Down-Regulation of Osteopontin Suppresses Growth and Metastasis of Hepatocellular Carcinoma Via Induction of Apoptosis
Jian Zhao,Li Dong,Bin Lu,Guobin Wu,Dongmei Xu,Jingjing Chen,Kai Li,Xin Tong,Jianxin Dai,Side Yao,Mengchao Wu,Yajun Guo
DOI: https://doi.org/10.1053/j.gastro.2008.05.025
IF: 29.4
2008-01-01
Gastroenterology
Abstract:Background & Aims: Expression of osteopontin correlates with tumor progression and metastasis. The mechanisms by which osteopontin promotes tumor cell survival remain unclear. Here we used short-hairpin RNA–mediated gene silencing to investigate the antitumor effects by osteopontin depletion in hepatocellular carcinoma (HCC). Methods: We applied polyethylenimine nanoparticles to deliver a short-hairpin RNA for depletion of osteopontin expression in HCC cells. Tumorigenicity and metastatic potentials of HCC cells were studied in vitro and in nude mice. Nuclear factor-κB (NF-κB) activation was analyzed by gel shift assay and luciferase analysis. The expressions of integrins were examined by real-time reverse-transcription polymerase chain reaction. Apoptosis was examined by terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling assay and mitochondrial membrane potential analysis. Results: Down-regulation of osteopontin inhibited HCC cell growth, anchorage-independent growth, adhesion with fibronectin and invasion through extracellular matrix in vitro, and suppressed tumorigenicity and lung metastasis in nude mice. Osteopontin silencing resulted in suppression of αv, β1, and β3 integrin expressions, blockade of NF-κB activation, inhibition of Bcl-2/Bcl-xL and XIAP expressions, increase of Bax expression, and induction of a mitochondria-mediated apoptosis. Furthermore, down-regulation of osteopontin inhibited drug-induced NF-κB activation and sensitized HCC cells to chemotherapeutic agents in vitro, which led to complete regression of HCC xenografts in nude mice. Conclusions: Osteopontin may facilitate tumorigenesis and metastasis through prevention of tumor cells from apoptosis. RNA interference–mediated depletion of osteopontin may be a promising strategy for the treatment of HCC by sensitizing the chemotherapeutic drugs. Background & Aims: Expression of osteopontin correlates with tumor progression and metastasis. The mechanisms by which osteopontin promotes tumor cell survival remain unclear. Here we used short-hairpin RNA–mediated gene silencing to investigate the antitumor effects by osteopontin depletion in hepatocellular carcinoma (HCC). Methods: We applied polyethylenimine nanoparticles to deliver a short-hairpin RNA for depletion of osteopontin expression in HCC cells. Tumorigenicity and metastatic potentials of HCC cells were studied in vitro and in nude mice. Nuclear factor-κB (NF-κB) activation was analyzed by gel shift assay and luciferase analysis. The expressions of integrins were examined by real-time reverse-transcription polymerase chain reaction. Apoptosis was examined by terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling assay and mitochondrial membrane potential analysis. Results: Down-regulation of osteopontin inhibited HCC cell growth, anchorage-independent growth, adhesion with fibronectin and invasion through extracellular matrix in vitro, and suppressed tumorigenicity and lung metastasis in nude mice. Osteopontin silencing resulted in suppression of αv, β1, and β3 integrin expressions, blockade of NF-κB activation, inhibition of Bcl-2/Bcl-xL and XIAP expressions, increase of Bax expression, and induction of a mitochondria-mediated apoptosis. Furthermore, down-regulation of osteopontin inhibited drug-induced NF-κB activation and sensitized HCC cells to chemotherapeutic agents in vitro, which led to complete regression of HCC xenografts in nude mice. Conclusions: Osteopontin may facilitate tumorigenesis and metastasis through prevention of tumor cells from apoptosis. RNA interference–mediated depletion of osteopontin may be a promising strategy for the treatment of HCC by sensitizing the chemotherapeutic drugs. Recent evidence has shown that the expression of osteopontin (OPN) is increased significantly in hepatocellular carcinoma (HCC), which associates closely with poor prognosis, early recurrence, tumor aggressiveness, and metastasis.1Pan H.W. Ou Y.H. Peng S.Y. et al.Overexpression of osteopontin is associated with intrahepatic metastasis, early recurrence, and poorer prognosis of surgically resected hepatocellular carcinoma.Cancer. 2003; 98: 119-127Crossref PubMed Scopus (226) Google Scholar, 2Ye Q.H. Qin L.X. Forgues M. et al.Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning.Nat Med. 2003; 9: 416-423Crossref PubMed Scopus (751) Google Scholar OPN, an extracellular matrix protein, binds to αvβ integrins and CD44 family of receptors to propagate cellular signals on extracellular matrix (ECM) degradation, angiogenesis, and tumor cell apoptosis.3Rittling S.R. Chambers A.F. Role of osteopontin in tumor progression.Br J Cancer. 2004; 90: 1877-1881Crossref PubMed Scopus (371) Google Scholar, 4Weber G.F. The metastasis gene osteopontin: a candidate target for cancer therapy.Biochim Biophys Acta. 2001; 1552: 61-85Crossref PubMed Scopus (136) Google Scholar, 5Rangaswami H. Bulbule A. Kundu G.C. Osteopontin: role in cell signaling and cancer progression.Trends Cell Biol. 2006; 16: 79-87Abstract Full Text Full Text PDF PubMed Scopus (589) Google Scholar The mechanisms by which OPN promotes tumor cell survival are still unclear. Soluble OPN inhibits growth factors and cytokine depletion induced apoptosis in adherent endothelial cells, which involves enhanced Bcl-xL expression.6Khan S.A. Lopez-Chua C.A. Zhang J. et al.Soluble osteopontin inhibits apoptosis of adherent endothelial cells deprived of growth factors.J Cell Biochem. 2002; 85: 728-736Crossref PubMed Scopus (113) Google Scholar OPN contributes to antiapoptotic signaling of melanocytes in dermal collagen through interaction with integrin αvβ3.7Geissinger E. Weisser C. Fischer P. et al.Autocrine stimulation by osteopontin contributes to antiapoptotic signalling of melanocytes in dermal collagen.Cancer Res. 2002; 62: 4820-4828PubMed Google Scholar OPN-deficient cardiac fibroblasts undergo increased cell death in response to hydrogen peroxide through a caspase-3–independent pathway.8Zohar R. Zhu B. Liu P. et al.Increased cell death in osteopontin-deficient cardiac fibroblasts occurs by a caspase-3-independent pathway.Am J Physiol Heart Circ Physiol. 2004; 287: 1730-1739Crossref PubMed Scopus (54) Google Scholar OPN null mice show a significant delay in 7,12-dimethylbenzanthracene–induced papilloma development as a result of enhanced apoptotic effects.9Hsieh Y.H. Juliana M.M. Hicks P.H. et al.Papilloma development is delayed in osteopontin-null mice: implicating an antiapoptosis role for osteopontin.Cancer Res. 2006; 66: 7119-7127Crossref PubMed Scopus (56) Google Scholar OPN treatment confers colon cancer and gastric cancer cells an increased resistance to ultraviolet-induced apoptosis via integrin activation.10Lee J.L. Wang M.J. Sudhir P.R. et al.Osteopontin promotes integrin activation through outside-in and inside-out mechanisms: OPN-CD44V interaction enhances survival in gastrointestinal cancer cells.Cancer Res. 2007; 67: 2089-2097Crossref PubMed Scopus (124) Google Scholar Efforts have been made to inhibit tumor progression and metastasis by targeting OPN. 4Weber G.F. The metastasis gene osteopontin: a candidate target for cancer therapy.Biochim Biophys Acta. 2001; 1552: 61-85Crossref PubMed Scopus (136) Google Scholar Suppression of OPN by antisense oligonucleotides or RNA-mediated interference and inhibition of OPN protein by antibodies or synthetic peptides have been used as the tools for cancer therapy.4Weber G.F. The metastasis gene osteopontin: a candidate target for cancer therapy.Biochim Biophys Acta. 2001; 1552: 61-85Crossref PubMed Scopus (136) Google Scholar OPN-neutralizing antibody efficiently blocked invasion and metastasis of HCC cells both in vitro and in vivo.2Ye Q.H. Qin L.X. Forgues M. et al.Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning.Nat Med. 2003; 9: 416-423Crossref PubMed Scopus (751) Google Scholar Blockade of OPN expression using small interfering RNA effectively suppressed metastasis of CT26 murine colon adenocarcinoma and human esophageal squamous cell carcinoma.11Wai P.Y. Mi Z. Guo H. et al.Osteopontin silencing by small interfering RNA suppresses in vitro and in vivo CT26 murine colon adenocarcinoma metastasis.Carcinogenesis. 2005; 26: 741-751Crossref PubMed Scopus (92) Google Scholar, 12Ito T. Hashimoto Y. Tanaka E. et al.An inducible short-hairpin RNA vector against osteopontin reduces metastatic potential of human esophageal squamous cell carcinoma in vitro and in vivo.Clin Cancer Res. 2006; 12: 1308-1316Crossref PubMed Scopus (58) Google Scholar Polymeric nanoparticles for gene delivery have drawn much attention for its resistance to nuclease degradation and sustained duration of plasmid DNA administration.13Cohen H. Levy R.J. Gao J. et al.Sustained delivery and expression of DNA encapsulated in polymeric nanoparticles.Gene Ther. 2000; 7: 1896-1905Crossref PubMed Scopus (353) Google Scholar, 14Vasir J.K. Labhasetwar V. Polymeric nanoparticles for gene delivery.Expert Opin Drug Deliv. 