Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology...

12
Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis Xiaoyu Fan 1,2 , Chunyan He 1 , Wei Jing 3 , Xuyu Zhou 3 , Rui Chen 1,2 , Lei Cao 1 , Minhui Zhu 3 , Rongjie Jia 1,2 , Hao Wang 1,2 , Yajun Guo 1,2,4 , and Jian Zhao 1,2,4 Abstract Osteopontin (OPN) has been implicated widely in tumor growth and metastasis, but the range of its contributions is not yet fully understood. In this study, we show that genetic ablation of Opn in mice sensitizes them to diethylnitrosamine (DEN)-induced hepatocarcinogenesis. Opn-decient mice (Opn / mice) exhibited enhanced production of proinammatory cytokines and compen- satory proliferation. Administering OPN antibody or recombinant OPN protein to wild-type or Opn / mice-derived macrophages, respectively, had little effect on cytokine production. In contrast, overexpression of intracellular OPN (iOPN) in Opn-decient macrophages strongly suppressed production of proinammatory cytokines. In addition, we found that iOPN was able to interact with the pivotal Toll-like receptor (TLR) signaling protein MyD88 in macrophages after stimulation with cellular debris, thereby disrupting TLR signaling in macrophages. Our results indicated that iOPN was capable of functioning as an endogenous negative regulator of TLR-mediated immune responses, acting to ameliorate production of proinammatory cytokines and curtail DEN- induced hepatocarcinogenesis. Together, our results expand the important role of OPN in inammation-associated cancers and deepen its relevance for novel treatment strategies in liver cancer. Cancer Res; 75(1); 8697. Ó2014 AACR. Introduction Hepatocellular carcinoma (HCC) is one of the most frequent malignant carcinomas in the world and is known for its high malignancy, quick progression, and poor prognosis. It leads to more than 500,000 deaths per year (1). Major HCC risk factors include viral infection, toxicant and drug metabolic intermedi- ates, and alcohol (2, 3). Diethylnitrosamine (DEN) is a hepatic carcinogen, which can be metabolized into an alkylating agent that induces DNA damage and mutation (4). Because DEN- induced HCC has histologic and genetic features similar to human HCC (5), it has been widely used as a hepatic carcinogen to explore human HCC. Osteopontin (OPN) is a phosphorylated glycoprotein that is expressed in various tissues (6). In liver, OPN is related to steatohepatitis (7), liver brosis progression (8), and liver cancer. In patients with HCC, elevated plasma OPN level is closely related to liver function deterioration and has a positive correlation to tumor stage. Therefore, plasma OPN levels are also regarded as a potent diagnosis biomarker (9). Meanwhile, its disturbance can hamper the growth and metastasis of HCC in vitro and in vivo (10). All these results reveal that OPN aggravates growth and metastasis of HCC. Nevertheless, the role of OPN during HCC initiation is poorly understood. Alternative splicing of OPN results in three isoforms, OPN-a, OPN-b, and OPN-c (11) and alternative translation of OPN generates two isoforms, secreted OPN (sOPN) and intracellular OPN (iOPN; ref. 12). sOPN has been recognized as an extracel- lular protein and participates in several physiologic and patho- logic events, including immune regulation (6), inammation (13), tumor progression, and metastasis (14). iOPN is a shortened protein that lacks the N-terminal signal sequence of sOPN and mainly localizes to cytoplasm (12). It was rst found in rat calvarial cells (15) and now has been found in other kinds of cells, such as dendritic cells (12, 16), macrophages (17), and nerve cells (18). iOPN takes part in several biologic functions. In the nucleus of 293 cells, iOPN colocalizes with polo-like kinase 1 and participates in cell duplication (19). In broblasts, iOPN coloca- lizes with the hyaluronanCD44ERM (ezrin/radixin/moesin) complex at perimembrane regions and plays a role in cell migra- tion (20). iOPN is also an adaptor molecule in innate immunity. In plasmacytoid dendritic cells, it interacts with myeloid differ- entiation primary response gene 88 (MyD88) and enhances interferon-a production through Toll-like receptor 9 (TLR9) sig- naling (16). In antifungal innate immunity, it participates in cluster formation of fungal receptors, as an adaptor molecule of TLR2 and dectin-1 signaling pathways, to enhance phagocytosis and clearance of fungus (21). TLRs are pattern recognition receptors, and participate in innate immune responses against microbial pathogens (22). TLRs acti- vate activator protein-1 (AP-1)/nuclear factor-kB (NF-kB) and 1 International Joint Cancer Institute, The Second Military Medical University, Shanghai, China. 2 School of Pharmacy, Liaocheng Univer- sity, Liaocheng, Shandong, China. 3 Changhai Hospital, The Second Military Medical University, Shanghai, China. 4 PLA General Hospital Cancer Center, PLA Postgraduate School of Medicine, Beijing, China. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). X. Fan, C. He, and W. Jing contributed equally to this article. Corresponding Authors: Yajun Guo, International Joint Cancer Institute, The Second Military Medical University, 800 Xiang Yin Road, Library Building 9-11th Floor, Shanghai 200433, China. Phone: 86-21-81870807; Fax: 86-21-81870801. E-mail: [email protected]; and Jian Zhao, [email protected]. doi: 10.1158/0008-5472.CAN-14-0615 Ó2014 American Association for Cancer Research. Cancer Research Cancer Res; 75(1) January 1, 2015 86 Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association for https://bloodcancerdiscov.aacrjournals.org Downloaded from

Transcript of Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology...

Page 1: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

Molecular and Cellular Pathobiology

Intracellular Osteopontin Inhibits Toll-likeReceptor Signaling and Impedes LiverCarcinogenesisXiaoyu Fan1,2, Chunyan He1,Wei Jing3, Xuyu Zhou3, Rui Chen1,2, Lei Cao1,Minhui Zhu3, Rongjie Jia1,2, Hao Wang1,2, Yajun Guo1,2,4, and Jian Zhao1,2,4

Abstract

Osteopontin (OPN) has been implicated widely in tumorgrowth and metastasis, but the range of its contributions is notyet fully understood. In this study,we show that genetic ablationofOpn inmice sensitizes them to diethylnitrosamine (DEN)-inducedhepatocarcinogenesis.Opn-deficientmice (Opn�/�mice) exhibitedenhanced production of proinflammatory cytokines and compen-satory proliferation. AdministeringOPN antibody or recombinantOPN protein to wild-type or Opn�/� mice-derived macrophages,respectively, had little effect on cytokine production. In contrast,overexpression of intracellular OPN (iOPN) in Opn-deficientmacrophages strongly suppressed production of proinflammatory

cytokines. In addition, we found that iOPN was able to interactwith the pivotal Toll-like receptor (TLR) signaling protein MyD88in macrophages after stimulation with cellular debris, therebydisrupting TLR signaling in macrophages. Our results indicatedthat iOPN was capable of functioning as an endogenous negativeregulator of TLR-mediated immune responses, acting to ameliorateproduction of proinflammatory cytokines and curtail DEN-induced hepatocarcinogenesis. Together, our results expand theimportant role of OPN in inflammation-associated cancers anddeepen its relevance for novel treatment strategies in liver cancer.Cancer Res; 75(1); 86–97. �2014 AACR.

IntroductionHepatocellular carcinoma (HCC) is one of the most frequent

malignant carcinomas in the world and is known for its highmalignancy, quick progression, and poor prognosis. It leads tomore than 500,000 deaths per year (1). Major HCC risk factorsinclude viral infection, toxicant and drug metabolic intermedi-ates, and alcohol (2, 3). Diethylnitrosamine (DEN) is a hepaticcarcinogen, which can be metabolized into an alkylating agentthat induces DNA damage and mutation (4). Because DEN-induced HCC has histologic and genetic features similar tohuman HCC (5), it has been widely used as a hepatic carcinogento explore human HCC.

