA repair proteinRad51 and of p53, as well as the phosphorylation of p53at serine 15. However, lamellarin D killed efficiently mutat-ed p53 or p53 null cancer cells, and sensitivity to lamellarinD was abrogated neither by cycloheximide nor in enu-cleated cells. Lamellarin D–induced cytochrome c releaseoccurs independently of nuclear factors in a cell-free sys-tem. These results suggest that lamellarin D exerts itscytotoxic effects primarily by inducing mitochondrial apo-ptosis independently of nuclear signaling. Thus, lamellarinD constitutes a new proapoptotic agent that may bypasscertain forms of apoptosis resistance that occur in tumorcells. [Mol Cancer Ther 2009;8(12): 3307–17]

IntroductionLamellarins represent a family of hexacyclic pyrrole alka-loids originally derived from marine invertebrates and havebeen shown to exert antineoplastic effects (1). Among lamel-larins, the natural compound lamellarin D (Fig. 1A) hasbeen shown to inhibit a large panel of tumor cell typesin vitro. At doses in the micromolar range, lamellarin D ex-hibits high apoptotic activities in leukemia cell lines (2, 3)characterized by the dissipation of the mitochondrial mem-brane potential (Δψm), the release of cytochrome c from mi-tochondria, and caspase-3 activation (2). The proapoptoticeffects of lamellarin D can result from several mechanisms.Lamellarin D was first described to inhibit nuclear topoi-somerase I (4). In vitro, lamellarin D induced DNA cleavagewith an efficiency just slightly lower than the prototype to-poisomerase I inhibitor campthotecin (5). Structure-activitystudies indicate a good correlation between topoisomerase Iinhibition and cytotoxicity of lamellarin D derivatives, lead-ing to the suggestion that one of the proapoptotic targets oflamellarin D is in the cell nucleus (5). Nevertheless, its rolein apoptosis still needs to be shown. Interestingly, topoi-somerase I-mutated cancer cells are not totally resistant to-ward lamellarin D–induced apoptosis, suggesting thatinduction of apoptosis could depend on other intracellulartargets. We have previously reported strong evidence thatlamellarin D and its amino synthetic derivative PM031379can trigger apoptosis through a direct increase in mitochon-drial permeability (2). These results were obtained using acell-free system of apoptosis in which supernatants of puri-fied mitochondria treated with lamellarin D were mixedwith isolated nuclei. However, we could not conclude thatapoptosis in intact cells results exclusively from this directaction on mitochondria. Thus, how lamellarin D triggers celldeath still deserves further investigation.Most of the conventional anticancer treatments are

thought to induce cell death through indirect activation ofthe mitochondria-dependent pathway of apoptosis, a path-way often found altered in drug-resistant cancer cells (6).

© 2009 American Association for Cancer Research.

Mitochondria, DNA damage, and Lamellarin D3308

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

In most cases, chemotherapeutic drugs first interact with anintracellular target resulting in stress signals that secondarilyconverge to mitochondria and finally result in apoptotic celldeath. The best-known intracellular target of conventionalanticancer drugs is nuclear DNA. Numerous cytotoxicagents, including topoisomerase I and II inhibitors, primar-ily damage nuclear DNA. As a consequence, DNA insults, inthe absence of efficient repair, lead to cell apoptosis throughactivation of complex molecular cascades (7). The tumorsuppressor protein p53 is at the center of the process cou-pling DNA damage to the initiation of apoptosis. In responseto DNA insults, intracellular levels of p53 increase andpromote the transcription of several proapoptotic genes ofthe bcl-2 family such as the BH3-only genes, noxa, puma,and bax (7). Additionally, the translocation of p53 from thenucleus to the mitochondria is associated with cell death ina transcription-independent manner (8).Following stress signals generated by conventional

treatments, the permeability of mitochondrial membranesis increased, leading to the release of proapoptotic pro-teins which in turn initiate the caspase cascade and finallyresult in cell death (7). The ratio of proapoptotic (e.g.,Bax, Bak, Noxa, Puma) and antiapoptotic members (e.g.,Bcl-2, Bcl-xl, Mcl-1) of the Bcl-2 family affects the perme-ability of mitochondrial membranes, thereby determiningthe sensitivity of cells to apoptotic signals (9). Thus, theexpression level of antiapoptotic Bcl-2 proteins correlateswith the resistance to a large spectrum of chemotherapeu-tic agents (9).In this study, we examined the molecular circuits through

which lamellarin D promotes cancer cell apoptosis. In par-ticular, we investigated the role of the cell nucleus in lamel-larin D–induced apoptosis. An understanding of themechanisms by which lamellarin D causes apoptosis wouldpermit identification of the most valuable molecularly tar-geted strategy for drug development.

Materials and MethodsChemicals

Camptothecin was purchased from Sigma, and lamellarinD was synthesized at Pharmamar. Several caspase inhibitorswere used: the pan–caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(Ome)-fluoromethylketone (z-VAD.fmk;Sigma), the caspase-1 inhibitor benzyloxycarbonyl-Tyr-Val-Ala-Asp-chloromethylketone (z-YVAD.cmk; Bachem), thecaspase-2 inhibitor benzyloxycarbonyl-Val-Asp-Val-Ala-Asp-fluoromethylketone (z-VDVAD.fmk; Calbiochem,Chemicon International), the caspase-3 inhibitor benzyloxy-carbonyl-Val-Glu-Val-Asp-chloromethylketone (z-DEVD.cmk; Pharmingen BD Biosciences), the caspase-8 inhibitorbenzyloxycarbonyl-Ile-Glu-Thr-Asp-fluoromethylketone(z-IETD.fmk; 50 μmol/L; Pharmingen BD Biosciences), andthe caspase-9 inhibitor benzyloxycarbonyl-Leu-Glu-His-Asp-fluoromethylketone (z-LEHD.fmk; Pharmingen BDBiosciences). 4′,6-Diamidino-2-phenylindole (DAPI) and tet-ramethylrhodamine methyl ester (TMRM) were purchasedfromMolecular Probes. Other compounds were from Sigma.

on May 15, 2020.mct.aacrjournals.org Downloaded from

Cell Lines

The mouse leukemia P388 cell line and P388CPT5, itstopoisomerase I–mutated subclone (kindly provided byPr. J.-F. Riou, Museum National d'Histoire Naturelle, Paris,France), were cultured as described (2). 2B411 T hybridomacells stably transfected with a SFFVneo vector containingthe human bcl-2 gene or the neomycin resistance gene(neo) only (kindly provided by Dr. Jonathan Ashwell,NIH) and the cell lines Jurkat, Jurkat I 9.2 (caspase-8 defi-cient), and Jurkat I 2.1 (FADD-deficient) were cultured inRPMI-1640 medium supplemented with 10% FCS, 2mmol/L L-glutamine, penicillin (100 U/mL), and strepto-mycin (100 mg/mL). Human colon carcinoma HCT116 celllines, wild-type HCT116-p53+/+, and its p53 null derivativeHCT116-p53−/− (a generous gift from Dr B. Vogelstein,Johns Hopkins University, Baltimore, MD) were culturedin DMEM supplemented with 10% heat-inactivated FCS,2mmol/L L-glutamine, penicillin (100 U/mL), and strepto-mycin (100 mg/mL). Osteosarcoma Saos-2 cells weregrown as previously described (10).Generation of Enucleated Cells (Cytoplasts)