2006; 3: 325-344Crossref PubMed Scopus (64) Google Scholar In this study, we applied spheric polyethylenimine nanoparticles (named M-PEI) of 80 nm prepared by photo-Fenton reaction in aqueous solution15Xu D.M. Yao S.D. Liu Y.B. et al.Size-dependent properties of M-PEIs nanogels for gene delivery in cancer cells.Int J Pharm. 2007; 338: 291-296Crossref PubMed Scopus (74) Google Scholar, 16Xu D.M. Hong J. Sheng K.L. et al.Preparation of polyethyleneimine nanogels via photo-Fenton reaction.Radiat Phys Chem. 2007; 76: 1606-1611Crossref Scopus (25) Google Scholar to deliver a short-hairpin RNA (shRNA) targeting osteopontin and investigated the therapeutic efficacy of this novel approach in the treatment of human HCC in a xenografted mouse model system. The sequence targeting human OPN for RNA interference was 5′ GGACAGTTATGAAACGAGT 3′. Sequence (5′ TTCTCCGAACGTGTCACGT 3′) targeting none of the known genes was used for negative control. Oligonucleotides of shRNA including a TTCAAGAGA loop motif were synthesized and inserted into pGCsi vector containing H1 promoter, T6 terminal sequences, and green fluorescent protein (GFP) reporter gene with a standard cloning procedure by GeneChem (Shanghai, China). Cells were grown in Dulbecco's modified Eagle medium supplemented with 10% fetal bovine serum in a humidified incubator at 37°C in a 5% CO2 atmosphere. Transfection of plasmids was performed using M-PEI in 6-well tissue culture plates as described previously.15Xu D.M. Yao S.D. Liu Y.B. et al.Size-dependent properties of M-PEIs nanogels for gene delivery in cancer cells.Int J Pharm. 2007; 338: 291-296Crossref PubMed Scopus (74) Google Scholar Briefly, 3.6 μg plasmid was dissolved in 100 μL 0.9% NaCl, 14.4 μg of M-PEI then was pipetted to plasmid. The plasmid/M-PEI mixture then was added to cells in serum-free Dulbecco's modified Eagle medium. IκBα kinase β mammalian–expressing plasmids were from GeneCopoeia (Guangzhou, China). For anchorage-independent growth assays, cells were seeded in 0.3% agarose over a 0.6% agarose bottom layer at a density of 500 cells per well in a 24-well plate. After 3 weeks, the numbers of colonies greater than 100 μm in diameter were counted. Invasion of cells through the ECM was determined by the Cell Invasion Assay Kit (Chemicon, Temecula, CA). The number of cells invading through the ECM was counted by 5 randomly selected visual fields, and the extent of invasion was expressed as the average number of cells per microscopic field at a magnification of 200. Ninety-six–well plates coated with 10 μg/mL fibronectin (Calbiochem, San Diego, CA) were used for adhesion assay as described previously.17Bisanz K. Yu J. Edlund M. et al.Targeting ECM-integrin interaction with liposome-encapsulated small interfering RNAs inhibits the growth of human prostate cancer in a bone xenograft imaging model.Mol Ther. 2005; 12: 634-643Abstract Full Text Full Text PDF PubMed Scopus (77) Google Scholar The fluorescein FragEL DNA Fragmentation Detection Kit (Calbiochem) was used for the terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay. The fluorescein of cells carrying DNA labeled with fluorescein isothiocyanate–deoxyuridine triphosphate (TUNEL-positive cells) was analyzed by Becton Dickinson (Franklin Lakes, NJ) FACscan. For in vivo apoptosis assay, the formalin-fixed paraffin sections were deparaffinized and incubated with TUNEL reaction mixture. Apoptotic cells were observed under a fluorescence microscope (Olympus, Tokyo, Japan). Cells were incubated with tetramethylrhodamine ethylester (TMRE) by the MitoShift Kit (Trevigen, Gaithersburg, MD) and observed under a fluorescence microscope. In healthy cells, the TMRE dye accumulates in the mitochondria and appears bright red by fluorescence microscopy, but in apoptotic cells the mitochondrial membrane potential collapses and the TMRE dye cannot accumulate within the mitochondria. Total RNA was isolated using the RNeasy mini kit (Qiagen, Hilden, Germany), and genomic DNA was removed using the RNase-Free DNase set (Promega, Madison, WI). First-strand cDNA was generated using the Reverse Transcription System kit (Promega). For quantification of integrin and CD44 messenger RNA expression, real-time polymerase chain reaction was performed as described previously.18Xu G. Nie H. Li N. et al.Role of osteopontin in amplification and perpetuation of rheumatoid synovitis.J Clin Invest. 