Osteopontin (OPN) is a phosphorylated glycoprotein that isexpressed in various tissues (6). In liver, OPN is related tosteatohepatitis (7), liver fibrosis progression (8), and liver cancer.In patients withHCC, elevated plasmaOPN level is closely relatedto liver function deterioration and has a positive correlation to

tumor stage. Therefore, plasma OPN levels are also regarded as apotent diagnosis biomarker (9). Meanwhile, its disturbance canhamper the growth andmetastasis ofHCC in vitro and in vivo (10).All these results reveal thatOPN aggravates growth andmetastasisof HCC. Nevertheless, the role of OPN during HCC initiation ispoorly understood.

Alternative splicing of OPN results in three isoforms, OPN-a,OPN-b, and OPN-c (11) and alternative translation of OPNgenerates two isoforms, secreted OPN (sOPN) and intracellularOPN (iOPN; ref. 12). sOPN has been recognized as an extracel-lular protein and participates in several physiologic and patho-logic events, including immune regulation (6), inflammation(13), tumor progression, andmetastasis (14). iOPN is a shortenedprotein that lacks the N-terminal signal sequence of sOPN andmainly localizes to cytoplasm (12). It was first found in ratcalvarial cells (15) and now has been found in other kinds ofcells, such as dendritic cells (12, 16),macrophages (17), andnervecells (18). iOPN takes part in several biologic functions. In thenucleus of 293 cells, iOPN colocalizes with polo-like kinase 1 andparticipates in cell duplication (19). In fibroblasts, iOPN coloca-lizes with the hyaluronan–CD44–ERM (ezrin/radixin/moesin)complex at perimembrane regions and plays a role in cell migra-tion (20). iOPN is also an adaptor molecule in innate immunity.In plasmacytoid dendritic cells, it interacts with myeloid differ-entiation primary response gene 88 (MyD88) and enhancesinterferon-a production through Toll-like receptor 9 (TLR9) sig-naling (16). In antifungal innate immunity, it participates incluster formation of fungal receptors, as an adaptor molecule ofTLR2 and dectin-1 signaling pathways, to enhance phagocytosisand clearance of fungus (21).

TLRs are pattern recognition receptors, and participate in innateimmune responses against microbial pathogens (22). TLRs acti-vate activator protein-1 (AP-1)/nuclear factor-kB (NF-kB) and

1International Joint Cancer Institute, The Second Military MedicalUniversity, Shanghai, China. 2School of Pharmacy, Liaocheng Univer-sity, Liaocheng, Shandong, China. 3Changhai Hospital, The SecondMilitary Medical University, Shanghai, China. 4PLA General HospitalCancer Center, PLA Postgraduate School of Medicine, Beijing, China.

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

X. Fan, C. He, and W. Jing contributed equally to this article.

Corresponding Authors: Yajun Guo, International Joint Cancer Institute, TheSecond Military Medical University, 800 Xiang Yin Road, Library Building 9-11thFloor, Shanghai 200433, China. Phone: 86-21-81870807; Fax: 86-21-81870801.E-mail: [email protected]; and Jian Zhao, [email protected].

doi: 10.1158/0008-5472.CAN-14-0615

�2014 American Association for Cancer Research.

CancerResearch

Cancer Res; 75(1) January 1, 201586

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 2: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

interferon-regulatory factor (IRF) to increase proinflammatorycytokine expression through adaptor molecules MyD88 andTIR-domain–containing adapter-inducing interferon-bb (TRIF;ref. 23). Here, we found that iOPN is recruited to MyD88 understimulation of cellular debris released by necrotic hepatocytes,and negatively regulates TLR signaling in macrophages, whichleads to reduced proinflammatory cytokine production and hepa-tocarcinogenesis in DEN-treatedmice. These findings suggest thatiOPN may function as an endogenous negative regulator of TLR-mediated immune responses to ameliorate inflammation-asso-ciated hepatocarcinogenesis.

Materials and MethodsMice

C57BL/6 mice were purchased from the Shanghai Experi-mental Animal Center, Chinese Academy of Sciences, and thestrain was introduced from The Jackson Laboratory in 2005.Opn�/� mice (B6.Cg-Spp1tm1blh/J; cat. no. 004936) werepurchased from The Jackson Laboratory. Mice in this studywere hosted in a pathogen-free facility under standard 12-hourlight-dark cycle, fed standard rodent chow, and water ad libitum.All animals were maintained in accordance with the guidelinesof the Committee on Animals of the Second Military MedicalUniversity (Shanghai, China).

Animal treatmentFor hepatocarcinogenesis, mice were injected intraperitoneally

(i.p.) with 25 mg/kg of DEN (Sigma) at 14 days of age and thensacrificed at the indicated times. For short-term studies of inflam-mation and liver injury, 6- or 8-week-old male mice were injectedi.p. with 100 mg/kg of DEN and sacrificed at the indicated times.For subcutaneous injection, 6- or 8-week-old male mice wereinjected i.p. with 50 mg/kg of DEN 24 hours before inoculation.

Isolation and cell culturePrimary hepatocyteswere isolated as described (24). Briefly, the

liver was perfused in situ with liver perfusion solution (Gibco).The cell suspensionwasfiltered througha70mmol/L cellfilter (BDFalcon) and the filtrate centrifuged three times at 50 � g for 1minute. The resultant cell pellets were hepatocyte-rich fraction.Hepatocytes were identified by periodic acid-Schiff staining andthe purity regularly exceeded 90%. Isolated hepatocytes were thenfrozen (�80�C), thawed (room temperature) three times, andfiltered to prepare cellular debris released by necrotic hepatocytes.The supernatant run on an agarose gel showed no sign of DNAfragmentation, as would be seen in apoptosis. The concentrationused to stimulate the macrophages was 106 necrotic hepatocytes/mL.

Bonemarrow–derivedmacrophages were prepared as described(25). Both femurs and tibiaswere dissected and flushed. Cells wereincubated with red cell lysis buffer (Beyotime Biotechnology) toobtain pure macrophages. After rinses, cell suspensions werecultured in DMEM (Gibco) supplemented with 10% (vol/vol)heat-inactivated fetal bovine serum (Hyclone), 20 ng/mL MurineMacrophage Colony Stimulating Factor (Peprotech), and penicil-lin–streptomycin solution (1�; Biolight) at 37�C in a humidifiedincubator containing 5% CO2. After overnight incubation, non-adherent cells were removed by three successive washes with PBS.Macrophages were identified by flow cytometry with PE anti-mouse F4/80 antibody and FITC anti-mouse/human CD11b anti-

body (BioLegend). The purity of the isolated subpopulationsregularly exceeded 85%.

HEK 293T and Hepa 1-6, a mouse hepatoma cell line derivedfrom C57BL/6 mice, were purchased from Cell Bank of ShanghaiInstitutes for Biological Sciences, Chinese Academy of Sciences.Cells were cultured at 37�C in a humidified atmosphere of 5%CO2 in DMEM supplemented with 10% FBS.

Construction of expression vectorsMurine full-length (secreted) Opn (Spp1) cDNA was generated

by RT-PCR. BamHI and EcoRI sites were introduced into the PCRprimers for cloning into the pcDNA3.0 vector (Invitrogen), pri-mers: forward, CAGGGATCCATGAGATTGGCAGTGATTTGC;reverse, CGCGAATTCTTAGTTGACCTCAGAAGATGAACTC. Theexpression plasmid for intracellular form of Opn (iOpn) deletingthe codons from 1 to 15 was generated from sOpn expressionplasmid by PCR-mediated mutagenesis with the QuikChange IIXL Site-directed Mutagenesis kit (Stratagene). Murine MyD88cDNA was generated by RT-PCR. EcoRI and XhoI sites wereintroduced into pcDNA3.0 vector, primers: forward, GTGAATT-CATGTCTGCGGGAGACCC; reverse, GACTCGAGTCAGGGCAG-GGACAAAG. The preceiver-myc-IRAK1 vector was purchasedfrom the Guangzhou Fulen Gene Company.

