11
Interferon-a Delays S-Phase Progression in Human Hepatocellular Carcinoma Cells Via Inhibition of Specific Cyclin-Dependent Kinases DEREK MURPHY,KATHARINA M. DETJEN,MARTINA WELZEL,BERTRAM WIEDENMANN, AND STEFAN ROSEWICZ The potential antiproliferative effects of interferon-a (IFN-a) in the treatment of hepatocellular carcinoma (HCC) are controversial, and the growth inhibitory mechanisms remain poorly understood. Therefore, the current study was designed to delineate the molecular mechanisms responsi- ble for direct antiproliferative actions of IFN-a in HCC cells. IFN-a receptor expression and signal transduction were ex- amined by RT-PCR, immunoprecipitation, Western analy- sis, and transient transactivation assays. Effects of IFN-a on cell growth and cell-cycle distribution were evaluated based on cell numbers and flow cytometry. Composition and ac- tivity of cyclin-dependent kinase complexes were deter- mined by immunoblotting and histone-H1-kinase assays. Expression of IFN-a receptors was found in all 3 HCC cell lines. IFN-a binding initiated phosphorylation of Jak1 and Tyk2 kinases leading to Stat1/Stat2 activation, nuclear translocation, and transactivation of an ISRE-luciferase re- porter gene construct. IFN-a treatment resulted in a time- and dose-dependent reduction of proliferation. Cell cycle analysis of G1-synchronized, IFN-a–treated HCC cells re- vealed a substantial delay in S-phase progression but no alteration of G 1 /S-phase transition or evidence of apoptotic cell death. Reflecting the time course of S-phase accumula- tion, cell cycle-dependent induction of Cyclin A and Cyclin B was impaired, resulting in reduced activity of Cdk2 and Cdc2 kinases. Furthermore, Cdc25C was selectively down- regulated. IFN-a treatment inhibits growth of HCC cells by specifically delaying S-phase progression, most likely be- cause of inhibition of Cyclin A induction, resulting in de- creased activity of the associated Cdk2 and Cdc2 kinases. (HEPATOLOGY 2001;33:346-356.) Hepatocellular carcinoma (HCC) is one of the most fre- quent malignancies worldwide. HCC commonly arises in liver tissue damaged by long standing chronic inflammation. Fac- tors able to induce and maintain a chronic injury are diverse and include hepatitis B virus (HBV) and hepatitis C virus (HCV) infection 1,2 as well as toxic agents such as alcohol 3 and aflatoxins. 4 Accordingly, a broad variety of genetic alterations contribute to a multistep transformation process leading to clinical manifestation of HCC. The molecular targets of HCC carcinogenesis encompass growth factors and their receptors, signaling cascades and effectors, as well as oncogenes, tumor suppressors, and factors controlling matrix interaction and angiogenesis. 5,6 Although several common alterations such as loss of the tumor suppressors p16 7 and p53 8,9 have been re- ported, tissue-specific molecular defects in HCC remained elusive and thus inaccessible to therapeutic strategies. Despite extensive clinical trials, the prognosis for patients with advanced HCC has remained poor unless curative sur- gery can be performed. 10 Because in most cases the disease is not amenable to surgical resection at the time of diagnosis, an urgent need for systemic treatment is obvious. Interferon-a (IFN-a) belongs to the interferon family of cytokines originally identified because of their function in the antiviral host-defense. 11 Further characterization of IFN-a mediated biological effects revealed a prominent role in the control of cellular proliferation and survival as well as potent immunomodulatory and antiangiogenic actions. Therefore, IFN-a has been successfully included in treatment protocols for malignancies such as hairy-cell leukemia, chronic myelog- enous leukemia, and Kaposi’s sarcoma. 12 IFN-a has proven effective in eliminating HBV and HCV in a considerable number of patients with chronic hepatitis. 13,14 Furthermore, IFN-a receptors are present and functional in the inflamed or cirrhotic liver tissue before transforma- tion. 15,16 Accordingly, considerable expectations to reduce the incidence of HCC were connected with the use of IFN-a in antiviral treatment of hepatitis B or C. By now, first clinical trials have indeed confirmed a reduced incidence of HCC in IFN-a treated patients with chronic active hepatitis C-associ- ated cirrhosis. 17-21 In contrast, the benefit derived from IFN-a treatment of established HCC remains controversial. 22,23 Research over the past decade has comprehensively delin- eated the signal transduction pathways initiated in response to IFN-a. IFN-a binds to specific type I IFN cell-surface re- ceptors. 24 Binding of IFN-a to the receptor causes activation of the receptor associated tyrosine kinases, Jak1 and Tyk2, which in turn recruit and activate the transcription factors Stat1 and Stat2 via tyrosine phosphorylation. On het- Abbreviations: HCC, hepatocellular carcinoma; HBV, hepatitis B virus; HCV, hepatitis C virus; IFN-a, interferon-alfa; DMEM, Dulbecco’s modified Eagle medium; PBS, phos- phate buffered saline; FCS, fetal calf serum; RT-PCR, reverse transcription-polymerase chain reaction; ISRE, interferon-stimulated response element; CMV, cytomegalovirus; SDS-PAGE, sodium dodecyl sulfate–polyacrylamide gel electrophoresis; ECL, enhanced chemiluminescence. From Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Uni- versita ¨tsklinikum Charite ´ , Campus Virchow-Klinikum, Humboldt Universita ¨ t zu Berlin. Received August 8, 2000; accepted November 16, 2000. Supported by the Deutsche Forschungsgemeinschaft, Mildred-Scheel Stiftung and Berliner Krebsgesellschaft (all to SR). Derek Murphy and Katharina M. Detjen contributed equally to this work. Address reprint requests to: Professor Dr. med. Stefan Rosewicz, Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Augustenburger Platz 13353, Ber- lin, Germany. E-mail: [email protected]; fax: (49) 30-450-53-940. Copyright © 2001 by the American Association for the Study of Liver Diseases. 0270-9139/01/3302-0006$35.00/0 doi:10.1053/jhep.2001.21749 346

Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

Embed Size (px)

Citation preview

Page 1: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

Interferon-a Delays S-Phase Progression in Human HepatocellularCarcinoma Cells Via Inhibition of Specific

Cyclin-Dependent Kinases

DEREK MURPHY, KATHARINA M. DETJEN, MARTINA WELZEL, BERTRAM WIEDENMANN, AND STEFAN ROSEWICZ

The potential antiproliferative effects of interferon-a(IFN-a) in the treatment of hepatocellular carcinoma (HCC)are controversial, and the growth inhibitory mechanismsremain poorly understood. Therefore, the current study wasdesigned to delineate the molecular mechanisms responsi-ble for direct antiproliferative actions of IFN-a in HCC cells.IFN-a receptor expression and signal transduction were ex-amined by RT-PCR, immunoprecipitation, Western analy-sis, and transient transactivation assays. Effects of IFN-a oncell growth and cell-cycle distribution were evaluated basedon cell numbers and flow cytometry. Composition and ac-tivity of cyclin-dependent kinase complexes were deter-mined by immunoblotting and histone-H1-kinase assays.Expression of IFN-a receptors was found in all 3 HCC celllines. IFN-a binding initiated phosphorylation of Jak1 andTyk2 kinases leading to Stat1/Stat2 activation, nucleartranslocation, and transactivation of an ISRE-luciferase re-porter gene construct. IFN-a treatment resulted in a time-and dose-dependent reduction of proliferation. Cell cycleanalysis of G1-synchronized, IFN-a–treated HCC cells re-vealed a substantial delay in S-phase progression but noalteration of G1/S-phase transition or evidence of apoptoticcell death. Reflecting the time course of S-phase accumula-tion, cell cycle-dependent induction of Cyclin A and CyclinB was impaired, resulting in reduced activity of Cdk2 andCdc2 kinases. Furthermore, Cdc25C was selectively down-regulated. IFN-a treatment inhibits growth of HCC cells byspecifically delaying S-phase progression, most likely be-cause of inhibition of Cyclin A induction, resulting in de-creased activity of the associated Cdk2 and Cdc2 kinases.(HEPATOLOGY 2001;33:346-356.)

