Co-culture of hepatoma cells with hepatocytic precursor (stem-like) cells inhibits tumor cell growth and invasion by downregulating Akt/NF-κB expression

  • Authors:
    • Cheng‑Jun Sui
    • Miao Xu
    • Wei‑Qing Li
    • Jia‑Mei Yang
    • Hong‑Zhu Yan
    • Hui‑Min Liu
    • Chun‑Yan Xia
    • Hong‑Yu Yu
  • View Affiliations

  • Published online on: September 13, 2016     https://doi.org/10.3892/ol.2016.5128
  • Pages: 4054-4060
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocytic stem cells (HSCs) have inhibitory effects on hepatocarcinoma cells. The present study investigated the effects of HSC activity in hepatocarcinoma cells in vitro. A Transwell co‑culture system of hepatocytic precursor (stem‑like) WB‑F344 cells and hepatoma CBRH‑7919 cells was used to assess HSC activity in metastasized hepatoma cells in vitro. Nude mouse xenografts were used to assess HSC activity in vivo. Co‑culture of hepatoma CBRH‑7919 cells with WB‑F344 cells suppressed the growth and colony formation, tumor cell migration and invasion capacity of CBRH‑7919 cells. The nude mouse xenograft assay demonstrated that the xenograft size of CBRH‑7919 cells following co‑culture with WB‑F344 cells was significantly smaller compared with that of control cells. Furthermore, the expression levels of the epithelial markers E‑cadherin and β‑catenin were downregulated, while the mesenchymal markers α‑SMA and vimentin were upregulated. Co‑culture of CBRH‑7919 cells with WB‑F344 cells downregulated NF‑κB and phospho‑Akt expression. In conclusion, hepatocytic precursor (stem‑like) WB‑F344 cells inhibited the growth, colony formation and invasion capacity of metastasized hepatoma CBRH‑7919 cells in vitro and in vivo by downregulating Akt/NF‑κB signaling.

Introduction

Hepatocellular carcinoma (HCC) is a significant health problem in the world and has a very poor prognosis (1). To date, a treatment strategy does not exist to effectively treat HCC patients (1,2). Although the overall 5-year survival rate of HCC patients receiving conventional chemotherapy after surgical resection or liver transplantation at a sufficiently early stage is increasing (3), the long-term prognosis remains unsatisfactory due to high recurrence rates. Therefore, novel approaches are needed to prevent and treat HCC.

Stem cell research may provide a novel insight into identifying a better strategy to treat HCC. Both embryonic and adult stem cells have self-renewal capacities and have the ability to produce differentiated progenitors. During tumorigenesis, stem cells from normal tissues are able to migrate into tumor lesions and suppress tumor cell growth (4,5). For example, a previous study demonstrated that human bone marrow-derived mesenchymal stem cells could be used to treat gliomas (4). These findings led to other studies that investigated whether modified stem cells can express anticancer molecules as targeted anticancer agents (69) and whether normal stem cells have antitumor activity.

In liver tissue, the ability of hepatocytic stem cells to migrate into HCC tissue has been reported previously (10). Thus, stem cells may be used as a vector to deliver therapeutic genes for targeted cancer therapy (1113). Indeed, the microenvironment of stem cells is considered to serve a role in regulating HCC cell growth (14,15) by downregulating multiple signal transduction pathways, such as the Wnt/β-catenin (14) and TGF-β/Smad (16) pathways. However, the molecular mechanisms of the inhibitory effect of stem cells on tumor cells remain to be defined.

Constitutive activation of Akt and nuclear factor-kappaB (NF-κB) are major cellular abnormalities associated with HCC pathogenesis and progression (17). Inactivation of NF-κB signaling inhibits growth and induces apoptosis of HCC cells (1821). Similarly, a previous study demonstrated that inhibition of Akt signaling resulted in HCC cell growth, arrest and apoptosis (22,23). Another previous study demonstrated that Akt activates NF-κB transcription by phosphorylating IκBκ (24,25). Thus, inhibiting Akt/NF-κB signaling may be a novel therapeutic approach for the control of HCC (17).