RNA isolation and quantitative RT-PCRTotal RNA was isolated using the Nucleospin RNA (Macherey-

Nagel). First-strand synthesis was performed with random pri-mers and reverse transcription with Quant Reverse Transcriptase(Tiangen Biotech). Quantitative RT-PCR was performed usingSYBRGreen reagent (TaKaRa) in a Light Cycler (Roche). Reactionswere performed twice in triplicate, and actin values were used tonormalize gene expression. The primer sequences canbe obtainedfrom Supplementary Table S1.

Biochemical analysis and ELISASerum alanine transaminase (ALT) levels in liver were mea-

sured with commercial kits (Nanjing Jiancheng). Levels of IL6and TNFa from serum and cell supernatant were quantitatedwith ELISA kits (eBioscience) according to the manufacturer'sinstructions. Cell supernatant of bone marrow–derived macro-phages for OPN qualification was quantitated with the mouseOPN ELISA Kit (R&D Systems). Preparation of cell supernatantwas as follows. 2 � 106 macrophages were seeded into 24-wellplates; after culturing for 5 days, medium was discarded, andreplaced by serum-freemediumwith cellular debris fromOpn�/

� necrotic hepatocytes. Supernatant was collected at the indi-catedtimes.

PathScan Inflammation Multi-Target Sandwich ELISA kit (CellSignaling Technology): liver tissues were lysed and prepared as 1mg/mL by the BCA Protein Assay Kit (Thermo Scientific) fordetecting endogenous levels of transcription factors: NF-kBp65, phospho-NF-kB p65 (Ser536), phospho-SAPK/JNK(Thr183/Tyr185), phospho-p38 MAPK (Thr180/Tyr182), phos-pho-STAT3 (Tyr705), and phospho-IkB-a (Ser32). Each phos-phoprotein absorbance was corrected by the negative control andnormalized by its relevant NF-kB p65 absorbance.

Western blottingCells were homogenized in 1� SDS lysis buffer (62.5 mmol/L

Tris–HCl, 2% w/v SDS, 10% glycerol, 50 mmol/L DTT, 0.01%

iOPN Inhibits Hepatocarcinogenesis in Mice

www.aacrjournals.org Cancer Res; 75(1) January 1, 2015 87

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 3: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

w/v bromophenol blue). Frozen livers were prepared in ice-coldRIPA lysis buffer (50 mmol/L Tris–HCl, 150 mmol/L NaCl, 1%v/v NP-40, 0.5% w/v sodium deoxycholate, 0.1%w/v SDS)supplemented with Mammalian Protease Inhibitor Mixture(100�; BioColors) and protein concentration of the extracts wasmeasured by the BCA Protein Assay Kit (Thermo Scientific) forWestern blotting. Proteins were separated by SDS-PAGE andtransferred onto polyvinylidene difluoride (PVDF) membranes(0.45 mm, Millipore). After probing with individual antibodies,the antigen–antibody complex was visualized by EnhancedChemiluminescence's Reagents Supersignal (Pierce Biotechnol-ogy). The antibodies used in this study are listed in the Supple-mentary Table S2.

Coimmunoprecipitation analysisFor detecting endogenous levels of interaction, with or without

stimulation by cellular debris released by necrotic hepatocytes,macrophages from WT or Opn�/� mice were lysed by RIPA lysisbuffer and incubated with anti-OPN antibody (Mouse monoclo-nal antibody 23C3, details in Supplementary Methods; 1:50),anti-MyD88 monoclonal antibody (Cell Signaling Technology;1:50), or normal IgG (Cell Signaling Technology; 1:50) overnightat 4�C. To detect interaction in vitro, HEK 293T cells were cotrans-fected with MyD88-pcDNA3.0, preceiver-myc-IRAK1, and iOpn-pcDNA3.0/pcDNA3.0 plasmid. At 48hours posttransfection, cellswere incubated with cellular debris for 4 hours and then thetotal cell lysateswere lysedbyRIPA lysis buffer and incubatedwithanti-MyD88 antibody or normal IgG overnight at 4�C. Preclearedprotein A/G-Sepharose (Santa Cruz Biotechnology) was usedto isolate antibody-bound proteins; precipitated complexeswere separated by SDS-PAGE and subjected to Western blottinganalysis.

Histologic, immunohistochemical assayFormalin-fixed, paraffin-embedded liver tissues were used for

hematoxylin and eosin (H&E), terminal deoxynucleotidyl trans-ferase-mediated dUTP nick end labeling (TUNEL), 5-bromo-20-deoxyuridine (BrdUrd), proliferating cell nuclear antigen (PCNA),and F4/80 staining. Apoptosis was assessed by TUNEL stainingwith the TUNEL Detection Kit (Calbiochem) according to themanufacturer's instructions. Proliferation was assessed by BrdUrd(Sigma) and PCNA monoclonal antibody (Cell Signaling Tech-nology). Mice were injected i.p. with BrdUrd (1 mg/mL) 2 hoursbefore sacrifice for proliferation assessment and paraffin sectionswere stained using the BrdU in situ Detection Kit (BD Biosciences)according to the manufacturer's instructions. PCNA was assessedby the PCNA antibody (1:50); more details are available inSupplementary Material and Methods.

Immunofluorescent assayFor the visualization of macrophages, immunofluorescence

analysis with F4/80 specific monoclonal antibody (Santa CruzBiotechnology) was used. The slides were dewaxed, hydrated,and washed. After microwave antigen retrieval, slides wereblocked and then incubated with the antibody against F4/80(1:50) overnight at 4�C, and Cy3-labeled anti-rat IgG (BeyotimeBiotechnology; 1:500) was used as a secondary antibody. Final-ly, the slides were washed and mounted with DAPI (DojindoLaboratories; 1:1,000) for 5 minutes. Images were capturedusing a Leica DMIRB Fluorescence Microscope (OLYMPUSIX71).

Confocal microscopyAfter culturing for 5 days in 24-well plates, macrophages were

treated with cellular debris released by necrotic hepatocytes forindicated times, and then fixed with 4% paraformaldehyde.OPN and MyD88 were detected with mouse anti-OPN (23C3;1:100) and rabbit anti-MyD88 polyclonal antibody (Santa CruzBiotechnology; 1:50), respectively, followed by secondary Alex-aFluor 488–conjugated anti-mouse IgG (OPN; Life Technolo-gies; 1:250) or AlexaFluor 555–conjugated anti-rabbit IgG(MyD88; Life Technologies; 1:250). Nuclei were stained withDAPI. Stained cells were viewed with a confocal microscope(Zeiss LSM 510).

Antibody arrayAccording to the manufacturer's instructions, serum of tumor-

bearing mice was obtained and incubated with the mouse angio-genesis antibody array membranes (Panomics) for 2 hours atroom temperature. Then serum was discarded and the biotin-labeled detection antibodies mixture was added and incubatedwith membranes for 2 hours at room temperature. Streptavidin–horseradish peroxidase was next added and then incubated for 1hour at room temperature. Chemiluminescence substrated andfilms (Kodak) were used for detection of spots. Films werescanned and spots were quantitated usingQuantityOne software.

Statistical analysisData expressed are means � SE. Differences were analyzed by

the Student t test, and P values < 0.05 were considered assignificant. For the overall survival analysis, the log-rank testwas used in assessing the significance seen in the Kaplan–Meiercurve.