Hepatocellular carcinoma (HCC) is one of the most fre-quent malignancies worldwide. HCC commonly arises in livertissue damaged by long standing chronic inflammation. Fac-tors able to induce and maintain a chronic injury are diverseand include hepatitis B virus (HBV) and hepatitis C virus(HCV) infection1,2 as well as toxic agents such as alcohol3 andaflatoxins.4 Accordingly, a broad variety of genetic alterationscontribute to a multistep transformation process leading toclinical manifestation of HCC. The molecular targets of HCCcarcinogenesis encompass growth factors and their receptors,signaling cascades and effectors, as well as oncogenes, tumorsuppressors, and factors controlling matrix interaction andangiogenesis.5,6 Although several common alterations such asloss of the tumor suppressors p167 and p538,9 have been re-ported, tissue-specific molecular defects in HCC remainedelusive and thus inaccessible to therapeutic strategies.

Despite extensive clinical trials, the prognosis for patientswith advanced HCC has remained poor unless curative sur-gery can be performed.10 Because in most cases the disease isnot amenable to surgical resection at the time of diagnosis, anurgent need for systemic treatment is obvious.

Interferon-a (IFN-a) belongs to the interferon family ofcytokines originally identified because of their function in theantiviral host-defense.11 Further characterization of IFN-amediated biological effects revealed a prominent role in thecontrol of cellular proliferation and survival as well as potentimmunomodulatory and antiangiogenic actions. Therefore,IFN-a has been successfully included in treatment protocolsfor malignancies such as hairy-cell leukemia, chronic myelog-enous leukemia, and Kaposi’s sarcoma.12

IFN-a has proven effective in eliminating HBV and HCV ina considerable number of patients with chronic hepatitis.13,14

Furthermore, IFN-a receptors are present and functional inthe inflamed or cirrhotic liver tissue before transforma-tion.15,16 Accordingly, considerable expectations to reducethe incidence of HCC were connected with the use of IFN-a inantiviral treatment of hepatitis B or C. By now, first clinicaltrials have indeed confirmed a reduced incidence of HCC inIFN-a treated patients with chronic active hepatitis C-associ-ated cirrhosis.17-21 In contrast, the benefit derived from IFN-atreatment of established HCC remains controversial.22,23

Research over the past decade has comprehensively delin-eated the signal transduction pathways initiated in responseto IFN-a. IFN-a binds to specific type I IFN cell-surface re-ceptors.24 Binding of IFN-a to the receptor causes activationof the receptor associated tyrosine kinases, Jak1 and Tyk2,which in turn recruit and activate the transcription factorsStat1 and Stat2 via tyrosine phosphorylation. On het-

Abbreviations: HCC, hepatocellular carcinoma; HBV, hepatitis B virus; HCV, hepatitisC virus; IFN-a, interferon-alfa; DMEM, Dulbecco’s modified Eagle medium; PBS, phos-phate buffered saline; FCS, fetal calf serum; RT-PCR, reverse transcription-polymerasechain reaction; ISRE, interferon-stimulated response element; CMV, cytomegalovirus;SDS-PAGE, sodium dodecyl sulfate–polyacrylamide gel electrophoresis; ECL, enhancedchemiluminescence.

From Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Uni-versitatsklinikum Charite, Campus Virchow-Klinikum, Humboldt Universitat zu Berlin.

Received August 8, 2000; accepted November 16, 2000.Supported by the Deutsche Forschungsgemeinschaft, Mildred-Scheel Stiftung and

Berliner Krebsgesellschaft (all to SR).Derek Murphy and Katharina M. Detjen contributed equally to this work.Address reprint requests to: Professor Dr. med. Stefan Rosewicz, Medizinische Klinik

mit Schwerpunkt Hepatologie und Gastroenterologie, Augustenburger Platz 13353, Ber-lin, Germany. E-mail: [email protected]; fax: (49) 30-450-53-940.

Copyright © 2001 by the American Association for the Study of Liver Diseases.0270-9139/01/3302-0006$35.00/0doi:10.1053/jhep.2001.21749

346

Page 2: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

erodimerization Stat1/Stat2 then translocate into the nucleus,where they control the expression of IFN-regulated genes orgene networks.25

Unlike these early well defined signaling events, growthregulatory pathways responsible for the antitumor action ofIFN-a are not well delineated and appear less conserved. Thismay in part result from the fact that multiple pathways con-tribute to the overall growth regulatory effects.26 Typically,direct antiproliferative action, combined with indirect effectssuch as immunmodulation and interference with angiogene-sis, participate in the biological outcome of IFN-a treatmentand may therefore considerably vary between different tumorentities. Even when focusing on the direct antiproliferativeactions of IFN-a in transformed cell models, various mecha-nisms have been observed. IFN-a has been described to altercell cycle progression by inducing a G0/G1 arrest, prolongingS-phase transition or arresting cells in G2/M.27-29 Further-more, induction of apoptosis has been reported as an alterna-tive response to IFN-a treatment.30-32 The variability in thesedirect responses may be influenced by cell type specific differ-ences in STAT complement and availability of coactivators.33

Alternatively, it reflects differences in the available spectrumof IFN-a–susceptible growth relevant effector molecules be-cause of specific oncogenic alterations present in the targetcell.

In an etiologically heterogeneous tumor entity such asHCC, a detailed molecular knowledge of the antiproliferativemechanisms appears crucial to critically redefine its potentialtherapeutic benefit. Importantly, delineating the componentsand targets of IFN-a growth-modulatory pathways should en-hance our understanding of its potential role and significancein HCC treatment. Therefore, the current study analyzed theaction of IFN-a in a panel of 3 human HCC cell lines. Specif-ically, the effects of IFN-a on central cell cycle regulatoryproteins were examined in detail in order to characterize thegrowth regulatory mechanisms and to identify relevant IFN-a-modulated target molecules in HCC.

MATERIALS AND METHODS

Materials. HepG2 and HuH7 cell lines were obtained from theATCC; SK-Hep-1 cells were a kind gift from D. Schuppan. Dulbecco’smodified Eagle medium (DMEM), RPMI 1640 medium and phos-phate buffered saline (PBS) were purchased from Gibco BRL (Berlin,Germany). Fetal calf serum (FCS), Trypsin/EDTA, penicillin, andstreptomycin were from Seromed (Berlin, Germany). The antibodiesfor Cdk2, Cdc2, Cdc25A, Cdc25C, Jak1, Tyk2, Stat1, and Stat2 werefrom Santa Cruz Biochemicals (Santa Cruz, CA), those for Cyclin A,Cyclin E, and Cyclin B were from Pharmingen (San Diego, CA) andthe a-phosphotyrosine antibody was from Transduction Laborato-ries (Lexington, KY). All secondary antibodies were purchased fomDianova GmbH (Hamburg, Germany). (g-32P)ATP was from Amer-sham (Braunschweig, Germany). Calf histone H1, DNase and ATPwere purchased from Boehringer Mannheim (Mannheim, Germany).Reagents for Western analysis were from BioRad Laboratories GmbH(Munich, Germany) and polyvinyl difluoride (PVDF)-membranesfrom NEN (Brussels, Belgium). IFN-a 2b (Roferon) was kindly sup-plied by Hofmann-LaRoche (Basel, Switzerland). PCR reagents werefrom Promega (Heidelberg, Germany). Protein A-sepharose beadsand all other reagents were from Sigma Chemical Co. (Deisenhofen,Germany).

Cell Lines and Culture. The HCC cell lines HepG2 and HuH7 weregrown in a culture medium consisting of Dulbecco’s modified Eaglemedium supplemented with 10% FCS, 100 U/mL penicillin, 100mg/mL streptomycin, and 2 mmol/L L-glutamine. SK-Hep-1 cellswere grown in RPMI 1640 medium supplemented with 20% FCS,

100 U/mL penicillin, 100 mg/mL streptomycin, and 2 mmol/L L-glutamine. Growth conditions were 37°C in a humidified atmo-sphere of 5% CO2. Cells were cultured for 16 hours before the ex-periments to allow the cells to enter their logarithmic growth phase.

Anchorage Dependent Growth Assays. Cells were grown in 96 wellculture dishes at a density of 2,000 cells/well. After an attachmentperiod of 16 hours, IFN-a was added at the indicated concentration(10-103 IU/mL). Control cells received vehicle only (PBS). After theincubation period cells were trypsinated and stained with trypanblue. Viable cells were then counted using a hemocytometer. Tripli-cate wells were analyzed for each time point and IFN-a dosage.

