opposing actions of endocannabinoids on cholangiocarcinoma growth is via the differential activation...

17
Opposing Actions of Endocannabinoids on Cholangiocarcinoma Growth RECRUITMENT OF Fas AND Fas LIGAND TO LIPID RAFTS * Received for publication, August 28, 2006, and in revised form, February 26, 2007 Published, JBC Papers in Press, February 28, 2007, DOI 10.1074/jbc.M608238200 Sharon DeMorrow , Shannon Glaser ‡§ , Heather Francis , Julie Venter § , Bradley Vaculin § , Shelley Vaculin , and Gianfranco Alpini §¶1 From the Central Texas Veterans Health Care System and the Departments of § Medicine and Systems Biology and Translational Medicine and Division of Research and Education, Scott and White Hospital and Texas A&M University System Health Science Center College of Medicine, Temple, Texas 76504 Cholangiocarcinomas are devastating cancers of biliary origin with limited treatment options. Modulation of the endocan- nabinoid system is being targeted to develop possible therapeu- tic strategies for a number of cancers; therefore, we evaluated the effects of the two major endocannabinoids, anandamide and 2-arachidonylglycerol, on numerous cholangiocarcinoma cell lines. Although anandamide was antiproliferative and proapo- ptotic, 2-arachidonylglycerol stimulated cholangiocarcinoma cell growth. Specific inhibitors for each of the cannabinoid receptors did not prevent either of these effects nor did pretreat- ment with pertussis toxin, a G i/o protein inhibitor, suggesting that anandamide and 2-arachidonylglycerol did not exert their diametric effects through any known cannabinoid receptor or through any other G i/o protein-coupled receptor. Using the lipid raft disruptors methyl--cyclodextrin and filipin, we demon- strated that anandamide, but not 2-arachidonylglycerol, requires lipid raft-mediated events to inhibit cellular prolifera- tion. Closer inspection of the lipid raft structures within the cell membrane revealed that although anandamide treatment had no observable effect 2-arachidonylglycerol treatment effectively dissipated the lipid raft structures and caused the lipid raft-associated proteins lyn and flotillin-1 to disperse into the surrounding membrane. In addition, anandamide, but not 2-arachidonylglycerol, induced an accumulation of ceramide, which was required for anandamide-induced sup- pression of cell growth. Finally we demonstrated that anand- amide and ceramide treatment of cholangiocarcinoma cells recruited Fas and Fas ligand into the lipid rafts, subsequently activating death receptor pathways. These findings suggest that modulation of the endocannabinoid system may be a tar- get for the development of possible therapeutic strategies for the treatment of this devastating cancer. Cholangiocarcinomas are devastating cancers of intrahe- patic and extrahepatic origin that are increasing in both their worldwide incidence and mortality rates (1, 2). The challenges posed by these often lethal biliary tract cancers are daunting; conventional treatment options are limited, and the only hope for long term survival is that of complete surgical resection of the tumor (1, 2). Conventional chemotherapy and radiation therapy are not effective in prolonging long term survival (1); therefore it is important to understand the cellular mechanisms of cholangiocarcinoma cell growth with a view to develop novel chemopreventive strategies. Marijuana and its derivatives have been used in medicine for many centuries, and presently there is an emerging renaissance in the study of the therapeutic effects of cannabinoids. Ongoing research is determining that regulation of the endocannabinoid system may be effective in the treatment of pain (3, 4), glaucoma (5), and neurodegenerative disorders such as Parkinson disease (6) and multiple sclerosis (7). In addition, cannabinoids might be effective antitumoral agents because of their ability to inhibit the growth of various types of cancer cell lines in culture (8 –11) and in laboratory animals (12). The finding in the early 1990s of specific G-protein-coupled receptors for the psychoactive component of Cannabis sativa ()- 9 -tetrahydrocannabinol (13) led to the discovery of a whole endogenous signaling system now known as the endo- cannabinoid system (14). This system consists of the cannabi- noid receptors (to date there are two, Cb1 2 and Cb2, as well as a putative involvement of the vanilloid receptor VR1), their endogenous ligands (endocannabinoids), and the proteins for their synthesis and inactivation (14). The cannabinoid recep- tors are seven-transmembrane domain proteins coupled to the G i/o type of G-proteins (14). Cb1 receptors are found predom- inantly in the central nervous system but also in most periph- eral tissues including immune cells, the reproductive system, * The study was supported by a grant award from Scott and White Hospital (to S. D.), the Dr. Nicholas C. Hightower Centennial Chair of Gastroenterol- ogy, a Veterans Affairs Research Scholar Award, a Veterans Affairs Merit Award, and National Institutes of Health Grant DK062975 (to G. A.). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertise- ment” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 To whom correspondence should be addressed: Depts. of Medicine and Systems Biology and Translational Medicine, Central Texas Veterans Health Care System, Texas A&M University System Health Science Center College of Medicine, Medical Research Bldg., 702 S. W. H. K. Dodgen Loop, Temple, TX 76504. Tel.: 254-742-7044; Fax: 254-724-8070; E-mail: [email protected]. 2 The abbreviations used are: Cb1, cannabinoid receptor 1; Cb2, cannabinoid receptor 2; 2-AG, 2-arachidonylglycerol; AEA, anandamide; BrdUrd, bro- modeoxyuridine; DAPI, 4,6-diamidino-2-phenylindole; dnFADD, domi- nant negative FADD; DR, death receptor; FADD, Fas-associated death domain protein; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy- phenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; PBS, phosphate-buff- ered saline; TNF, tumor necrosis factor; VR1, vanilloid receptor; FasL, Fas ligand; MES, 4-morpholineethanesulfonic acid; shRNA, short hairpin RNA; BSA, bovine serum albumin; MEK, mitogen-activated protein kinase/extra- cellular signal-regulated kinase kinase; ERK, extracellular signal-regulated kinase. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 282, NO. 17, pp. 13098 –13113, April 27, 2007 Printed in the U.S.A. 13098 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007 by guest on August 5, 2016 http://www.jbc.org/ Downloaded from

Upload: sistemaucem

Post on 22-Feb-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Opposing Actions of Endocannabinoids onCholangiocarcinoma GrowthRECRUITMENT OF Fas AND Fas LIGAND TO LIPID RAFTS*

Received for publication, August 28, 2006, and in revised form, February 26, 2007 Published, JBC Papers in Press, February 28, 2007, DOI 10.1074/jbc.M608238200

Sharon DeMorrow‡, Shannon Glaser‡§, Heather Francis‡, Julie Venter§, Bradley Vaculin§, Shelley Vaculin¶,and Gianfranco Alpini§¶�1

From the ¶Central Texas Veterans Health Care System and the Departments of §Medicine and �Systems Biology and TranslationalMedicine and ‡Division of Research and Education, Scott and White Hospital and Texas A&M University SystemHealth Science Center College of Medicine, Temple, Texas 76504

Cholangiocarcinomas are devastating cancers of biliary originwith limited treatment options. Modulation of the endocan-nabinoid system is being targeted to develop possible therapeu-tic strategies for a number of cancers; therefore, we evaluatedthe effects of the twomajor endocannabinoids, anandamide and2-arachidonylglycerol, on numerous cholangiocarcinoma celllines. Although anandamide was antiproliferative and proapo-ptotic, 2-arachidonylglycerol stimulated cholangiocarcinomacell growth. Specific inhibitors for each of the cannabinoidreceptors didnot prevent either of these effects nor did pretreat-ment with pertussis toxin, a Gi/o protein inhibitor, suggestingthat anandamide and 2-arachidonylglycerol did not exert theirdiametric effects through any known cannabinoid receptor orthrough any otherGi/o protein-coupled receptor.Using the lipidraft disruptors methyl-�-cyclodextrin and filipin, we demon-strated that anandamide, but not 2-arachidonylglycerol,requires lipid raft-mediated events to inhibit cellular prolifera-tion. Closer inspection of the lipid raft structures within thecell membrane revealed that although anandamide treatmenthad no observable effect 2-arachidonylglycerol treatmenteffectively dissipated the lipid raft structures and caused thelipid raft-associated proteins lyn and flotillin-1 to disperseinto the surrounding membrane. In addition, anandamide,but not 2-arachidonylglycerol, induced an accumulation ofceramide, which was required for anandamide-induced sup-pression of cell growth. Finally we demonstrated that anand-amide and ceramide treatment of cholangiocarcinoma cellsrecruited Fas and Fas ligand into the lipid rafts, subsequentlyactivating death receptor pathways. These findings suggestthat modulation of the endocannabinoid systemmay be a tar-get for the development of possible therapeutic strategies forthe treatment of this devastating cancer.