ResultsOpn ablation greatly enhances chemically inducedhepatocarcinogenesis in mice

Upon DEN injection on postnatal day 14, all wild-type (WT)and Opn knockout (Opn�/�) males developed HCCs within 9months (Fig. 1A). Strikingly, we observed a significant increase intumor numbers inOpn�/�mice, about three times comparedwiththat in the WT counterparts (Fig. 1B). The percentages of tumoroccupied areawere approximately 5%and40% inWTandOpn�/�

mice, respectively (Fig. 1C). The maximal tumor diameters werealso notably larger in Opn�/� mice compared with WT controls,17.5� 2.5 mm vs. 6.4 � 2 mm (Fig. 1D). To analyze the survivaldifference between WT and Opn�/�mice, another group of micewas sacrificed at 16 months after DEN injection. Kaplan–Meiersurvival curves (Fig. 1E) clearly showed that Opn�/� mice had asignificantly shorter survival time.

To further determine whether the enhanced hepatocarcino-genesis was due to the alteration of host microenvironment,Hepa1-6, a mouse hepatoma cell line derived from C57BL/6mice, was injected subcutaneously in DEN-pretreated Opn�/�

and WT mice. Hepa1-6 cells were transfected with siControl orsiOpn and extracted for detecting OPN expression (Supplemen-tary Fig. S1A). All mice were sacrificed 14 days postinoculation;gross appearance, tumor weight, and tumor volume weremonitored (Supplementary Fig. S1B). Tumor tissues werelysed and then analyzed for expression of OPN and GAPDH(as internal control) protein (Supplementary Fig. S1C). Inter-estingly, Hepa1-6-xenografted tumors were much bigger in

Fan et al.

Cancer Res; 75(1) January 1, 2015 Cancer Research88

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 4: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

Opn�/� mice than those in the WT mice. Moreover, when Opnwas silenced in Hepa1-6 cells to eliminate endogenous tumor-derived OPN, the xenografted-tumors were still bigger inOpn�/� mice than those in WT mice. Analysis of macrophageinfiltration by F4/80 staining revealed less infiltration in siOpn-transfected xenografts than that in siControl-transfected xeno-grafts, but not significantly. And no difference was observed inmacrophage infiltration between the xenografts formed fromthe same cells in WT andOpn�/�mice (Supplementary Fig. S1Dand S1E). These findings suggest that host-derived OPN mighthave a negative effect on the inflammatory microenvironmentin DEN-induced hepatocarcinogenesis.

Deletion of Opn exhibits enhanced cell turnover and survivalsignaling

Besides a high incidence of HCC, liver tumors in Opn�/� micedisplayed strongly elevated apoptotic (TUNEL; Fig. 2A andC) andproliferating (BrdUrd and PCNA) tumor cells compared withthose in WT mice (Fig. 2B and C), indicating enhanced cellturnover inOpn�/� tumors. Consistently, malignant liver tumorsin Opn�/� mice displayed strongly increased levels of cyclin D1and c-Myc, which are needed for cell proliferation, comparedwithWT controls (Fig. 2D).

We next used the Inflammation Multi-Target Sandwich ELISAKit to examine several key regulatory proteins in signaling path-ways controlling the stress and inflammation response, includingphospho-NF-kB p65, phospho-p38 mitogen-activated proteinkinase (MAPK), phospho-signal transducer and activator of tran-scription 3 (STAT3), phospho-stress-activated protein kinase/c-jun N-terminal kinase (SAPK/JNK) and phospho-inhibitor-kBa(IkB-a; Fig. 2E). Tumor tissues of livers were prepared. Opn�/�

mice exhibited a larger degree of increase in phospho-NF-kB p65,in which activation correlates with proliferation, apoptosis, andinflammation (26). In Opn�/� tumors, there were also obviouselevations inphospho-p38 andphospho-STAT3 levels, aswell as aslight increase in phospho-SAPK/JNK. p38 MAPK and SAPK/JNKare activated by a variety of cellular stresses, including inflamma-tory cytokines, lipopolysaccharides (LPS), and growth factors (27,28). STAT3 is activated in response to various cytokines andgrowth factors and mediates the expression of a variety of genescontrolling cell growth and apoptosis (29). Phospho-IkB-a, aninhibitory protein of NF-kB, was too low to be detected (data notshown). Elevated activation of these proliferation and inflamma-tion-related key regulators indicated that enhanced proliferationof hepatocytes and proinflammatory response could be inducedby Opn deficiency.

Figure 1.Opn ablation promotes DEN-inducedhepatocarcinogenesis in mice. A,gross and histologic appearance (H&Estaining) of 9-month-old DEN-treatedmaleWT andOpn�/�mice. Scale bars,50 mm. B, tumor numbers; C, tumorincidence; and D, maximum tumorsizes (diameters) in livers of male WT(n ¼ 14) and Opn�/� (n ¼ 14) mice 9months after DEN injection. Data,mean � SD; �� , P < 0.01. E, Kaplan–Meier survival graph of 16-month-oldDEN-treated male WT (n ¼ 10) andOpn�/� (n ¼ 10) mice. Survival of theOpn�/� mice was significantly lowerthan the other group. Log-rank testP < 0.001.

iOPN Inhibits Hepatocarcinogenesis in Mice

www.aacrjournals.org Cancer Res; 75(1) January 1, 2015 89

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 5: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

Opn deficiency aggravates liver cell death and compensatoryproliferation after DEN treatment

Because the deficiency of Opn could increase the susceptibilityof mice to DEN-induced hepatocarcinogenesis, which might berelated to enhanced cell turnover, we therefore examined theshort-term response elicited by DEN in vivo. Opn�/� mice exhib-ited higher ALT level in serum, indicative of liver injury, than thatin WT mice 24 and 48 hours after DEN injection (Fig. 3A). Therewas also an elevated number of TUNEL-positive cells in Opn�/�

mice compared with WT mice (Fig. 3B and D). These resultsindicated more hepatocyte death in Opn�/� mice. Liver hasregenerative capacity, and cell death might lead to compensatoryproliferation of surviving hepatocytes.Differences in proliferationat 24 and 48 hours after DEN injection matched the degree ofinjury (Fig. 3C and D). The levels of phosphorylated MAPKs,including ERK and p38, were slightly increased in liver tissuesfrom Opn-deficient mice, although phosphorylation of JNK hadno significant change (Fig. 3E), confirming stronger proliferativesignaling pathways in livers from Opn�/� mice.

To better understand how the absence ofOpn promoted tumorpromotion, we further examined phosphorylation of NF-kB p65in liver tissues, which is known to be a link of inflammation andcancer (26). Consistentwith the observation in tumor tissues (Fig.2E), absence ofOpn exhibited a great increase in phospho-NF-kBp65 relative to WT mice (Fig. 3F). Therefore, during the processof DEN-induced HCC, higher susceptibility to chemical hepato-

carcinogenesis inOpn�/�micemight be due to both enhanced cellapoptosis and compensatory proliferation ofDEN-initiated hepa-tocytes, and enhanced NF-kB activation might be an importantpromoter under the circumstance of Opn deficiency.

Opn�/� mice exhibit elevated expression of proinflammatorycytokines

NF-kB–dependent production of proinflammatory cytokineshas been demonstrated to be able to promote DEN-inducedhepatocarcinogenesis through compensatory proliferation (30).Therefore, we examined expression of NF-kB–targeted proinflam-matory cytokines such as interleukin-6 (IL6), tumor necrosisfactor-a (TNFa), and interleukin-1b (IL1b) after DEN adminis-tration. Significantly higher mRNA levels of IL6 and TNFa couldbe detected at 4 hours after DEN treatment in livers from Opn�/�

mice than those fromWT mice (Fig. 4A). Tumor tissues fromWTandOpn�/�micewere then separated. ThemRNA levels of IL6 andTNFa in tumors from Opn�/� mice were also significantly higherthan those fromWTmice (Fig. 4B). Next, we tested the change oftumor microenvironment through detecting proinflammatorycytokines in circulating serum of tumor-bearingmice by antibodyarrays. Besides IL6 and TNFa, other NF-kB–targeted genes likeIL1b, IL12, granulocyte colony stimulating factor (G-CSF), andtissue inhibitor of metalloproteinase-1 (TIMP-1) were also sig-nificantly increased in Opn�/� tumor-bearing mice comparedwith WT mice (Fig. 4C and D). All the results revealed that