2005; 115: 1060-1067Crossref PubMed Scopus (140) Google Scholar Cytosol and mitochondria fractionation was performed by the centrifugation technique as described previously.19Zhao J. Jin J. Zhang X. et al.Transfection of Smac sensitizes tumor cells to etoposide-induced apoptosis and eradicates established human hepatoma in vivo.Cancer Gene Ther. 2006; 13: 420-427Crossref PubMed Scopus (26) Google Scholar Nuclear extracts were made using NE-PER Nuclear and Cytoplasmic Extraction Reagents (Pierce Biotechnology, Rockford, IL). Total cell lysate was prepared in 1 × sodium dodecyl sulfate buffer. Proteins at the same amount were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis and transferred onto polyvinylidene difluoride membranes. After probing with individual antibodies, the antigen–antibody complex was visualized by the enhanced chemiluminescence reagent Supersignal (Pierce Biotechnology). The antibodies used were anti–caspase-3, anti–caspase-9, anti-X-linked inhibitor of apoptosis protein (XIAP), anti-poly (ADP-ribose) polymerase (PARP), anti–cytochrome c, and anti-OPN (R&D Systems, Minneapolis, MN), anti–Bcl-2, anti–Bcl-xL, and anti-Bax (Santa Cruz Biotechnology, Santa Cruz, CA), anti–p-Akt, anti–nuclear factor κB (NF-κB) p65, and anti-IκBα (Boster Biotechnology, Wuhan, China), anti-GFP and anti-human matrix matallopeptidase 2 (MMP2; Beyotime, Haimen, China). Cells were deprived of serum 24 hours after transfection. The conditioned media was collected and viable cell numbers were assessed by trypan-blue staining every 12 hours. OPN concentrations in conditioned media were measured using a quantitative immunoassay enzyme-linked immunosorbent assay kit (QuantiKine Assay; R&D Systems). OPN levels were normalized to viable cell numbers and calculated according to the following formula: OPN levels (ng/mL) = OPN concentrations in conditioned media (ng/mL) × (cell numbers in mock wells/cell numbers in shRNA-transfected wells). Triplicate samples were analyzed. Electrophoretic mobility shift assay was performed using the LightShift chemiluminescent electrophoretic mobility shift assay kit (Pierce Biotechnology). Double-stranded gel shift probes corresponding to the human consensus NF-κB sequences20Luan B. Zhang Z. Wu Y. et al.Beta-arrestin2 functions as a phosphorylation-regulated suppressor of UV-induced NF-kappaB activation.EMBO J. 2005; 24: 4237-4246Crossref PubMed Scopus (67) Google Scholar (5′-AGTTGAGGGGACTTTCCCAGGC-3′) were end-labeled with biotin. Cells were transfected with NF-κB–driven luciferase plasmid together with the pRL-TK, which contains the herpes simplex virus thymidine kinase (HSK-TK) promoter to provide low to moderate levels of Renilla luciferase expression, vector as a control for transfection efficiency using Lipofectamine 2000 (Invitrogen, Carlsbad, CA). The luciferase activities were measured using the Dual Luciferase Reporter Assay System (Promega). MMP-2 activity was detected by zymography as described previously.21Ellerbroek S.M. Fishman D.A. Kearns A.S. et al.Ovarian carcinoma regulation of matrix metalloproteinase-2 and membrane type 1 matrix metalloproteinase through beta1 integrin.Cancer Res. 1999; 59: 1635-1641PubMed Google Scholar Briefly, the conditioned medium and tumor lysates were collected and the equal amounts of protein were separated by 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis containing 0.1% gelatin without reducing agent. The gels were washed with 2.5% Triton X-100 and incubated in 20 mmol/L glycine, 10 mmol/L CaCl2, and 1 mmol/L ZnCl2 (pH 8.3) at 37°C for 24 hours to allow the gelatinases to digest the gelatin structure. The gels were stained with Coomassie Blue and destained to visualize the results. Six-week-old male BALB/c nude mice were purchased from the Shanghai Experimental Animal Center of Chinese Academy of Sciences (Shanghai, China). All mice were maintained under pathogen-free conditions in laminar flow boxes in accordance with established institutional guidance and approved protocols. Two days after transfection with shRNA plasmids, 5 × 106 HCC-LM3 cells were injected subcutaneously into the right flanks of nude mice. The tumor volume was calculated according to the following formula: V = length × width2 × 0.5. Animals were killed for histopathology examination at day 35 after cell inoculation. For in vivo treatment experiments, mice inoculated with 5 × 106 HCC-LM3 cells were divided randomly into 6 groups (6 mice/group) when tumor volume reached 100 mm3. Mice were treated daily for 5 days by intratumoral injection of 20 μg shRNA plasmids mixed with 80 μg