Cholangiocarcinomas are devastating cancers of intrahe-patic and extrahepatic origin that are increasing in both theirworldwide incidence and mortality rates (1, 2). The challengesposed by these often lethal biliary tract cancers are daunting;conventional treatment options are limited, and the only hopefor long term survival is that of complete surgical resection ofthe tumor (1, 2). Conventional chemotherapy and radiationtherapy are not effective in prolonging long term survival (1);therefore it is important to understand the cellularmechanismsof cholangiocarcinoma cell growthwith a view to develop novelchemopreventive strategies.Marijuana and its derivatives have been used in medicine for

many centuries, and presently there is an emerging renaissancein the study of the therapeutic effects of cannabinoids. Ongoingresearch is determining that regulation of the endocannabinoidsystemmaybe effective in the treatment of pain (3, 4), glaucoma(5), and neurodegenerative disorders such as Parkinson disease(6) and multiple sclerosis (7). In addition, cannabinoids mightbe effective antitumoral agents because of their ability to inhibitthe growth of various types of cancer cell lines in culture (8–11)and in laboratory animals (12).The finding in the early 1990s of specific G-protein-coupled

receptors for the psychoactive component of Cannabis sativa(�)-�9-tetrahydrocannabinol (13) led to the discovery of awhole endogenous signaling system now known as the endo-cannabinoid system (14). This system consists of the cannabi-noid receptors (to date there are two, Cb12 and Cb2, as well as aputative involvement of the vanilloid receptor VR1), theirendogenous ligands (endocannabinoids), and the proteins fortheir synthesis and inactivation (14). The cannabinoid recep-tors are seven-transmembrane domain proteins coupled to theGi/o type of G-proteins (14). Cb1 receptors are found predom-inantly in the central nervous system but also in most periph-eral tissues including immune cells, the reproductive system,

* The study was supported by a grant award from Scott and White Hospital(to S. D.), the Dr. Nicholas C. Hightower Centennial Chair of Gastroenterol-ogy, a Veterans Affairs Research Scholar Award, a Veterans Affairs MeritAward, and National Institutes of Health Grant DK062975 (to G. A.). Thecosts of publication of this article were defrayed in part by the payment ofpage charges. This article must therefore be hereby marked “advertise-ment” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1 To whom correspondence should be addressed: Depts. of Medicine andSystems Biology and Translational Medicine, Central Texas VeteransHealth Care System, Texas A&M University System Health Science CenterCollege of Medicine, Medical Research Bldg., 702 S. W. H. K. Dodgen Loop,Temple, TX 76504. Tel.: 254-742-7044; Fax: 254-724-8070; E-mail:[email protected].

2 The abbreviations used are: Cb1, cannabinoid receptor 1; Cb2, cannabinoidreceptor 2; 2-AG, 2-arachidonylglycerol; AEA, anandamide; BrdUrd, bro-modeoxyuridine; DAPI, 4�,6-diamidino-2-phenylindole; dnFADD, domi-nant negative FADD; DR, death receptor; FADD, Fas-associated deathdomain protein; MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; PBS, phosphate-buff-ered saline; TNF, tumor necrosis factor; VR1, vanilloid receptor; FasL, Fasligand; MES, 4-morpholineethanesulfonic acid; shRNA, short hairpin RNA;BSA, bovine serum albumin; MEK, mitogen-activated protein kinase/extra-cellular signal-regulated kinase kinase; ERK, extracellular signal-regulatedkinase.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 282, NO. 17, pp. 13098 –13113, April 27, 2007Printed in the U.S.A.

13098 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

the gastrointestinal tract, and the lung (15–17). On the otherhand, Cb2 receptors are found predominantly in theimmune system, i.e. in tonsils, spleen, macrophages, andlymphocytes (15–17). To date, many endocannabinoids havebeen identified with varying affinities for the receptors (18).Anandamide (AEA) was the first endogenous ligand to beidentified (15); it acts as a partial Cb1 agonist and weak Cb2agonist. Another endocannabinoid is 2-arachidonylglycerol(2-AG), which activates both Cb1 and Cb2 receptors (19, 20).The chemical structures of these two endocannabinoids areshown in Fig. 1. Although the physiological role of the manyother ligands has not yet been fully clarified, AEA and 2-AGhave been implicated in a wide variety of physiological andpathological processes.In addition to their cannabinoid receptor-mediated effects,

endocannabinoids, and in particular AEA, are capable of medi-ating a plethora of receptor-independent cell signaling effects(21–26), most of which are via an interaction with cholesterol-rich microdomains of the cell membrane, also known as lipidrafts (21–23), and the synthesis of the raft-associated, sphingo-lipidmoiety ceramide (24). These lipid rafts are thought to pro-vide a platform for signaling molecules to concentrate and effi-ciently interact thereby facilitating the subsequent signaltransduction process (27, 28). Indeed the subcellular localiza-tion of death receptors such as tumor necrosis factor (TNF)receptor 1, Fas, Fas-associated death domain protein (FADD),and other receptors of the TNF-related apoptosis-inducingligand pathway are redistributed to lipid rafts in response tomany apoptosis-inducing factors including a number of che-motherapeutic agents (29–32).In the present study, we show the opposing effects of AEA

and 2-AG on cholangiocarcinoma cell growth that were canna-binoid receptor-independent. In addition, we show that AEAmediated its inhibitory effects on cell growth by stimulating theaccumulation of ceramide, stabilization of lipid rafts, and therecruitment of Fas and Fas ligand (FasL) to the lipid rafts. Con-versely 2-AG increased cholangiocarcinoma cell growth anddisrupted the organization of the lipid raft structures in the cellmembrane.

EXPERIMENTAL PROCEDURES

Materials—Reagentswere purchased fromSigmaunless oth-erwise stated. AEA, 2-AG, and AM9810 were purchased fromTocris (Ellisville, MO). The cannabinoid receptor inhibitorsSR141716 and SR144528 were obtained from the NationalInstitute on Drug Abuse drug supply program (Bethesda, MD).Antibodies against Cb1 and Cb2 as well as C2-ceramide, C6-ceramide, and filipin III were purchased from Cayman Chemical(Ann Arbor, MI). Spiroepoxide and an anti-ceramide antibodywere obtained from Axxora, LLC (San Diego, CA). BiotinylatedAnnexin V was from Roche Applied Science, anti-bromode-oxyuridine (BrdUrd) antibody was purchased from ChemiconInternational (Temecula, CA), and anti-flotillin-1 and anti-FasLantibodies were from BD Biosciences. Specific antibodies againstthe intra- and extracellular domains of the Fas receptor and lynwerepurchased fromSantaCruzBiotechnology (SantaCruz,CA).Theexpressionvector fordominantnegativeFADDwasakindgiftfromDr.G. J.Gores (MayoClinic,Rochester,MN), andthehumanFADD shRNA plasmids and PCR primers were purchased fromSuperArray (Frederick, MD).Cell Culture—We used four human cholangiocarcinoma cell

lines (Mz-ChA-1, HuH-28, HuCC-T1, and SG231) with differ-ent origins.Mz-ChA-1 cells, fromhuman gallbladder (33), werea gift from Dr. G. Fitz (University of Texas Southwestern Med-ical Center, Dallas, TX). HuH-28 cells, from human intrahe-patic bile duct (34), were acquired fromCancerCell Repository,Tohoku University, Sendai, Japan. These cells were maintainedat standard conditions as described previously (35). In additionHuCC-T1 (36) and SG231 cells (37), also from intrahepatic bileducts, were a kind gift from Dr. A. J. Demetris (University ofPittsburgh, Pittsburgh, PA) andwere cultured as described pre-viously (36, 37). The human immortalized, nonmalignantcholangiocyte cell line H69 (from Dr. G. J Gores, Mayo Clinic)was cultured as described previously (38).Reverse Transcription-PCR Analysis—We evaluated the

expression of the cannabinoid receptors by reverse transcrip-tion-PCR using total RNA (1 �g) from H69, Mz-ChA-1, HuH-28, HUCC-T1, and SG231 cholangiocarcinoma cells. cDNAsynthesis was performed using the Reaction ReadyTM firststrand cDNA synthesis kit from SuperArray. For PCR, we usedprimers specific for humanCb1,Cb2,VR1, and glyceraldehyde-3-phosphate dehydrogenase (Cb1: forward, 5�-ctggttctgatcctg-gtggt-3�; reverse, 5�-tgtctcaggtccttgctcct-3�; product size, 185bp; Cb2: forward, 5�-gattggcagcgtgactatga-3�; reverse, 5�-gct-gagaggacccacatgat-3�; product size, 159 bp; VR1: forward,5�-gcctggagctgttcaagttc-3�; reverse, 5�-tctcctgtgcgatcttgttg-3�;product size, 177 bp). PCR was performed using the followingthermocycler conditions: 5 min at 94 °C followed by 30cycles of 30 s at 94 °C, 30 s at 60 °C, and 45 s at 72 °C followedby a prolonged extension step for 5 min at 72 °C. Ten micro-liters of each PCR product were separated on a 1.5% agarosegel and stained with ethidium bromide and visualized using aChemiImager low light imaging system (Alpha InnotechCorp., San Leandro, CA). Sequence analysis (Davis Sequenc-ing, Davis, CA) indicated that the reverse transcription-PCRfragments for all cannabinoid receptors were homologous totheir corresponding published sequence (GenBankTM acces-

FIGURE 1. Chemical structures of AEA and 2-AG.