The present authors previous study demonstrated that hepatocytic precursor (stem-like) cells reduced the tumorigenicity and induced apoptosis of hepatoma cells in a co-culture system (16). In the present study, the effect of co-culturing hepatoma cells with hepatocytic precursor (stem-like) cells on the regulation of HCC phenotypic and gene expression was assessed in vitro and in nude mice. The present study aimed to provide useful insight into the development of stem cell-initiated antitumor therapy for HCC patients.

Materials and methods

Cell lines and culture

The diploid rat liver epithelial cell line WB-F344 was obtained from the Shanghai Cell Bank (Shanghai, China). These cells are oval in shape and have the capacity to differentiate into hepatocyte and biliary lineages under suitable conditions (26); thus, these cells are considered to be hepatocytic precursor (stem-like) cells. The rat hepatoma cell line CBRH-7919 was also obtained from the Shanghai Cell Bank.

WB-F344 and CBRH-7919 cells were co-cultured using a Transwell chamber culture system containing 4.0 µm pore polycarbonate membrane inserts (Corning, NY, USA). The WB-F344 to CBRH-7919 cell ratio was 5:1 (2×105:4×104 cells/well). CBRH-7919 cells cultured alone in the Transwell chamber culture system were used as a control. All the cells were cultured in serum-free conditioned medium from established cultures at 37°C with 95% air, 5% CO2, and 100% humidity for 7 days before the cells were used for subsequent assays. The serum-free conditioned medium was composed of Dulbecco's modified Eagle's medium/Ham's F12 medium (DMEM/F12, Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 20 ng/ml of basic fibroblast growth factor (Sigma-Aldrich, St. Louis, MO, USA), 20 ng/ml of epidermal growth factor (Sigma-Aldrich), and 20 µl/ml of B27 supplement (Invitrogen; Thermo Fisher Scientific, Inc.).

Nude mouse experiments

The present study was approved by the Institutional Animal Care and Use Committee (IACUC) of the Second Military Medical University (Shanghai, China). The mice used in the experiment were maintained under specific pathogen-free conditions and handled in accordance with the procedures and guidelines set by the Institutional Animal Care and Use Committee of The Second Military Medical University (Shanghai, China). Co-cultured WB-F344 and CBRH-7919 cells and single culture CBRH-7919 cells (1×106 cell/mouse) were subcutaneously inoculated into the axillary fossae of female nude mice (age, 6–8 weeks old). The tumor size was monitored every 3 days by measuring the length and width with calipers. The tumor volume was calculated using the formula: [(L × W2) × 0.5 mm3], in which L was the length and W was the width of each tumor. At day 35 post-injection, mice were sacrificed for pathological analysis.

Cell proliferation and clonogenic assays

Cell counting kit-8 (CCK-8) is a sensitive, nonradioactive colorimetric assay that assesses cell proliferation and detects the number of living cells. In the present study, a CCK-8 (Dojindo Molecular Technologies, Inc., Tokyo, Japan) assay was performed to assess the effect of rat WB-F344 stem cells on CBRH-7919 cell proliferation. In brief, after co-culturing these cell lines for 7 days in serum-free conditioned medium, CBRH-7919 cells were trypsinized, counted, and 5×104 cells were seeded in 24-well plates in triplicate and cultured for up to 8 days. At the end of each experiment, the cells were further incubated with an additional equal amount of fresh medium containing 10% CCK-8 at 37°C for 4 h, and the cell number was then counted. The data are presented as the mean cell number of each count in the curve diagrams.