P388 cells (2 × 107/mL) were cultured in the presenceof 10 μg/mL cytochalasin B and 20 U/mL of DNase for45 min at 37°C following procedures described previously(11), with minor modifications. The cell suspension waslayered onto a discontinuous Ficoll density gradient(2 mL of 25%, 2 mL of 17%, 0.5 mL of 16%, 0.5 mL of15%, and 2 mL of 12.5% Ficoll in complete medium con-taining 7.5 μg/mL cytochalasin B and 40 U/mL of DNasepreequilibrated in ultracentrifuge tubes at 37°C in a CO2

incubator for 24 h). Gradients containing cell suspensionswere centrifuged in a prewarmed SW41 rotor (Beckman)at 25,000 rpm for 30 min at 30°C. Cytoplasts were collect-ed from the interface between 15% and 17% Ficoll layers,washed in complete medium (supplemented with insulintransferrin and selenium), and cultured overnight at 37°Cbefore treatment with 5 μmol/L lamellarin D for 14 h.Cells were then stained with TMRM or FITC-VAD.fmkprior to the cytofluorometric analysis.Determination of Nuclear Apoptosis

The frequency of hypoploid (sub-G1) cells was assessed asdescribed (12). DNA fragmentation was determined by aga-rose gel electrophoresis as already described (13).Caspase Activity Assay

The activity of caspases was determined with specific pro-luminescent caspase-3, caspase-2, caspase-8, and caspase-9substrates (Caspase-Glo luminescent kits from PromegaCorp.) in triplicate following the manufacturer's instruc-tions. Luminescence was recorded with a Lumicount lumin-ometer (Packard Instrument Company).Immunoblot Analysis

Whole cell lysates were prepared as described (12). Equalprotein quantities (50 μg) were subjected to SDS-PAGE.Membranes were blocked in 5% powdered milk in TBSTween 0.05% for 1 h at room temperature and then incubat-ed with primary antibodies specific for poly(ADP-ribose)polymerase (PARP) 1/2 (1:500; Santa Cruz, H-250), Puma(1:1,000; Cell Signaling), Noxa (1:1,000; Pro-science), Bid

Mol Cancer Ther 2009;8(12). December 2009

© 2009 American Association for Cancer Research.

Molecular Cancer Therapeutics 3309

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

(1:250, Santa Cruz, C-20), Bad (1:1,000; Sigma), Bim (1:1,000;Sigma), Bax (1:500; Santa Cruz, N20), Bcl-2 (1:500; SantaCruz, Δc21), Mcl-1 (1:5,000; Rockland), Bcl-xl (1:1,000; Sig-ma, 2H12), caspase-9 (1:500, Santa Cruz, H-83), cleavedcaspase-3 (1:500; Cell signaling, 5A1), caspase-2 (1:500; San-ta Cruz, H-145), caspase-8 (1:500; Santa Cruz, D-8), XIAP(1:500; Stressgen, 2F1), cIAP2 (1:500; Santa Cruz, H-85), sur-vivin (1:250; Santa Cruz, D-8), Fas-L (1:500; Santa Cruz, C-178), Fas (1:500; Santa Cruz, A20), GADD45-α (1:500; SantaCruz, C-4), p21 (1:500; Santa Cruz, F-5), p53 (1:500; SantaCruz, FL-393), and phospho-p53 (Ser 15; 1:1,000; Cell Sig-naling). Secondary horseradish peroxidase–conjugated anti-bodies (BioRAD) were used at 1:2,000 for 1 h at roomtemperature, and detection was carried out by enhancedchemiluminescence. Anti-actin (1:5,000; Sigma) or anti-G3PDH antibodies (1:2,000; Trevigen) were used for stan-dardization of protein loading. For analysis of cytochromec release, cytosolic fractions were prepared using a methoddescribed previously (14).Cytofluorometric Analysis of Apoptosis

To detect total activated caspases by flow cytometry, thecell-permeant fluorochrome FITC-VAD.fmk (Promega) wasused as described (15). To evaluate mitochondrial trans-membrane potential (Δψm), cells (5 × 105/mL) were incu-bated for 20 min at 37°C with TMRM (125 nmol/L inPBS; λem = 573 nm). Samples were stored on ice prior to cy-tofluorometric analysis on a FACScan cytofluorometer(Becton Dickinson). For flow cytometric analysis of Bax ac-tivation, we used a procedure described elsewhere (13).Immunofluorescence Microscopy of p53

P388 cells were fixed with 4% paraformaldehyde in PBSfor 10 min and permeabilized with 0.1% Triton X-100 for 10min, and washed twice in PBS with 2% FCS before incuba-tion with a rabbit anti-p53 antibody (1:100; Santa Cruz, sc-6243) on ice overnight. After three washes in PBS, the cellswere incubated with AlexaFluor488-conjugated antirabbitantibody (1:500; Molecular Probes) for 1 h at room temper-ature. DAPI was used for fluorescence counterstaining ofnuclei. All samples were viewed and photographed witha fluorescence microscope (DMLR, Leica).γH2AX Analysis

The H2AX Phosphorylation Assay kit (Upstate) was usedfollowing the manufacturer's instructions. After stainingwith the anti-phospho-histone H2AX-FITC conjugate,which recognizes H2AX phosphorylated at serine 139, orwith normal mouse IgG conjugate as control, cells werecounterstained with propidium iodide for analysis of cellcycle (14). Fluorescence was measured by flow cytometryas described (16).Cell-Free System

Nuclei were purified from P388 cells and mitochondriafrom rat liver following a method previously described(11). Mitochondria (25 μg of protein) and/or nuclei (10 ×106) were resuspended in 50 μL of reaction buffer containing20 mmol/L Hepes (pH 7.4), 10 mmol/L KCl, 2 mmol/LMgCl2, 1 mmol/L EGTA, 1 mmol/L DTT, 250 mmol/L su-crose, 10 mmol/L succinate, 2 mmol/L ATP, 10 mmol/Lcreatine phosphate, 50 μg/mL creatine kinase, and protease

Mol Cancer Ther 2009;8(12). December 2009

on May 15, 2020.mct.aacrjournals.org Downloaded from

inhibitor cocktail (Sigma). The reaction mix was exposed todrugs at 37°C for 3 h after which mitochondria and nucleiwere removed by centrifugation at 12,500 × g for 10 min at4°C before immunoblot analysis.