Figure 2.Liver tumors in Opn�/� mice exhibitenhanced cell turnover and survivalsignaling. A, in WT and Opn�/�

HCC-bearing mice, Opn deficiencypromotes cell apoptosis in livers asdetermined by in situ TUNELapoptosis analysis. Slides werecounterstained with DAPI. Scale bars,100 mm. The percentage of apoptoticcells was calculated by countinggreen-stained nuclei (the apoptoticnuclei) versus blue-stained nuclei (thetotal nuclei) from six randomly chosenfields in each section. B, BrdUrd (top)and PCNA (bottom) assays of liversections to assess proliferative cellsfrom WT and Opn�/� HCC-bearingmice. Scale bars, 50mm.C, frequenciesof apoptotic cells and proliferatingcells. Data, mean � SD; � , P < 0.05. D,mRNA levels of cyclin D1 and c-Myc inthe livers of WT and Opn�/� HCC-bearing mice (n ¼ 4 mice per group).Data, mean� SD; � , P < 0.05. E, tumortissues from WT and Opn�/� mice(n ¼ 4 per group) were lysed andquantitated as 1 mg/mL, and thendetection of p-NF-kB P65, p-p38MAPK, p-STAT3, and p-SAPK/JNK bythe PathScan Inflammation Multi-Target Sandwich ELISA kit. Data,means � SD; � , P < 0.05.

Fan et al.

Cancer Res; 75(1) January 1, 2015 Cancer Research90

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 6: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

Opn�/� mice suffered from a more severe inflammatory responsethanWTmice, whichmight favor the survival and proliferation ofhepatocytes.

Intracellular OPN acts as a negative regulator for inflammatoryresponse in macrophages

Because OPN is expressed in a range of immune cells andreported to act as an immune modulator through its chemotacticproperties (6), we first detected infiltration ofmacrophages by F4/80 staining inWT andOpn�/�mice. In both tumor and nontumor

tissues, there were no obvious differences of macrophage infil-tration between WT and Opn�/� mice (Fig. 5A and B). Wetherefore investigated whether Opn deficiency caused enhancedinflammation response in macrophages. Activation of NF-kB inmacrophages is a critical event during progress of tumorigenesis,which links inflammation response to cancer (26). Stimulated byLPS (10 ng/mL) or cellular debris from Opn�/� necrotic hepato-cytes, which could exclude the disturbance of exogenous OPNprotein, macrophages from Opn�/� mice exhibited a higherexpression of phospho-NF-kB p65 (Fig. 5C), as well as IL6 and

Figure 3.Opn deficiency aggravates liver celldeath and compensatory proliferationafter DEN treatment. A, ALT levels inserum were determined at theindicated times after DEN injection(n ¼ 4 mice per time point). Data,mean � SD; � , P < 0.05. B, TUNELassay of liver sections from WT andOpn�/� mice at 24 and 48 hours afterDEN administration. Scale bars, 50mm. C, representative images ofimmunohistochemistry staining forincorporating BrdUrd in the livers ofDEN-treated WT and Opn�/� mice at24 and 48 hours. Scale bars, 50 mm.D, frequencies of apoptotic cells (left)andBrdUrd-positive proliferating cells(right) after DEN injection,respectively. Data, mean � SD;� , P < 0.05. E and F, the livers ofDEN-treated WT and Opn�/� mice(n ¼ 4 mice per time point) wereremoved and lysed to assessactivation of phosphoproteins byWestern blotting, includingp-ERK, total ERK, p-p38, totalp38, p-JNK, total JNK, andp-NF-kB p65; GAPDH was used asan internal control for analysis in F.

iOPN Inhibits Hepatocarcinogenesis in Mice

www.aacrjournals.org Cancer Res; 75(1) January 1, 2015 91

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 7: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

TNFa thanmacrophages fromWTmice did (Fig. 5D). In additionto activation of NF-kB, TLR/MyD88 signal pathway can activatemembers of MAPKs (23). Macrophages fromWT orOpn�/� micewere treated with cellular debris (NEC) for the indicated times.Then the cells were lysed and assessed for activation of MAPKs,including ERK, p38, and JNK. The levels of phosphorylatedMAPKs showed no obvious difference between macrophagesfrom Opn�/� mice and WT mice (Supplementary Fig. S2). Thus,Opn deficiency might enhance production of proinflammationcytokines through regulating NF-kB activity in macrophages.

Alternative splicing of OPN results in three isoforms, OPN-a,OPN-b, and OPN-c (11), and alternative translation of OPNgenerates two isoforms, sOPN and iOPN (12). Our results havesuggested that Opn deficiency causes enhanced inflammationresponse in macrophages, which is contradictory to the effectinduced by sOPN (6). We therefore investigated whether iOPN iscritical for Opn deficiency-induced inflammation response. First,cellular debris fromOpn�/�necrotic hepatocyteswas added toWTmacrophages and cell supernatant was collected at the indicatedtimes. The change trends of sOPN in debris-inducedmacrophageswere the same as those in controls (Fig. 6A). Macrophages werethen lysed, and expressions of iOPN, around 55 kDa, were greatlyincreased, whereas sOPN, around 60 kDa, were not significantlychanged (Fig. 6B). In addition, macrophages fromWT or Opn�/�

mice were treated with cellular debris and different amounts ofanti-OPN antibody or recombinant OPN protein, respectively, atthe same time for 4 hours. RT-PCR showed that extracellular OPNhad little impact on cytokine production in macrophages (Sup-plementary Fig. S3). Whereas, accompanied with elevated expres-

sion of iOPN, levels of IL6 and TNFa in cell supernatant weredecreased (Fig. 6C and 6D). iOpn or sOpn coding sequences werethen introduced intoOpn�/�macrophages. iOpn could efficientlyinhibit IL6 expression both in cell supernatant and mRNA level,whereas sOpn had no obvious effect (Fig. 6E and F). Thus, iOPNmay act as a negative regulator for inflammation response inmacrophages.

iOPN interacts with MyD88 to block IRAK1 dissociationTLR can activate NF-kB to increase proinflammatory cytokine

expression via adaptor molecule MyD88 and plays an importantrole in innate immune responses (23). We therefore investigatedwhether iOPN inhibited NF-kB activation and cytokine produc-tion by affecting the TLR/MyD88 signaling pathway. Coimmu-noprecipitation of iOPN and MyD88 in macrophages demon-strated their interaction under stimulation of cellular debris (Fig.7A). Confirmed by confocal microscopy, in WT macrophages,these two molecules showed few colocalization under no stim-ulation. But the circumstance changed when stimulated by cel-lular debris for 2 hours: colocalization was observed at peri-nuclear regions and when stimulated for 4 hours, they were bothin the nucleus (Fig. 7B). IL1 receptor-associated kinase 1 (IRAK1)is involved in TLR signaling; phosphorylation of IRAK1 by IRAK4can result in the activation of IRAK1 kinase activity and thus theformation of hyperphosphorylated and then phosphorylatedIRAK1 is released from MyD88 and the receptor complex toactivate downstream signaling (31). We detected the dissociationof IRAK1 from MyD88 complex by coimmunoprecipitation. Inendogenous level, after stimulated by cellular debris for 4 hours,

Figure 4.Expression of proinflammatorycytokines is elevated in Opn-deficientmice. A, livers ofWT andOpn�/�mice(n ¼ 4 mice per time point) wereremoved at the indicated hours afterDEN injection. B, tumor tissues fromWT orOpn�/� HCC-bearingmice (n¼4 mice per group) were removed.Expression of IL6, TNFa, and IL1bmRNA was determined by RT-PCR.Expression levels were normalized forb-actin. Data, mean � SD; � , P < 0.05.C, serum from WT and Opn�/� HCC-bearing mice (n ¼ 4 mice per group)was incubated with the mouseangiogenesis antibody arraymembranes. The serum levels ofcytokines were shown on films; WT,top left, Opn�/�, top right. Cytokines,represented by the spots, are listedin the lower table. D, the spots in Cwere scanned for densitometricanalyses and quantitated usingQuantity One software. Data,mean � SD; � , P < 0.05.

