FH535 suppresses the proliferation and motility of hepatocellular carcinoma cells

  • Authors:
    • Minoru Tomizawa
    • Fuminobu Shinozaki
    • Yasufumi Motoyoshi
    • Takao Sugiyama
    • Shigenori Yamamoto
    • Naoki Ishige
  • View Affiliations

  • Published online on: October 29, 2015     https://doi.org/10.3892/ijo.2015.3220
  • Pages: 110-114
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The Wnt signaling pathway is activated in hepatocellular carcinoma (HCC). This study investigated the effects of FH535, an inhibitor of the Wnt signaling pathway, on the proliferation and motility of HCC cells. HLF cells and PLC/PRF/5 cells, HCC cells, were subjected to 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay with the addition of FH535. RNA was isolated from the cells and subjected to real-time quantitative PCR. Hematoxylin and eosin (H&E) staining was performed to analyze apoptosis. A scratch assay was performed to analyze cell motility. Cell proliferation significantly decreased (P<0.05). The expression levels of cyclin D1 significantly decreased in both cell lines (P<0.05). Pyknotic nuclei were observed in the cells cultured with FH535 (50 µM). In the scratch assay, the distance between the growing edges of cells and the scratched line significantly decreased with the addition of FH535 at 50 µM (P<0.05). The expression levels of matrix metalloproteinase 9 significantly decreased at 50 µM (P<0.05). FH535 suppressed the proliferation of HCC cells by downregulating the expression of cyclin D1 and by inducing apoptosis. Further, it suppressed cell motility by downregulating the expression of matrix metalloproteinase.

Introduction

Hepatocellular carcinoma (HCC) is a cancer originating from the liver. Its prognosis is poor despite the advancements in treatment (1). Treatments for HCC include local ablation, surgery, transcatheter arterial chemoembolization, and chemotherapy (2,3). Molecular therapy has also been established as a treatment option (4). To develop a new molecular therapy, research has focused on signaling pathways (5).

The Wnt pathway is involved in cell proliferation and differentiation (6). Wnt proteins bind to their receptor, frizzled, and its co-receptors, low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6), to form a complex (7,8). β-catenin is degraded by the glycogen synthase kinase-3β complex (9). When Wnt binds to its receptor complex, the degradation of β-catenin is inhibited. β-catenin then accumulates in the cytoplasm and the Wnt pathway is activated. β-catenin is a co-factor of the T-cell factor (TCF)/lymphoid enhancer factor (LEF). When the Wnt pathway is activated, the accumulated β-catenin translocates to the nucleus, binds the promoter of target genes with TCF/LEF (10). In HCC, β-catenin is mutated and overexpressed, which suggests that the Wnt pathway is constitutively activated (11). Therefore, β-catenin is a potential target in the exploration of molecular therapy (12). The inhibition of frizzled-9 suppresses the proliferation and motility of HCC cells (13). Niclosamide is a drug used for the treatment of tapeworm infections. It is an inhibitor of the Wnt signaling pathway and it suppresses the proliferation of HCC cells (14). Previous reports indicate that the Wnt pathway is a promising target in the treatment of HCC.

FH535 is a small molecule that inhibits the Wnt signaling pathway and peroxisome proliferator-activator receptor signaling (15). One of its unique characteristics is that it inhibits the recruitment of β-catenin. Therefore, FH535 is expected to be a potent inhibitor of the Wnt signaling pathway.

In this study, we investigated the effects of FH535 on motility and proliferation of HCC cells.

Materials and methods

Cell culture

HLF cells and PLC/PRF/5 cells, human HCC cells, were obtained from RIKEN Cell Bank (Tsukuba, Japan) and cultured in Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS; Life Technologies, Grand Island, NY, USA). They were cultured in 10-cm dishes (Asahi Techno Glass, Funabashi, Japan) with 5% carbon dioxide at 37°C in a humidified chamber.

Cell proliferation assay

Cells were trypsinized, harvested, and spread onto 96-well plates (Asahi Techno Glass) at a density of 1,000 cells/well. They were cultured in DMEM supplemented with 10% FBS. The cells were cultured for 72 h with 0, 0.5, 1.5, 5, 15 or 50 μM FH535 (Merck, Darmstadt, Germany) and subjected to 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay, according to the manufacturer's instructions (Promega Corp., Madison, WI, USA). MTS is reduced by cells to a colored formazan product that has an absorbance maximum at 490 nm. Absorbance was measured using an iMark Microplate Absorbance Reader (Bio-Rad, Hercules, CA, USA).