For the clonogenic assay, CBRH-7919-only cultured cells and CBRH-7919 cells co-cultured with WB-F344 stem cells were seeded in 12-well plates in triplicate at a density of 100 cells/well and grown for 14 days. Subsequently, cell colonies were stained with 0.5% crystal violet and images were captured (EOS 600D Digital SLR; Canon, Inc., Tokyo, Japan) using an Olympus 1×71 inverted microscope (Olympus Corp., Tokyo). The number of colonies was counted 14 days after seeding. A colony was counted only if it contained ≥50 cells. The rate of colony formation was calculated with the following equation: colony formation rate = (number of colonies/number of seeded cells) ×100%.

Tumor cell migration and invasion assay

The ability of CBRH-7919-only cultured cells and CBRH-7919 cells co-cultured with WB-F344 stem cells to invade through Matrigel-coated filters was investigated using the 8-µm BD Falcon™ cell culture insert (BD Biosciences, San Jose, CA, USA). Briefly, 1×105 cell were suspended in 500 µl serum-free DMEM/F12 and then seeded into the upper compartments of each chamber. The lower compartments were filled with 1 ml DMEM/F12 supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.). After 24 h of incubation at 37°C in 5% CO2, non-invading cells were removed by scrubbing the upper surface of the membrane. Cells that invaded into the bottom surface of the membrane were fixed with methanol and stained with 0.5% crystal violet. The cells were then reviewed and counted under a microscope (Olympus IX71; Olympus Corporation, Tokyo, Japan) in 5 microscopic fields (x× magnification).

Wound healing assay

Scratch assays were performed to assess the effect of WB-F344 stem cells on the migration of hepatoma cells. Briefly, cells were seeded in 6-well plates at a density of 2×105 cells/well. When the cells were 90–100% confluent, the monolayer was scratched manually with a plastic pipette tip across each plate. The cells were then washed twice with phosphate buffered saline (PBS) and further incubated for up to 96 h. At the end of each experiment, images of each plate were captured to demonstrate ‘wound healing’ using a PowerShot G10 camera (Canon, Inc.). The distance of each wound was measured and the mean was calculated for the capacity of tumor cell migration.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total cellular RNA was isolated from the cultured cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and then reverse transcribed into cDNA using a Two-Step RT-PCR kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The PCR cycling conditions were as follows: One cycle at 95°C for 30 sec, followed by 30 cycles at 95°C for 5 sec, one cycle at 60°C for 30 sec, with a final extension step at 72°C for 1 min, using the SYBR Green Realtime PCR Master Mix (Takara Bio, Inc., Otsu, Japan) according to the manufacturer's protocol. The samples were held at 4°C until required. To determine the levels of E-cadherin, β-catenin, α-SMA and vimentin, qPCR was performed with an ABI 7300 instrument (Thermo Fisher Scientific, Inc.) using a SYBR PrimeScript RT-PCR kit (Takara Suzo, Kyoto, Japan) according to the manufacturer's instructions. The mRNA β-actin level was used as an internal control. The primer sequences used for amplification were as follows: E-cadherin, 5′-TGAGGTCGGTGCCCGTATTG-3′ and 5′-GAATGCCCTCGTTGGTCTTGG-3′; β-catenin, 5′-ACTCTAGTGCAGCTTGGTTC-3′ and 5′-ATGGCAGGCTCGGTAATGTC-3′; α-SMA, 5′-ATGACCCAGATTATGTTTGAGACC-3′ and 5′-CCAGAGTCCAGCACAATACCAG-3′; vimentin, 5′-CGCAGCCTCTATTCCTCGTC-3′ and 5′-TCTTGAACTCGGTGTTGATGG-3′; and β-actin, 5′-ACGTTGACATCCGTAAAGAC-3′ and 5′-GAAGGTGGACAGTGAGGC-3′. The relative mRNA levels were calculated based on the Ct values and normalized to the level of β-actin mRNA according to the equation: 2−ΔΔCt [ΔCt=Ct (Gene)-Ct (β-actin)]. All of the experiments were performed in triplicate and repeated once.