Figure 1. Nuclear apoptosis induced by lamellarin D in P388 cells. A,structure of lamellarin D. B, kinetics of nuclear signs of apoptosis. P388cells were cultured in the presence of lamellarin D (0.2 μmol/L, 1 μmol/L,5 μmol/L) and at the end of the indicated times, the percentage of subdi-ploid cells (sub-G1) was determined using flow cytometric analysis. Dataare representative of three independent experiments (mean ± SD).C, kineticsof the effects of proapoptotic doses of lamellarin D on P388 cell cycle distri-bution.Cellswere incubatedwith 5 μmol/L lamellarinD for the indicated timesand DNA content was analyzed by flow cytometry using propidium iodidestaining. Data are mean values of four independent experiments(SD <10%). D, P388 cells were cultured for 18 h with 5 μmol/L lamellarinD alone or in combination with 50 μmol/L z-VAD.fmk. After treatment, cellswere harvested, lysed, then immunoblotted using anti-PARP antibody.

© 2009 American Association for Cancer Research.

Mitochondria, DNA damage, and Lamellarin D3310

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

ResultsCell Cycle Distribution and Nuclear Apoptosis Induced

by Lamellarin D in P388 Cells

We previously reported that lamellarin D triggered thecommitment step of the mitochondrial apoptotic cascadein leukemia cells (2). At 1 or 5 μmol/L, concentrations com-patible with an in vivo antitumor activity (2), lamellarin Dprovoked a time-dependent accumulation of P388 leukemiacells in sub-G1 (Fig. 1B) and internucleosomal DNA frag-mentation, a hallmark for apoptosis (SupplementaryFig. S1). Similar results were obtained in human Jurkat T

on May 15, 2020.mct.aacrjournals.org Downloaded from

cells (not shown). At lower doses (<0.5 μmol/L), we havepreviously shown that lamellarin D can perturb cell cycledistribution with a marked arrest of the cells in S and G2-M phase, a phenomenon presumably attributable to its inhi-bition of topoisomerase I (3, 4). The G2-M arrest induced by0.2 μmol/L lamellarin Dwas not followed by any increase ofcells in sub-G1 (Fig. 1B) even after protracted exposure(<10% sub-G1 after 96 hours of exposure). We next evaluat-ed the effect of a proapoptotic concentration of lamellarin Don cell cycle (Fig. 1C). The appearance of cells in sub-G1 wasnot preceded by a prominent cell cycle arrest, although a

Mol Cancer T

© 2009 American Association

Figure 2. Lamellarin D activatesthe intrinsic pathway of apoptosis.A, P388 cells were incubated forthe indicated times with or without5 μmol/L lamellarin D. Whole-cell ly-sates were subjected to SDS-PAGE,blotted, and probed with antibodiesrecognizing caspase-2, cleaved cas-pase-3, caspase-9, and caspase-8.Arrows, cleaved products. The sameblot was stripped and reblotted forcaspase-2 and caspase-8. Actinserved as a loading control. B, effectof caspase inhibitors on lamellarin D–induced apoptosis. P388 cells werecultured in the presence of 5 μmol/Llamellarin D and various inhibitors ofproteases, i.e., phenylmethylsulfo-nylfluoride (1 mmol/L), pepstatine(10 μmol/L), z-VAD.fmk (50 μmol/L),z-VDVAD.fmk (50 μmol/L), z-IETD.fmk (50 μmol /L ) , z -LEHD.fmk(50 μmol/L), and z-DEVD.fmk (50μmol/L). At 24 h posttreatment,Δψm and sub-G1 were determinedby flow cytometry. Data are means ±SD of three independent experi-ments. C, Bcl-2 overexpression de-lays lamellarin D–induced viabilityloss. The effect of lamellarin D(5 μmol/L) on viability of bcl-2–(white square) or neo-transfected(black square) 2B411 T-cell hybrid-oma cells was evaluated over time.At the indicated time points (2, 4,8, 18, 24, and 48 h of lamellarin Dexposure), the percentage of viablecells was determined by flow cyto-metry staining with propidium iodide.D, the small-molecule Bcl-2 inhibitorHA14-1 acts with lamellarin D to in-crease mitochondria-dependent apo-ptosis. P388 cells were treated withlamellarin D (at indicated concentra-tions) and/or 10 μmol/L HA14-1 for20 h, after which the percentage ofcells displaying a loss of Δψm andsub-G1 was determined by flow cyto-metry (means ± SD of three separateexperiments in triplicate).

her 2009;8(12). December 2009

for Cancer Research.

Molecular Cancer Therapeutics 3311

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

slight increase of cells in S phase was observed after 14hours of incubation with 5 μmol/L lamellarin D. During ap-optosis, activated effector caspases cleave various substrateproteins, including the nuclear enzyme PARP (17). Lamel-larin D promoted PARP cleavage into a p85 fragment andthis effect was inhibited by the pan-caspase inhibitorz-VAD.fmk (Fig. 1D). Similar protection was observed whenapoptosis was detected by the percentage of cells in sub-G1

(2). Taken together, these results suggest that micromolardoses of lamellarin D induce caspase-mediated apoptosisof leukemia cells, independently of cell cycle arrest.Activation of the Intrinsic Caspase Cascade in

Lamellarin D–Induced Cell Death

To explore which caspases might be involved in lamel-larin D–induced cell death, we analyzed caspases proces-sing by immunoblotting. Lamellarin D induced cleavage ofpro–caspase-9 as observed by the appearance of severalbands of lower molecular weights (Fig. 2A). The time-dependent processing of caspase-9 was followed by theprocessing of caspase-3, occurring after 18 hours of treat-ment with lamellarin D. This result was consistent with thecanonical concept of the intrinsic pathway placing caspase-3 downstream of caspase-9. In contrast, incubation of cellswith lamellarin D failed to process caspase-8, the apicalcaspase of the extrinsic pathway, and caspase-2, the initia-tor caspase of apoptosis induced by cellular stress. We nextcorrelated the processing of caspases with their activity(Supplementary Fig. S2). DEVDase (caspase-3/-7) andLEHDase (caspase-9) activities were detected in lamellarinD–treated P388 cells in a dose-dependent manner (Supple-mentary Fig. S2, left), confirming the results of immunoblotassays (Fig. 2A). We also detected a weak IETDase activityafter lamellarin D exposure, but no VDVDase (caspase-2)activity (Supplementary Fig. S2, right). The DEVDase andLEHDase activities were markedly inhibited in the pres-ence of the corresponding inhibitors, showing the specific-ity of these results. This was not observed for IETDaseactivity, putting the validity its fluorimetric detection inour model in question. The pretreatment of cells withz-VAD.fmk was also effective against lamellarin D–inducedcaspase-9 and -3 activities, as it was against lamellarin D–induced nuclear apoptosis (Fig. 1D). Together, these resultsclearly implicate the involvement of the intrinsic caspase cas-cade in lamellarin D–triggered apoptosis.Lamellarin D Promotes Apoptosis through Activation

of Mitochondria

To determine the molecular orders between lamellarin D–mediated apoptotic events, P388 cells were pretreated withseveral caspase inhibitors, and apoptosis was assessedusing the determination of Δψm and cells in sub-G1