Fan et al.

Cancer Res; 75(1) January 1, 2015 Cancer Research92

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 8: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

association between MyD88 and IRAK1 was greatly reduced inOpn�/� macrophages (Fig. 7C). To further confirm the endoge-nous data in macrophages, HEK 293T cells were cotransfectedwith MyD88 and IRAK1, with or without iOpn. IRAK1 was notable to dissociate fromMyD88whenMyD88was bound to iOPN(Fig. 7D). These observations indicated that under cellular debrisstimulation, iOPN could inhibit activation of the TLR/MyD88signaling through interaction with MyD88 and blocking thedissociation of IRAK1 from the MyD88–IRAK1 complex.

DiscussionPrevious studies have suggested that OPN might play different

roles in tumorigenesis when diverse carcinogens were used (32–34). In N-methyl-N-nitro-N-nitrosoguanidine–induced cutane-ous squamous cell carcinoma,Opn-nullmicehad increased tumorgrowth, progression, and metastasis. Because host-derived OPNacted as a macrophage chemoattractant and at the tumor site, itcould recruit or maintain macrophages, the degree of whichcorrelated inversely with tumor growth in this model (32). Inthe 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphor-bol-13-acetate–induced skin papilloma model, Opn-null miceexhibited a decreased tumor/papilloma incidence by preventionof apoptosis (34). In MMTV-c-myc/MMTV-v-Ha-ras transgenicmice, OPN was not required for mammary primary tumor for-mation (33). In the DEN-induced HCC model, the role ofOPN was largely unknown. Our previous study found that OPNwas related to estrogen-mediated hepatoprotection in DEN-induced liver injury by enhancement of hepatocyte survival andinhibition of DEN biotransformation (35). Here, in accordancewith liver protection at an early stage, we found OPN had aprotective effect during hepatocarcinogenesis. Most chemicalcarcinogens act through interacting with the genetic material ofthe cell, especially with the DNA template (36), but there is

evidence that DEN-induced HCC depends on inflammationresponse (24, 30). Up to 16 months of age, no Opn�/� miceexhibited spontaneous HCC. However, Opn�/� mice exhibitedheavier production of proinflammatory cytokines and tumori-genesis than WT controls when DEN was administrated. Thesefindings indicate thatOpn deficiency may promote DEN-inducedhepatocarcinogenesis through a robust inflammatory response.

Macrophages, which are pivotal members within the solidtumor microenvironment, play a significant role in tumor initi-ation when inflammation is a causal factor (37). OPN is reportedto regulate function of macrophages and lead to cytokine pro-duction (13), phagocytosis and clearance of fungus (21), orbacterial infections (38). During these processes, sOPN and iOPNplay different roles in adaptive and innate immunity, respectively(39). sOPN is considered as amacrophage chemoattractant and isexpressed in cancer-infiltrating macrophages (9). iOPN is consti-tutively expressed bymacrophages and participates in chemotaxis(40). Tumor-associated macrophages (TAM) provide an inflam-mation microenvironment for tumor progression. Previous evi-dence has shown thatOPN is oneof themost upregulated genes inTAM, which may contribute to migration of macrophages (41).Data from exogenous OPN support that extracellular OPN is ableto promote macrophage migration. However, the role of endog-enous OPN in macrophage migration using Opn-deficient miceremains inconsistent. Macrophages are recruited into the perito-neum after intraperitoneal thioglycollate injection. However, thenumber of recruited macrophages is either fewer (42) or greater(43) inOpn-deficient mice as compared withWTmice. Moreover,in transgenic mice expressing c-myc and v-Ha-ras specifically inthe mammary gland, a model of spontaneous tumor develop-ment, expression of OPN was greatly enhanced in these tumors.However, when the transgenic mice were crossed with Opn�/�

mice, macrophage accumulation was found to be independent ofOPN status (33). Here, we found there were no differences

Figure 5.Opn�/�macrophages show increasedsensitivity to cell death and endotoxinstimulation and produce moreproinflammatory cytokines. A and B,F4/80 staining in liver sections of WTand Opn�/� HCC-bearing mice; theexpression of F4/80 in mouse liverswas detected by rat anti-mouse F4/80 antibody and Cy3-labeled anti-ratIgG as primary and secondaryantibody, respectively. Nuclei werevisualized by DAPI staining. Scalebars, 50 mm. C and D, macrophageswere isolated from WT and Opn�/�

mice (n ¼ 3 mice per group). Afterculturing for 5 days, cells werestimulated with cellular debrisreleased by necrotic hepatocytes(Nec) or LPS for 4 hours, and thenlysed for Western blotting, includingp-NF-kB p65 and GAPDH, or for RT-PCR. Data, mean � SD; � , P < 0.05.

iOPN Inhibits Hepatocarcinogenesis in Mice

www.aacrjournals.org Cancer Res; 75(1) January 1, 2015 93

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 9: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

between numbers of infiltrated macrophages in intratumoral ormarginal tissues of WT and Opn�/� mice in DEN-induced hepa-tocarcinogenesis. Our data and previous reports suggest thatregulation of macrophage migration may be complex when bothtumors and hosts are Opn deficient.

Here, OPN might modulate function instead of accumulationof macrophages in DEN-induced HCC. Indeed, Opn�/� macro-phages exhibited more production of proinflammatory cytokinessuch as IL6 and TNFa. Our results further demonstrated thatiOPN, rather than sOPN, acted as a negative regulator for inflam-mation response in macrophages. When WT macrophages werestimulated by cellular debris released by necrotic hepatocytes,protein level of iOPN was elevated, accompanied by decreasedrelease of IL6 and TNFa. Park and colleagues (44) have suggestedthat both TNFa and IL6 contribute to HCC development inmice.As the major proinflammatory cytokines in the microenviron-ment, IL6 and TNFa might promote survival and compensatoryproliferation of hepatocytes through activation of NF-kB,MAPKs,and IL6/STAT3 signaling (45). Indeed, in addition to enhancedactivation of NF-kB, activation of p38 MAPK and STAT3 was

detected in tumors developed from Opn�/� mice compared withthat from WT mice (Fig. 2E). Enhanced proliferation of hepato-cytes was also observed in tumors fromOpn�/�mice (Fig. 2B–D).However, the cooperation of NF-kB, p38 MAPK, and STAT3signaling in the regulation of tumor progression is not clear,which needs further investigation.

Kupffer cells, as resident liver macrophages, express most TLRs(46). TLR/MyD88 signaling has a strong contribution to inflam-mation and hepatocarcinogenesis. After incubation with eitherLPS or cellular debris released by necrotic hepatocytes, IL6 pro-duction in Kupffer cells from WT mice was increased, but inKupffer cells fromMyD88�/�mice, its production was elevated ata relatively low level (24). TLR4 is the major receptor recognizingendogenous ligands released from damaged or dying cells; itparticipates in cytokine expression mainly in myeloid cells andpromotes liver tumorigenesis (47). Mice deficient in MyD88 orTLR4 had a significant reduction in tumor incidence in DEN-induced liver cancer (24, 48). These findings suggest that in DEN-induced HCC, production of cytokines by resident liver macro-phages is mainly via the TLR4/MyD88 signaling.