Protein extraction and western blot analysis

The levels of NF-κB (p65), phospho-Akt, phospho-IKKα/β and phospho-IκBα proteins were assayed in CBRH-7919 cells co-cultured with WB-F344 cells and CBRH-7919-only cultured cells using western blot analysis. Briefly, total cell extracts were prepared using RIPA lysis buffer (50 mM Tris-HCl, 1% NP-40, 2 mM EDTA, 10 mM NaCl, 2 mg/ml aprotinin, 5 mg/ml leupeptin, 2 mg/ml pepstatin, 1 mM DTT, 0.1% SDS and 1 mM phenylmethylsulfonyl fluoride). Protein concentrations were determined using the BCA assay (Pierce Biotechnology, Inc., Rockford, IL, USA). Equal amounts of protein (20 µg) were then separated by SDS-PAGE and transferred onto PVDF membranes (EMD Millipore, Billerica, MA, USA) by electroblotting. The membranes were then blocked with 5% non-fat milk in PBS-T (PBS with 0.05% Tween-20) for 1 h and then incubated with specific primary antibodies against rat NF-κB (p65; rabbit monoclonal; dilution, 1:500; catalog no., CST-8242), phospho-Akt (rabbit monoclonal; dilution, 1:1,000; catalog no., CST-12178), phospho-IKKα/β (rabbit monoclonal; dilution, 1:1,000; catalog. no., CST-2697), phospho-IκBα (rabbit monoclonal; dilution, 1:500; catalog no., CST-2859; all purchased from Cell Signaling Technology, Inc., Danvers, MA, USA) and GAPDH (rabbit polyclonal; dilution, 1:500; catalog no., SC-25778; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) overnight at 4°C. The membranes were then washed with PBS-T and incubated with a horseradish peroxidase conjugated goat anti-rabbit immunoglobulin G (dilution, 1:2,000; catalog no., SC-2004; Santa Cruz Biotechnology, Inc.) for 1 h. The protein bands were visualized using an enhanced chemiluminescence kit (Pierce Biotechnology, Inc.) and exposed on X-ray films. The level of protein expression was normalized to GAPDH protein.

Statistical analysis

Data are expressed as the mean ± standard deviation. SPSS version 19 (IBM SPSS, Armonk, NY, USA) was used to analyze the data. Statistical analysis was performed using the two-tailed Student's t-test and P<0.05 was considered to indicate a statistically significant difference.

Results

Hepatocytic precursor WB-F344 cells suppressed the growth of hepatoma cell xenografts in nude mice

In the present study, CBRH-7919 cells were co-cultured with WB-F344 cells or were cultured alone for 7 days and then injected into nude mice. On day 22 post-injection, nude mice bearing co-cultured or control CBRH-7919 cells had similar tumor xenograft volumes. From ~4 weeks, mice bearing co-cultured CBRH-7919 cells had a significantly reduced tumor volume compared with mice bearing control xenografts (Fig. 1, P<0.05). The data indicated that WB-F344 cells inhibited the growth of hepatoma CBRH-7919 cell xenografts in nude mice.

Hepatocytic precursor (stem-like) WB-F344 cells suppressed the clonogenic growth of CBRH-7919 cells

After co-culture with hepatocytic precursor (stem-like) WB-F344 cells, hepatoma CBRH-7919 cells exhibited a reduced cell growth and proliferation rate; there was a significant reduction in the number of CBRH-7919 cells in both monolayer and colony formation (P<0.05, Fig. 2A and B).

Hepatocytic precursor (stem-like) WB-F344 cells suppressed the migration and invasion of CBRH-7919 cells

As presented in Fig. 3A, the Transwell assay demonstrated that co-cultured CBRH-7919 cells demonstrated a reduced invasion capacity compared with that of the control cells (P<0.05).

In addition, epithelial-mesenchymal transition (EMT), a key event in tumor invasion and metastasis (27,28), was also characterized in the present study. The RT-qPCR analysis results demonstrated that the mRNA expression levels of the epithelial markers E-cadherin and β-catenin were downregulated in co-cultured CBRH-7919 cells, while the mesenchymal markers α-SMA and vimentin were upregulated when compared with control cells (P<0.05; Fig. 3B).