M-PEI. One day after injection, the mice were dosed intraperitoneally with or without 5-fluorouracil (5-FU) at a concentration of 10 mg/kg body weight daily for 5 days. Tumor volume was monitored. Statistical analysis was performed using the Analysis ToolPack provided by Microsoft Excel (Beijing, China). A 2-sample Student t test, assuming unequal variances, was used to determine the equality of the means of 2 samples. Results were considered statistically significant at a P value of less than .05. About 50%–60% of cells were transfected with shRNA plasmids by M-PEI, as monitored by GFP expression in HCC cells (Figure 1A). Transfection with shRNA targeting OPN (shOPN), but not nonsilencing shRNA (shNon), reduced OPN expression by about 50% as detected by reverse-transcription polymerase chain reaction and Western blotting analyses compared with mock controls (Figure 1B and C). Expression of GFP and inhibition of OPN expression lasted up to 14 days posttransfection (Figure 1D), which might be owing to plasma DNA entrapped in PEI polymeric particles showing a sustained release pattern.13Cohen H. Levy R.J. Gao J. et al.Sustained delivery and expression of DNA encapsulated in polymeric nanoparticles.Gene Ther. 2000; 7: 1896-1905Crossref PubMed Scopus (353) Google Scholar, 15Xu D.M. Yao S.D. Liu Y.B. et al.Size-dependent properties of M-PEIs nanogels for gene delivery in cancer cells.Int J Pharm. 2007; 338: 291-296Crossref PubMed Scopus (74) Google Scholar The secreted OPN in the culture medium was reduced significantly in shOPN-transfected HCC-LM3 cells (Figure 1E). Thus, M-PEI–mediated shOPN transfection efficiently inhibits OPN expression in HCC cells. shOPN, but not shNon, significantly inhibited the growth and anchorage-independent growth of HCC cells (Figure 2A and B). Addition of recombinant human OPN generated from mouse myeloma cell NSO (rhOPN; R&D Systems) or HCC-LM3 conditioned media could not reverse the inhibitory effect of shOPN on cell growth (Figure 2A). A marked reduction of adhesion and invasion abilities was observed in shOPN-transfected cells, which could be reversed by addition of rhOPN (Figure 2C and D). Moreover, shOPN-transfected HCC-LM3 cells showed a drastic reduction of MMP-2 expression (Figure 2E). Thus, depletion of OPN has an inhibitory effect on the growth and invasion of HCC cells. Eight days after tumor cell inoculation, all mice injected with shNon-transfected or mock-treated HCC-LM3 cells developed tumors in nude mice. However, it took 13 to 28 days for shOPN-transfected cells to develop tumors (Figure 3A). Growth of shOPN-modified HCC-LM3 cells was suppressed significantly compared with that of shNon-transfected or mock-treated cells (Figure 3B). The tumor volumes and weights of shOPN-modified HCC-LM3 xenografts were only about 20% and 25% of that of mock-treated xenografts 35 days after implantation (Table 1). Furthermore, the incidence of lung metastasis dropped to zero in shOPN-modified HCC-LM3 xenografted mice, which is significantly different from that in the mock-treated group (83.3%) or the shNon-modified group (71.4%) (Figure 3C, Table 1). Inhibition of OPN expression was confirmed by detection of OPN levels in plasma (Figure 3D), as well as detection of OPN and GFP expression by Western blotting in tumor samples 35 days after implantation (Figure 3E). Inhibition of MMP2 expression by shOPN was observed in tumor lysates (Figure 3F). These data suggest that OPN-depletion leads to significant inhibition of tumorigenesis and metastasis of HCC in vivo.Table 1Osteopontin shRNA Inhibits Tumorigenesis and Metastasis of HCC-LM3 Cells in Nude MiceTumor sizeLung metastasisTreatment groupsVolume, mm3Weight, gCases (%)Metastasis, no.Mock2167 ± 4282.37 ± 0.235 of 6 (83.3%)5.2 ± 1.3shNon1971 ± 5002.12 ± 0.345 of 7 (71.4%)4.4 ± 2.3shOPN416 ± 314⁎P < .05, compared to mock treatment.0.605 ± 0.27⁎P < .05, compared to mock treatment.0 of 7 (0)⁎P < .05, compared to mock treatment.0⁎P < .05, compared to mock treatment.NOTE. Evaluation of lung metastasis in mice was performed on day 35 after subcutaneous implantation of HCC-LM3 cells. Tumor sizes are presented by both volume in cubic millimeters and weight in grams (mean ± SD). Lung metastasis is denoted as both the positive metastases in the lungs (the percentages are indicated in parentheses) and the average number of metastases (no.) in lungs per positive case (mean ± SD). P < .05, compared to mock treatment. Open table in a new tab NOTE. Evaluation of lung metastasis in mice was performed on day 35 after subcutaneous implantation of