Flow Cytometry. After trypsination, approximately 106 cells werecollected by centrifugation at 1,000g for 5 minutes. Cells were thenwashed in PBS followed by resuspension and fixation in 70% ethanolfor approximately 2 hours. Next, cells were briefly centrifuged,washed in PBS, and resuspended in 500 mL PBS containing 100 mgRNase and incubated for 30 minutes. Cellular DNA was then stainedby the addition of 10 mg propidium iodide, and a total of 10,000cells/condition were analyzed on a FACScan utilizing Cellquest soft-ware (Becton Dickinson).

Analysis of IFN-a Receptor Expression Using Reverse-Transcription Poly-merase Chain Reaction. Total RNA was prepared using RNAzol R re-agent (WAK Chemie, Bad Soden, Germany) according to the manu-facturer’s instructions. RNA was submitted to DNAse digestion, andcDNA synthesis was carried out using 5 mg total RNA, which wasincubated for 45 minutes in 20 mL reverse-transcriptase (RT) bufferwith 40 U RNAsin and 2mmol/L poly-dT primer and 200 units Molo-ney murine leukemia virus (MMLV) RT. The RT reaction wasstopped by heating the mixture at 95°C for 5 minutes. One tenth ofthis reaction was used as DNA template for the polymerase chainreaction (PCR) of the human IFN-a receptor, using specific primers(59-AGC GAT GAG TCT GTC GGG; 39-GGC GTG GAG CCA CTGAAC). Amplification was carried out in 10 mmol/L Tris HCl (pH 9)containing 50 mmol/L KCl, 0.01% Triton X-100, 1.5 mmol/L MgCl2,200 mmol/L dNTPs each, 50 pmol/L of each primer, and 2.5 U Ther-mus aquaticus polymerase for 30 cycles. Amplification parameterswere as follows: 30 seconds denaturation at 94°C, 60 seconds anneal-ing at 60°C, and 90 seconds extension at 72°C. An additional 10-minute extension period was added to the final cycle. Ten microlitersof the PCR products were separated by electrophoresis in a 1.2%agarose gel, which was subsequently stained in a 0.1% ethidiumbromide solution for 10 minutes. PCR products were then visualizedon a ultraviolet transilluminator. Size determination of the amplifi-cates was deduced from a DNA ladder electrophoresed in parallel.

Transactivation Assay. The ability of IFN-a to stimulate transacti-vation of an interferon-stimulated response element (ISRE) was de-termined in transient transfection assays with an ISRE-Luciferasereporter gene construct. The ISRE-luc construct, containing residues–206/286 of the human 29,59oligo-A synthetase enhancer, and amutated variant were a generous gift from Shoumo Bhattacharya(Boston, MA) and were subcloned into the pTGL2 promoter vector(Promega, Madison, WI). Transfection efficiency was monitored us-ing a b-Gal reporter construct containing the constitutive activeCMV promoter. Two 3 105 cells/well were plated in 6 well tissueculture dishes and transfected with the constructs (1.5 mg) by meansof the calcium phosphate precipitation technique using the “DNATransfection Kit” (5 Prime-3 Prime, Boulder, CO) according to man-ufacturer’s instructions. Transfection was carried out for 6 hours.Cells were allowed to recover overnight followed by stimulation for24 hours with the indicated IFN-a concentrations. Lysates of thecells were prepared, and luciferase and b-Gal activity were measuredaccording to manufacturer’s instructions (Promega, Madison, WI).

Western Analysis. Proteins were isolated in ice cold lysis buffer(150 mmol/L NaCL, 20 mmol/L Tris/HCl [pH 7.8], 2 mmol/L EDTA,50 mmol/L b-glycerophosphate, 0.5% TNP 40, 1% glycerol, 1mmol/L DTT, 10 mmol/L NaF, 10 mg/mL leupeptin, 10 mg/mL apro-tinin, 2 mmol/L phenylmethylsulfonyl fluoride, and 1 mmol/L so-dium orthovanadate). Laemmli buffer was added, the samples wereboiled for 5 minutes, and aliquots were separated by SDS-PAGE.

HEPATOLOGY Vol. 33, No. 2, 2001 MURPHY ET AL. 347

Page 3: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

Samples were electroblotted onto PVDF membranes and blocked inPBST containing either 5% BSA (for Cdk2 and Cdc2 antibodies ) or5% nonfat dry cow milk (all other antibodies) for 2 hours at roomtemperature. Incubation with the primary antibody was carried outovernight at 4°C. After washing in PBST, the blots were incubatedwith horseradish peroxidase-conjugated second antibody, followedby washing. Bands were visualized using the enhanced chemilumi-nescence (ECL) system from Amersham. Quantification of band in-tensity was carried out with help of the computer program ScionImage (Scion Corporation, Frederick, MD).

Determination of Phosphorylation of Stat1, Stat2, Jak1, and Tyk2 by Im-munoprecipitation Analysis. After treatment with/without 1,000 IU/mLIFN-a for the indicated times, cells were rinsed in ice cold phos-phate-buffered saline buffer and lysed in IP lysis buffer (150 mmol/LNaCL, 20 mmol/L Tris/HCl [pH 7.8], 2 mmol/L EDTA, 50 mmol/LGlycerophosphate, 0.5% TNP 40, 1% glycerol, 1 mmol/L DTT, 10mmol/L NaF, 10 mg/mL leupeptin, 10 mg/mL aprotinin, 2 mmol/Lphenylmethylsulfonyl fluoride). The lysates were sonicated, and celldebris was removed by centrifugation (10,000 rpm at 4°C for 15minutes). Aliquots of equal volume and protein content were pre-cleared for 1 hour with protein A-sepharose beads and then incu-bated overnight at 4°C with 10 mg of primary antibody (Stat1, Stat2,Jak1, or Tyk2) precoated onto protein A-sepharose beads under gen-tle rotation. Immunoprecipitates were collected, washed 4 timeswith lysis buffer and subsequently eluted by boiling in Laemmlibuffer. Tyrosin-phosphorylation of the precipitated proteins was an-alyzed by immunoblotting using an a-phosphotyrosine antibody(0,2 mg/mL). To confirm that equal amounts of immuncomplexeswere analyzed, the membranes were stripped in a solution of 62.5mmol/L Tris-HCl, pH 6.8, 2% SDS, and 100 mmol/L 2-mercaptoetha-nol for 30 minutes at 55°C and resubmitted to immunoblotting withthe antibody utilized for the immunoprecipitation.

Determination of Cdk2 and Cdc2 Activity. Activity of the cyclin de-pendent kinases was determined after immunoprecipitation. Cellswere lysed by mild sonication in ice-cold ELB buffer (50 mmol/LHEPES pH 7.5, 250 mmol/L NaCl, 5 mmol/L EDTA, 0.1% NP-40, 1mmol/L DTT, 1 mmol/L NaF, 0.1 mmol/L Na3VO4, 2mg/mL aproti-nin, 5 mg/mL leupeptin, 0.1 mmol/L PMSF) followed by a 30-minuteincubation on ice with occasional vortexing. After brief centrifuga-tion (10 min, 4°C, 13,000g) lysates (500 mg/sample) were preclearedfor 1 hour with protein A-Sepharose beads. Immuncomplexes werethen precipitated by addition of protein A-Sepharose beads pre-coated with an excess of the Cdc2 or Cdk2 antibody (10 mg). Sampleswere incubated with gentle agitation at 4°C for 4 hours. Immuncom-

plexes were subsequently washed 4 times in ice cold ELB buffer andtwice in 50 mmol/L HEPES pH 7.5 containing 1 mmol/L DTT. Kinaseactivity was determined by addition of 30 mL kinase buffer contain-ing 50 mmol/L HEPES pH 7.5, 1 mmol/L DTT, 10 mmol/L MgCl2, 1mg calf histone H1/sample, 50 mmol/L ATP, and 5 mCi of (g-32P)ATP/sample. Kinase reactions were stopped after 30 and 5 minutes forCdc2 and Cdk2, respectively, by boiling the samples in Laemmlibuffer. The reactions were separated by 10% SDS-PAGE, and kinaseactivity was determined by autoradiography of the dried gels.

Statistics. Statistical differences between control and treatmentgroups were calculated by ANOVA (Newman-Keuls) and consideredsignificant at P , .05. Values are given as mean 6 SEM unless oth-erwise stated.

RESULTS

IFN-a Signal Transduction in HCC Cell Lines. Three humanHCC cell lines were utilized throughout the study (HepG2,HuH7, and SK-Hep-1), reflecting the well-differentiated(HepG2) and the poorly differentiated (SK-Hep-1) phenotypeencountered in HCC.