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13099

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

sion numbers: human Cb1, AY995204; Cb2, NM_001841;and VR1, NM_080704).Immunoblots—Following trypsinization, H69, Mz-ChA-1,

HuCC-T1, SG231, and HuH-28 cells (1 � 106) were resus-pended in lysis buffer as described previously (35, 39) and son-icated six times (30-s bursts). Immunoblots to detect Cb1, Cb2,and VR1 receptor expression were performed as described pre-viously (35, 39).Immunofluorescence—We detected Cb1, Cb2, and VR1

receptor expression in H69, Mz-ChA-1, HuCC-T1, HuH-28,and SG231 cells. Cells were seeded into 6-well dishes contain-ing a sterile coverslip on the bottom of each well. Cells wereallowed to adhere overnight, washed once in cold PBS, fixed tothe coverslip with 4% paraformaldehyde (in 1� PBS) at roomtemperature for 5 min, permeabilized in PBS containing 0.2%Triton X-100 (PBST), and blocked in 4% bovine serum albumin(BSA) in PBST for 1 h. Cb1, Cb2, and VR1 receptor antibodieswere diluted (1:50 for Cb1 and VR1 antibodies and 1:100 forCb2) in 1%BSA in PBST, added to the coverslips, and incubatedovernight at 4 °C. Cells were washed 3 � 10min in PBST, and a1:50 dilution (in 1%BSA in PBST) of Cy3-conjugated secondaryantibodies (Jackson Immunochemicals, West Grove, PA) wasadded for 2 h at room temperature. Cells were washed againand mounted into microscope slides with Prolong AntifadeGold containing 4�,6-diamidino-2-phenylindole (DAPI) as acounterstain (Molecular Probes, Eugene, OR). Negative con-trols were done with the omission of the respective primaryantibodies. Sections were visualized using an Olympus IX-71inverted confocal microscope.Cell Proliferation Assays—Cholangiocarcinoma growth was

evaluated by a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxyme-thoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt(MTS) proliferation assay in the Mz-ChA-1, HuCC-T1, HuH-28, and SG231 cell lines (40). After trypsinization, cells wereseeded into 96-well plates (10,000 cells/well) in a final volumeof200 �l of medium. Cells were stimulated for 24, 48, and 72 hwith AEA or 2-AG (1 nM to 10 �M). Cell proliferation wasassessed using a colorimetric cell proliferation assay (CellTiter96Aqueous, Promega Corp., Madison, WI), and absorbancewas measured at 490 nm by a microplate spectrophotometer(Versamax, Molecular Devices, Sunnyvale, CA).To determine possible mechanisms of action, Mz-ChA-1

cellswere first preincubated for 1 hwith various inhibitors priorto the addition of AEA or 2-AG for 48 h. The inhibitors utilizedwere SR141716 (aCb1 antagonist, 10 nM (41)); SR144528 (aCb2antagonist, 10 nM) (42); AMG9810 (a VR1 antagonist, 10 nM(43)); pertussis toxin (Gi/o protein inhibitor, 100 ng/ml (44));methyl-�-cyclodextrin (0.1 mM), which depletes intracellularcholesterol and hence disrupts lipid rafts (45); filipin III (1�g/ml), which binds and cross-links intracellular cholesteroland hence disrupts lipid rafts (46, 47); L-cycloserine (0.5 mM), ade novo ceramide synthesis inhibitor (48); or spiroepoxide (100nM), which inhibits ceramide synthesis by hydrolysis of sphin-gomyelin (49).In all cases, datawere expressed as the -fold change of treated

cells as compared with vehicle-treated controls. Statistical sig-nificance was determined using a t test, and p � 0.05 was con-sidered significant.

Annexin V Labeling—Apoptosis was evaluated usingAnnexin V labeling of cells (50). Mz-ChA-1 cells were seededin 6-well plates (500,000 cells/well) containing sterile cover-slips on the bottom of each well and allowed to adhere over-night. Cells were stimulated with AEA (10 �M) or 2-AG (10�M) for 24 h after which time coverslips were removed andrinsed in cold incubation buffer (10 mM HEPES, pH 7.4, 140mM NaCl, 5 mM CaCl2) to remove excess media. Coverslipswere then incubated in a solution containing Annexin V-bi-otin complex (1:50 dilution in incubation buffer) for 30 minat room temperature. After washing the coverslips to removeexcess Annexin V-biotin, the cells were fixed in cold 4%paraformaldehyde (in PBS) for 10 min. Bound Annexin V-bi-otin complex was detected with Cy2-labeled streptavidin(1:200 dilution in PBS), mounted onto microscope slideswith Prolong Antifade Gold containing DAPI, and visualizedusing an Olympus IX-71 inverted confocal microscope. Thenumber of Annexin V-positive cells was counted andexpressed as a percentage of total cells in five random fieldsfor each treatment group. Data are the average � S.E. of fivefields in three independent experiments.BrdUrd Labeling—BrdUrd labeling was used to evaluate the

effects of AEA and 2-AG on cell cycle progression. Mz-ChA-1cells were seeded in 6-well plates (500,000 cells/well) contain-ing sterile coverslips on the bottom of each well and allowed toadhere overnight. Cells were stimulated with AEA (10 �M) or2-AG (10 �M) for 48 h after which time BrdUrd (10 �M finalconcentration) was added to each well and incubated for a fur-ther 3 h at 37 °C. The coverslips were removed, rinsed in PBSto remove excess media, and fixed in 4% paraformaldehyde(in PBS) for 10 min at room temperature. DNA was dena-tured in 2 MHCl for 1 h at 37 °C and then neutralized with 0.5M borate buffer (pH 9.5). The cell membranes were thenpermeabilized with PBST for 3 � 5 min. Nonspecific proteinbinding was then blocked with 4% BSA (in PBST) for 1 h atroom temperature followed by a 2-h incubation with a 1:200dilution (in 1% BSA in PBST) of anti-BrdUrd antibody. Cov-erslips were washed 3 � 5 min in PBST, and a 1:50 dilution(in 1% BSA in PBST) of Cy3-labeled anti-mouse secondaryantibody was added and incubated for 2 h at room tempera-ture. Excess antibody was removed by further washing inPBST, and the coverslips were mounted onto microscopeslides with Prolong Antifade Gold containing DAPI andvisualized using an Olympus IX-71 inverted confocal micro-scope. The number of BrdUrd-positive nuclei was countedand expressed as a percentage of total cells in five randomfields for each treatment group. Data are the average � S.E.of five fields in three independent experiments.Lipid Raft Labeling—Lipid rafts were visualized using the

Vybrant� lipid raft labeling kit (Molecular Probes) with modi-fications. Briefly Mz-ChA-1 cells were seeded onto sterile cov-erslips placed in the bottom of 6-well culture dishes andallowed to adhere overnight. Cells were treated with vehicle,AEA, or 2-AG (both at 10 �M) for 4 h at 37 °C. Coverslips werethenmoved into new6-well dishes for staining. Cells were incu-bated in the presence of 1 �g/ml (diluted in growth media) offluorescent cholera toxin B subunit (Alexa Fluor 594 was thefluorochrome) for 15 min at 4 °C. This cholera toxin B subunit

Endocannabinoids and Cholangiocarcinoma Growth

13100 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

conjugate binds to the pentasaccharide chain of plasma mem-brane ganglioside GM1, which selectively partitions into lipidrafts (51, 52). After this incubation, the cells were gentlywashedwith chilled PBS, fixed in 4% paraformaldehyde (in PBS) for 15min at 4 °C, washed again in PBS, mounted onto microscopeslides with Prolong Antifade Gold containing DAPI, and visu-alized using an Olympus IX-71 inverted confocal microscope.In the lipid raft/Fas co-localization studies, the lipid rafts were

stained and fixed in 4% paraformal-dehyde as described above. The cellswere then counterstained with anantibody against the extracellulardomain of Fas overnight at 4 °Cunder non-permeabilizing condi-tions (i.e. all washes were performedin PBS instead of PBST). Immun-ofluorescent staining of membrane-bound Fas was continued as de-scribed above using a Cy2-labeledanti-rabbit secondary antibody.Analysis of the degree of co-local-ization between lipid rafts and Fasimmunoreactivity was performedusing a Pearson’s correlation analy-sis that is part of the FluoView imag-ing software from Olympus. Usingthis analysis, complete co-localiza-tion of the two stains is representedby a Pearson’s coefficient of 1.0,whereas complete non-colocaliza-tion gives a Pearson’s coefficient of�1.0.Isolation of Detergent-resistant

Lipid Raft Fractions—Mz-ChA-1cells (5 � 106 cells) were seededonto 10-cm cell culture plates andallowed to adhere overnight. Plateswere then stimulated with vehicle,AEA (10�M), 2-AG (10�M),C6-cer-amide (10 �M), spiroepoxide (100nM), or spiroepoxide � AEA for 4 h(two plates for each treatment) afterwhich time cells were scraped intoice-cold PBS, like treatments werepooled, and cells were pelleted bybrief centrifugation. Cells were thenlysed in 300�l of 1�MES buffer (25mM MES, 150 mM NaCl, 1 mMEDTA) containing 1% Triton X-100as well as 1 mM phenylmethylsulfo-nyl fluoride, 1 mM leupeptin, 1 mMaprotinin, and 1 mM sodiumorthovanadate. The cell lysate wasmixedwith 600�l of 60%OptiPrep�(in 1� MES buffer). The resulting40% OptiPrep solution was overlaidby a step gradient of 2.5 ml of 30%OptiPrep (in 1� MES buffer) and

1.0 ml of 5% OptiPrep and centrifuged at 20,000 � g for 20 h at4 °C. Fractionswere collected and analyzed byWestern blottingas described previously (35, 39) using 20 �l of each fraction andspecific antibodies against lyn, �-tubulin, flotillin-1, Fas, andFasL or �-actin.Cholesterol Assays—Cholesterol content was determined

in total cell extracts as well as in each of the lipid raft frac-tions (50-�l sample volume) after basal, AEA (10 �M), and

FIGURE 2. Expression of cannabinoid receptors Cb1, Cb2, and VR1 in cholangiocarcinoma cell lines aswell as H69, SV-40-transformed normal cholangiocytes as shown by PCR (A), Western blotting (B), andimmunofluorescence (C). Receptors were predominantly found in the cytoplasm, and no observable differ-ences in receptor expression levels were seen between the cholangiocarcinoma cell lines and the H69 cell line.Cannabinoid receptor expression is shown in red with nuclei counterstained with DAPI (blue). Original magni-fication for immunofluorescence, �60. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13101

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

2-AG (10 �M) treatment using the cholesterol determinationkit from Molecular Probes according to the manufacturer’sinstructions. For total cholesterol content, results were cor-rected for protein content in each sample and expressed asaverage � S.E. in three samples. For the cholesterol contentin lipid raft fractions, results were expressed as the average(�S.E.) of the percentage of the total cholesterol content inthree samples.Ceramide Labeling—Ceramide synthesis was measured

using the immunofluorescence technique described previouslywith a specific anti-ceramide monoclonal antibody that inphysiological in vitro and in vivo conditions is highly specific forceramide and does not cross-react with sphingomyelin, choles-terol, or other phospholipids (53). Ceramide levels were visual-ized using anOlympus IX-71 inverted confocalmicroscope andquantitated using Adobe Photoshop where the number of cer-amide-positive pixels (red) per nuclei (blue) was assessed. Dataare the average � S.E. in at least five random fields per treat-ment from three independent experiments.

