Recombinant Escherichia coli Trx-JZTX-III represses the proliferation of mouse hepatocellular carcinoma cells through induction of cell cycle arrest

  • Authors:
    • Mei-Na Sun
    • Xue-Jiao Zhao
    • Han-Dong Zhao
    • Wei-Guang Zhang
    • Feng-Lan Li
    • Ming-Zi Chen
    • Hui Li
    • Guangchao Li
  • View Affiliations

  • Published online on: April 16, 2013     https://doi.org/10.3892/mmr.2013.1432
  • Pages: 1800-1804
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects of recombinant Escherichia coli (E. coli) Trx-jingzhaotoxin (JZTX)-III on cell growth in the mouse hepatocellular carcinoma (HCC) cell line Hepa1-6. The JZTX-III gene sequence was synthesized and cloned into the pET-32a(+) vector to construct the recombinant fusion protein Trx-JZTX-III, which was subsequently purified. Hepa1-6 cells were treated with 0 to 1,000-µg/ml concentrations of Trx-JZTX-III; this was demonstrated to affect cell viability, as determined by the 3-(4,5-dimethylthiazol‑2-yl)-2,5-diphenyltetra-zolium bromide (MTT) assay. The expression of the proliferating cell nuclear antigen (PCNA) protein was investigated using western blot analysis. A colony formation assay was used to determine Hepa1-6 cell proliferation, and the migration ability of cells was determined using a wound‑healing assay. Additionally, flow cytometry was employed to observe changes in the cell cycle. The MTT assay and quantification of PCNA expression indicated that recombinant E. coli Trx-JZTX-III significantly repressed the proliferation of Hepa1-6 cells. Colony formation and the migration of malignant cells was inhibited following treatment with recombinant E. coli Trx-JZTX-III. Flow cytometry showed that recombinant E. coli Trx-JZTX-III induced G0/G1 cell cycle arrest. In conclusion, recombinant E. coli Trx-JZTX-III functions as a tumor suppressor drug in mouse HCC and its underlying mechanism may involve the induction of G0/G1 cell cycle arrest.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common malignancies and the third most common cause of cancer-related mortality worldwide (1). The treatment of HCC is currently challenging and may remain difficult in the future (2). Although a few novel methods have been applied (3), pharmacotherapy remains the routine treatment for HCC.

Venom from natural toxins has been considered for development into anticancer drugs (4) and the value of their potential biomedical application is promising. Animal toxins have also been identified to have antitumor activity, including bee (5), snake (6) and scorpion venom (7). Recently, scorpion venom was shown to induce cell apoptosis (8) and snake toxin was demonstrated to inhibit the proliferation of PA-1 and SK-OV3 cells (human ovarian cancer cells) (9).

Spider venom isolated from Lycosa singorensis has exhibited an ability to inhibit cancer cell growth in vitro(10). Spider venom is a liquid with offensive, defensive and digestive functions. Furthermore, different species of spiders vary in toxicity; homology is limited even in primary structures. There are a large number of spider species in China; Chilobrachy jingzhao is one of these and is mainly located in the Guangxi Province of China, with the following classification: Phylum, Arthropoda; class, Arachnids; order, Araneae; family, Theraphosidae. Chilobrachy jingzhaotoxin (JZTX)-III is a common biological resource that may play a role in directly killing tumor cells. Therefore, JZTX-III has the potential to be used in the development of anticancer drugs and has received increasing attention from pharmaceutical scientists and molecular biologists.

The present study aimed to investigate the potential use of recombinant Escherichia coli (E. coli) Trx-JZTX-III in the treatment of HCC by examining the proliferation of Hepa1-6 cells, a HCC cell line from mice, following treatment with or without recombinant E. coli Trx-JZTX-III.

Materials and methods

Preparation of recombinant E. coli Trx-JZTX-III

In this study, genetic engineering was utilized to inhibit the tetrodotoxin-resistant sodium channel. JZTX-III was inserted in the prokaryotic expression vector pET-32a(+) to create a recombination plasmid which expressed the fusion protein Trx-JZTX-III in E. coli, using Trx as a protein tag. The recombinant E. coli Trx-JZTX-III was freeze-dried and stored at −80°C until use. The study was approved by the ethics committee of Harbin Medical University, Harbin, China.

Cell culture

The HCC cell line Hepa1-6 was obtained from the Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai Branch Cell Bank, Shanghai, China). The cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Carlsbad, CA, USA) containing 10% (v/v) fetal bovine serum (FBS; Gibco), 100 IU/ml penicillin and 100 μg/ml streptomycin at 37°C in a humidified atmosphere containing 5% CO2.

MTT assay

The growth and viability of Hepa1-6 cells were evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra-zolium bromide (MTT; Sigma, St. Louis, MO, USA) assay. The cells were plated into 96-well microtiter plates at a density of 5×103 cells/well. Following incubation for 24 h, recombinant E. coli Trx-JZTX-III was serially diluted to various concentrations (1,000, 900, 800, 700, 600, 500, 400, 300, 200 and 100 μg/ml) and added to each well. A control group was treated with phosphate-buffered saline (PBS; Gibco) alone. After incubation for 24 and 48 h, 200 μl MTT (5 mg/ml) solution was added to each well and cultured for 4 h at 37°C. Formazan crystals were solubilized using 150 μl DMSO (Sigma) and plates were agitated for 10 min. The absorbance was measured at 490 nm and a colorimetric MTT assay was performed to investigate cell growth. All the experiments were repeated three times.