Because loss of expression or function of the IFN-a recep-tor and/or components of the subsequent signaling cascademay account for the IFN-resistance encountered in individualtumors, we initially attempted to confirm functionally intactIFN-a–dependent signal transduction in the HCC cell lines.First, the expression of IFN-a receptors was determined byRT-PCR using IFN-a receptor cDNA-specific primers. Thepredicted 639 base pair (bp) amplificate was detected in eachof the HCC cell lines tested (Fig. 1).

To examine the integrity of IFN-a receptor-initiated signaltransduction, we next evaluated activation of downstream ef-fectors in the IFN-a signal transduction pathway. Accord-ingly, Jak1, Tyk2, Stat1, and Stat2 were immunoprecipitatedfrom cells incubated with IFN-a (103 IU/mL) for various timeperiods. Phosphorylation on tyrosine-residues, which reflectsthe activation state of these signaling components, was thenevaluated by immunoblotting with an antiphosphotyrosineantibody. IFN-a treatment resulted in a rapid and time-de-pendent phosphorylation of the receptor-associated kinasesJak1 and Tyk2 (Fig. 2, upper panels), with maximal phos-phorylation being observed after 15 minutes. As a conse-

FIG. 1. RT-PCR analysis ofIFN-a receptors in human HCC celllines. RT-PCR products were sepa-rated on a 2% agarose gel. The re-verse transcription reaction was car-ried out in the presence (1) andabsence (2) of reverse transcriptaseto exclude artefacts resulting fromgenomic contamination. The arrowindicates the predicted band of 639bp as deduced from the DNA ladderelectrophoresed in parallel.

348 MURPHY ET AL. HEPATOLOGY February 2001

Page 4: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

quence, enhanced phosphorylation was detected for both the91-kd and the 84-kd splice variants of Stat1, with the strongersignal observed for the 91-kd form most likely reflecting itshigher abundance in HCC cells. An analogous, time-depen-dent phosphorylation of Stat2 was also noted in the 3 celllines.

All phosphotyrosine-blots were subsequently stripped andreprobed with the respective primary antibody to verify thatequal quantities of immuncomplexes were analyzed (Fig. 2,lower panels).

IFN-a Is Capable of Transactivating Specific DNA Elements inHCC Cell Lines. On phosphorylation, Stat1 and Stat2 formtranscriptionally active heterodimers that bind to interferon-stimulated response elements (ISREs), DNA motifs found inthe promoter region of target genes. To directly determineIFN-a dependent transactivation of ISREs in HCC cells, re-portergene studies were performed. In cells transiently trans-fected with an ISRE-luciferase reporter construct, IFN-a treat-ment induced a dose-dependent increase in luciferase activity,ranging from 3- to 5-fold in HepG2 and SK-Hep-1 to approx-imately sevenfold in HuH7 cells (Fig. 3). In contrast, cellstransfected with a mutant ISRE construct revealed no change

in the level of luciferase activity (data not shown). Thus,IFN-a induced JAK/STAT-signaling resulted in specific trans-activation of IFN regulated genes in HCC cell lines.

IFN-a Inhibits Anchorage-Dependent Growth. Having establishedan intact IFN-a signaling machinery in all three HCC cell lines, wenext evaluated the growth regulatory effects of IFN-a. Prolifera-tion of HepG2, HuH7, and SK-Hep-1 cells was determined over aperiod of 96 hours in the presence or absence of 1,000 IU/mLIFN-a. In all 3 cell lines, IFN-a-treatment resulted in a time-de-pendent reduction of cell numbers (Fig. 4), with first significanteffects observed at 72 hours in HepG2 and SK-Hep-1 cells. Growthinhibition of HuH7 was slower in onset and did not reach signifi-cance before 96 hours, which most likely reflected the comparablyslower growth rate of this cell line under control conditions. At 96hours of treatment, cell numbers had decreased to 69.7 62.5% of control in HepG2, 55.3 6 10% in HuH7 and to 52. 7 62.4% in SK-Hep-1 cells. The antiproliferative action of IFN-awas also dose-dependent (Fig. 5) with a half maximal inhibi-tion of proliferation calculated at 96 6 1.8 IU/mL for HepG2,at 90 6 1.4 IU/mL for SK-Hep-1, and at 362 6 1.5 IU/mL forHuH7 cells.

FIG. 2. Activation of the Jak-STAT signal transduction pathway in HCC cell lines. Cell lines were stimulated with IFN-a (103 IU/mL) for the indicated timeperiods and immunoprecipitations were performed with antibodies to Jak1, Tyk2, Stat1, and Stat2. Phosphorylation status of these proteins was thendetermined by Western blot analysis using the a-phosphotyrosine antibody PY-20 (upper band). Blots were subsequently stripped and reprobed with theprimary antibody (lower band) to control the amount of immunoprecipitated protein analyzed.

HEPATOLOGY Vol. 33, No. 2, 2001 MURPHY ET AL. 349

Page 5: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

Cell Cycle Analysis of IFN-a–Treated HCC Cells Reveals S-PhaseAccumulation. To gain first insights into the mechanisms ofIFN-a–induced growth inhibition, we next analyzed cell-cy-cle distribution in response to IFN-a treatment by flow cy-tometry. Based on the earlier onset of the antiproliferativeactions of IFN-a in HepG2 and SK-Hep-1 cells, these cell lines

FIG. 4. IFN-a inhibits growth in HCC cells: time course. Subconfluentcell monolayers were treated with IFN-a (103 IU/mL) for the indicated timeperiods and cell numbers were determined. Data represent the means 6 SEMof 3 experiments carried out in triplicate. (*P , .05.)

FIG. 3. IFN-a transactivates an ISRE luciferase reporter construct. Each ofthe HCC cell lines was transfected with 1.5 mg of the ISRE luciferase reporterconstruct. After a recovery period of 24 hours, cells were treated with IFN-aat the indicated concentrations for an additional 24 hours, harvested and thenluciferase activity (RLU) was measured. The data shown represent means 6SEM of 3 experiments conducted in triplicates.

350 MURPHY ET AL. HEPATOLOGY February 2001

Page 6: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

were chosen as representative models for a detailed analysis ofcell cycle effects. In addition, cell populations were synchro-nized in G1 to allow a more distinct representation of theirprogression through the individual cell cycle phases. Syn-chronization was achieved using serum starvation, which rou-tinely retained approximately 70% to 85% of cells in the G0/

G1-phase. Cells were then stimulated to re-enter the cell cycleby addition of fetal calf serum, and cell cycle progression wasmonitored for up to 30 hours both in the presence or absenceof IFN-a (shown in Fig. 6A for SK-Hep-1 cells). For a quan-titative evaluation of cell cycle redistribution, the percentageof cells in the G1-, S-, and G2/M-phase of IFN-a–treated cul-tures were determined and compared with the respective frac-tions in untreated time-matched controls (Fig. 6B).

Serum-stimulated synchronized SK-Hep-1 cultures exitedG0/G1 between 12 and 20 hours following addition of FCS,irrespective of IFN-a treatment (Fig. 6B, first panel). In con-trol cells, the subsequent passage through S-phase was com-pleted at 24 hours as evidenced by a decrease in the S-phasefraction (Fig. 6B, second panel, open circles) paralleled by anincreased G2/M phase population (Fig. 6B, third panel, opencircles). In contrast, S-phase progression was delayed in IFN-a–treated SK-Hep-1 cells, resulting in a significantly increasedS-phase fraction after 20 hours of IFN-a treatment (Fig. 6B,second panel, closed circles). At 30 hours, a decrement in thestill significant S-phase accumulation was noted accompaniedby an increase in the G2/M phase, suggesting that treatedSK-Hep-1 cells resumed cell-cycle progression in spite of thecontinued presence of IFN-a.

A very similar pattern of cell-cycle redistribution was ob-served in HepG2 cells (Fig. 6B). However, the time course ofcell-cycle transition appeared faster such that the G1 exit wasalmost completed at 12 hours after release from serum starva-tion. Again, IFN-a treatment resulted in a pronounced in-crease of the S-phase population.