(Fig. 2B). z-DEVD.fmk, z-LEHD.fmk, and the pan caspaseinhibitor z-VAD.fmk largely suppressed lamellarin D–induced sub-G1, confirming that both caspase-3 and caspase-9 are functionally involved in lamellarin D–mediatedapoptosis. Under the same conditions, none of the caspaseinhibitors succeeded to inhibit the lamellarin D–induceddrop in Δψm (Fig. 2B), nor did the pan–caspase inhibitorz-VAD.fmk inhibit the release of cytochrome c from the

Mol Cancer Ther 2009;8(12). December 2009

on May 15, 2020.mct.aacrjournals.org Downloaded from

mitochondria (Supplementary Fig. S3). These data indicatethat in lamellarin D–induced apoptosis, the caspase cascade(caspase-9 then caspase-3) operates downstream ofmitochondria. Lamellarin D–induced apoptosis was not in-hibited by the broad-spectrum aspartyl and serine proteaseinhibitors pepstatin and phenylmethylsulfonylfluoride,suggesting that only caspases were involved in lamellarinD–mediated apoptosis. Experiments were done to explorethe role of Bcl-2, a prototypical suppressor of the mitochon-drial intrinsic pathway of apoptosis. 2B411 hybridoma Tcellsstably transfected with the bcl-2 gene or a neo control vectorwere treated with lamellarin D followed by determination ofviability. As shown in Fig. 2C, loss of viability was signifi-cantly delayed by overexpression of Bcl-2, at least for the first18 hours of incubation. At later time points, the protectiveeffect of Bcl-2 was lost (Fig. 2C). We next determined the

Figure 3. Lamellarin D induces cell death through a Fas-independentpathway. A, expression of Fas and Fas ligand (Fas L) in response to lamel-larin D exposure. P388 cells were incubated for the indicated times with orwithout 5 μmol/L lamellarin D, and whole-cell lysates were subjected toimmunoblotting for detection of Fas and Fas L. G3PDH served as a loadingcontrol. B, wild-type, FADD-deficient and caspase-8 deficient Jurkat Tcells were treated with anti-Fas antibody (500 ng/mL) or lamellarin D(5 μmol/L) for 24 h. The cells were stained for sub-G1 and analyzed byflow cytometry. C, lamellarin D induced the mitochondrial pathway of ap-optosis in wild-type, FADD-deficient and caspase-8–deficient Jurkat Tcells. After 12 h of treatment with 5 μmol/L lamellarin D, cytosolic frac-tions were analyzed by immunoblotting using cytochrome c antibody andthe percentage of cells with a decrease in Δψm (Δψmlow) were measuredby flow cytometry. G3PDH served as a loading control.

© 2009 American Association for Cancer Research.

Mitochondria, DNA damage, and Lamellarin D3312

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

ability of a small molecule Bcl-2 inhibitor, HA14-1, to reversethe effects of Bcl-2 on apoptosis induced by lamellarin D.P388 cells synergistically incubated with 10 μmol/L HA14-1 and lamellarin D for 24 hours had higher levels of cells insub-G1 (Fig. 2D, rings) and a more pronounced drop inΔψm(Fig. 2D, bars) than those treated with lamellarin D alone.Altogether, these data support the contention that lamellarinD triggers the intrinsic, mitochondria-mediated pathway ofapoptosis, which can be modulated by the Bcl-2 protein.The Fas-Dependent Extrinsic Pathway Is Not

Required for Lamellarin D–Induced Apoptosis

Several anticancer drugs activate both the intrinsic as wellas the extrinsic pathway of apoptosis, particularly in leuke-mia cells (18). Therefore, we tested whether the apoptotic re-sponse triggered by lamellarin D also involves the activationof the extrinsic, death receptor–mediated, pathway. We firstevaluated the expression of Fas and FasL after lamellarin Dtreatment. Time-course immunoblot analysis revealed thatboth Fas and FasL protein levels were not elevated after la-mellarin D treatment (Fig. 3A). However, anticancer drugscan activate the extrinsic pathway independently of anychange in Fas/FasL expression by promoting the intracellu-lar recruitment of Fas-associated protein with death domain(FADD) to Fas and thereby favor caspase-8 activation (19).Thus, we used human Jurkat T leukemia cells lacking thecaspase-8 adaptor protein FADD (FADD−/−) and Jurkat cells

on May 15, 2020.mct.aacrjournals.org Downloaded from

deficient of caspase 8 (caspase-8−/−) to determine whetherthe Fas pathway was required for lamellarin D–inducedapoptosis. Lamellarin D promoted nuclear apoptosis inFADD−/− cells as well as in caspase-8−/− cells (Fig. 3B). Incontrast, apoptosis was not induced by the Fas agonisticmonoclonal antibody CH-11 used as control (Fig. 3B). Therewas no significant difference in the kinetics or dose responseof apoptotic death induction by lamellarin D when compar-ing FADD−/− cells, caspase-8−/− cells, and wild-type Jurkatcells (data not shown). In addition,Δψmdissipation and mi-tochondrial cytochrome c release were observed in FADD−/−

cells and caspase-8−/− cells after treatment with lamellarin D(Fig. 3C). These results indicate that the Fas/FasL death re-ceptor pathway is not critical for apoptosis by lamellarin D.Lamellarin D Downregulates Expression of the

Antiapoptotic Proteins Bcl-2 and cIAP2, and Activates

the Proapoptotic Protein Bax in P388 Cells

Because Bcl-2 protein is a critical regulator of lamellarin D–induced apoptosis (Fig. 2C and D), we investigated the ex-pression of Bcl-2 family members at 2, 4, 8, and 18 hours oflamellarin D exposure (i.e., before completion of nuclear ap-optosis). Figure 4A shows that the levels of the proapoptoticproteins Puma, Noxa, Bid, Bim, and Bad were not modifiedafter treatment with lamellarin D, excluding a role of theseproteins in lamellarin D–induced apoptosis. Although noupregulation of Bax was found (Fig. 4A), we observed that

© 2009 America

Figure 4. Lamellarin D shifts the balance of Bcl-2–related antiapoptotic and proapoptotic proteins to fa-vor apoptotic demise. A, expression of proapoptoticand antiapoptotic proteins was monitored by immu-noblot. P388 cells were treated with 5 μmol/L of la-mellarin D or left untreated (control; Co.). After theindicated times, immunoblot analyses for Puma,Noxa, Bax, Bid, Bad, Bim, Bcl-2, Mcl-1, and Bcl-xlwere done. Alternatively, P388 cells were treatedfor 18 h with 5 μmol/L lamellarin D in the presenceor absence of 50 μmol/L z-VAD.fmk and immuno-blotted using Bcl-2 antibody. Equal protein loadingwas controlled by actin antibody. B, Bax activationwas assessed by flow cytometry using conforma-tion-specific anti-Bax antibody (solid line). Secondaryantibody alone was used as negative control (dashedline). Numbers, mean fluorescence intensity valuesof Bax-stained cells. C, expression of inhibitors ofapoptosis proteins (XIAP, cIAP2, survivin) were ex-amined by immunoblotting as in A.