HCC-LM3 cells. Tumor sizes are presented by both volume in cubic millimeters and weight in grams (mean ± SD). Lung metastasis is denoted as both the positive metastases in the lungs (the percentages are indicated in parentheses) and the average number of metastases (no.) in lungs per positive case (mean ± SD). After transfection with shOPN, most HCC cells had shrunk and detached from the plate under serum starvation. In contrast, the shNon-transfected cells remained attached to the dish and showed normal morphology (Figure 4A). The cell death in the shOPN-treated group was significantly higher than that in the shNon or mock-treated groups, which was strongly enhanced by serum starvation in HCC cells (Figure 4B). Addition of rhOPN could reverse anti-OPN antibody-induced but not shOPN-induced cell death (Figure 4C).The percentage of TUNEL-positive apoptotic cells greatly increased by OPN depletion compared with shNon-transfection, which was greatly enhanced by serum starvation (Figure 4D). Another shOPN construct targeting 5'-GCATTCCGATGTGATTGAT-3' resulted in similar effects (Figure 4E). Addition of rhOPN could not reverse shOPN-induced apoptosis (Figure 4E). Apoptosis was examined further by in situ TUNEL assay on HCC-LM3 xenografted tumor tissues. The apoptotic nuclei, seen as green color excited under fluorescence microscope, were greatly increased in shOPN-transfected HCC-LM3 xenografts (Figure 4F). The percentage of apoptotic cells in the shOPN group was 2 or 3 times higher than that in the shNon or mock groups. Procaspase-3 and -9 were greatly decreased at 48 hours posttransfection with shOPN (Figure 5A). PARP, the substrate of caspase-3, was found cleaved with an 85-kilodalton fragment after shOPN transfection (Figure 5A). Furthermore, the broad-spectrum caspases inhibitor N-tert-butoxy-carbonyl-Val-Ala-Asp-fluoromethylketone significantly inhibited shOPN-induced cell death (Figure 5B). Thus, shOPN induced apoptosis is caspase-dependent in HCC cells. Transfection with shOPN resulted in a significant loss of mitochondrial membrane potential indicated by a substantial reduction of TMRE staining (Figure 5C), as compared with surrounding nontransfected cells. Down-regulation of OPN caused a dramatic accumulation of Bax and obvious elimination of Bcl-2 at mitochondria, as well as release of cyto-c from mitochondria to cytosol (Figure 5D). OPN silencing resulted in an obvious decrease of Bcl-xL, Bcl-2, and XIAP, and a dramatic increase of Bax expression, which could not be reversed by addition of rhOPN (Figure 5E). However, IκBα kinase β, which activates NF-κB through phosphorylation and degradation of IκBα, blocked shOPN-induced decrease of Bcl-2 and induction of Bax (Figure 5F), indicating that the alterations of Bcl-2 and Bax expression might be the result of inhibition of NF-κB activity by OPN depletion. Thus, OPN depletion might induce mitochondria-mediated apoptosis through regulation of the expression of Bcl-2 and IAPs family members. Expressions of integrin αv, β1, and β3, and CD44 were much higher in HCC-LM3 cells than HL7702 cells (Figure 6A). OPN depletion greatly decreased the expressions of αv, β1, and β3 integrin in HCC-LM3 cells (Figure 6A). Addition of rhOPN attenuated anti-OPN antibody–induced but not shOPN-induced αv, β3 integrin suppression (Figure 6A). However, the mechanism for down-regulation of the expressions of integrins by OPN depletion still need to be investigated further. OPN could activate both Akt and NF-κB activities on interaction with αv β1 and αvβ3 integrins to promote cell survival and motility.5Rangaswami H. Bulbule A. Kundu G.C. Osteopontin: role in cell signaling and cancer progression.Trends Cell Biol. 2006; 16: 79-87Abstract Full Text Full Text PDF PubMed Scopus (589) Google Scholar We first measured the activity of Akt by assessing phosphorylation of Akt after OPN depletion. Phosphorylation of Akt was not affected by OPN knockdown (Figure 6B). However, OPN depletion greatly inhibited the amount of p65 subunit of NF-κB and prevented the nuclear accumulation of p65 induced by serum starvation or 5-FU stimulation (Figure 6B). NF-κB transcriptional activity was greatly enhanced in HCC-LM3 cells compared with HL7702 cells (Figure 6C). Down-regulation of OPN significantly inhibited NF-κB activity and cisplatin-induced NF-κB activation in HCC-LM3 cells (Figure 6C). NF-κB DNA-binding activities induced by serum starvation or cisplatin and 5-FU treatment also were strongly attenuated by OPN depletion, which could not be reversed by addition of rhOPN (Figure 6D). Next, the