Real-time quantitative polymerase chain reaction

Total RNA (5 μg), which was isolated using Isogen (Nippon Gene, Tokyo, Japan), was used for the first-strand cDNA synthesis with SuperScript III and oligo(dT) following the manufacturer's instructions (Life Technologies). Real-time quantitative PCR was performed using Fast SYBR Green Master Mix (Life Technologies) with MiniOpticon (Bio-Rad). The results were analyzed using the MiniOpticon system. Real-time quantitative PCR was performed for 40 cycles, with 5 sec of denaturation and 5 sec of annealing/extension. The primer sequences are listed in Table I. RPL19 was used as an internal control as the target gene is a constitutively expressed house-keeping gene (16).

Table I

The primer sequences.

Table I

The primer sequences.

Primer nameSequenceDescriptionProduct size (bp)Annealing temperatureCycleGenBank
OMC355 5′-AGAGGCGGAGGAGAACAAACAG-3′Cyclin D1, forward1806040NM_053056
OMC356 5′-AGGCGGTAGTAGGACAGGAAGTTG-3′Cyclin D1, reverse
OMC749 5′-CCTGGGCAGATTCCAAACCT-3′MMP9, forward896040NM_004994
OMC750 5′-GCAAGTCTTCCGAGTAGTTTTGGAT-3′MMP9, reverse
OMC321 5′-CGAATGCCAGAGAAGGTCAC-3′RPL19, forward1576040BC095445
OMC322 5′-CCATGAGAATCCGCTTGTTT-3′RPL19, reverse

[i] MMP9, matrix metalloproteinase 9; RPL19, ribosomal protein L (RPL) 19.

Scratch assay and hematoxylin and eosin staining

Cells were plated on 4-well chamber slides (Becton-Dickinson, Franklin Lakes, NJ, USA). When the cells reached confluence, they were scratched with a sterile razor. The cells were incubated with 0 or 50 μM FH535 for 48 h and stained with hematoxylin and eosin (H&E). For the analysis of apoptosis, the cells were plated in 4-well chamber slides (Becton-Dickinson). The cells were incubated with 0 or 50 μM FH535 for 48 h and then stained with H&E. The stained slides were observed under an AX80 microscope (Olympus, Tokyo, Japan) for the apoptosis analysis and scratch assay. In the scratch assay, the distance between the scratched line and the growing edges of the cells was measured at five points.

Statistical analysis

Data were analyzed by one-way analysis of variance (ANOVA) using JMP 10.0.2 software (SAS Institute, Cary, NC, USA). P-values <0.05 were considered statistically significant.

Results

To analyze the suppression of cell proliferation, HLF cells (Fig. 1A) and PLC/PRF/5 cells (Fig. 1B) were cultured with FH535. After 72 h, the cells were subjected to MTS assay. Proliferation was found to be significantly suppressed in both cell lines (P<0.05).

Cylin D1 is involved in the regulation of cell cycle progression (17). To evaluate the expression levels of cyclin D1, HLF cells (Fig. 2A) and PLC/PRF/5 cells (Fig. 2B) were incubated with FH535. After 48 h, RNA was isolated from the cells and subjected to real-time quantitative PCR. The expression levels of cyclin D1 were significantly suppressed in both cell lines (P<0.05).

To clarify the involvement of apoptosis in the suppression of cell proliferation, HLF (Fig. 3A and B) and PLC/PRF/5 cells (Fig. 3C and D) were incubated with FH535 at 0 μM (Fig. 3A and C) or 50 μM (Fig. 3B and D) and subjected to H&E staining. Pyknotic nuclei (arrows) were observed in the cells cultured with 50 μM FH535. These results indicate that the cells underwent apoptosis with FH535 at 50 μM.

To address the possibility that FH535 suppressed cell motility, HLF cells (Fig. 4A and C) and PLC/PRF/5 cells (Fig. 4B and D) were cultured with FH535 at 0 μM (Fig. 4A and C) or 50 μM (Fig. 4B and D). The distance between the growing edges of the cells and the scratched line significantly decreased with the addition of 50 μM FH535 (Fig. 4E) (P<0.05).