The tumor cell wound healing assay demonstrated that CBRH-7919 cells co-cultured with WB-F344 cell demonstrated a reduced invasion capacity compared with that of the control cells (P<0.05; Fig. 3C and D).

These results indicated that co-culture with WB-F344 cells suppressed the migration and invasion of CBRH-7919 cells via regulation of tumor cell EMT activity.

Akt/NF-κB signaling mediated the migration and invasion capacity of hepatoma CBRH-7919 cells

Since AKT and NF-κB are essential regulators of tumor cell properties, including metastasis (29,30), the present authors proposed that the Akt/NF-κB signaling pathway may be involved in the inhibition of hepatoma CBRH-7919 cell migration and invasion capacity through its pro-apoptotic role. As illustrated in Fig. 4, western blot analyses demonstrated that co-culture of hepatoma CBRH-7919 cells with WB-F344 cells resulted in the downregulation of NF-κB and phospho-Akt expression in CBRH-7919 cells.

Discussion

HCC is a leading cause of cancer-associated mortality in the world and has an extremely poor prognosis (31,32). The development and identification of novel therapeutic strategies to effectively control HCC metastasis may increase patient survival. Previous studies have indicated that stem cells may be useful to treat different types of human cancer (415). Regardless of their distinct origins, stem cells and tumor cells share a number of common properties, including the loss of contact inhibition and immortality, and therefore, similar signaling pathways may be involved in their control. It has been reported that the stem cell microenvironment is essential in preventing carcinogenesis by providing signals to inhibit cell proliferation and to promote differentiation (33). In the current study, it was demonstrated that co-culture with WB-F344 cells suppressed the growth, colony formation and tumor cell migration and invasion capacity of CBRH-7919 cells. Co-cultured CBRH-7919 cells also expressed downregulated levels of the epithelial markers E-cadherin and β-catenin and upregulated expression levels of the mesenchymal markers α-SMA and vimentin via downregulating NF-κB and phospho-Akt expression. In addition, the co-culture of CBRH-7919 cells with WB-F344 cells inhibited xenograft formation and growth in a nude mouse model. The current study therefore indicates that hepatocytic precursor (stem-like) WB-F344 cells regulate HCC cell phenotype and gene expression. However, this study is a proof-of-principle study and additional studies are needed to understand the underlying molecular mechanisms by which stem cells act on HCC cells.

The results of the present study are consistent with the effect of human mesenchymal stem cells on HCC cells, as previously reported (34). In the current study, suppression of the Akt/NF-κB signaling pathway after co-culturing CBRH-7919 cells with WB-F344 cells contributed to the suppression of HCC cell growth and invasion. Indeed, aberrant activation of the Akt/NF-κB signaling pathway has been widely investigated and reported in the literature. Akt and NF-κB proteins regulate a variety of cellular processes, including cell proliferation, apoptosis, invasion and angiogenesis (3538). Furthermore, Akt and NF-κB are being extensively investigated as molecular targets for cancer therapeutics (38), as studies have demonstrated that repression of Akt and/or NF-κB activity inhibit cancer cell growth, migration and invasion, and sensitizes cancer cells to apoptosis (3941). In agreement with these previous studies, the current data demonstrated that Akt-mediated phosphorylation of IKKα/β and IκBα was necessary for the activation of NF-κB, further mediating the metastasis potential of hepatoma CBRH-7919 cells in this co-culture system. In the resting state, the activity of NF-κB is sequestered in the cytoplasm as an inactive precursor complex with inhibitory κB (IκB). The biological activity of NF-κB is tightly regulated by IκBα. It appears that newly synthesized IκB rapidly re-associates with newly released NF-κB, thereby markedly reducing the amount of NF-κB translocated into the nucleus for the activation of cytokine genes. The activity of the NF-κB transcription factor is regulated by p-IκB through multiple intracellular signal transduction pathways (42).