expression of NF-κB inhibitor IκBα was determined by Western blotting. Treatment of cisplatin caused a decrease of IκBα within 30 minutes and then the amount of IκBα returned to basal levels after 60 minutes because of its NF-κB–dependent do novo synthesis. OPN depletion greatly enhanced the amount of IκBα and completely blocked cisplatin-induced turnover of IκBα (Figure 6E). These data suggest that depletion of OPN might block chemotherapy drug–induced NF-κB activation. Depletion of OPN significantly enhanced the sensitivities of HCC-LM3 cells to topoisomerase II inhibitors etoposide and Adriamycin (Merck, San Diego, CA), thymidylate synthase inhibitor 5-FU, as well as the DNA-reactive agent cisplatin, as compared with shNon or mock treatment (Figure 7A). TNF-related apoptosis-inducing ligand (TRAIL) induced less than 20% of cell death in HCC-LM3 and HepG2 cells at a dose as high as 100 ng/mL. Down-regulation of OPN dramatically increased TRAIL-mediated cell death (Figure 7B). Thus, down-regulation of OPN sensitizes HCC cells to both death receptor and DNA damage–induced cell death. Intratumoral administration of shOPN significantly inhibited growth of HCC-LM3 xenografts in nude mice compared with treatment with shNon or M-PEI (Figure 7C). Notably, intraperitoneal administration of 5-FU in combination with intratumoral administration of shOPN resulted in complete eradication of all 6 HCC-LM3 xenografts (Figure 7C). Thus, down-regulation of OPN might be a promising adjuvant for chemotherapy of HCC. Apoptosis plays a central role in tissue homeostasis during both development and adult life. Tumor cells often are deficient in apoptotic regulation, which leads to the expansion of tumor cells, metastasis, and resistance to chemotherapy.22Mehlen P. Puisieux A. Metastasis: a question of life or death.Nat Rev Cancer. 2006; 6: 449-458Crossref PubMed Scopus (1456) Google Scholar, 23Igney F.H. Krammer P.H. Death and anti-death: tumour resistance to apoptosis.Nat Rev Cancer. 2002; 2: 277-288Crossref PubMed Scopus (1701) Google Scholar, 24Johnstone R.W. Ruefli A.A. Lowe S.W. Apoptosis: a link between cancer genetics and chemotherapy.Cell. 2002; 108: 153-164Abstract Full Text Full Text PDF PubMed Scopus (2051) Google Scholar In the current study, we show that depletion of OPN by RNA-mediated interference promotes HCC cell apoptosis, leading to inhibition of tumorigenesis and metastasis both in vitro and in vivo. Previous studies have shown that OPN may prevent apoptosis through interaction with integrins and/or CD44 or its variants. OPN-CD44 interaction induced the phosphorylation of Akt in the survival pathway of interleukin-3,25Lin Y.H. Yang-Yen H.F. The osteopontin-CD44 survival signal involves activation of the phosphatidylinositol 3-kinase/Akt signaling pathway.J Biol Chem. 2001; 276: 46024-46030Crossref PubMed Scopus (209) Google Scholar whereas OPN-integrin αvβ1 and αvβ3 interaction induced endothelial cell survival as a result of the activation of NF-κB.26Scatena M. Almeida M. Chaisson M.L. et al.NF-kappaB mediates alphavbeta3 integrin-induced endothelial cell survival.J Cell Biol. 1998; 141: 1083-1093Crossref PubMed Scopus (449) Google Scholar In exploring the molecular mechanism responsible for OPN-depletion–induced apoptosis, a significantly decreased expressions of αν, β3, and β1 integrin were found. Given that integrins are a large family of cell-surface receptors that bind to ECM components to activate signaling pathways for tumor invasion and metastasis,27Guo W. Giancotti F.G. Integrin signalling during tumour progression.Nat Rev Mol Cell Biol. 2004; 5: 816-826Crossref PubMed Scopus (1240) Google Scholar inhibiting αν, β3, and β1 integrin expressions and their signaling pathway by OPN depletion might contribute to the inhibitory effect on tumor metastasis and apoptosis. However, how the OPN silencing might inhibit integrin expression still needs to be identified. Supplement of rhOPN could not reverse shOPN-induced apoptosis in HCC-LM3, which is related directly to the decreased expression of integrins by OPN depletion. The shOPN-treated tumor cells with significantly decreased expression of integrins had a dysfunction to bind and use the secreted or exogenous OPN, which directly contributed to cell apoptosis. However, the importance of intracellular OPN in the regulation of cell survival still needs to be investigated. Because most tumor cells contain high levels of intracellular OPN, the exogenous or tumor cell–secreted OPN might integrate with intracellular OPN to regulate cell survival. Several reports have indicated the vital roles of intracellular OPN. Intracellular OPN is essential for interferon-alfa production by plasmacytoid dendritic cells,28Shinohara M.L. Lu L. Bu J. et al.Osteopontin expression is essential for interferon-alpha production by plasmacytoid dendritic cells.Nat Immunol. 