Western blot analysis

Hepa1-6 cells (5×105) were seeded in 6-well plates and treated with 0, 600, 800 and 1,000 μg/ml recombinant E. coli Trx-JZTX-III for 48 h, and proteins were then extracted from each group of cells. The proteins were separated by 10% SDS-PAGE and subsequently transferred to PVDF membranes. Nonfat milk (5%) in TBST was used to block the membranes at room temperature for 2 h. The membranes were then incubated with rabbit anti-proliferating cell nuclear antigen (PCNA; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) or mouse anti-β-actin antibodies (Santa Cruz Biotechnology, Inc.) at 4°C overnight. Next, the membranes were incubated with anti-rabbit or anti-mouse secondary antibodies for 2 h. Finally, the membranes were washed three times with TBST and exposed to X-ray film.

Colony formation assay

A colony formation assay was conducted by plating Hepa1-6 cells into 6-well plates (200 cells/plate). Following a 24-h incubation, recombinant E. coli Trx-JZTX-III at various concentrations (600, 800 and 1,000 μg/ml) was added; the control group received no recombinant E. coli Trx-JZTX-III. After 2 weeks, the cells were stained with Giemsa. Colonies were counted only when a single clone contained >50 cells.

Cell migration assay

Cell migration was determined using a wound-healing assay. Hepa1-6 cells were seeded at a density of 5×105 cells/6-well plate in DMEM containing 10% FBS. A scratch wound was created on the confluent cell monolayer using a 200-μl pipette tip, and then the cells were treated with various concentrations of recombinant E. coli Trx-JZTX-III (0, 600, 800 and 1,000 μg/ml). The wounded areas were observed and images were captured using a microscope at 0 and 48 h after scraping.

Flow cytometry

Hepa1-6 cells were seeded into 6-well plates in DMEM containing 10% FBS and treated with various concentrations of recombinant E. coli Trx-JZTX-III (0, 600, 800 and 1,000 μg/m) for 48 h. The cells were harvested, washed in cold PBS and then fixed in 75% alcohol at 4°C for ≥12 h. The fixed cells were resuspended in PBS containing 50 g/l RNase A and 50 mg/l propidium iodide (PI) for 30 min, and analyzed using flow cytometry (Becton Dickinson, USA).

Statistical analysis

The data are presented as the mean ± SEM. P<0.05 was considered to indicate a statistically significant difference. The results were analyzed by Student’s t-test using SPSS version 10.0 (SPSS, Inc., Chicago, IL, USA).

Results

Recombinant E. coli Trx-JZTX-III inhibits Hepa1-6 cell proliferation

Hepa1-6 cells were exposed to various concentrations of E. coli Trx-JZTX-III (1,000, 900, 800, 700, 600, 500, 400, 300, 200 and 100 μg/ml). Growth inhibition was most significant at a treatment time of >48 h (Fig. 1). Therefore, we further investigated the in vitro inhibitory effects of recombinant E. coli Trx-JZTX-III using a treatment time of 48 h (P<0.05).

In order to obtain additional evidence, western blot analysis was performed; PCNA was used as a reporter for proliferation. Hepa1-6 cells were treated with recombinant E. coli Trx-JZTX-III for 48 h and the expression of PCNA protein was determined using western blot analysis. The expression of PCNA was significantly lower with 600, 800 and 1,000 μg/ml E. coli Trx-JZTX-III treatment compared with cells in the control group (Fig. 2; P<0.05).

Recombinant E. coli Trx-JZTX-III represses colony formation and cell migration

A colony formation assay was used to assess the colony-forming ability of Hepa1-6 cells following treatment with or without recombinant E. coli Trx-JZTX-III. Compared with cells in the control group (Fig. 3D), cells treated with 600 (Fig. 3C), 800 (Fig. 3B) and 1,000 μg/ml (Fig. 3A) recombinant E. coli Trx-JZTX-III formed fewer and smaller colonies.

Compared with the control group (Fig. 4A), a significant decrease in Hepa1-6 cell migration was treated with recombinant E.coli Trx-JZTX-III (Fig. 4B–D).

Recombinant E. coli Trx-JZTX-III induces Hepa1-6 G0/G1 cell cycle arrest

As shown in Fig. 5, cells treated with recombinant E. coli Trx-JZTX-III at concentrations of 600 (Fig. 5C), 800 (Fig. 5B) or 1,000 μg/ml (Fig. 5A) exhibited a significantly increased cell population in the G0/G1 phase when compared with the control group (Fig. 5D). However, cells treated with recombinant E. coli Trx-JZTX-III demonstrated a significantly decreased cell population in the G2/M or S phases (P<0.01; Table I).

Table I

Cell cycle distribution of Hepa1-6 cells (mean ± SEM).