Figure 6.iOPN suppresses proinflammatorycytokine production. WTmacrophages were stimulated withnecrotic debris at the indicated times(n¼ 4mice per time point). A, the cellsupernatant was collected and theexpression level of sOPN wasquantitated by ELISA. B, the cellswerelysed to assess the expression of sOPN(60 kDa) and iOPN (55 kDa) byWestern blotting, using OPN mAb(AKm2A1), with GAPDH serving as aloading control. C and D, supernatantof A was also collected for detectingthe level of IL6 and TNFa by ELISA. Eand F, macrophages were transfectedwith different plasmids for 48 hours,and then stimulated with necroticdebris for 4 hours; the expression ofIL6 was quantitated by ELISA and RT-PCR. Data, mean � SD; � , P < 0.05.

Fan et al.

Cancer Res; 75(1) January 1, 2015 Cancer Research94

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 10: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

Previous studies have suggested that iOPN might perform asone of the intracellular regulators of TLR2/TLR9 signaling tobalance the cytokine milieu (16, 21). In plasmacytoid dendriticcells, shortly after TLR9 engagement, iOPN interacted withMyD88 and enhanced interferon-a production (16). In antifun-gal innate immunity, iOPN acted as an adaptor molecule in TLR2and dectin-1 signaling pathways and increased zymosan-inducedcytokine production such as IL1b and IL10 (21). Following TLRstimulation inmacrophages, iOPNwas induced in vivo and in vitroand negatively regulated interferon-b production in murinemacrophages (17).Here,macrophageswere incubatedwith eitherLPS or cellular debris released by necrotic hepatocytes; by per-forming coimmunoprecipitation analysis and confocal micros-copy, we demonstrated that iOPN was able to interact withMyD88 and block dissociation of IRAK1 from MyD88 complex,which led to the activation of NF-kB and enhanced production ofcytokines such as IL6 and TNFa. In addition to activation of NF-kB, the TLR/MyD88 signal pathway can activate members ofMAPKs (23). However, levels of phosphorylated MAPKs had noobvious difference between macrophages from Opn�/� mice andWTmice. The mechanism by which iOPN regulates TLR/MyD88-

mediatedNF-kB activation selectively needs further investigation.Several molecules, such as MyD88s (the short form of MyD88)and IRAKM, have been found to be able to negatively regulateTLR-mediated immune responses (49). Our results suggest thatiOPN acts as a negative regulator of TLR4/MyD88 signaling inmacrophages and plays an important role in inflammation-relat-ed hepatocarcinogenesis.

OPN has long been considered as a prosurvival factor of tumorcells via inhibitionof apoptosis and inflammation regulation (50).Here, combining our observations and previous publications, wepropose that in the DEN-induced hepatocarcinogenesis murinemodel, iOPN is able to interactwithMyD88 inmacrophages understimulationof cellular debris, and thusblockdissociationof IRAK1from the MyD88 complex and subsequent NF-kB activation. Lossof OPN causes dissociation of IRAK1 from the MyD88 complex,activation of NF-kB, enhanced production of IL6 and TNFa inmacrophages, which promote survival and proliferation of pre-malignant hepatocytes and leads to tumorigenesis eventually. Ourfindings demonstrate a new mechanism by which iOPN plays acritical role in the anti-inflammation response inmacrophages anda novel insight into hepatocarcinogenesis.

Figure 7.iOPN interacts with MyD88 to blockIRAK dissociation. A, macrophagesfrom WT mice were stimulated withor without necrotic debris for4 hours, and the endogenousassociation of MyD88 and OPN wascoimmunoprecipitated and detectedwith the indicated antibodies; theexpression levels of MyD88 and OPNin lysates were examined by Westernblotting using anti-MyD88 and anti-OPN antibodies, respectively. B,confocal microscopy of OPN (green)and MyD88 (red) in macrophages.After treatment with necrotic debrisfor the indicated hours, cells werefixed. OPN and MyD88 were detectedwith mouse anti-OPN, rabbit anti-MyD88, followed by secondaryantibodies. Scale bars, 5 mm. C,WT and Opn�/� macrophageswere stimulated with or withoutnecrotic debris for 4 hours. Thecell lysates were subjected tocoimmunoprecipitation and Westernblotting, using anti-IRAK1 and anti-MyD88 antibodies. D, HEK 293T cellswere cotransfected with MyD88-pcDNA3.0 and pReceiver-myc-IRAK1,with or without iOpn-pcDNA3.0. At48 hours posttransfection, cells wereincubated with necrotic debris for4 hours. The cell lysates weresubjected to coimmunoprecipitationand Western blotting, using anti-IRAK1, anti-MyD88, and anti-OPNantibodies.

iOPN Inhibits Hepatocarcinogenesis in Mice

www.aacrjournals.org Cancer Res; 75(1) January 1, 2015 95

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 11: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: J. ZhaoDevelopment of methodology: X. ZhouAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): X. Fan, C. He, W. JingWriting, review, and/or revision of the manuscript: X. Fan, J. ZhaoAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): R. Chen, L. Cao, M. Zhu, R. Jia, H. WangStudy supervision: Y. Guo

Grant SupportThis work is supported in part by grants from Ministry of Science and

Technology of China "973" and "863" programs (2010CB945600,2010CB833600, and 2011CB966200), National Nature Science Foundationof China, State Key Project for Infection Disease and New Drug Development,and Programs of Shanghai Subject Chief Scientists, Municipal Commission ofEducation and Municipal Commission of Science and Technology.

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received March 4, 2014; revised October 18, 2014; accepted October 21,2014; published OnlineFirst November 14, 2014.

References1. Bruix J, Boix L, Sala M, Llovet JM. Focus on hepatocellular carcinoma.

Cancer Cell 2004;5:215–9.2. Bosch FX, Ribes J, Diaz M, Cleries R. Primary liver cancer: worldwide

incidence and trends. Gastroenterology 2004;127:S5-S16.3. Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet 2003;

362:1907–17.4. Verna L,Whysner J,WilliamsGM.N-nitrosodiethylaminemechanistic data

and risk assessment: bioactivation, DNA-adduct formation, mutagenicity,and tumor initiation. Pharmacol Ther 1996;71:57–81.

5. Lee JS, Chu IS,MikaelyanA,CalvisiDF,Heo J, Reddy JK, et al. Applicationofcomparative functional genomics to identify best-fit mouse models tostudy human cancer. Nat Genet 2004;36:1306–11.

6. WangKX,DenhardtDT.Osteopontin: role in immune regulation and stressresponses. Cytokine Growth Factor Rev 2008;19:333–45.

7. Sahai A, Malladi P, Melin-Aldana H, Green RM, Whitington PF. Upregula-tion of osteopontin expression is involved in the development of nonal-coholic steatohepatitis in a dietary murine model. Am J Physiol Gastro-intest Liver Physiol 2004;287:G264–73.

8. Syn WK, Choi SS, Liaskou E, Karaca GF, Agboola KM, Oo YH, et al.Osteopontin is induced by hedgehog pathway activation and promotesfibrosis progression in nonalcoholic steatohepatitis. Hepatology 2011;53:106–15.

9. Kim J, Ki SS, Lee SD, Han CJ, Kim YC, Park SH, et al. Elevated plasmaosteopontin levels in patients with hepatocellular carcinoma. Am J Gastro-enterol 2006;101:2051–9.

10. Zhao J, Dong L, Lu B, Wu G, Xu D, Chen J, et al. Down-regulation ofosteopontin suppresses growth and metastasis of hepatocellular car-cinoma via induction of apoptosis. Gastroenterology 2008;135:956–68.

11. Saitoh Y, Kuratsu J, Takeshima H, Yamamoto S, Ushio Y. Expression ofosteopontin in human glioma. Its correlation with the malignancy. LabInvest 1995;72:55–63.

12. Shinohara ML, Kim HJ, Kim JH, Garcia VA, Cantor H. Alternative trans-lation of osteopontin generates intracellular and secreted isoforms thatmediate distinct biological activities in dendritic cells. Proc Natl Acad SciU S A 2008;105:7235–9.

13. Ramaiah SK, Rittling S. Pathophysiological role of osteopontin in hepaticinflammation, toxicity, and cancer. Toxicol Sci 2008;103:4–13.