Mol Cancer Ther 2009;8(12). December 2009

n Association for Cancer Research.

Molecular Cancer Therapeutics 3313

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

lamellarin D led to significant Bax activation in a time- anddose-dependent manner (Fig. 4B). Lamellarin D induced atime-dependent reduction in the steady-state level of Bcl-2beginning within 8 hours of treatment (Fig. 4A). A bandcorresponding to the full-length Bcl-2 disappeared, but nocleaved fragments could be detected. The degradation ofBcl-2 was caspase dependent because pretreatment of P388cells with z-VAD.fmkmaintained a high level of Bcl-2 expres-sion (Fig. 4A). Conversely, lamellarin D did not modulate theexpression of the other Bcl-2 familymembers, Bcl-xl orMcl-1.These results suggest that lamellarin D shifts the balance ofBcl-2–related antiapoptotic and proapoptotic proteins to fa-vor apoptotic demise.The inhibitor of apoptosis protein (IAP) family members

XIAP, cIAP1, cIAP2, and survivin are antiapoptotic proteinsthat inhibit caspases directly (20). These IAPs are expressed athigh levels in P388 cells (Fig. 4C). Notably, lamellarin D–

Mol Cancer Ther 2009;8(12). December 2009

on May 15, 2020.mct.aacrjournals.org Downloaded from

induced caspase-9 and caspase-3 activation correlated witha reduction in the level of cIAP2, detected already 4 hourspost-lamellarin D exposure (Fig. 4C). The level of other IAPsremained unchanged (Fig. 4C).DNA Damage following Lamellarin D Exposure

Topoisomerase I inhibitors like campthotecin are knownto cause DNA damage, involving the formation of DNAdouble-strand breaks that trigger apoptosis (for review,see ref. 21). We used flow cytometry to detect the phosphor-ylated form of histone H2AX (γH2AX), which is a good in-dicator of double-strand DNA breaks in chromatin. Cellswere costained with propidium iodide for cell cycle analysisto correlate expression of γH2AX with cell cycle position(Fig. 5A). P388 cells exposed to lamellarin D displayed a sig-nificant increase in γH2AX fluorescence after a short incu-bation time (within 15 minutes) with a high response at 60minutes (Fig. 5A). Nearly identical changes in DNA damage

Figure 5. Lamellarin D–induced DNA damage andp53 expression depend on the inhibition of nucleartopoisomerase I. A, multiparameter distribution re-presenting expression of γH2AX versus DNA con-tent of wild-type P388 cells (top) or topoisomeraseI mutated P388cells, P388 CPT5 (bottom), keptuntreated (control; Co.), treated with 5 μmol/L la-mellarin D for 15 or 60 min, or treated with thewell-known DNA topoisomerase I inhibitor camp-tothecin (5 μmol/L) for 60 min. Based on differencein DNA content, G1, S, and G2-M cell populationswere defined. Numbers in percentage representthe γH2AX positive cells gated. Data are represen-tative of three independent experiments. B, immu-noblot analysis of the DNA repair protein Rad51 inP388 and P388 CPT5 cells treated with 5 μmol/Llamellarin D for indicated times. Actin served asloading control. C, immunoblot analysis of p53 pro-tein in P388 and P388 CPT5 cells treated with5 μmol/L lamellarin D for indicated times. Actinserved as loading control. D, left, immunofluores-cence staining of p53 in untreated P388 cells (con-trol cells; Co.) and in P388 cells 20 h post-lamellarinD treatment (5 μmol/L). Cells were stained withDAPI to display nuclear morphology. Original magni-fication ×40. Right, time course of p53 activationduring lamellarin D treatment. P388 cells were trea-ted with 5 μmol/L lamellarin D for 0 to 18 h andwhole-cell lysates were subjected to immunoblot-ting with anti-phospho-Ser15 p53 antibody.

© 2009 American Association for Cancer Research.

Mitochondria, DNA damage, and Lamellarin D

Mol Cancer Ther 2009;8(12). December 2009

3314

on May 15, 2020. © 2009 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

Molecular Cancer Therapeutics 3315

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

were observed for cells treated with the prototypic topoi-somerase I inhibitor camptothecin (Fig. 5A). It is evidentfrom these multiparameter analyses that the increase inγH2AX expression was most prominent for cells in S phase,a situation classically observed in response to topoisomeraseI inhibitors (ref. 22 and Fig. 5A). In contrast, lamellarin D, aswell as camptothecin, did not promote any nuclear damagein the topoisomerase I–mutated cells, P388 CPT5 (Fig. 5A),even upon long-term exposure (data not shown). Underthese conditions, the double-strand breaks repair proteinRad51 was induced by lamellarin D at 8 hours exclusivelyin wild-type P388 cells (Fig. 5B). Collectively, these data indi-cate that lamellarin D may act as a classical nuclear topoi-somerase I inhibitor to promote DNA damage and repair inP388 cells.Role of p53 in Lamellarin D–Induced Apoptosis

The tumor suppressor p53 is generally activated in re-sponse to DNA damage in cancer cells. As shown inFig. 5C, incubation of P388 cells with lamellarin D resultedin a time-dependent increase in the basal expression of thep53 protein not observed in P388 CPT5 cells. We also ana-lyzed the subcellular localization of p53 by immunofluores-cence microscopy (Fig. 5D). After lamellarin D treatment,p53 predominantly accumulated in the nucleus and nop53 immunoreactivity was detected in the cytoplasm or inmitochondria (Fig. 5D). This nuclear accumulation wasmore evident in apoptotic cells with condensed chromatin.Incubation of P388 cells with lamellarin D resulted in phos-phorylation of p53 at Ser 15 (Fig. 5D, right). Nevertheless,the p53-inducible gene products Puma, Noxa, Bax(Fig. 4A), GADD45, and p21 (Supplementary Fig. S4) werenot induced in P388 cells. Consistent with these data, wefound that pifithrin-α, a chemical inhibitor of p53 transcrip-tional activity, was unable to impede apoptosis triggered bylamellarin D (data not shown). To rule out a transcription-independent proapoptotic function of p53, we tested the ef-fect of lamellarin D on cancer cell lines possessing differentp53 status (Fig. 6A and B). The wild-type HCT116 p53+/+

colon carcinoma cells, as well as the HCT116 p53−/− cells,having a disrupted p53 gene due to homologous recombina-tion, both displayed a reduction in mitochondrial Δψm andan increased number of cells in sub-G1 upon lamellarin Dexposure (Fig. 6A). Although not statistically significant,