Neither one of the HCC cell lines presented an accumula-tion of cells in the G1-phase, indicating an unhindered pas-sage of IFN-a–treated cells through the G1 checkpoint. Fur-thermore, no significant increase of cells with subdiploidDNA-content was noted in IFN-a–treated cultures (Fig 6Aand B), suggesting that the antiproliferative action did notresult from apoptosis. Taken together, the cell-cycle analysesindicated that IFN-a–induced cell growth inhibition was de-termined by S-phase regulatory factors.

IFN-a Inhibits Cdk2 and Cdc2 Kinase in SK-Hep-1 Cells. For amore detailed study on the mechanisms of IFN-a–dependentS-phase delay, SK-Hep-1 cells were chosen, because of themore persistent synchronization achieved in these cells. Dur-ing late G1, Rb phosphorylation permits derepression of E2Ffamily transcription factors, which in turn direct the tran-scription of genes required for S-phase progression, includingCyclin A and Cyclin E. Therefore, Rb expression and phos-phorylation were determined in whole cell lysates of SK-Hep-1 cells synchronized and treated as described earlier (Fig.7A). As expected, the hypophosphorylated, faster migratingform of Rb was present in starved control cultures before theaddition of serum. On serum stimulation, this faster migratingband disappeared as cells exited from G1 and, irrespective ofIFN-a treatment, was no longer detectable 12 hours after mi-togen exposure, indicating that the cytokine did not preventRb-phosphorylation in HCC cells. Similarly, the level of theG1-phase associated Cyclin E was not affected by IFN-a until20 to 24 hours after stimulation, when an increase was occa-sionally noted in treated cultures. Taken together, these re-sults are in good agreement with the FACS analyses and placethe IFN-a–dependent cell cycle regulatory events beyond thepassage of the G1 restriction point.

FIG. 5. IFN-a inhibits growth in HCC cells: dose response. Subconfluentmonolayers were treated for 96 hours with the indicated concentrations ofIFN-a. Cell numbers were then determined. Data shown is the mean 6 SEMof 3 experiments, each conducted in triplicate. (*P , .05.)

HEPATOLOGY Vol. 33, No. 2, 2001 MURPHY ET AL. 351

Page 7: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

The subsequent progression of cells through the S-phaseand exit into G2/M are propagated by the sequential activationof Cdk2-Cyclin A, Cdc2-Cyclin A, and Cdc2-Cyclin B com-plexes, which were therefore examined next (Fig. 7). Westernanalysis of total protein lysates showed an induction in cellu-lar Cyclin A levels as control cells progressed from G1 intoS-phase. This Cyclin A induction was invariably reduced intime-matched IFN-a–treated cultures. Similarly, IFN-a treat-ment blunted the increase of cellular Cyclin B content preced-ing G2/M phase. In contrast, Cdk2 expression was not signif-icantly affected throughout the whole experiment, whereas areduction of Cdc2 expression was noted in individual exper-iments (Fig. 7A and B).

Additional control of CDK activity is exerted by inhibitorsof cyclin dependent kinases (CKIs). To examine if an induc-tion of p21cip1 or p27kip1 CKIs participates in IFN-a–mediatedgrowth inhibition in HCC cells, Western blot analyses wereperformed. As expected, the overall cellular level of p27kip1

decreased slightly whereas p21cip1 levels increased as cells re-entered the cell cycle. However, the abundance of both p21cip1

and p27kip1 were not affected by IFN-a treatment when com-pared with their levels in time-matched controls. Thus, theIFN-a–induced S-phase accumulation in HCC cells occursindependent of cellular CKI levels.

The CDC25 family of dual specificity phosphatases pro-vides a third mechanism of controlling CDK activity withCdc25A required for full activation of Cdk2 and Cdc25C forCdc2 activity. We therefore examined Cdc25 expression insynchronized mitogen-stimulated SK-Hep-1 cultures in thepresence or absence of IFN-a (Fig. 7). As expected, expres-sion of Cdc25A was regulated in a cell-cycle dependent man-ner in control cells, whereas Cdc25C levels remained un-changed. IFN-a treatment had no effect on Cdc25Aexpression, but reduced cellular Cdc25C levels at 12, 20, and24 hours. Thus, IFN-a–dependent regulation of Cdc25 ex-pression could participate in the cell cycle effects via inhibi-tion of Cdc2, but not Cdk2 activity.

To directly examine the effects of IFN-a on the activity ofCDK complexes acting at S- and G2/M-phase transitions,histone H1 kinase assays were performed on Cdk2 and Cdc2complexes from synchronized SK-Hep-1 cells. These kinaseassays revealed a marked early inhibition of Cdk2 activity inIFN-a treated cultures compared with time-matched controlcells (Fig. 8A), which persisted for up to 20 hours. Similarly,Cdc2 activity was reduced by IFN-a (Fig. 8B); however thisinhibition was first observed 20 to 24 hours after IFN-a incu-bation. Thus, IFN-a inhibited both Cdk2 and Cdc2 activity.

FIG. 6. Effects of IFN-a on cell cycle distribution of HCC cells. (Upper panels) Cell cycle progression of synchronized SK-Hep-1 populations grown in theabsence (control) or presence of IFN-a (103 IU/mL) at the indicated times after release from serum starvation (A). (Lower panels, B) A summarized time courseof IFN-a-induced cell cycle redistribution in Sk-Hep-1 and HepG2 cells (E, control conditions;F, 1,000 IU/mL IFN-a). Data shown represent the mean 6 SEMof 5 to 10 independent experiments.

352 MURPHY ET AL. HEPATOLOGY February 2001

Page 8: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

Taken together, the observed changes in cell cycle regula-tory molecules were consistent with a significant, but tran-sient inhibition of Cdk2 activity resulting from reduced Cy-clin A levels. Subsequent cell cycle events required for theactivation of Cdc2, such as induction of Cyclin B and Cdc2 aswell as expression of Cdc25C were inhibited (Fig. 7), eithersecondary to the impairment of Cdk2 or because of indepen-dently acting mechanisms.

DISCUSSION

In view of the diverse genetic background of HCC, anygiven therapeutic approach may conceivably yield inconsis-tent and varying responses. Without the identification of rel-evant target molecules, it will be difficult to understand andinterpret the results of clinical trials in such a hetergenouspatient population. This is clearly an issue in the case ofIFN-a, which may affect multiple aspects of HCC, i.e., riskfactors, the hepatocyte transformation process, and later stepsin tumor progression via distinct and separate mechanisms.Thus, the current study undertook a detailed attempt to elu-

cidate one of these aspects, the mechanisms of direct antipro-liferative actions of IFN-a in HCC cells.

Initially, the expression of functionally intact IFN-a recep-tors was established, and the activation of STAT transcriptionfactors was confirmed. Stat1 has been critically implicated inthe growth effects of type I interferons in studies utilizingStat1-deficient cells34 and targeted gene disruption in trans-genic mice.35 Furthermore, molecular defects in the STATactivation pathway are observed in some human tumors.36

However, all 3 cell lines readily showed Stat1 phosphorylationand ISGF3 transactivation on IFN-a treatment, suggestingthat IFN-a–dependent signal transduction is retained inHCC.

With respect to the most common genetic lesions impli-cated in HCC, i.e., loss of the tumor suppressors p16ink47,37

and p53,8 the panel of cell lines that we used reflected some ofthe heterogeneity in the genetic background: HepG2 cells ex-press functionally intact p16ink438 and p53.9,39 Conversely, SK-Hep-1 are p16ink4 deficient (data not shown) but possess func-

FIG. 7. Effects of IFN-a on cell cycle regulatory proteins in SK-Hep-1 cells. (A) Whole cell lysates were isolated from control and IFN-a–treatedsynchronized SK-Hep-1 cells after the indicated time and subjected to Western analysis. (B) Band intensity was determined by densitometry in 2 to 3independent experiments. Values obtained in IFN-a–treated cell populations after 24 hours of culture with 103 IU/mL IFN-a are expressed as a percentage ofthe values in time-matched controls.