The expression levels of matrix metalloproteinase 9 were analyzed because this gene is involved in cancer metastasis (18). The expression levels of matrix metalloproteinase 9 were significantly suppressed in the HLF cells (Fig. 5A) and PLC/PRF/5 cells (Fig. 5B) (P<0.05).

Discussion

FH535 suppresses the proliferation of cancer cells. Specifically, in this study, it was observed to suppress the proliferation of HLF cells and PLC/PRF/5 cells. The expression levels of cyclin D1 decreased in both cell types after incubation with FH535. In the HCC cells, FH535 decreased the expression levels of cyclin D1 and suppressed the cell cycle (19). These results were consistent with those of the previous reports. The data clearly showed that the cells underwent apoptosis. The results indicated that FH535 suppressed cell proliferation by suppressing the cell cycle and inducing apoptosis.

FH535 suppresses cell motility as evidenced in the present study. In addition, FH535 is known to suppress the metastasis of HCC and pancreatic cancer cells (19,20). In this study, the expression levels of matrix metalloproteinase 9 decreased in HLF cells and PLC/PRF/5 cells. The previous reports and our data indicated that FH535 suppresses the motility of cancer cells by decreasing the expression levels of matrix metalloproteinase 9.

One possible limitation of this study is that the concentration of FH535 was relatively high. FH535 suppressed the proliferation and migration of HCC cells at 50 μM with statistical significance. In breast cancer cells, FH535 suppresses proliferation and migration at 1 μM (21). A higher concentration might be hazardous to cells, leading to adverse effects. To reduce this risk, using a combination of FH535 and other reagents would be desirable.

FH535 and sorafenib synergistically inhibit the proliferation of Huh-7 cells, another HCC cell line, and cancer stem cells (22). Another application of FH535 is in irradiation therapy (23). In the future, FH535 should be combined with other chemotherapeutic agents or small molecules.

In conclusion, FH535 suppressed cell proliferation by decreasing the expression of cyclin D1 and by inducing apoptosis. In addition, it suppressed cell motility by decreasing the expression of matrix metalloproteinase.

References

1 

Tejeda-Maldonado J, García-Juárez I, Aguirre-Valadez J, González-Aguirre A, Vilatobá-Chapa M, Armengol-Alonso A, Escobar-Penagos F, Torre A, Sánchez-Ávila JF and Carrillo-Pérez DL: Diagnosis and treatment of hepatocellular carcinoma: An update. World J Hepatol. 7:362–376. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Lencioni R, Petruzzi P and Crocetti L: Chemoembolization of hepatocellular carcinoma. Semin Intervent Radiol. 30:3–11. 2013. View Article : Google Scholar :

3 

Kim HY and Park JW: Clinical trials of combined molecular targeted therapy and locoregional therapy in hepatocellular carcinoma: Past, present, and future. Liver Cancer. 3:9–17. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Furuse J, Ishii H, Nakachi K, Suzuki E, Shimizu S and Nakajima K: Phase I study of sorafenib in Japanese patients with hepatocellular carcinoma. Cancer Sci. 99:159–165. 2008.

5 

Chen C and Wang G: Mechanisms of hepatocellular carcinoma and challenges and opportunities for molecular targeted therapy. World J Hepatol. 7:1964–1970. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Bogaerts E, Heindryckx F, Vandewynckel YP, Van Grunsven LA and Van Vlierberghe H: The roles of transforming growth factor-β, Wnt, Notch and hypoxia on liver progenitor cells in primary liver tumours (Review). Int J Oncol. 44:1015–1022. 2014.PubMed/NCBI

7 

Tanaka SS, Kojima Y, Yamaguchi YL, Nishinakamura R and Tam PP: Impact of WNT signaling on tissue lineage differentiation in the early mouse embryo. Dev Growth Differ. 53:843–856. 2011. View Article : Google Scholar : PubMed/NCBI