Sphingomyelinase Assays—Sphingomyelinase activity wasdetermined in samples 1, 2, and 4 h after AEA (10 �M) treat-ment using the sphingomyelinase assay kit purchased fromMolecular Probes according to the instructions supplied by thevendor. This assay was performed under neutral and acidicconditions to assess both neutral and acidic sphingomyelinaseactivity. Data were expressed as -fold change in activity of foursamples (average � S.E.).Transient Transfection—Mz-ChA-1 cells were seeded into

96-well plates (5,000 cells/well) and allowed to adhere over-night. Expression vectors containing dominant negative FADDor the control vector was transfected into the cells (0.1 �g ofDNA/well) with 0.28�l/well TransIT-LT1 transfection reagent(Mirus, Madison, WI) overnight at 37 °C. The cells were thenincubated under basal conditions or stimulated with AEA (10�M) for 48 h, and cell viability was determined byMTS assay asdescribed above.In addition, FADD expression was knocked down using

shRNA plasmids containing a green fluorescent protein label

FIGURE 3. Endocannabinoids exert opposing effects on cholangiocarcinoma cell growth. AEA (10 �M) caused a decrease in cell growth as shown by MTSassays in all cholangiocarcinoma cell lines studied (A), whereas 2-AG increased cellular proliferation in all cholangiocarcinoma cell lines studied (B). * denotesp � 0.05 and is considered significant using a t test (n � 7).

Endocannabinoids and Cholangiocarcinoma Growth

13102 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

from SuperArray. Four plasmids containing shRNA sequencesagainst different regions of the human FADD gene were tested.Mz-ChA-1 cells were plated into 96-well plates and transientlytransfected with the shRNA plasmids as described above.Transfection efficiency was monitored using fluorescencemicroscopy and determined to be equal in every well and at anefficiency of 60%. MTS assays were then performed asdescribed above. In a parallel experimentMz-ChA-1 cells wereseeded into 6-well plates and transfected with the shRNA plas-mids. After 48 h, RNA was extracted as described above, andFADD expression was determined by real time PCR. Brieflyreverse transcription was performed using the Reaction Readyfirst strand cDNA synthesis kit (SuperArray). The real timePCRs were set up in 96-well plates using the SYBR Green PCRMaster Mix kit (SuperArray) and either specific primers forhuman FADD or glyceraldehyde-3-phosphate dehydrogenase(SuperArray). The fluorescence was monitored continuouslythroughout 40 cycles of amplification (95 °C for 15 s and 60 °Cfor 1 min) using the ABI Prism 7900HT sequence detectionsystem. A ��CT analysis was performed, and data wereexpressed as -fold change of FADD mRNA expression after

correction for glyceraldehyde-3-phosphate dehydrogenaseexpression.

RESULTS

Cannabinoid Receptor Expression—All of the cholangiocar-cinoma cell lines as well as the SV-40-transformedH69 cholan-giocyte cell line expressed the RNAmessage (Fig. 2A) and pro-tein (Fig. 2B) for the cannabinoid receptors Cb1, Cb2, and VR1.Therewas no obvious difference in levels of expression betweenthe H69 cells and the carcinoma cell lines (Fig. 2, A and B).Immunofluorescence staining of these receptors showed a pre-dominantly cytoplasmic expression in all cell lines consistentwith previous studies (Fig. 2C) (54, 55).Effect of Endocannabinoids on Cholangiocarcinoma Cell

Growth—MTS proliferation assays revealed that AEA (10�M) significantly decreased cell growth 48 h after stimula-tion (Fig. 3A) and up to 72 h (data not shown) in all cholan-giocarcinoma cell lines studied. This concentration farexceeded the Ki values for any of the cannabinoid receptors(Ki � 89 and 371 nM at Cb1 and Cb2, respectively (56, 57)).Concentrations below 10 �M had no effect on growth. In

FIGURE 3—continued

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13103

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

contrast, the other most abundant endocannabinoid, 2-AG(10 �M), had a stimulatory effect on cholangiocarcinoma cellgrowth at 48 h (Fig. 3B) and 72 h (data not shown). Onceagain, there was no effect at concentrations below 10 �M.The fact that these endocannabinoid-induced effects on cellgrowth were seen at concentrations that far exceed the bind-ing affinities for any of the cannabinoid receptors suggeststhat these effects are probably receptor-independent. Indeedspecific cannabinoid antagonists failed to block the AEA-and 2-AG mediated effects (Fig. 4, A and B). In addition,because these cannabinoid receptors are coupled to Gi/o pro-teins, we used pertussis toxin, an inhibitor of these proteins

(44), to determine whether AEA or 2-AG is functioningthrough a Gi/o protein-coupled receptor. As seen in Fig. 4, Aand B, pertussis toxin failed to inhibit either AEA-mediatedinhibition or 2-AG stimulation of cholangiocarcinoma cellgrowth. These inhibitors, however, did effectively block theAEA-mediated suppression of forskolin-induced cAMP pro-duction in these cells (data not shown) in a manner similar tothat in previous studies (58–60), indicating that the receptorantagonists are indeed functional at the concentrations usedhere. This indicates that neither endocannabinoid is exert-ing its effects through the cannabinoid receptors or anyother Gi/o-coupled receptor.

FIGURE 4. The effects of AEA and 2-AG on cholangiocarcinoma cell growth are cannabinoid receptor-independent. Mz-ChA-1 cells were pretreated with10 nM SR141716 (Cb1 antagonist), 10 nM SR144528 (Cb2 antagonist), 10 nM AMG 9810 (VR1 antagonist), or 100 ng/ml pertussis toxin (G-protein antagonist) for1 h prior to the addition of 10 �M AEA (A) or 10 �M 2-AG (B). Cell proliferation was determined by MTS assays. * denotes significance (p � 0.05) when comparedwith basal treatment using a t test (n � 7).

Endocannabinoids and Cholangiocarcinoma Growth

13104 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

Endocannabinoid-induced Effects on Apoptosis and CellCycle Progression—Because the MTS proliferation assay onlygives information on changes in cell number and not themech-anism of any observed change (changes in cell death versus cellcycle progression), we evaluated the effects of AEA and 2-AGon apoptosis, as shown with Annexin V staining, and cell cycleprogression, as indicated by BrdUrd incorporation. As seen inFig. 5A, 27% of the cholangiocarcinoma cells stained positivefor Annexin-V under the basal conditions of our experiment.This value increased up to 60% after AEA treatment for 48 h,whereas 2-AG treatment decreased the incidence of apoptosisslightly but significantly down to 20% Annexin V-positive cells.In contrast, during the 2 h of incubation in the presence of

BrdUrd, 40% of the cells took up BrdUrd into their DNA

under the control conditions ofthese experiments (Fig. 5B). AEAtreatment, however, decreased theamount of BrdUrd-positive cellsand hence the rate of cell cycle pro-gression, and 2-AG treatmentincreased the rate of cell cycleprogression.Endocannabinoids Exert Their

Effects through Cholesterol-rich LipidRafts—Cannabinoids have beenshown previously to have recep-tor-independent effects throughinteraction with lipid rafts (21–23). Therefore, we evaluatedwhether the receptor-independ-ent effects of AEA and 2-AGshown here are also mediatedthrough lipid rafts. Treatment ofMz-ChA-1 cells with the lipid raftdisrupter methyl-�-cyclodextrin(45) had no effect on basal prolif-eration (Fig. 6A). However, pre-treatment with methyl-�-cyclodex-trin inhibited the AEA-mediatedsuppression of cell proliferation butfailed to prevent the 2-AG-inducedstimulation of proliferation (Fig.6A). Similar results were observedfollowing treatment of Mz-ChA-1cells with filipin III, which disruptslipid raft structure by a mechanismdifferent from that of methyl-�-cy-clodextrin (46) (Fig. 6B). Takentogether these data suggest thatAEA requires the integrity of thelipid raft membrane structure to beintact to induce the signal transduc-tion pathways that ultimately resultin increased apoptosis/cell cycleinhibition, whereas this membranestructure is not required for 2-AG-mediated increase in proliferation.We then evaluated the effects of

both AEA and 2-AG on the integrity of the lipid raft membranestructures by staining the GM1 gangliosides, which are an inte-gral component of lipid rafts (51, 52). Fig. 7A, shows the lipidraft structures on the membranes of Mz-ChA-1 cells underbasal conditions. The distribution and morphology of theselipid rafts did not appear to change significantly after AEAtreatment. Stimulation of the cells with 2-AG, however,resulted in marked changes in the lipid raft staining patterns(Fig. 7A). The staining becamemore diffuse and apparently lessmembranous suggesting that perhaps 2-AG effectively disruptsthe lipid raft structure and hence any lipid raft-mediated con-trol of the intracellular machinery responsible for the initiationof apoptosis and/or cell cycle progression. To further demon-strate this phenomenon, we isolated the detergent-resistant