14. Rittling SR, Chambers AF. Role of osteopontin in tumour progression.Br J Cancer 2004;90:1877–81.

15. Zohar R, Lee W, Arora P, Cheifetz S, McCulloch C, Sodek J. Single cellanalysis of intracellular osteopontin in osteogenic cultures of fetal ratcalvarial cells. J Cell Physiol 1997;170:88–100.

16. Shinohara ML, Lu L, Bu J, Werneck MB, Kobayashi KS, Glimcher LH, et al.Osteopontin expression is essential for interferon-alpha production byplasmacytoid dendritic cells. Nat Immunol 2006;7:498–506.

17. Zhao W, Wang L, Zhang L, Yuan C, Kuo PC, Gao C. Differentialexpression of intracellular and secreted osteopontin isoforms by murinemacrophages in response to toll-like receptor agonists. J Biol Chem2010;285:20452–61.

18. Wung JK, Perry G, Kowalski A, Harris PL, Bishop GM, Trivedi MA, et al.Increased expression of the remodeling- and tumorigenic-associated factorosteopontin in pyramidal neurons of the Alzheimer's disease brain. CurrAlzheimer Res 2007;4:67–72.

19. Junaid A,MoonMC,HardingGE, Zahradka P.Osteopontin localizes to thenucleus of 293 cells and associates with polo-like kinase-1. Am J PhysiolCell Physiol 2007;292:C919–26.

20. Zohar R, Suzuki N, Suzuki K, Arora P, Glogauer M, McCulloch CA, et al.Intracellular osteopontin is an integral component of the CD44-ERMcomplex involved in cell migration. J Cell Physiol 2000;184:118–30.

21. InoueM,Moriwaki Y, Arikawa T, Chen YH,Oh YJ, Oliver T, ShinoharaML.Cutting Edge: Critical Role of Intracellular Osteopontin in AntifungalInnate Immune Responses. J Immunol 2011;186:19–23.

22. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol 2004;4:499–511.

23. Kawai T, Akira S. TLR signaling. Cell Death Differ 2006;13:816–25.24. Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM, et al.

Gender disparity in liver cancer due to sex differences inMyD88-dependentIL-6 production. Science 2007;317:121–4.

25. Celada A, Gray PW, Rinderknecht E, Schreiber RD. Evidence for a gamma-interferon receptor that regulates macrophage tumoricidal activity. J ExpMed 1984;160:55–74.

26. Karin M. NF-kappaB as a critical link between inflammation and cancer.Cold Spring Harb Perspect Biol 2009;1:a000141.

27. Ip YT, Davis RJ. Signal transduction by the c-Jun N-terminal kinase (JNK)–from inflammation to development. Curr Opin Cell Biol 1998;10:205–19.

28. Schieven GL. The biology of p38 kinase: a central role in inflammation.Curr Top Med Chem 2005;5:921–8.

29. Hirano T, Ishihara K, Hibi M. Roles of STAT3 in mediating the cell growth,differentiation and survival signals relayed through the IL-6 family ofcytokine receptors. Oncogene 2000;19:2548–56.

30. Maeda S, KamataH, Luo JL, Leffert H, KarinM. IKKbeta couples hepatocytedeath to cytokine-driven compensatory proliferation that promotes chem-ical hepatocarcinogenesis. Cell 2005;121:977–90.

31. Gottipati S, RaoNL, Fung-LeungWP. IRAK1: a critical signalingmediator ofinnate immunity. Cell Signal 2008;20:269–76.

32. Crawford HC, Matrisian LM, Liaw L. Distinct roles of osteopontin in hostdefense activity and tumor survival during squamous cell carcinomaprogression in vivo. Cancer Res 1998;58:5206–15.

33. Feng F, Rittling SR. Mammary tumor development in MMTV-c-myc/MMTV-v-Ha-ras transgenic mice is unaffected by osteopontin deficiency.Breast Cancer Res Treat 2000;63:71–9.

34. Hsieh YH, JulianaMM,Hicks PH, FengG, Elmets C, Liaw L, et al. Papillomadevelopment is delayed in osteopontin-null mice: implicating an anti-apoptosis role for osteopontin. Cancer Res 2006;66:7119–27.

35. HeC, FanX,ChenR, Liang B,Cao L,GuoY, et al.Osteopontin is involved inestrogen-mediated protection against diethylnitrosamine-induced liverinjury in mice. Food Chem Toxicol 2012;50:2878–85.

36. Lawley PD. Carcinogenesis by alkylating agents. Chem Carcinog1976;1:325–484.

37. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progressionand metastasis. Cell 2010;141:39–51.

38. Yang H, Guo H, Fan K, Zhang B, Zhao L, Hou S, et al. Clearance ofPropionibacterium acnes by kupffer cells is regulated by osteopontinthrough modulating the expression of p47phox. Mol Immunol2011;48:2019–26.

39. Cantor H, Shinohara ML. Regulation of T-helper-cell lineage developmentby osteopontin: the inside story. Nat Rev Immunol 2009;9:137–41.

Cancer Res; 75(1) January 1, 2015 Cancer Research96

Fan et al.

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from

Page 12: Intracellular Osteopontin Inhibits Toll-like Receptor …...Molecular and Cellular Pathobiology Intracellular Osteopontin Inhibits Toll-like Receptor Signaling and Impedes Liver Carcinogenesis

40. Zhu B, Suzuki K, Goldberg HA, Rittling SR, Denhardt DT, McCulloch CA,et al. Osteopontin modulates CD44-dependent chemotaxis of peritonealmacrophages through G-protein-coupled receptors: evidence of a role foran intracellular form of osteopontin. J Cell Physiol 2004;198:155–67.

41. Rittling SR. Osteopontin in macrophage function. Expert Rev Mol Med2011;13:e15.

42. Bruemmer D, Collins AR, Noh G, Wang W, Territo M, Arias-Magallona S,et al. Angiotensin II-accelerated atherosclerosis and aneurysm formation isattenuated in osteopontin-deficientmice. J Clin Invest 2003;112:1318–31.

43. Nau GJ, Liaw L, Chupp GL, Berman JS, Hogan BL, Young RA. Attenuatedhost resistance againstMycobacteriumbovis BCG infection inmice lackingosteopontin. Infect Immun 1999;67:4223–30.

44. Park EJ, Lee JH, Yu GY, He G, Ali SR, Holzer RG, et al. Dietary and geneticobesity promote liver inflammation and tumorigenesis by enhancing IL-6and TNF expression. Cell 2010;140:197–208.

45. He G, Karin M. NF-kappaB and STAT3 - key players in liver inflammationand cancer. Cell Res 2011;21:159–68.

46. Schwabe RF, Seki E, Brenner DA. Toll-like receptor signaling in the liver.Gastroenterology 2006;130:1886–900.

47. Maeda S. NF-kappaB, JNK, and TLR signaling pathways in hepatocarcino-genesis. Gastroenterol Res Pract 2010;2010:367694.

48. Yu LX, Yan HX, Liu Q, Yang W, Wu HP, Dong W, et al. Endo-toxin accumulation prevents carcinogen-induced apoptosis andpromotes liver tumorigenesis in rodents. Hepatology 2010;52:1322–33.

49. Liew FY, Xu D, Brint EK, O'Neill LA. Negative regulation of toll-likereceptor-mediated immune responses. Nat Rev Immunol 2005;5:446–58.

50. Rangaswami H, Bulbule A, Kundu GC. Osteopontin: role in cell signalingand cancer progression. Trends Cell Biol 2006;16:79–87.

www.aacrjournals.org Cancer Res; 75(1) January 1, 2015 97

iOPN Inhibits Hepatocarcinogenesis in Mice

Cancer Research. by guest on August 28, 2020. Copyright 2014 American Association forhttps://bloodcancerdiscov.aacrjournals.orgDownloaded from