Mol Cancer Ther 2009;8(12). December 2009

on May 15, 2020.mct.aacrjournals.org Downloaded from

the kinetic and extent of sub-G1 were more pronounced inp53+/+ cells, suggesting that a functional p53 pathwaymightparticipate in lamellarin D–mediated apoptosis in HCT116cells. The p53-null SAOS2 cells, although resistant to the in-duction of apoptosis by camptothecin, were sensitive to la-mellarin D–induced apoptosis (Fig. 6B). Altogether theseresults indicate that intact p53 is not necessary for the apo-ptotic response triggered by lamellarin D.We next examined the possibility that protein synthesis

could be required for apoptosis induced by lamellarin D.Inhibition of protein synthesis by cycloheximide did notaffect the appearance of nuclear and mitochondrial signsof apoptosis upon lamellarin D exposure, whereas a sig-nificant inhibition was observed for the dexamethasone-induced apoptosis, used as control (Fig. 6C). Finally, westudied the requirement of cell nucleus in lamellarin D–induced apoptosis. Upon lamellarin D treatment, both in-tact and enucleated P388 cells (cytoplasts) displayed aloss of mitochondrial Δψm to the same extent (Fig. 6D,left). Moreover, lamellarin D also triggered caspase activi-ty irrespective of the presence of nucleus (Fig. 6D, right).To further document that the cell nucleus was dispensablefor lamellarin D–induced apoptosis, we did a cell-freesystem experiment consisting of purified mitochondriacombined with isolated P388 cell nuclei. Consistent withprevious data (2), lamellarin D directly stimulated cy-tochrome c release from isolated mitochondria in adose-dependent manner, even in the absence of nuclei(Fig. 6E). As a control, the DNA damaging drug etoposide(10 μmol/L) needed the presence of nuclei to promote mi-tochondrial release of cytochrome c in this reconstitutedcell-free system. Together, these results show that apopto-sis induced by lamellarin D is strictly dependent on mito-chondria and can occur without protein synthesis andnuclear involvement.

DiscussionThe discovery of the antitumor activity of lamellarin Dencouraged molecular studies aimed at deciphering howthis natural product and its synthetic derivatives were ableto inhibit the growth of different cancer cells. This growthinhibition involves both decreased cell division (4) and

Figure 6. p53-dependent and nuclear-dependent signaling pathways are not required for lamellarin D–induced apoptosis. A, p53+/+ and p53−/−

HCT116 colon carcinoma cells were incubated with lamellarin D (1 or 5 μmol/L) for the indicated times before determination of cells in sub-G1. Camp-tothecin (5 μmol/L 24 h) was used as a control for p53-dependent apoptosis. Results (mean ± SD) are representative of at least five independent experi-ments. B, The SAOS2 osteosarcoma cell line (p53 null) was incubated with lamellarin D for 24 h before determination of sub-G1 and Δψm. Camptothecin(5 μmol/L 24 h) was used as a control for p53-dependent apoptosis. Results (mean ± SD) are representative of three independent experiments. C, proteinsynthesis is dispensable for lamellarin D–triggered apoptosis. P388 cells were incubated with either 5 μmol/L lamellarin D or 1 μmol/L dexamethasone(DEX) alone or in the presence of 1 μmol/L cycloheximide (CHX). After 4 h of culture, changes in mitochondrial membrane potential (Δψm) were assessedby the use of TMRM. Alternatively, after 12 h of lamellarin D treatment, the percentage of cells in sub-G1 was determined. Results (mean ± SD) arerepresentative of three independent experiments. D, comparison of enucleated P388 cells (cytoplasts) and intact P388 cells in response to lamellarinD. Cytoplasts or intact P388 cells were treated with lamellarin D at indicated concentrations for 6 h. Left, mitochondrial membrane potential was deter-mined by flow cytometry using TMRM and the values are relative mean fluorescence intensity ± SD of three independent experiments. Right, detection ofpan-caspase activity with FITC-VAD.fmk. Cytoplasts and P388 control cells were incubated for 15 h with or without 5 μmol/L lamellarin D, and caspaseactivity was assessed by flow cytometry. Results are representative of two independent experiments. E, lamellarin D induces cytochrome c release in acell-free system. Upper box, mitochondria isolated from P388 cells (0.5 mg/mL final concentration) were incubated for 90 min at 37°C in the absence(control; Co.) or presence of lamellarin D (at 0.5, 2 and 5 μmol/L) or 10 μmol/L etoposide (Eto.). Lower box, conditions as in upper box, except that nuclei(2 × 106/mL) isolated from p388 cells were added to the samples. At the end of the incubation time the reaction mixture was centrifuged and supernatantswere immunoblotted for the detection of cytochrome c release.

© 2009 American Association for Cancer Research.

Mitochondria, DNA damage, and Lamellarin D3316

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

induction of apoptosis (2), and it seems that these two anti-tumor actions are dose dependent. When used at very lowdoses (nanomolar range), lamellarin D did not kill cancercells but induced an accumulation of cells in S and G2-Mphases, dependent on its inhibition of topoisomerase I(Fig. 1B and ref. 2). Conversely, at higher doses of lamellarinD (micromolar range compatible with the xenograft assays;ref. 23), cancer cells displayed a potent apoptotic responsewithout major effects on cell cycle (2). In cancer, the thera-peutic goal is to trigger tumor-selective cell death (24), andthe response of tumors to therapy mainly depends on theirability to undergo cell death (24). Nowadays, the role of ap-optosis in the cytotoxicity of anticancer drugs has becomeclearer (6). Different classes of anticancer drugs exert theircytotoxicities through a variety of primary molecular targetsto ultimately complete cell death via common apoptotic sig-naling pathways (24). Our data clearly indicate that lamel-larin D activates the intrinsic, mitochondria-dependent,apoptotic pathway in cancer cells. This is based on severalpieces of evidence: (a) proteolytic activation of caspase-9; (b)induction of caspase-independent cytochrome c release; (c)modulation of the Bcl-2 protein, which influences the levelof apoptosis induced by lamellarin D; and (d) the occurrenceof lamellarin D–induced cell death independently of the ex-trinsic death receptor–mediated pathway. We consideredthe possibility that lamellarin D, by reducing the expressionof antiapoptotic proteins (e.g., Bcl-2), favors apoptosisthrough amplification of the mitochondria-dependent path-way. We observed a reduced expression of the Bcl-2 proteinassociated with a conformational activation of Bax. TheseBcl-2 family proteins normally function to control the mito-chondrial release of cytochrome c (25). However, our resultsshow a reduction in Bcl-2 level long after the onset of cyto-chrome c release (2). Based on these results, we can assumethat caspase-mediated cleavage of Bcl-2 represents a feedbackloop for the amplification of mitochondrial cytochrome crelease during lamellarin D–induced apoptosis. In additionto Bcl-2 family proteins, we also noticed that lamellarin Dreduced levels of the antiapoptotic protein, cIAP2. IAPfamily members are generally overexpressed in cancer andtheir downregulation contributes to an increased sensitivityto chemotherapeutic drugs. cIAP2 inhibits caspase-9 andcaspase-3, thereby blocking the intrinsic caspase cascadedownstream of mitochondria. It is conceivable that the ca-pacity of lamellarin D to activate the intrinsic pathwaydownstream of mitochondria was also amplified by its in-hibitory effect on cIAP2. The mechanisms of cIAP2 reduc-tion were independent of caspases (data not shown).cIAP2 is subjected to regulation by the second mitochon-drial activator of caspase (Smac/Diablo), a protein that isreleased from mitochondria during apoptosis (20). Thus, itis possible that the reduced levels of cIAP2 might be adownstream consequence of mitochondrial membrane per-meabilization, and hence the release of Smac/Diablo. Fur-ther studies should clarify the mechanisms responsible forcIAP2 reduction.Most conventional chemotherapeutic drugs are nucleus-