HEPATOLOGY Vol. 33, No. 2, 2001 MURPHY ET AL. 353

Page 9: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

tional p53 (in spite of a rearrangement in one allele).9,39

Finally, HuH7 cells lack both functional p16ink440 and p53.9,39

Despite their genetic heterogeneity, IFN-a treatment uni-formly inhibited proliferation of all 3 HCC cell lines. Al-though cell doubling times were significantly prolonged,IFN-a did not completely block proliferation as cells contin-ued to grow even at maximal concentrations of IFN-a. Inagreement with these observations, cell cycle analyses per-formed in synchronized HCC-cell populations did not reveal adistinct cell cycle block, but rather a prolongation of the timeperiod required to progress through S-phase. In unsynchro-nized cell populations, this corresponded to a distinct in-crease in the S-phase population (data not shown), and thusreproduced the IFN-a–dependent cell cycle redistributionpreviously reported by Yano et al.,29 who recorded an in-creased S-phase fraction in 10 out of 11 human HCC cell lines.However, in contrast to their results, we could not observeapoptosis in IFN-a–treated HCC cell cultures. BecauseHepG2 and SK-Hep-1 cells readily undergo apoptosis in re-sponse to chemotherapeutic drugs such as 5-fluorouracil orcisplatin,41 this discrepancy does not reflect an apoptosis-re-sistant phenotype of the cell lines utilized in the current study.Also, both studies obtained comparable EC50s for the growthinhibitory effects of IFN-a, excluding relevant differences inthe sensitivity of the cell lines to the cytokine. Furthermore,the failure of IFN-a to induce apoptosis was not the result ofp53 deficiency, as HepG2 and SK-Hep-1 cells analyzed in thecurrent study express wild-type p53 and, inversely, the HAK1cells reported to undergo apoptosis in response to IFN-a carrya p53 mutation.42

Irrespective of these differences, both studies consistentlyidentified an IFN-a–induced S-phase accumulation as theprevalent cell cycle alteration, suggesting that the molecularmechanism of IFN-a action is highly conserved between dif-ferent HCC tumor collectives. This is noteworthy becauseIFN-a–dependent cell cycle effects in IFN-a–sensitive leuke-

mia and lymphoma cell lines43-46 are directed primarily at theG1/S transition and suggests that IFN-a differentially targetsspecific cell cycle components in different tumor entities. InG1-arrest models, the cell cycle effects are generally Rb-depen-dent and mediated by an increase in the cellular comple-ment of CKIs, such as p15,47 p19,44,49 p21cip1,47,49,50 andp27kip1.47,51,52

Based on the IFN-a–induced cell cycle redistribution ob-served in the current study, neither p15 nor p16ink4 were in-vestigated because these CKIs do not inhibit kinases involvedin later cell cycle phases.53 However, regulation of the lessrestricted CDK inhibitors p21cip1 and/or p27kip154 was exam-ined in SK-Hep-1 cells. Upregulation of both CKIs by gin-sengoid compounds has previously been reported to result ina G1-phase arrest in SK-Hep-1 cells,55,56 indicating that theCDK inhibitors are functionally competent in HCC cells whenadequately stimulated. In the current study, p21cip1 and p27kip1

were found to be regulated cell cycle dependent but not inresponse to IFN-a treatment.

Although most studies on IFN-a–induced cell cycle regu-lation have observed inhibitory effects on G1 to S-phase tran-sition, other studies revealed S-phase accumulation in re-sponse to IFN-a treatment in vivo57 and in vitro.58-61 S-phaseregulatory pathways were proposed to prevail under condi-tions of a severely disrupted Rb-dependent G1 checkpointcontrol, as is often encountered in epithelial tumors.28 How-ever, the mechanisms that operate in the transformed cellmodels to cause S-phase accumulation have remained elusive.Our data on cell cycle distribution show that IFN-a treatmentprevented neither G1-exit nor entry into the S-phase of thecell cycle in synchronized HCC cultures. In sharp contrast,induction of Cyclin A, which occurred in the synchronizedcontrol populations as the cells traversed the restriction point,was distinctly reduced by IFN-a stimulation. Because CyclinA expression is absolutely required for passage through S-phase,62 the decrease of cellular Cyclin A content represents a

FIG. 8. Effects of IFN-a on the activity of the cyclin dependent kinases Cdk2 and Cdc2 in SK-Hep-1 cells. The activity of Cdk2 (A) and Cdc2 (B) wasdetermined after the indicated times using Histone-H1 kinase assays. The upper panel shows a representative autoradiograph for Cdk2 (A) and Cdc2 (B) kinaseassays and the bar graph in the lower half of each figure summarizes the results from 3 independent experiments with the mean 6 SEM of kinase activity aftertreatment with IFN-a expressed as percentage of time-matched controls.

354 MURPHY ET AL. HEPATOLOGY February 2001

Page 10: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

likely cause for IFN-a–dependent growth inhibition in HCC.Supporting a critical role of Cyclin A repression for IFN-amediated cell cycle inhibition, a selective down-regulation ofCyclin A in HCC cell lines similarly resulted in S-phase pro-longation or S-phase arrest.63 The capacity of IFN-a to inter-fere with Cyclin A expression in HCC is particularly notewor-thy in view of the reported overexpression of Cyclin A inapproximately 40% of tumor tissues from HCC-patients.64

Furthermore, an HBV integration into the human Cyclin Agene has been documented in an early HCC that resulted inthe production of a chimeric, degradation-resistant Cyclin Aprotein, responsible for enhanced expression of Cyclin A inthe tumor tissue.65,66

As a consequence of Cyclin A inhibition, subsequent cellcycle events were impaired in IFN-a–treated HCC cell popu-lations, as evidenced by significant downregulation of CyclinB. Cyclin B is synthesized to function as a regulatory subunitin Cdc2 kinase complexes as cells progress from S- into G2/M,67 and the peak of cellular Cyclin B levels observed at theG2/M transition is largely dependent on Cdk2 activity.68

Thus, the lack of Cyclin B induction observed in the IFN-a–treated HCC cells likely reflects the inhibition of Cdk2 activ-ity. Similarly, the cellular Cdc2 content, which analogous toCyclin B depends on Cdk2/E2F mediated induction,69 ap-peared reduced under these conditions but this effect did notreach statistical significance. The comparatively strong inhi-bition of Cyclin B expression might therefore be driven byadditional mechanisms specifically affecting this cyclin. Sucha Cdk-2–independent, IFN-a–induced inhibition of Cyclin Bexpression also appears to operate in human neuroendocrinetumor cells.70

Although reduced Cyclin B levels in IFN-a–treated SK-Hep-1 cells could account for an inhibition of Cdc2 kinaseactivity, additional downregulation of Cdc25C phosphatasemight well contribute to the inhibitory mechanism. Cdc25Cphosphatase dephosphorylates Cdc2 on both Thr-14 andTyr-15 in late G2, leading to the activation of Cdc2-Cyclin Bcomplexes.71,72 As Cdc25C expression is thought to be cellcycle independent,73,74 the reduction of Cdc25C cannot beexplained as a mere consequence of reduced Cdk2 activity,but would represent a second, independent pathway of cellcycle inhibition at the G2/M transition. Despite the fact thatthis mechanism was not rate limiting in SK-Hep-1 cells al-ready arrested in the S-phase of the cell cycle, it might operatein cells that arrest in the G2/M phase in response to type 1interferons.

In summary, the current study provides for the first time adetailed analysis of the direct, cell cycle regulatory effects ofIFN-a in HCC cells. We show a direct antiproliferative actionof IFN-a that results in a substantial but transient delay inS-phase progression. We have identified Cyclin A, Cyclin B,and Cdc25C as relevant, cell cycle regulatory target moleculesof IFN-a in HCC. Via down-regulation of these targets, IFN-ais capable of modulating 2 independent cell cycle controlpathways affecting either the S-phase or the G2/M transition.

REFERENCES

1. Shiratori Y, Shiina S, Imamura M, Kato N, Kanai F, Okudaira T, TerataniT, et al. Characteristic difference of hepatocellular carcinoma betweenhepatitis B- and C- viral infection in Japan. HEPATOLOGY 1995;22:1027-1033.

2. Colombo M. Hepatitis C virus and hepatocellular carcinoma. BaillieresBest Pract Res Clin Gastroenterol 1999;13:519-528.

3. Degos F. Hepatitis C and alcohol. J Hepatol 1999;31(Suppl 1):113-118.

4. Jackson PE, Groopman JD. Aflatoxin and liver cancer. Baillieres BestPract Res Clin Gastroenterol 1999;13:545-555.

5. Ozturk M. Genetic aspects of hepatocellular carcinogenesis. Semin LiverDis 1999;19:235-242.

6. Abou-Shady M, Baer HU, Friess H, Zimmermann A, Buchler MW. Mo-lecular aspects of hepatocellular carcinoma. Swiss Surg 1999;5:102-106.