FIGURE 5. A, AEA, but not 2-AG, induces apoptosis in cholangiocarcinoma cells. Mz-ChA-1 cells were treatedwith AEA or 2-AG for 24 h, and apoptosis was measured using Annexin V staining. The number of Annexin-positive cells was expressed as a percentage of total cells. Data are the average � S.E. from five random fieldsfrom three independent experiments. * denotes significance (p � 0.05) when compared with basal treatment.B, AEA suppresses and 2-AG stimulates cell cycle progression. BrdUrd (BrdU) incorporation was used as amarker of cell cycle progression in Mz-ChA-1 cells after AEA or 2-AG treatment. The number of BrdUrd-positivecells was expressed as a percentage of total cells. Data are the average � S.E. from five random fields from threeindependent experiments. * denotes significance (p � 0.05) when compared with basal treatment.

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13105

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

membrane fractions on an OptiPrep density gradient. Previousstudies have shown that using a similar method the lipid raftsmigrate to the interface between the 5 and 30%OptiPrep layers(61). Under the conditions used in the experiments outlinedhere, this corresponds to fractions 3 and 4. Using lyn and flotil-lin-1 as lipid raft markers, it can be seen that the lipid rafts didindeed migrate to the interface between the 5 and 30% Opti-Prep layers under both basal conditions and after AEA treat-ment (i.e. lyn and flotillin-1 were found predominantly in frac-tions 3 and 4, Fig. 7B). However, 2-AG treatment resulted in thedetection of lyn and flotillin-1 in many other fractions besidesfractions 3 and 4 (Fig. 7B) suggesting that the integrity of thelipid rafts was compromised and that components that are usu-ally found exclusively in the raft fraction diffused into the sur-rounding membrane. In these experiments, the non-raft-asso-ciated �-tubulin was used as a negative control to show thatunder our experimental conditions not all proteins are found infractions 3 and 4 and that 2-AG does not disrupt the intracel-lular organization of all proteins (Fig. 7B).Neither AEA nor 2-AG Effects the Total Cholesterol Content,

but 2-AG Effects the Distribution—There was no observabledifference in cholesterol content after 4 h ofAEAor 2-AG treat-ment (Fig. 8). However, upon closer inspection, the distributionof membrane cholesterol in each of the membrane fractionsused in Fig. 7B was markedly different after 2-AG treatment.In cells under basal conditions and AEA-treated cells, themajority of the cholesterol could be seen in fractions 3 and 4corresponding to the lipid raft fractions, whereas after 2-AGtreatment, less cholesterol was observed in fractions 3 and 4and more was observed in the non-raft fractions 7 and 8 (Fig.8). This once again suggests that 2-AGmay be disrupting thelipid raft structures.AEA, but Not 2-AG, Increases Intracellular Ceramide Levels—

AEA has been shown previously to increase ceramide levelswithin the cell (24), and ceramide is an integral component ofthe lipid raft-mediated signaling cascade (62). For this reasonwe determined the effects of AEA and 2-AG on intracellularceramide levels using a specific antibody. As can be seen in Fig.

9A, AEA increased the intracellular ceramide levels 2-foldfrom basal conditions, whereas 2-AG had no effects. Further-more both the acid and neutral sphingomyelinase activitieswere significantly induced from 1 to 4 h after AEA treatment(Fig. 9B). By treating Mz-ChA-1 cells with various concentra-tions of C2- and C6-ceramide, we showed that ceramide causesa decrease in cell proliferation similar to that seen after AEAtreatment as shown by MTS assay (Fig. 9C). In addition, pre-treatment of the cells with spiroepoxide and L-cycloserine,inhibitors of the two pathways leading to the accumulation ofceramide, resulted in an inhibition of the AEA-mediated sup-pression of cell growth but no effect on the 2-AG-mediatedenhancement of cell growth (Fig. 9D). Taken together, thesedata suggest that AEA, but not 2-AG, causes an increase inintracellular ceramide and that this ceramide accumulation isessential for the decrease in cellular proliferation seen afterAEA treatment.Involvement of Fas Activation in AEA-mediated Proapoptotic

Pathway—Because AEA appears to activate proapoptoticevents in cholangiocarcinoma cell lines, we wanted to deter-mine what involvement the death receptor complexes of theTNF superfamily has in this pathway. These death receptors areall capable of using FADD as a downstream effector molecule(63–65). Therefore, we transiently transfected a dnFADDexpression vector into the Mz-ChA-1 cells to determinewhether blocking the death receptor cascade has any effect onthe AEA-induced cell death. Indeed transient dnFADD expres-sion significantly inhibited the AEA-mediated suppression ofcell growth as shown by MTS assay (Fig. 10A). In a similarexperiment, we used shRNA vectors to knock down FADDexpression. Transient transfection of the four different vectorsknocked down the expression of FADD to 25–60% of theexpression levels after transfection of the empty vector (Fig.10B). This specific gene knockdown was enough to block theantiproliferative effects of AEA on Mz-ChA-1 cells. Totalblockage of this antiproliferative effect by either dnFADD orthe shRNA vectors was not achieved because the transfectionefficiency was only 50–60%.

FIGURE 6. Disruption of lipid rafts inhibits AEA- but not 2-AG-mediated effects on cholangiocarcinoma cell growth. Mz-ChA-1 cells were pretreated withlipid raft disrupters 0.1 mM methyl-�-cyclodextrin (A) or 1 �g/ml filipin III (B) for 1 h prior to the addition of 10 �M AEA or 10 �M 2-AG. Cell proliferation wasdetermined my MTS assays. Data are expressed as average � S.E., and * denotes significance (p � 0.05) compared with basal treatment (n � 7).

Endocannabinoids and Cholangiocarcinoma Growth

13106 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

In addition, using the lipid raft plasma membrane fractions,we showed that under basal conditions Fas and FasL were pre-dominantly found in fractions 6 and 7, i.e. the non-lipid raft-containing fractions (Fig. 10C). Upon AEA stimulation thesereceptor components could also be detected in the lipid raftfractions (fractions 3 and 4; Fig. 10C), which presumably facil-itates in the death receptor complex formation and subsequentactivation of proapoptotic pathways. In addition, the recruit-ment of Fas and FasL into the lipid raft fractions was alsoobserved after C6-ceramide treatment (Fig. 10C) suggestingthat the accumulation of ceramide may indeed be responsiblefor the effects of AEA on lipid raft proteins. Furthermore pre-treatment with the ceramide synthesis inhibitor spiroepoxideprior to the addition of AEA blocked the recruitment of bothFas and FasL into the lipid raft fractions.

FIGURE 7. 2-AG but not AEA disrupts the distribution of lipid raftmicrodomains. A, Mz-ChA-1 cells were treated with AEA or 2-AG for 4 h. Lipidrafts were labeled with Alexa Fluor 594-conjugated cholera toxin B subunit(red), and nuclei were counterstained with DAPI (blue) and visualized by con-focal microscopy. Scale bar, 20 �m. Lipid raft structures are indicated with asingle arrowhead. Disrupted lipid raft structures are indicated with a doublearrowhead. B, detergent-resistant lipid rafts from Mz-ChA-1 cells were iso-lated on a discontinuous 5– 40% OptiPrep gradient. The lipid rafts would

normally elute in fractions 3 and 4, as indicated by the boxed area. Antibodiesagainst the lipid raft-associated proteins lyn and flotillin-1 were used in West-ern blotting to determine the fraction in which the lipid rafts eluted.

FIGURE 8. Neither AEA nor 2-AG effects intracellular concentration ofcholesterol, but 2-AG effects the intracellular cholesterol distribution.Total cholesterol content was assayed in Mz-ChA-1 cells 4 h after either AEA or2-AG treatment and corrected for total protein content of each sample. Dataare expressed as the amount of cholesterol (ng)/�g of protein (average � S.E.of three samples). The distribution of cholesterol within the cell membranewas assessed using the lipid raft fractions isolated from Mz-ChA-1 cells 4 hafter AEA or 2-AG treatment. The majority of cholesterol was found in the lipidraft fractions (fractions 3 and 4) after basal and AEA treatment, whereas after2-AG treatment, the majority of cholesterol could be found in the non-raftfractions (fractions 7 and 8). Data are expressed as the percentage of totalcholesterol (average � S.E. of four experiments).