In conclusion, hepatocytic precursor (stem-like) WB-F344 cells inhibited the growth, colony formation and invasion capacity of hepatoma CBRH-7919 cells in vitro and in vivo by downregulating Akt/NF-κB signaling. However, further studies are required to confirm the results of the present study. Subsequent studies by the present authors will assess whether overexpressing genes in this signaling pathway overcome the activity of co-culturing CBRH-7919 cells with WB-F344 cells.

Acknowledgements

The present study was supported in part by grants from the National Natural Sciences Fund Project of China (nos. 81370553 and 39670699 to Dr. Hong-Yu Yu). The authors would like to thank Medjaden Bioscience Limited (Hong Kong, China) for editing and improving English of this manuscript.

References

1 

He J, Gu D, Wu X, et al: Major causes of death among men and women in China. N Engl J Med. 353:1124–1134. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, et al: Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 359:378–390. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Nakamizo A, Marini F, Amano T, Khan A, Studeny M, Gumin J, Chen J, Hentschel S, Vecil G, Dembinski J, et al: Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 65:3307–3318. 2005.PubMed/NCBI

5 

Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W, Small JE, Herrlinger U, Ourednik V, Black PM, et al: Neural stem cells display extensive tropism for pathology in adult brain: Evidence from intracranial gliomas. Proc Natl Acad Sci U S A. 97:12846–12851. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Chen X, Lin X, Zhao J, Shi W, Zhang H, Wang Y, Kan B, Du L, Wang B, Wei Y, et al: A tumor-selective biotherapy with prolonged impact on established metastases based on cytokine gene-engineered MSCs. Mol Ther. 16:749–756. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Uchibori R, Okada T, Ito T, Urabe M, Mizukami H, Kume A and Ozawa K: Retroviral vector-producing mesenchymal stem cells for targeted suicide cancer gene therapy. J Gene Med. 11:373–381. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Ren C, Kumar S, Chanda D, Chen J, Mountz JD and Ponnazhagan S: Therapeutic potential of mesenchymal stem cells producing interferon-alpha in a mouse melanoma lung metastasis model. Stem Cells. 26:2332–2338. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Nakamura K, Ito Y, Kawano Y, Kurozumi K, Kobune M, Tsuda H, Bizen A, Honmou O, Niitsu Y and Hamada H: Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther. 11:1155–1164. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Zhong XG, He S, Yin W, Deng JY and Cheng B: Selective tropism of liver stem cells to hepatocellular carcinoma in vivo. World J Gastroenterol. 13:3886–3891. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Chen XC, Wang R, Zhao X, Wei YQ, Hu M, Wang YS, Zhang XW, Zhang R, Zhang L, Yao B, et al: Prophylaxis against carcinogenesis in three kinds of unestablished tumor models via IL12-gene-engineered MSCs. Carcinogenesis. 27:2434–2441. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Gao Y, Yao A, Zhang W, et al: Human mesenchymal stem cells overexpressing pigment epithelium-derived factor inhibit hepatocellular carcinoma in nude mice. Oncogene. 29:2784–2794. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Aquino JB, Bolontrade MF, García MG, Podhajcer OL and Mazzolini G: Mesenchymal stem cells as therapeutic tools and gene carriers in liver fibrosis and hepatocellular carcinoma. Gene Ther. 17:692–708. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Qiao L, Xu Z, Zhao T, et al: Suppression of tumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res. 18:500–507. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Qiao L, Zhao TJ, Wang FZ, Shan CL, Ye LH and Zhang XD: NF-kappaB downregulation may be involved the depression of tumor cell proliferation mediated by human mesenchymal stem cells. Acta Pharmacol Sin. 29:333–340. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Campos B, Wan F, Farhadi M, Ernst A, Zeppernick F, Tagscherer KE, Ahmadi R, Lohr J, Dictus C, Gdynia G, et al: Differentiation therapy exerts antitumor effects on stem-like glioma cells. Clin Cancer Res. 16:2715–2728. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Omar HA, Sargeant AM, Weng JR, Wang D, Kulp SK, Patel T and Chen CS: Targeting of the Akt-nuclear factor-kappaB signaling network by [1-(4-chloro-3-nitrobenzenesulfonyl)-1H-indol-3-yl]-methanol (OSU-A9), a novel indole-3-carbinol derivative, in a mouse model of hepatocellular carcinoma. Mol Pharmacol. 76:957–968. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Wu LF, Li GP, Su JD, Pu ZJ, Feng JL, Ye YQ and Wei BL: Involvement of NF-kappaB activation in the apoptosis induced by extracellular adenosine in human hepatocellular carcinoma HepG2 cells. Biochem Cell Biol. 88:705–714. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Kim MO, Moon DO, Kang CH, Kwon TK, Choi YH and Kim GY: beta-Ionone enhances TRAIL-induced apoptosis in hepatocellular carcinoma cells through Sp1-dependent upregulation of DR5 and downregulation of NF-kappaB activity. Mol Cancer Ther. 9:833–843. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Ren K, Zhang W, Shi Y and Gong J: Pim-2 activates API-5 to inhibit the apoptosis of hepatocellular carcinoma cells through NF-kappaB pathway. Pathol Oncol Res. 16:229–237. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Cho HA, Park IS, Kim TW, Oh YK, Yang KS and Kim JS: Suppression of hepatitis B virus-derived human hepatocellular carcinoma by NF-kappaB-inducing kinase-specific siRNA using liver-targeting liposomes. Arch Pharm Res. 32:1077–1086. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Fei HR, Chen G, Wang JM and Wang FZ: Perifosine induces cell cycle arrest and apoptosis in human hepatocellular carcinoma cell lines by blockade of Akt phosphorylation. Cytotechnology. 62:449–460. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Buontempo F, Ersahin T, Missiroli S, Senturk S, Etro D, Ozturk M, Capitani S, Cetin-Atalay R and Neri ML: Inhibition of Akt signaling in hepatoma cells induces apoptotic cell death independent of Akt activation status. Invest New Drugs. 29:1303–1313. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Nair AS, Shishodia S, Ahn KS, Kunnumakkara AB, Sethi G and Aggarwal BB: Deguelin, an Akt inhibitor, suppresses IkappaBalpha kinase activation leading to suppression of NF-kappaB-regulated gene expression, potentiation of apoptosis and inhibition of cellular invasion. J Immunol. 177:5612–5622. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Ozes ON, Mayo LD, Gustin JA, Pfeffer SR, Pfeffer LM and Donner DB: NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase. Nature. 401:82–85. 1999. View Article : Google Scholar : PubMed/NCBI