targeted (genotoxic) agents that have been designed to in-

on May 15, 2020.mct.aacrjournals.org Downloaded from

teract with DNA or DNA-associated proteins such as topoi-somerases (26). As a true topoisomerase I inhibitor,lamellarin D induced a time-dependent DNA damage re-sponse assessed by staining for γ-H2AX and Rad51 inP388 cells. In response to lamellarin D–induced DNA dam-age, levels of activated, Ser15-phosphorylated p53 increasedmarkedly. However, no activation of the p53 downstreamtargets GADD45, p21, Bax, Puma, and Noxa, was observed.One possible explanation is that P388 cells do not respondto p53-mediated apoptosis due to mutations in the p53binding site, as it has been suggested (27). Transcription-independent activities of p53 are important for the apoptoticresponse (28). Thus, p53 can be translocated to mitochon-dria to cause cell death in response to DNA damage (for re-view see ref. 8). It is hardly probable, however, that the earlymitochondrial permeability observed after lamellarin Dexposure is a consequence of p53 translocation to mitochon-dria because there was no p53 translocation to mitochondria(Fig. 5D). Furthermore, in p53 mutated or null cells, mito-chondria in themselves seem sufficient to trigger apoptosisupon lamellarin D exposure. Indeed, although we wereable to detect a moderate dose-dependent increase of theproapoptotic effect of lamellarin D in wild-type p53-expres-sing cells when compared with p53−/− cells (Fig. 6A), thiseffect of p53 was limited and transient because no further in-crease in apoptosis was observed after 48 hours of treatmentwith lamellarin D. These results indicate that p53 is not crit-ical for the completion of lamellarin D–induced apoptosis.Interestingly, this characteristic distinguishes lamellarin Dfrom camptothecin, the prototypic topoisomerase I poison.These results prompted us to further explore whether

lamellarin D–induced DNA damage might generate othernuclear signaling pathways contributing to apoptosis.Using enucleated cells and cell-free system assays, our re-sults show that when lamellarin D was targeted to mito-chondria, cells underwent apoptosis. Hence, its nucleartargeting seemed dispensable. These results raise the pos-sibility of “double hits” of lamellarin D, one directly onthe mitochondria to trigger apoptosis and the other loc-ated in the cell nucleus, resulting in DNA damage, cellcycle arrest, and DNA repair. Like lamellarin D, betulinicacid is a mitochondria-targeting drug and an inhibitor ofnuclear topoisomerase I (29). Moreover, betulinic acidtriggers apoptosis by a direct effect on mitochondria, in-dependent of its nuclear action (30). Thus, lamellarin Dtriggers an apoptotic pathway different from those previ-ously identified for standard genotoxic chemotherapeuticdrugs such as topoisomerase I inhibitors (26). Unlike camp-tothecin, lamellarinD exerts a secondmode of action, bypass-ing the nucleus and directly triggering apoptosis via themitochondria. In theory, direct targeting of the mitochondriamay circumvent drug resistance caused by most of the apo-ptotic defects. Lamellarin D kills a variety of tumor cells in-cluding several leukemia and carcinoma cell lines (2, 4),suggesting that themechanism of apoptosis is not cell type de-pendent. This broad antitumor activitymay result from its mi-tochondrial targeting. As a result, mitochondriophilic agents,like lamellarin D, may prove to be highly efficient in killing

Mol Cancer Ther 2009;8(12). December 2009

© 2009 American Association for Cancer Research.

Molecular Cancer Therapeutics 3317

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639

cancer cells normally resistant to apoptosis (31). In recentyears, mitochondria have emerged as a promising target forcancer therapy (32), and clinical evaluation of several drugstargeting mitochondria-related proteins of the Bcl-2 family(GX15-070, oblimersen sodium, ABT-737) are ongoing (33).In summary, this article not only contributes to decipher-

ing the apoptotic pathways activated by lamellarin D butalso provides evidence that lamellarin D is still active evenif the classical apoptotic pathways are blocked in cancercells (e.g., when p53 is null/mutated, when topoisomerase1 is mutated or Bcl-2 overexpressed). This study providesthe basis for new strategies establishing mitochondria as avaluable target in cancer cells, reinforcing the potential in-terest of mitochondriophilic drugs for cancer therapy. Tar-geting mitochondria may confer a solid advantage againstdrug resistance in cancer cells, provided that a selectivity fortumor versus normal mitochondria can be delineated. It willbe therefore essential to identify the molecular target(s) oflamellarin D in mitochondria. Studies are ongoing in ourlaboratories.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank Drs. Bert Vogelstein, J.-F. Riou and J. Ashwell for providing thecell lines, and Dr. Steve Lancel (EA2689, Faculté de Médecine, Lille) forcritical reading.

References

1. Bailly C. Lamellarins, from A to Z: a family of anticancer marine pyrrolealkaloids. Curr Med Chem Anticancer Agents 2004;4:363–78.

2. Kluza J, Gallego MA, Loyens A, et al. Cancer cell mitochondria aredirect proapoptotic targets for the marine antitumor drug lamellarin D.Cancer Res 2006;66:3177–87.

3. Vanhuyse M, Kluza J, Tardy C, et al. Lamellarin D: a novel pro-apoptoticagent from marine origin insensitive to P-glycoprotein-mediated drug ef-flux. Cancer Lett 2005;221:165–75.

4. Facompre M, Tardy C, Bal-Mahieu C, et al. Lamellarin D: a novel potentinhibitor of topoisomerase I. Cancer Res 2003;63:7392–9.