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13107

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

In support of these data, double staining experiments (Fig.10D) showed that under basal conditions there was a moderateamount of overlap between the lipid raft staining (red) and Fas(green) indicated by the yellow color. Pearson’s correlation anal-ysis of the degree of co-localization revealed a Pearson’s coeffi-cient in the range of 0.48–0.65 in six random regions.However,after both AEA and C6-ceramide treatment, there was a greaterdegree of correlation between Fas immunoreactivity and lipidraft staining (Fig. 10D) with a Pearson’s coefficient in the rangeof 0.85–0.95. Other receptors in the TNF superfamily were notstudied here; however, because they can also function in anFADD-dependent manner (63–65) and have been shown pre-viously to be associated with lipid rafts (30, 32), their involve-ment in theAEA-mediated antiproliferative pathway cannot beruled out.

DISCUSSION

The major findings of this study relate to the endocannabi-noid system as a potential therapeutic target aimed to regulatecholangiocarcinoma cell growth. Using our different humancholangiocarcinoma cell lines we showed that 1) all cannabi-noid receptors were expressed to a similar level in cholangio-carcinoma cells versus the SV-40-transformed H69 humancholangiocyte cell line, 2) the two major endocannabinoids,AEA and 2-AG, exert differential effects on cholangiocarci-noma cell growth via receptor-independent mechanisms, 3)AEA causes an antiproliferative and proapoptotic responsethrough stabilization of lipid rafts and recruitment of Fas andFasL into the lipid rafts, and 4) the proliferative effect of 2-AGon cholangiocarcinoma cell growth may be due to the disrup-tion of the lipid raft structure and hence disturbance of thesubsequent lipid raft-dependent cell signaling cascades. Thesedata suggest that therapies aimed at either modulating the rel-ative levels of these two abundant endocannabinoids in vivo ormimicking the antiproliferative effects of AEA may prove ben-eficial in the treatment of cholangiocarcinoma.Consistent with our observation that AEA has antiprolifera-

tive and proapoptotic properties, cannabinoids of various ori-gins (endogenous, plant-derived, or synthetic analogues) havebeen shown to suppress cancer cell growth in vitro (8–11) aswell as in vivo (12). Indeed recently a clinical pilot study todetermine the safety of intracranial administration of (�)-�9-tetrahydrocannabinol in patients with glioblastoma demon-

FIGURE 9. A, AEA, but not 2-AG, increases the intracellular production of cer-amide. Mz-ChA-1 cells were treated with AEA (10 �M) or 2-AG (10 �M), andceramide levels were determined semiquantitatively using a specific anti-body against ceramide in an immunofluorescence technique. Data are

expressed as a ratio of the pixel intensity of the red channel (ceramide) overthe blue channel (DAPI) (average � S.E. in five different fields from threeindependent experiments) * denotes significance (p � 0.05) from basal treat-ment. B, AEA treatment can increase both acid and neutral sphingomyelinaseactivity. Mz-ChA-1 cells were treated with AEA (10 �M) for various time points,and sphingomyelinase activity was assayed using a commercially availablekit. Data are expressed as -fold change in activity over basal activity (aver-age � S.E. of four samples). * denotes significance (p � 0.05). C, syntheticderivatives of ceramide can induce apoptosis in cholangiocarcinoma celllines in a manner similar to that of AEA. Mz-ChA-1 cells were treated withvarious concentrations of C2- and C6-ceramide for 48 h. Cell proliferation wasdetermined by MTS assay. Data are expressed as -fold change from basaltreatment (average � S.E.; n � 7). * denotes significance (p � 0.05). D, inhib-itors of ceramide synthesis block the antiproliferative effects of AEA.Mz-ChA-1 cells were pretreated for 1 h with 100 nM spiroepoxide or 0.5 mM

L-cycloserine prior to the addition of 10 �M AEA or 10 �M 2-AG. Cell prolifera-tion was determined by MTS assay. Data are expressed as -fold change frombasal treatment (average � S.E.; n � 7). * denotes significance (p � 0.05).

Endocannabinoids and Cholangiocarcinoma Growth

13108 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

strated a marked reduction in Ki67 staining within the tumor(66). This observation together with the very promising resultsobserved in cell cultures and laboratory animals (8–12) suggestthat administering cannabinoids alone or in conjunction withexisting chemotherapeutic agents may be a promising treat-ment strategy for aggressive cancers of various origins. Anotherstrategy may be to administer drugs that inhibit endocannabi-noid uptake and subsequent degradation. Bifulco et al. (67)showed that administration of inhibitors of endocannabinoidcellular reuptake and endocannabinoid enzymatic hydrolysisinto athymic mice with thyroid tumor xenografts effectivelyenhanced the endocannabinoid concentrations within thetumor and suppressed the tumor growth. In the present study,we also observed that, in contrast to AEA, 2-AG treatment

resulted in an increase in cholangiocarcinoma cell growth.Although growth-promoting effects of cannabinoids in tumorsof various origins have been demonstrated previously (26), toour knowledge, this is the first demonstration of the opposingeffects of these two endocannabinoids within the one tumortype. To date, 2-AG is the only cannabinoid assayed that hasthis growth-promoting effect in cholangiocarcinoma cells.3 Inlight of this, as well as the fact that 2-AG is the most abundantendocannabinoid in tissues (14), caution must be taken whendeveloping a chemotherapeutic strategy that results in an“across the board” increase in endocannabinoid levels. A treat-

3 S. DeMorrow and G. Alpini, unpublished observation.

FIGURE 9 —continued

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13109

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

ment that specifically targets AEA levels without effecting2-AG levels would obviously be preferable for this tumor type.In the present study, we demonstrated that the effects of

both AEA and 2-AG are mediated via cannabinoid receptor-independent mechanisms, consistent with previous observa-tions (21–26). In fact, both growth-promoting and growth-suppressing actions of cannabinoids in a number of tumortypes are receptor-independent and in the majority of caseswere shown to be via interaction with lipid raft structures inthe plasma membrane and/or regulation of ceramide levels(23–26, 68) in complete agreement with the data presentedhere. Many other anticancer drugs claim the plasma mem-brane as their site of action (69–71). However, little is knownabout the signaling mechanisms downstream from theplasma membrane that are involved in chemotherapy-in-duced cell death; this is an object of ongoing research in ourlaboratory.In our studies, we demonstrated that 1) AEA induced an

accumulation of ceramide within the cell, and 2) this ceramideaccumulation was essential for the subsequent AEA-inducedcell death. Ceramide is an integral component of the lipid raftstructure and dramatically changes the biophysical propertiesof the rafts, allowing very tight packing of lipids within the rafts(72) and hence strongly stabilizing this membrane structure(73). This then facilitates the clustering or aggregation of vari-ous receptor molecules (74), which may theoretically have sev-eral functions including bringing many receptor molecules inclose proximity to facilitate the transactivation between differ-ent receptor systems (74). It may also enhance the interactionbetween a receptor and its intracellular signaling molecules(75). In addition, raft ceramide may directly activate signaltransduction pathways such as the Raf-1/MEK/ERK pathway(12, 76). The ceramide accumulation seen here after AEA treat-ment, we believe, is functioning to stabilize the lipid raft struc-ture; however, the direct effect of ceramide on signal transduc-tion pathways ultimately resulting in cell death cannot be ruledout and is a subject of our ongoing research. Indeed the tumornecrosis factor receptor molecule contains activator domains

FIGURE 10. Death receptors are required for the antiproliferative effectsof AEA and are recruited into lipid rafts by AEA stimulation. A, overexpres-sion of dnFADD blocks the antiproliferative actions of AEA. Mz-ChA-1 cellswere transiently transfected with empty vector or an expression vector con-taining dnFADD for 24 h prior to the addition of 10 �M AEA. Cell proliferationwas determined by MTS assay. Data are expressed as -fold change from basaltreatment (average � S.E.; n � 14). * denotes significance from basal treat-ment (p � 0.05); # denotes significance from empty vector-transfected, AEA-treated samples (p � 0.05). B, transfection of shRNA vectors against human

FADD blocks the antiproliferative actions of AEA. Mz-ChA-1 cells were trans-fected with shRNA vectors or a control vector for 24 h. FADD expression wasdetermined by real time PCR and expressed as relative FADD mRNA aftercorrecting for changes in glyceraldehyde-3-phosphate dehydrogenase(GAPDH) expression. Cell viability was determined by MTS assay after AEAtreatment. MTS data are expressed as -fold change from basal treatment(average � S.E.; n � 14). * denotes significance from basal treatment (p �0.05); # denotes significance from empty vector-transfected, AEA-treatedsamples (p � 0.05). Lane M, molecular size markers. C, detergent-resistantlipid rafts from Mz-ChA-1 cells were isolated on a discontinuous 5– 40% Opti-Prep gradient. The lipid rafts elute in fractions 3 and 4, as indicated by theboxed area. Antibodies against Fas, FasL, flotillin, and �-actin were used inimmunoblotting to show the recruitment of these proteins into the lipid raft-containing fractions after AEA (10 �M), C6-ceramide (10 �M), spiroepoxide(100 nM), and spiroepoxide � AEA stimulation. D, co-localization of Fas immu-noreactivity and lipid raft staining after AEA and C6-ceramide treatment. Mz-ChA-1 cells were treated with AEA (10 �M) or C6-ceramide (10 �M), and thelipid rafts were stained with Alexa Fluor 594-conjugated cholera toxin B sub-unit (red) as well as Fas immunoreactivity (green). Co-localization is indicatedby yellow areas. Nuclei were counterstained with DAPI (blue). Under basalconditions, areas that are clearly Fas-positive but not lipid raft-positive or viceversa are indicated with the single arrowhead. After AEA and C6-ceramidetreatment, areas that are both lipid raft-positive and Fas-positive are indi-cated with the double arrowhead. Scale bar, 10 �m.