26 

Tsao MS, Smith JD, Nelson KG and Grisham JW: A diploid epithelial cell line from normal adult rat liver with phenotypic properties of 'oval' cells. Exp Cell Res. 154:38–52. 1984. View Article : Google Scholar : PubMed/NCBI

27 

Huber MA, Kraut N and Beug H: Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol. 17:548–558. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Kang Y and Massagué J: Epithelial-mesenchymal transitions: Twist in development and metastasis. Cell. 118:277–279. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Lin HH, Chen JH, Chou FP and Wang CJ: Protocatechuic acid inhibits cancer cell metastasis involving the down-regulation of Ras/Akt/NF-kappaB pathway and MMP-2 production by targeting RhoB activation. Br J Pharmacol. 162:237–254. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Park YP, Choi SC, Kim BY, Kim JT, Song EY, Kang SH, Yoon DY, Paik SG, Kim KD, Kim JW and Lee HG: Induction of Mac-2BP by nerve growth factor is regulated by the PI3K/Akt/NF-kappaB-dependent pathway in the HEK293 cell line. BMB Rep. 41:784–789. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Yuki K, Hirohashi S, Sakamoto M, Kanai T and Shimosato Y: Growth and spread of hepatocellular carcinoma. A review of 240 consecutive autopsy cases. Cancer. 66:2174–2179. 1990. View Article : Google Scholar : PubMed/NCBI