7. Liew CT, Li HM, Lo KW, Leow CK, Chan JY, Hin LY, Lau WY, et al. Highfrequency of p16INK4A gene alterations in hepatocellular carcinoma.Oncogene 1999;18:789-795.

8. Puisieux A, Ozturk M. TP53 and hepatocellular carcinoma. Pathol Biol(Paris) 1997;45:864-870.

9. Bressac B, Galvin KM, Liang TJ, Isselbacher KJ, Wands JR, Ozturk M.Abnormal structure and expression of p53 gene in human hepatocellularcarcinoma. Proc Natl Acad Sci U S A 1990;87:1973-1977.

10. Cance WG, Stewart AK, Menck HR. The National Cancer Data BaseReport on treatment patterns for hepatocellular carcinomas: improvedsurvival of surgically resected patients, 1985-1996. Cancer 2000;88:912-920.

11. Isaacs A, Lindenmann J. Virus interference. I. The interferon. By A. Isaacsand J. Lindenmann, 1957. J Interferon Res 1987;7:429-438.

12. Gutterman JU. Cytokine therapeutics: lessons from interferon alpha.Proc Natl Acad Sci U S A 1994;91:1198-1205.

13. Davis GL, Balart LA, Schiff ER, Lindsay K, Bodenheimer HC, Jr., PerrilloRP, Carey W, et al. Treatment of chronic hepatitis C with recombinantinterferon alfa. A multicenter randomized, controlled trial. Hepatitis In-terventional Therapy Group. N Engl J Med 1989;321:1501-1506.

14. Malik AH, Lee WM. Chronic hepatitis B virus infection: treatment strat-egies for the next millennium. Ann Intern Med 2000;132:723-731.

15. Zavaglia C, Airoldi A, Pinzello G. Antiviral therapy of HBV- and HCV-induced liver cirrhosis. J Clin Gastroenterol 2000;30:234-241.

16. Di Bisceglie AM, Martin P, Kassianides C, Lisker-Melman M, Murray L,Waggoner J, Goodman Z, et al. Recombinant interferon alfa therapy forchronic hepatitis C. A randomized, double-blind, placebo-controlledtrial. N Engl J Med 1989;321:1506-1510.

17. Ikeda K, Saitoh S, Suzuki Y, Kobayashi M, Tsubota A, Fukuda M, KoidaI, et al. Interferon decreases hepatocellular carcinogenesis in patientswith cirrhosis caused by the hepatitis B virus: a pilot study. Cancer1998;82:827-835.

18. Serfaty L, Aumaitre H, Chazouilleres O, Bonnand AM, Rosmorduc O,Poupon RE, Poupon R. Determinants of outcome of compensated hepa-titis C virus-related cirrhosis. HEPATOLOGY 1998;27:1435-1440.

19. Fattovich G, Giustina G, Realdi G, Corrocher R, Schalm SW. Long-termoutcome of hepatitis B e antigen-positive patients with compensatedcirrhosis treated with interferon alfa. European Concerted Action onViral Hepatitis (EUROHEP). HEPATOLOGY 1997;26:1338-1342.

20. Fattovich G, Giustina G, Sanchez-Tapias J, Quero C, Mas A, Olivotto PG,Solinas A, et al. Delayed clearance of serum HBsAg in compensated cir-rhosis B: relation to interferon alpha therapy and disease prognosis. Eu-ropean Concerted Action on Viral Hepatitis (EUROHEP). Am J Gastro-enterol 1998;93:896-900.

21. Kasahara A, Hayashi N, Mochizuki K, Takayanagi M, Yoshioka K,Kakumu S, Iijima A, et al. Risk factors for hepatocellular carcinoma andits incidence after interferon treatment in patients with chronic hepatitisC. Osaka Liver Disease Study Group. HEPATOLOGY 1998;27:1394-1402.

22. Locker GJ, Mader RM, Steiner B, Wenzl E, Zielinski CC, Steger GG.Benefit of interferon-alpha2b in a patient with unresectable hepatomaand chronic infection with hepatitis C virus [In Process Citation]. Eur JGastroenterol Hepatol 2000;12:251-253.

23. Llovet JM, Sala M, Castells L, Suarez Y, Vilana R, Bianchi L, Ayuso C, etal. Randomized controlled trial of interferon treatment for advanced hep-atocellular carcinoma. HEPATOLOGY 2000;31:54-58.

24. Pestka S. The interferon receptors. Semin Oncol 1997;24:S9-S9.25. Ihle JN. The Janus protein tyrosine kinase family and its role in cytokine

signaling. Adv Immunol 1995;60:1-35.26. Grander D, Sangfelt O, Erickson S. How does interferon exert its cell

growth inhibitory effect? Eur J Haematol 1997;59:129-135.27. Creasey AA, Bartholomew JC, Merigan TC. Role of G0-G1 arrest in the

inhibition of tumor cell growth by interferon. Proc Natl Acad Sci U S A1980;77:1471-1475.

28. Qin XQ, Runkel L, Deck C, DeDios C, Barsoum J. Interferon-beta inducesS phase accumulation selectively in human transformed cells. J Inter-feron Cytokine Res 1997;17:355-367.

29. Yano H, Iemura A, Haramaki M, Ogasawara S, Takayama A, Akiba J,Kojiro M. Interferon alfa receptor expression and growth inhibition byinterferon alfa in human liver cancer cell lines. HEPATOLOGY 1999;29:1708-1717.

HEPATOLOGY Vol. 33, No. 2, 2001 MURPHY ET AL. 355

Page 11: Interferon-α delays S-phase progression in human hepatocellular carcinoma cells via inhibition of specific cyclin-dependent kinases

30. Giandomenico V, Vaccari G, Fiorucci G, Percario Z, Vannuchi S, Matar-rese P, Malorni W, et al. Apoptosis and growth inhibition of squamouscarcinoma cells treated with interferon-alpha, IFN-beta and retinoic acidare associated with induction of the cyclin-dependent kinase inhibitorp21. Eur Cytokine Netw 1998;9:619-631.

31. Sangfelt O, Erickson S, Castro J, Heiden T, Einhorn S, Grander D. Induc-tion of apoptosis and inhibition of cell growth are independent responsesto interferon-alpha in hematopoietic cell lines. Cell Growth Differ 1997;8:343-352.

32. Caraglia M, Abbruzzese A, Leardi A, Pepe S, Budillon A, Baldassare G,Selleri C, et al. Interferon-alpha induces apoptosis in human KB cellsthrough a stress-dependent mitogen activated protein kinase pathwaythat is antagonized by epidermal growth factor. Cell Death Differ 1999;6:773-780.

33. Bromberg J, Darnell Jr JE. The role of STATs in transcriptional controland their impact on cellular function. Oncogene 2000;19:2468-2473.

34. Bromberg JF, Horvath CM, Wen Z, Schreiber RD, Darnell JE, Jr. Tran-scriptionally active Stat1 is required for the antiproliferative effects ofboth interferon alpha and interferon gamma. Proc Natl Acad Sci U S A1996;93:7673-7678.

35. Meraz MA, White JM, Sheehan KC, Bach EA, Rodig SJ, Dighe AS, KaplanDH, et al. Targeted disruption of the Stat1 gene in mice reveals unex-pected physiologic specificity in the JAK-STAT signaling pathway. Cell1996;84:431-442.

36. Kaplan DH, Shankaran V, Dighe AS, Stockert E, Aguet M, Old LJ, Schrei-ber RD. Demonstration of an interferon gamma-dependent tumor sur-veillance system in immunocompetent mice. Proc Natl Acad Sci U S A1998;95:7556-7561.

37. Hui AM, Sakamoto M, Kanai Y, Ino Y, Gotoh M, Yokota J, Hirohashi S.Inactivation of p16INK4 in hepatocellular carcinoma. HEPATOLOGY 1996;24:575-579.

38. Okamoto A, Demetrick DJ, Spillare EA, Hagiwara K, Hussain SP, BennettWP, Forrester K, et al. Mutations and altered expression of p16INK4 inhuman cancer. Proc Natl Acad Sci U S A 1994;91:11045-11049.

39. Yoshikawa H, Nagashima M, Khan MA, McMenamin MG, Hagiwara K,Harris CC. Mutational analysis of p73 and p53 in human cancer cell lines.Oncogene 1999;18:3415-3421.