Endocannabinoids and Cholangiocarcinoma Growth

13110 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

capable of inducing ceramide production by the activation ofsphingomyelinase (77, 78), and this is FADD-dependent (79).Therefore it is also possible that instead of the AEA-inducedaccumulation of ceramide resulting in the stabilization of lipidrafts and recruitment of death receptors, AEAmay first recruitthe death receptor complex, which then results in the accumu-lation of ceramide. Regardless of the sequence of events, ourdata demonstrate a requirement for ceramide synthesis in theantiproliferative effects of AEA. The precise role of this phe-nomenon will be dissected further in our laboratory.Data, such as those presented here, demonstrating the

recruitment of protein components of the death receptor com-plexes into lipid raft structures are voluminous. It is widelyaccepted that both the TNF-receptor complex and the Fas-receptor complex are both recruited into lipid rafts in responseto apoptosis-inducing drugs; this recruitment facilitates thereceptor complex formation and is critical for their function(80–82). However, the stimulation of this recruitment into

lipid rafts by cannabinoids has notbeen demonstrated previously inany cell type. In addition, thesereceptor complexes all requireFADD to function (65). By overex-pressing dominant negative FADD,we blocked the antiproliferativeeffects of AEA, suggesting that theformation of these death receptorcomplexes is required for the AEA-mediated effects on cholangiocarci-noma cell growth.The effects of death receptor

activation on cholangiocarcinomacell growth have been demon-strated previously (83, 84). Tanakaet al. (83) showed that althoughthe expression of TNF-relatedapoptosis-inducing ligand recep-tors (including DR4 and DR5) isnot changed between resectedcholangiocarcinoma samples andthe surrounding normal liver tissue,exposure of cholangiocarcinomacells to TNF-related apoptosis-in-ducing ligand (ligand of DR5)increased the incidence of apoptosisin vitro and in vivo and reduced thevolume of a subcutaneous xenograftof human cholangiocarcinoma cellsin vivo. In addition, Fas expressionhas been shown to negatively regu-late cholangiocarcinoma cellgrowth in a xenograft model (84).Taken together, we propose thatAEA facilitates the activation ofthese receptor complexes, in partic-ular the Fas receptor, by theirrecruitment into lipid rafts, result-ing in a proapoptotic phenomenon.

In conclusion, we have clearly demonstrated opposingactions of the endocannabinoids AEA and 2-AG on cholangio-carcinoma cell proliferation and have shown that these actionsare via a cannabinoid receptor-independent but lipid raft-me-diated pathway. Furthermore we have shown that the antipro-liferative/proapoptotic actions of AEA are mediated via anaccumulation of ceramide and the recruitment of the Fas deathreceptor into the lipid rafts. Cholangiocarcinoma has a verypoor prognosis and survival rate; therefore we propose that thedevelopment of novel therapeutic strategies aimed at modulat-ing the endocannabinoid system or mimicking the mode ofaction of AEA would prove beneficial for the treatment of thisdevastating disease.

Acknowledgments—We thank Dr. Andreea Trache and Anna Webbof the Texas A&M University Health Science Center MicroscopyImaging Center for assistance with confocal microscopy.

FIGURE 10 —continued

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13111

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

REFERENCES1. Alpini, G., Prall, R. T., and LaRusso, N. F. (2001) The Liver: Biology and

Pathobiology, pp. 421–435, Lippincott, Williams and Wilkins,Philadelphia

2. Sirica, A. E. (2005) Hepatology 41, 5–153. Calignano, A., La Rana, G., Giuffrida, A., and Piomelli, D. (1998) Nature

394, 277–2814. Manzanares, J., Corchero, J., Romero, J., Fernandez-Ruiz, J. J., Ramos, J. A.,

and Fuentes, J. A. (1999) Trends Pharmacol. Sci. 20, 287–2945. Voth, E. A., and Schwartz, R. H. (1997) Ann. Intern. Med. 126, 791–7986. Piomelli, D., Giuffrida, A., Calignano, A., and Rodriguez de Fonseca, F.

(2000) Trends Pharmacol. Sci. 21, 218–2247. Baker, D., Pryce, G., Croxford, J. L., Brown, P., Pertwee, R. G., Huffman,

J. W., and Layward, L. (2000) Nature 404, 84–878. De Petrocellis, L., Melck, D., Palmisano, A., Bisogno, T., Laezza, C., Bi-

fulco, M., and Di Marzo, V. (1998) Proc. Natl. Acad. Sci. U. S. A. 95,8375–8380

9. Sanchez, C., Galve-Roperh, I., Canova, C., Brachet, P., and Guzman, M.(1998) FEBS Lett. 436, 6–10

10. Sanchez, C., de Ceballos, M. L., del Pulgar, T. G., Rueda, D., Corbacho, C.,Velasco, G., Galve-Roperh, I., Huffman, J. W., Ramon y Cajal, S., andGuzman, M. (2001) Cancer Res. 61, 5784–5789

11. Ruiz, L., Miguel, A., and Diaz-Laviada, I. (1999) FEBS Lett. 458, 400–40412. Galve-Roperh, I., Sanchez, C., Cortes, M. L., del Pulgar, T. G., Izquierdo,

M., and Guzman, M. (2000) Nat. Med. 6, 313–31913. Gaoni, Y., and Mechoulam, R. (1971) J. Am. Chem. Soc. 93, 217–22414. Bisogno, T., Ligresti, A., and Di Marzo, V. (2005) Pharmacol. Biochem.

Behav. 81, 224–23815. Devane,W.A., Dysarz, F. A., III, Johnson,M. R.,Melvin, L. S., andHowlett,

A. C. (1988)Mol. Pharmacol. 34, 605–61316. Matsuda, L. A., Lolait, S. J., Brownstein, M. J., Young, A. C., and Bonner,

T. I. (1990) Nature 346, 561–56417. Munro, S., Thomas, K. L., and Abu-Shaar, M. (1993) Nature 365, 61–6518. Di Marzo, V., and Deutsch, D. G. (1998) Neurobiol. Dis. 5, 386–40419. Mechoulam, R., Ben-Shabat, S., Hanus, L., Ligumsky, M., Kaminski, N. E.,

Schatz, A. R., Gopher, A., Almog, S., Martin, B. R., Compton, D. R., Pert-wee, R. G., Griffin, G., Bayewitch, M., Barg, J., and Vogel, Z. (1995) Bio-chem. Pharmacol. 50, 83–90

20. Sugiura, T., Kondo, S., Sukagawa, A., Nakane, S., Shinoda, A., Itoh, K.,Yamashita, A., andWaku, K. (1995)Biochem. Biophys. Res. Commun. 215,89–97

21. Siegmund, S. V., Uchinami, H., Osawa, Y., Brenner, D. A., and Schwabe,R. F. (2005) Hepatology 41, 1085–1095

22. Hinz, B., Ramer, R., Eichele, K., Weinzierl, U., and Brune, K. (2004) Bio-chem. Biophys. Res. Commun. 324, 621–626

23. Hinz, B., Ramer, R., Eichele, K., Weinzierl, U., and Brune, K. (2004) Mol.Pharmacol. 66, 1643–1651

24. Mimeault, M., Pommery, N., Wattez, N., Bailly, C., and Henichart, J. P.(2003) Prostate 56, 1–12

25. Vaccani, A., Massi, P., Colombo, A., Rubino, T., and Parolaro, D. (2005)Br. J. Pharmacol. 144, 1032–1036

26. Gardner, B., Zhu, L. X., Sharma, S., Tashkin, D. P., and Dubinett, S. M.(2003) FASEB J. 17, 2157–2159

27. Simons, K., and Ikonen, E. (1997) Nature 387, 569–57228. Foster, L. J., De Hoog, C. L., and Mann, M. (2003) Proc. Natl. Acad. Sci.

U. S. A. 100, 5813–581829. Gajate, C., Del Canto-Janez, E., Acuna, A. U., Amat-Guerri, F., Geijo, E.,

Santos-Beneit, A.M., Veldman, R. J., andMollinedo, F. (2004) J. Exp.Med.200, 353–365

30. Gajate, C., and Mollinedo, F. (2005) J. Biol. Chem. 280, 11641–1164731. Delmas, D., Rebe, C., Lacour, S., Filomenko, R., Athias, A., Gambert, P.,

Cherkaoui-Malki, M., Jannin, B., Dubrez-Daloz, L., Latruffe, N., and So-lary, E. (2003) J. Biol. Chem. 278, 41482–41490

32. Delmas, D., Rebe, C., Micheau, O., Athias, A., Gambert, P., Grazide, S.,Laurent, G., Latruffe, N., and Solary, E. (2004) Oncogene 23, 8979–8986

33. Knuth, A., Gabbert, H., Dippold, W., Klein, O., Sachsse, W., Bitter-Suer-mann, D., Prellwitz, W., and Meyer zum Buschenfelde, K. H. (1985)

J. Hepatol. 1, 579–59634. Kusaka, Y., Muraoka, A., Tokiwa, T., and Sato, J. (1988) Hum. Cell 1,

92–9435. Kanno, N., Glaser, S., Chowdhury, U., Phinizy, J. L., Baiocchi, L., Francis,