32 

Genda T, Sakamoto M, Ichida T, Asakura H, Kojiro M, Narumiya S and Hirohashi S: Cell motility mediated by rho and Rho-associated protein kinase plays a critical role in intrahepatocytic metastasis of human hepatocellular carcinoma. Hepatology. 30:1027–1036. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Li L and Neaves WB: Normal stem cells and cancer stem cells: The niche matters. Cancer Res. 66:4553–4557. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Li GC, Ye QH, Xue YH, Sun HJ, Zhou HJ, Ren N, Jia HL, Shi J, Wu JC, Dai C, et al: Human mesenchymal stem cells inhibit metastasis of a hepatocellular carcinoma model using the MHCC97-H cell line. Cancer Sci. 101:2546–2553. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Karin M, Cao Y, Greten FR and Li ZW: NF-kappaB in cancer: From innocent bystander to major culprit. Nat Rev Cancer. 2:301–310. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Haefner B: NF-kappaB: Arresting a major culprit in cancer. Drug Discov Today. 7:653–663. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Testa JR and Bellacosa A: AKT plays a central role in tumorigenesis. Proc Natl Acad Sci U S A. 98:10983–10985. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Cheng JQ, Lindsley CW, Cheng GZ, Yang H and Nicosia SV: The Akt/PKB pathway: Molecular target for cancer drug discovery. Oncogene. 24:7482–7492. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Wendel HG, De Stanchina E, Fridman JS, Malina A, Ray S, Kogan S, Cordon-Cardo C, Pelletier J and Lowe SW: Survival signalling by Akt and eIF4E in oncogenesis and cancer therapy. Nature. 428:332–337. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Clark AS, West K, Streicher S and Dennis PA: Constitutive and inducible Akt activity promotes resistance to chemotherapy, trastuzumab, or tamoxifen in breast cancer cells. Mol Cancer Ther. 1:707–717. 2002.PubMed/NCBI

41 

Li Y, Ahmed F, Ali S, Philip PA, Kucuk O and Sarkar FH: Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells. Cancer Res. 65:6934–6942. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Konrath F, Witt J, Sauter T and Kulms D: Identification of new IκBα complexes by an iterative experimental and mathematical modeling approach. PLoS Comput Biol. 10:e10035282014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 12 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sui CJ, Xu M, Li WQ, Yang JM, Yan HZ, Liu HM, Xia CY and Yu HY: Co-culture of hepatoma cells with hepatocytic precursor (stem-like) cells inhibits tumor cell growth and invasion by downregulating Akt/NF-κB expression. Oncol Lett 12: 4054-4060, 2016
APA
Sui, C., Xu, M., Li, W., Yang, J., Yan, H., Liu, H. ... Yu, H. (2016). Co-culture of hepatoma cells with hepatocytic precursor (stem-like) cells inhibits tumor cell growth and invasion by downregulating Akt/NF-κB expression. Oncology Letters, 12, 4054-4060. https://doi.org/10.3892/ol.2016.5128
MLA
Sui, C., Xu, M., Li, W., Yang, J., Yan, H., Liu, H., Xia, C., Yu, H."Co-culture of hepatoma cells with hepatocytic precursor (stem-like) cells inhibits tumor cell growth and invasion by downregulating Akt/NF-κB expression". Oncology Letters 12.5 (2016): 4054-4060.
Chicago
Sui, C., Xu, M., Li, W., Yang, J., Yan, H., Liu, H., Xia, C., Yu, H."Co-culture of hepatoma cells with hepatocytic precursor (stem-like) cells inhibits tumor cell growth and invasion by downregulating Akt/NF-κB expression". Oncology Letters 12, no. 5 (2016): 4054-4060. https://doi.org/10.3892/ol.2016.5128