5. Tardy C, Facompre M, Laine W, et al. Topoisomerase I-mediated DNAcleavage as a guide to the development of antitumor agents derived fromthe marine alkaloid lamellarin D: triester derivatives incorporating aminoacid residues. Bioorg Med Chem 2004;12:1697–712.

6. Call JA, Eckhardt SG, Camidge DR. Targeted manipulation of apoptosisin cancer treatment. Lancet Oncol 2008;9:1002–11.

7. Roos WP, Kaina B. DNA damage-induced cell death by apoptosis.Trends Mol Med 2006;12:440–50.

8. Moll UM, Wolff S, Speidel D, Deppert W. Transcription-independentpro-apoptotic functions of p53. Curr Opin Cell Biol 2005;17:631–6.

9. Reed JC. Bcl-2 family proteins: regulators of apoptosis and chemoresis-tance in hematologic malignancies. Semin Hematol 1997;34:9–19.

10. Martoriati A, Doumont G, Alcalay M, Bellefroid E, Pelicci PG, MarineJC. dapk1, encoding an activator of a p19ARF-p53-mediated apoptoticcheckpoint, is a transcription target of p53. Oncogene 2005;24:1461–6.

11. Marchetti P, Zamzami N, Joseph B, et al. The novel retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2- naphtalene carboxylic acid can triggerapoptosis through a mitochondrial pathway independent of the nucleus.Cancer Res 1999;59:6257–66.

Mol Cancer Ther 2009;8(12). December 2009

on May 15, 2020.mct.aacrjournals.org Downloaded from

12. Kluza J, Marchetti P, Gallego MA, et al. Mitochondrial proliferationduring apoptosis induced by anticancer agents: effects of doxorubicinand mitoxantrone on cancer and cardiac cells. Oncogene 2004;23:7018–30.

13. Gallego MA, Ballot C, Kluza J, et al. Overcoming chemoresistance ofnon-small cell lung carcinoma through restoration of an AIF-dependent ap-optotic pathway. Oncogene 2008;27:1981–92.

14. Gallego MA, Joseph B, Hemstrom TH, et al. Apoptosis-inducing fac-tor determines the chemoresistance of non-small-cell lung carcinomas.Oncogene 2004;23:6282–91.

15. Marchetti C, Gallego MA, Defossez A, Formstecher P, Marchetti P.Staining of human sperm with fluorochrome-labeled inhibitor of caspasesto detect activated caspases: correlation with apoptosis and sperm param-eters. Hum Reprod 2004;19:1127–34.

16. Huang X, Darzynkiewicz Z. Cytometric assessment of histone H2AXphosphorylation: a reporter of DNA damage. Methods Mol Biol 2006;314:73–80.

17. Timmer JC, Salvesen GS. Caspase substrates. Cell Death Differ2007;14:66–72.

18. Kaufmann SH, Earnshaw WC. Induction of apoptosis by cancer che-motherapy. Exp Cell Res 2000;256:42–9.

19. Gajate C, Del Canto-Janez E, Acuna AU, et al. Intracellular triggeringof Fas aggregation and recruitment of apoptotic molecules into Fas-enriched rafts in selective tumor cell apoptosis. J Exp Med 2004;200:353–65.

20. Zhang HG, Wang J, Yang X, Hsu HC, Mountz JD. Regulation ofapoptosis proteins in cancer cells by ubiquitin. Oncogene 2004;23:2009–15.

21. Pommier Y. Topoisomerase I inhibitors: camptothecins and beyond.Nat Rev Cancer 2006;6:789–802.

22. Huang X, Traganos F, Darzynkiewicz Z. DNA damage induced by DNAtopoisomerase I- and topoisomerase II-inhibitors detected by histoneH2AX phosphorylation in relation to the cell cycle phase and apoptosis.Cell Cycle 2003;2:614–9.

23. Elices M, Grant W, Luber-Narod J, et al. Lamellarins, novel marinecompounds showing efficacy in a panel of human tumor xenografts. AmAssoc Cancer Res 2005, Poster#645.

24. Viktorsson K, Lewensohn R, Zhivotovsky B. Apoptotic pathways andtherapy resistance in human malignancies. Adv Cancer Res 2005;94:143–96.

25. Gupta S, Kass GE, Szegezdi E, Joseph B. The mitochondrial deathpathway: a promising therapeutic target in diseases. J Cell Mol Med20092009 Feb 9. Epub ahead of print.

26. Sordet O, Khan QA, Kohn KW, Pommier Y. Apoptosis induced bytopoisomerase inhibitors. Curr Med Chem Anticancer Agents 2003;3:271–90.

27. Ishiguro K, ShyamK, Penketh PG, Sartorelli AC. Role ofO6-alkylguanine-DNA alkyltransferase in the cytotoxic activity of cloretazine. Mol CancerTher 2005;4:1755–63.

28. Steele AJ, Prentice AG, Hoffbrand AV, et al. p53-mediated apoptosisof CLL cells: evidence for a transcription-independent mechanism. Blood2008;112:3827–34.

29. Eiznhamer DA, Xu ZQ. Betulinic acid: a promising anticancer candi-date. IDrugs 2004;7:359–73.

30. Ganguly A, Das B, Roy A, et al. Betulinic acid, a catalytic inhibitor oftopoisomerase I, inhibits reactive oxygen species mediated apoptotic to-poisomerase I DNA cleavable complex formation in prostate cancer cellsbut does not affect the process of cell death. Cancer Res 2007;67:11848–58.

31. Pommier Y, Sordet O, Antony S, Hayward RL, Kohn KW. Apoptosisdefects and chemotherapy resistance: molecular interaction maps and net-works. Oncogene 2004;23:2934–49.

32. Armstrong JS. Mitochondria: a target for cancer therapy. Br J Pharma-col 2006;147:239–48.

33. Kang MH, Reynolds CP. Bcl-2 Inhibitors: targeting mitochondrialapoptotic pathways in cancer therapy. Clin Cancer Res 2009;15:1126–32.

© 2009 American Association for Cancer Research.

2009;8:3307-3317. Published OnlineFirst December 1, 2009.Mol Cancer Ther   Caroline Ballot, Jérome Kluza, Alain Martoriati, et al.   induced by the marine alkaloid Lamellarin DEssential role of mitochondria in apoptosis of cancer cells

  Updated version

  10.1158/1535-7163.MCT-09-0639doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2009/12/15/1535-7163.MCT-09-0639.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/8/12/3307.full#ref-list-1

This article cites 31 articles, 8 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/8/12/3307.full#related-urls

This article has been cited by 1 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] at

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

  Permissions

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

.http://mct.aacrjournals.org/content/8/12/3307To request permission to re-use all or part of this article, use this link

on May 15, 2020. © 2009 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst December 1, 2009; DOI: 10.1158/1535-7163.MCT-09-0639