H., LeSage, G., and Alpini, G. (2001) J. Hepatol. 34, 284–29136. Miyagiwa,M., Ichida, T., Tokiwa, T., Sato, J., and Sasaki, H. (1989) In Vitro

Cell. Dev. Biol. 25, 503–51037. Storto, P. D., Saidman, S. L., Demetris, A. J., Letessier, E., Whiteside, T. L.,

and Gollin, S. M. (1990) Genes Chromosomes Cancer 2, 300–31038. Grubman, S. A., Perrone, R. D., Lee, D. W., Murray, S. L., Rogers, L. C.,

Wolkoff, L. I., Mulberg, A. E., Cherington, V., and Jefferson, D. M. (1994)Am. J. Physiol. 266, G1060–G1070

39. Kanno, N., Lesage, G., Phinizy, J. L., Glaser, S., Francis, H., and Alpini, G.(2002) Hepatology 35, 1329–1340

40. Fava, G., Marucci, L., Glaser, S., Francis, H., DeMorrow, S., Benedetti, A.,Alvaro, D., Venter, J., Meininger, C., Patel, T., Taffetani, S., Marzioni, M.,Summers, R., Reichenbach, R., and Alpini, G. (2005) Cancer Res. 65,11437–11446

41. Rinaldi-Carmona, M., Barth, F., Heaulme, M., Shire, D., Calandra, B.,Congy, C., Martinez, S., Maruani, J., Neliat, G., Caput, D., Ferrara, P.,Soubrie, P., Breliere, J. C., and Le Fur, G. (1994) FEBS Lett. 350, 240–244

42. Griffin, G., Wray, E. J., Tao, Q., McAllister, S. D., Rorrer, W. K., Aung,M. M., Martin, B. R., and Abood, M. E. (1999) Eur. J. Pharmacol 377,117–125

43. Doherty, E. M., Fotsch, C., Bo, Y., Chakrabarti, P. P., Chen, N., Gavva, N.,Han, N., Kelly, M. G., Kincaid, J., Klionsky, L., Liu, Q., Ognyanov, V. I.,Tamir, R., Wang, X., Zhu, J., Norman, M. H., and Treanor, J. J. (2005)J. Med. Chem. 48, 71–90

44. Bokoch, G.M., Katada, T., Northup, J. K., Hewlett, E. L., andGilman, A. G.(1983) J. Biol. Chem. 258, 2072–2075

45. Scheiffele, P., Roth, M. G., and Simons, K. (1997) EMBO J. 16, 5501–550846. Orlandi, P. A., and Fishman, P. H. (1998) J. Cell Biol. 141, 905–91547. Smart, E. J., Ying, Y. S., Conrad, P. A., and Anderson, R. G. (1994) J. Cell

Biol. 127, 1185–119748. Sundaram, K. S., and Lev, M. (1984) J. Neurochem. 42, 577–58149. Arenz, C., and Giannis, A. (2000) Angew. Chem. Int. Ed. Engl. 39,

1440–144250. LeSage, G., Alvaro, D., Benedetti, A., Glaser, S., Marucci, L., Baiocchi, L.,

Eisel,W., Caligiuri, A., Phinizy, J. L., Rodgers, R., Francis, H., andAlpini, G.(1999) Gastroenterology 117, 191–199

51. Janes, P. W., Ley, S. C., and Magee, A. I. (1999) J. Cell Biol. 147, 447–46152. Parton, R. G. (1994) J. Histochem. Cytochem. 42, 155–16653. Cowart, L. A., Szulc, Z., Bielawska, A., and Hannun, Y. A. (2002) J. Lipid

Res. 43, 2042–204854. Sarnataro, D., Grimaldi, C., Pisanti, S., Gazzerro, P., Laezza, C., Zurzolo,

C., and Bifulco, M. (2005) FEBS Lett. 579, 6343–634955. Ashton, J. C., Friberg, D., Darlington, C. L., and Smith, P. F. (2006) Neu-

rosci. Lett. 396, 113–11656. Devane, W. A., Hanus, L., Breuer, A., Pertwee, R. G., Stevenson, L. A.,

Griffin, G., Gibson, D., Mandelbaum, A., Etinger, A., and Mechoulam, R.(1992) Science 258, 1946–1949

57. Pertwee, R. G. (1999) Curr. Med. Chem. 6, 635–66458. Ishii, I., and Chun, J. (2002) Neuroreport 13, 593–59659. Melck, D., Rueda, D., Galve-Roperh, I., De Petrocellis, L., Guzman,M., and

Di Marzo, V. (1999) FEBS Lett. 463, 235–24060. Shoemaker, J. L., Ruckle, M. B., Mayeux, P. R., and Prather, P. L. (2005)

J. Pharmacol. Exp. Ther. 315, 828–83861. Kern, A., Roempp, B., Prager, K., Walter, J., and Behl, C. (2006) J. Biol.

Chem. 281, 2405–241362. Bollinger, C. R., Teichgraber, V., and Gulbins, E. (2005) Biochim. Biophys.

Acta 1746, 284–29463. Newton, K., Harris, A.W., Bath,M. L., Smith, K. G., and Strasser, A. (1998)

EMBO J. 17, 706–71864. Yeh, W. C., Pompa, J. L., McCurrach, M. E., Shu, H. B., Elia, A. J., Shahin-

ian, A., Ng, M., Wakeham, A., Khoo, W., Mitchell, K., El-Deiry, W. S.,Lowe, S. W., Goeddel, D. V., and Mak, T. W. (1998) Science 279,1954–1958

65. Ashkenazi, A., and Dixit, V. M. (1998) Science 281, 1305–1308

Endocannabinoids and Cholangiocarcinoma Growth

13112 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 17 • APRIL 27, 2007

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

66. Guzman, M., Duarte, M. J., Blazquez, C., Ravina, J., Rosa, M. C., Galve-Roperh, I., Sanchez, C., Velasco, G., and Gonzalez-Feria, L. (2006) Br. J.Cancer 95, 197–203

67. Bifulco,M., Laezza, C., Valenti,M., Ligresti, A., Portella, G., andDiMarzo,V. (2004) FASEB J. 18, 1606–1608

68. Giuliano, M., Calvaruso, G., Pellerito, O., Portanova, P., Carlisi, D., Vento,R., and Tesoriere, G. (2006) Int. J. Mol. Med. 17, 811–819

69. Arancia, G., and Donelli, G. (1991) Pharmacol. Res. 24, 205–21770. Burns, C. P. (1988) Cancer Investig. 6, 439–45171. Tritton, T. R., and Hickman, J. A. (1990) Cancer Cells 2, 95–10572. Kolesnick, R. N., Goni, F. M., and Alonso, A. (2000) J. Cell. Physiol. 184,

285–30073. Xu, X., Bittman, R., Duportail, G., Heissler, D., Vilcheze, C., and London,

E. (2001) J. Biol. Chem. 276, 33540–3354674. Gulbins, E., and Grassme, H. (2002) Biochim. Biophys. Acta 1585,

139–14575. Field, K. A., Holowka, D., and Baird, B. (1995) Proc. Natl. Acad. Sci. U. S. A.

92, 9201–9205

76. Blazquez, C., Galve-Roperh, I., and Guzman, M. (2000) FASEB J. 14,2315–2322

77. Adam, D., Wiegmann, K., Adam-Klages, S., Ruff, A., and Kronke, M.(1996) J. Biol. Chem. 271, 14617–14622

78. Tartaglia, L. A., Ayres, T. M.,Wong, G. H., and Goeddel, D. V. (1993) Cell74, 845–853

79. Wiegmann, K., Schwandner, R., Krut, O., Yeh, W. C., Mak, T. W., andKronke, M. (1999) J. Biol. Chem. 274, 5267–5270

80. Aouad, S. M., Cohen, L. Y., Sharif-Askari, E., Haddad, E. K., Alam, A., andSekaly, R. P. (2004) J. Immunol. 172, 2316–2323

81. Muppidi, J. R., Tschopp, J., and Siegel, R.M. (2004) Immunity 21, 461–46582. O’Reilly, L. A., Divisekera, U., Newton, K., Scalzo, K., Kataoka, T.,

Puthalakath, H., Ito, M., Huang, D. C., and Strasser, A. (2004) Cell DeathDiffer. 11, 724–736

83. Tanaka, S., Sugimachi, K., Shirabe, K., Shimada, M., and Wands, J. R.(2000) Hepatology 32, 523–527

84. Pickens, A., Pan, G., McDonald, J. M., and Vickers, S. M. (1999) J. Gastro-intest. Surg. 3, 374–382

Endocannabinoids and Cholangiocarcinoma Growth

APRIL 27, 2007 • VOLUME 282 • NUMBER 17 JOURNAL OF BIOLOGICAL CHEMISTRY 13113

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from

Shelley Vaculin and Gianfranco AlpiniSharon DeMorrow, Shannon Glaser, Heather Francis, Julie Venter, Bradley Vaculin,

RECRUITMENT OF Fas AND Fas LIGAND TO LIPID RAFTSOpposing Actions of Endocannabinoids on Cholangiocarcinoma Growth:

doi: 10.1074/jbc.M608238200 originally published online February 28, 20072007, 282:13098-13113.J. Biol. Chem. 

  10.1074/jbc.M608238200Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

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

  http://www.jbc.org/content/282/17/13098.full.html#ref-list-1

This article cites 83 references, 30 of which can be accessed free at

by guest on August 5, 2016

http://ww

w.jbc.org/

Dow

nloaded from