2006; 7: 498-506Crossref PubMed Scopus (311) Google Scholar and also exists as an integral component of a hyaluronan-CD44-ezrim/radixim/moesin (ERM) attachment complex involved in the migration of embryonic fibroblasts, activated macrophages, and metastatic tumor cells.29Zohar R. Suzuki N. Suzuki K. et al.Intracellular osteopontin is an integral component of the CD44-ERM complex involved in cell migration.J Cell Physiol. 2000; 184: 118-130Crossref PubMed Scopus (242) Google Scholar Localization of OPN to the nucleus is associated with polo-like kinase-1, a mitosis regulator, in 293 cells.30Junaid A. Moon M.C. Harding G.E. et al.Osteopontin localizes to the nucleus of 293 cells and associates with polo-like kinase-1.Am J Physiol Cell Physiol. 2007; 292: C919-C926Crossref PubMed Scopus (67) Google Scholar NF-κB promotes cell survival through enhancing the expression of genes encoding anti-apoptotic proteins, such as Bcl-xL and BFL1 acting at the mitochondria, or IAP1, IAP2, and XIAP, blocking caspase activation.31Karin M. Nuclear factor-kappaB in cancer development and progression.Nature. 2006; 441: 431-436Crossref PubMed Scopus (3077) Google Scholar, 32Nakanishi C. Toi M. Nuclear factor-kappaB inhibitors as sensitizers to anticancer drugs.Nat Rev Cancer. 2005; 5: 297-309Crossref PubMed Scopus (702) Google Scholar Recent data have shown that p50/p65 dimers inhibited bax promoter activity, whereas p50/p50 dimers might enhance its activity.33Grimm T. Schneider S. Naschberger E. et al.EBV latent membrane protein-1 protects B cells from apoptosis by inhibition of BAX.Blood. 2005; 105: 3263-3269Crossref PubMed Scopus (80) Google Scholar Our present study suggests that the proapoptotic function of shOPN may result from, at least in part, impaired NF-κB activation and subsequent inhibition of anti-apoptotic proteins such as Bcl-2/Bcl-xL and XIAP, and an increase of pro-apoptotic protein Bax. NF-κB is activated in response to several types of chemotherapeutic drugs, which blunts the ability of the cancer therapy to induce cell death.32Nakanishi C. Toi M. Nuclear factor-kappaB inhibitors as sensitizers to anticancer drugs.Nat Rev Cancer. 2005; 5: 297-309Crossref PubMed Scopus (702) Google Scholar Inhibition of NF-κB activity has been shown as an adjuvant approach to chemotherapy for certain cancers.32Nakanishi C. Toi M. Nuclear factor-kappaB inhibitors as sensitizers to anticancer drugs.Nat Rev Cancer. 2005; 5: 297-309Crossref PubMed Scopus (702) Google Scholar, 34Wang C.Y. Cusack Jr, J.C. Liu R. et al.Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB.Nat Med. 1999; 5: 412-417Crossref PubMed Scopus (0) Google Scholar Here, we found that OPN depletion significantly blocked NF-κB activation induced by various chemotherapy drugs in HCC cells. Thus, shOPN might sensitize HCC cells to chemotherapy through inhibition of NF-κB activity. This hypothesis was authenticated by the finding that shOPN promoted chemotherapeutic agents and TRAIL-induced cell death in HCC cells. Importantly, the combination of intratumoral injection of shOPN with systemic administration of 5-FU completely eradicated established human hepatoma in nude mice. Given that HCC is refractory to chemotherapeutic drugs,23Igney F.H. Krammer P.H. Death and anti-death: tumour resistance to apoptosis.Nat Rev Cancer. 2002; 2: 277-288Crossref PubMed Scopus (1701) Google Scholar, 35Johnson P.J. Hepatocellular carcinoma: is current therapy really altering outcome?.Gut. 2002; 51: 459-562Crossref PubMed Scopus (100) Google Scholar our finding provides a new clue for the treatment of HCC by sensitizing HCC to chemotherapeutic drug–mediated cell death. In conclusion, our data presented here show that down-regulation of OPN expression in HCC cells significantly inhibits tumorigenesis and metastasis, which might be ascribed to the pro-apoptotic effects induced by OPN depletion. The enhanced sensitivity of HCC cells to chemotherapeutic drugs through blockade of NF-κB activation by OPN silencing may have a great potential in the development of therapeutic regimens for cancer treatment. The authors thank Dr Hongyang Wang for providing the NF-κB–driven luciferase plasmid, and also are grateful to Yunchao Ma, Huaizu Guo, Ling Wang, Qirui Liu, and Yan Huang for technical support.