8 

MacDonald BT, Tamai K and He X: Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev Cell. 17:9–26. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Takahashi-Yanaga F: Activator or inhibitor? GSK-3 as a new drug target. Biochem Pharmacol. 86:191–199. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Jamieson C, Sharma M and Henderson BR: Targeting the β-catenin nuclear transport pathway in cancer. Semin Cancer Biol. 27:20–29. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Rizvi S and Gores GJ: Molecular profiling and research of therapeutic targets. Dig Dis. 33:586–589. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Monga SP: β-catenin signaling and roles in liver homeostasis, Injury, and tumorigenesis. Gastroenterology. 148:1294–1310. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Fujimoto T, Tomizawa M and Yokosuka O: SiRNA of frizzled-9 suppresses proliferation and motility of hepatoma cells. Int J Oncol. 35:861–866. 2009.PubMed/NCBI

14 

Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Sueishi M and Yoshida T: Niclosamide suppresses hepatoma cell proliferation via the Wnt pathway. Onco Targets Ther. 6:1685–1693. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Handeli S and Simon JA: A small-molecule inhibitor of Tcf/beta-catenin signaling down-regulates PPARgamma and PPARdelta activities. Mol Cancer Ther. 7:521–529. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Davies B and Fried M: The L19 ribosomal protein gene (RPL19): Gene organization, chromosomal mapping, and novel promoter region. Genomics. 25:372–380. 1995. View Article : Google Scholar : PubMed/NCBI

17 

Casimiro MC, Velasco-Velázquez M, Aguirre-Alvarado C and Pestell RG: Overview of cyclins D1 function in cancer and the CDK inhibitor landscape: Past and present. Expert Opin Investig Drugs. 23:295–304. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Vandooren J, Van den Steen PE and Opdenakker G: Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9): The next decade. Crit Rev Biochem Mol Biol. 48:222–272. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Gedaly R, Galuppo R, Daily MF, Shah M, Maynard E, Chen C, Zhang X, Esser KA, Cohen DA, Evers BM, et al: Targeting the Wnt/β-catenin signaling pathway in liver cancer stem cells and hepatocellular carcinoma cell lines with FH535. PLoS One. 9:e992722014. View Article : Google Scholar

20 

Wu MY, Liang RR, Chen K, Shen M, Tian YL, Li DM, Duan WM, Gui Q, Gong FR, Lian L, et al: FH535 inhibited metastasis and growth of pancreatic cancer cells. Onco Targets Ther. 8:1651–1670. 2015.PubMed/NCBI

21 

Iida J, Dorchak J, Lehman JR, Clancy R, Luo C, Chen Y, Somiari S, Ellsworth RE, Hu H, Mural RJ, et al: FH535 inhibited migration and growth of breast cancer cells. PLoS One. 7:e444182012. View Article : Google Scholar : PubMed/NCBI

22 

Galuppo R, Maynard E, Shah M, Daily MF, Chen C, Spear BT and Gedaly R: Synergistic inhibition of HCC and liver cancer stem cell proliferation by targeting RAS/RAF/MAPK and WNT/β-catenin pathways. Anticancer Res. 34:1709–1713. 2014.PubMed/NCBI

23 

Su H, Jin X, Zhang X, Zhao L, Lin B, Li L, Fei Z, Shen L, Fang Y, Pan H, et al: FH535 increases the radiosensitivity and reverses epithelial-to-mesenchymal transition of radioresistant esophageal cancer cell line KYSE-150R. J Transl Med. 13:1042015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 48 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S and Ishige N: FH535 suppresses the proliferation and motility of hepatocellular carcinoma cells. Int J Oncol 48: 110-114, 2016
APA
Tomizawa, M., Shinozaki, F., Motoyoshi, Y., Sugiyama, T., Yamamoto, S., & Ishige, N. (2016). FH535 suppresses the proliferation and motility of hepatocellular carcinoma cells. International Journal of Oncology, 48, 110-114. https://doi.org/10.3892/ijo.2015.3220
MLA
Tomizawa, M., Shinozaki, F., Motoyoshi, Y., Sugiyama, T., Yamamoto, S., Ishige, N."FH535 suppresses the proliferation and motility of hepatocellular carcinoma cells". International Journal of Oncology 48.1 (2016): 110-114.
Chicago
Tomizawa, M., Shinozaki, F., Motoyoshi, Y., Sugiyama, T., Yamamoto, S., Ishige, N."FH535 suppresses the proliferation and motility of hepatocellular carcinoma cells". International Journal of Oncology 48, no. 1 (2016): 110-114. https://doi.org/10.3892/ijo.2015.3220