40. Sandig V, Brand K, Herwig S, Lukas J, Bartek J, Strauss M. Adenovirallytransferred p16INK4/CDKN2 and p53 genes cooperate to induce apopto-tic tumor cell death. Nat Med 1997;3:313-319.

41. Jiang S, Song MJ, Shin EC, Lee MO, Kim SJ, Park JH. Apoptosis in humanhepatoma cell lines by chemotherapeutic drugs via Fas-dependent andFas-independent pathways. HEPATOLOGY 1999;29:101-110.

42. Yano H, Iemura A, Fukuda K, Mizoguchi A, Haramaki M, Kojiro M.Establishment of two distinct human hepatocellular carcinoma cell linesfrom a single nodule showing clonal dedifferentiation of cancer cells.HEPATOLOGY 1993;18:320-327.

43. Tiefenbrun N, Melamed D, Levy N, Resnitzky D, Hoffman I, Reed SI,Kimchi A. Alpha interferon suppresses the cyclin D3 and cdc25A genes,leading to a reversible G0-like arrest. Mol Cell Biol 1996;16:3934-3944.

44. Resnitzky D, Tiefenbrun N, Berissi H, Kimchi A. Interferons and inter-leukin 6 suppress phosphorylation of the retinoblastoma protein ingrowth-sensitive hematopoietic cells. Proc Natl Acad Sci U S A 1992;89:402-406.

45. Zhang K, Kumar R. Interferon-alpha inhibits cyclin E- and cyclin D1-dependent CDK-2 kinase activity associated with RB protein and E2F inDaudi cells. Biochem Biophys Res Commun 1994;200:522-528.

46. Kumar R, Atlas I. Interferon alpha induces the expression of retinoblas-toma gene product in human Burkitt lymphoma Daudi cells: role ingrowth regulation. Proc Natl Acad Sci U S A 1992;89:6599-6603.

47. Sangfelt O, Erickson S, Castro J, Heiden T, Gustafsson A, Einhorn S,Grander D. Molecular mechanisms underlying interferon-alpha-inducedG0/G1 arrest: CKI-mediated regulation of G1 Cdk-complexes and acti-vation of pocket proteins. Oncogene 1999;18:2798-2810.

48. Arora T, Jelinek DF. Differential myeloma cell responsiveness to inter-feron-alpha correlates with differential induction of p19(INK4d) andcyclin D2 expression. J Biol Chem 1998;273:11799-11805.

49. Matsuoka M, Tani K, Asano S. Interferon-alpha-induced G1 phase arrestthrough up-regulated expression of CDK inhibitors, p19Ink4D andp21Cip1 in mouse macrophages. Oncogene 1998;16:2075-2086.

50. Subramaniam PS, Johnson HM. A role for the cyclin-dependent kinaseinhibitor p21 in the G1 cell cycle arrest mediated by the type I interfer-ons. J Interferon Cytokine Res 1997;17:11-15.

51. Mandal M, Bandyopadhyay D, Goepfert TM, Kumar R. Interferon-in-duces expression of cyclin-dependent kinase-inhibitors p21WAF1 and

p27Kip1 that prevent activation of cyclin-dependent kinase by CDK-activating kinase (CAK). Oncogene 1998;16:217-225.

52. Moro A, Santos A, Arana MJ, Perea SE. Activation of the humanp27(Kip1) promoter by IFNalpha 2b. Biochem Biophys Res Commun2000;269:31-34.

53. Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and itsrelatives. Biochim Biophys Acta 1998;1378:F115-F177.

54. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators ofG1-phase progression. Genes Dev 1999;13:1501-1512.

55. Kim SE, Lee YH, Park JH, Lee SK. Ginsenoside-Rs4, a new type of ginsengsaponin concurrently induces apoptosis and selectively elevates proteinlevels of p53 and p21WAF1 in human hepatoma SK-HEP-1 cells. Eur JCancer 1999;35:507-511.

56. Kim SE, Lee YH, Park JH, Lee SK. Ginsenoside-Rs3, a new diol-typeginseng saponin, selectively elevates protein levels of p53 and p21WAF1leading to induction of apoptosis in SK-HEP-1 cells. Anticancer Res1999;19:487-491.

57. Ohwada S, Kobayashi I, Maemura M, Satoh Y, Ogawa T, Iino Y, MorishitaY. Interferon potentiates antiproliferative activity of CPT-11 against hu-man colon cancer xenografts. Cancer Lett 1996;110:149-154.

58. Panniers LR, Clemens MJ. Inhibition of cell division by interferon:changes in cell cycle characteristics and in morphology of Ehrlich ascitestumour cells in culture. J Cell Sci 1981;48:259-279.

59. Lundblad D, Lundgren E. Block of glioma cell line in S by interferon. IntJ Cancer 1981;27:749-754.

60. Genka S, Shitara N, Tsujita Y, Kosugi Y, Takakura K. Effect of interferon-beta on the cell cycle of human glioma cell line U- 251 MG: flowcytometric two-dimensional (BrdU/DNA) analysis. J Neurooncol 1988;6:299-307.

61. Killander D, Lindahl P, Lundin L, Leary P, Gresser I. Relationship be-tween the enhanced expression of histocompatibility antigens on inter-feron-treated L 1210 cells and their position in the cell cycle. Eur J Im-munol 1976;6:56-59.

62. Zindy F, Lamas E, Chenivesse X, Sobczak J, Wang J, Fesquet D, HengleinB, et al. Cyclin A is required in S phase in normal epithelial cells. BiochemBiophys Res Commun 1992;182:1144-1154.

63. Chao Y, Shih YL, Chen HJ, Lee SD, Huang TS. Inhibition of DNA syn-thesis by downregulation of cyclin A but not Skp 2 overexpression inhuman hepatocellular carcinoma cells. Cancer Lett 1999;139:1-6.

64. Chao Y, Shih YL, Chiu JH, Chau GY, Lui WY, Yang WK, Lee SD, et al.Overexpression of cyclin A but not Skp 2 correlates with the tumorrelapse of human hepatocellular carcinoma. Cancer Res 1998;58:985-990.

65. Wang J, Zindy F, Chenivesse X, Lamas E, Henglein B, Brechot C. Modi-fication of cyclin A expression by hepatitis B virus DNA integration in ahepatocellular carcinoma. Oncogene 1992;7:1653-1656.

66. Wang J, Chenivesse X, Henglein B, Brechot C. Hepatitis B virus integra-tion in a cyclin A gene in a hepatocellular carcinoma. Nature 1990;343:555-557.

67. Sherr CJ. Cancer cell cycles. Science 1996;274:1672-1677.68. Moro A, Zerfass K, Joswig S, Jansen-Duerr P. Effect of cyclins and Cdks

on the cyclin B1 promoter activation. Biochem Mol Biol Int 1997;41:919-924.

69. DeGregori J, Kowalik T, Nevins JR. Cellular targets for activation by theE2F1 transcription factor include DNA synthesis- and G1/S-regulatorygenes [published erratum appears in Mol Cell Biol 1995 Oct;15(10):5846-7]. Mol Cell Biol 1995;15:4215-4224.

70. Detjen KM, Welzel M, Farwig K, Brembeck FH, Kaiser A, Riecken EO,Wiedenmann B, et al. Molecular mechanism of interferon alfa-mediatedgrowth inhibition in human neuroendocrine tumor cells. Gastroenterol-ogy 2000;118:735-748.

71. Gautier J, Solomon MJ, Booher RN, Bazan JF, Kirschner MW. cdc25 is aspecific tyrosine phosphatase that directly activates p34cdc2. Cell 1991;67:197-211.

72. Strausfeld U, Labbe JC, Fesquet D, Cavadore JC, Picard A, Sadhu K,Russell P, et al. Dephosphorylation and activation of a p34cdc2/cyclin Bcomplex in vitro by human CDC25 protein. Nature 1991;351:242-245.

73. Gabrielli BG, Clark JM, McCormack AK, Ellem KA. Hyperphosphoryla-tion of the N-terminal domain of Cdc25 regulates activity toward cyclinB1/Cdc2 but not cyclin A/Cdk2. J Biol Chem 1997;272:28607-28614.

74. Millar JB, Blevitt J, Gerace L, Sadhu K, Featherstone C, Russell P.p55CDC25 is a nuclear protein required for the initiation of mitosis inhuman cells. Proc Natl Acad Sci U S A 1991;88:10500-10504.

356 MURPHY ET AL. HEPATOLOGY February 2001