Table I

Cell cycle distribution of Hepa1-6 cells (mean ± SEM).

Cell cycle distribution (%)

Group G0/G1aSa G2/Ma
Trx-JZTX-III (μg/ml)
 100072.2±3.513.96±1.928.4±1.3
 80045.7±2.029.45±3.754.3±2.4
 60042.1±2.931.18±4.257.9±2.0
Control40.8±1.935.10±1.459.24±1.2

a P<0.05 compared with the control group, indicating a significant increase.

{ label (or @symbol) needed for fn[@id='tfn2-mmr-07-06-1800'] } JZTX-III, jingzhaotoxin-III.

Discussion

Currently, several types of JZTXs are derived from the spider Chilobrachys jingzhao, including JZTX-II (11), -IX (12), -V (13), -XI (14), -IV (15) and -XIII (16). JZTX-III (molecular weight, 3919.3 Da) is a peptide toxin, containing 36 residues and three pairs of intracellular disulfide bridges (I–IV, II–V, and III–VI) (17).

The venom from this spider has been shown to induce apoptosis in the myelogenous leukemia K562 cell line (18) and in MCF-7 cells (19). Spider venom has been demonstrated to be a novel tumor suppressor drug in HeLa cells (20) and was also shown to inhibit the proliferation of HepG2 cells (21) and induce BEL-7402 cell apoptosis (22) in HCC.

Spider venom is usually obtained by direct extraction and artificial chemical synthesis. In this study, our aim was to obtain a significant amount of recombinant fusion protein of Trx-JZTX-III. Compared with natural JXTX-III, recombinant E. coli Trx-JZTX-III has advantages with regard to mass production and cost efficiency.

The MTT assay and quantification of PCNA expression indicated that recombinant E. coli Trx-JZTX-III significantly repressed the proliferation of Hepa1-6 cells. Furthermore, the colony formation ability and migration of the malignant cells were inhibited following treatment with recombinant E. coli Trx-JZTX-III. Thus, recombinant E. coli Trx-JZTX-III functioned as a tumor suppressor drug.

To explain the underlying mechanism of recombinant E. coli Trx-JZTX-III in suppressing Hepa1-6 cell proliferation, we investigated the cell cycle of Hepa1-6 cells following treatment with or without recombinant E. coli Trx-JZTX-III. Recombinant E. coli Trx-JZTX-III was shown to induce G0/G1 cell cycle arrest; this may be one of the mechanisms that mediate the antitumor function of recombinant E. coli Trx-JZTX-III.

The present study demonstrated that recombinant E. coli Trx-JZTX-III functioned as a tumor suppressor drug in mouse HCC cell proliferation, potentially via the induction of G0/G1 cell cycle arrest. These results suggest that the antitumor activity of recombinant E. coli Trx-JZTX-III may be associated with the proliferation of the mouse cell line Hepa1-6. However, the effect of recombinant E. coli Trx-JZTX-III on tumors in vivo remains unknown. Future investigation with regard to the association between recombinant E. coli Trx-JZTX-III and human hepatoma cells is required to identify the specific molecules which are involved in the antitumor activity and synthesis of recombinant E. coli Trx-JZTX-III. Results of the present study suggest that recombinant E. coli Trx-JZTX-III may be a promising candidate for further development as a therapeutic agent due to its anticancer efficacy. In conclusion, recombinant E. coli Trx-JZTX-III represents a potentially important tool for the development of novel drugs and antitumor agents in the treatment of HCC.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (nos. 81172616, 30771863 and 30910007).

References

1 

Schütte K, Bornschein J and Malfertheiner P: Hepatocellular carcinoma - epidemiological trends and risk factors. Dig Dis. 27:80–92. 2009.PubMed/NCBI

2 

Taieb J, Barbare JC, Boussaha T, et al: Management of hepatocellular carcinoma. Where are we now? What’s next? Bull Cancer. 96:19–34. 2009.(In French).

3 

May BJ, Murthy R and Madoff DC: What’s new in transarterial therapies for hepatocellular carcinoma? Gastrointest Cancer Res. 5(Suppl 1): S14–S19. 2012.

4 

Harvey A: Strategies for discovering drugs from previously unexplored natural products. Drug Discov Today. 5:294–300. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Orsolić N, Sver L, Verstovsek S, Terzić S and Basić I: Inhibition of mammary carcinoma cell proliferation in vitro and tumor growth in vivo by bee venom. Toxicon. 41:861–870. 2003.PubMed/NCBI

6 

de Carvalho DD, Schmitmeier S, Novello JC and Markland FS: Effect of BJcuL (a lectin from the venom of the snake Bothrops jararacussu) on adhesion and growth of tumor and endothelial cells. Toxicon. 39:1471–1476. 2001.PubMed/NCBI

7 

Liu YF, Ma RL, Wang SL, et al: Expression of an antitumor-analgesic peptide from the venom of Chinese scorpion Buthus martensii Karsch in Escherichia coli. Protein Expr Purif. 27:253–258. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Ning YN, Zhang WD and Wu LC: Study on the mechanism of polypeptide extract from scorpion venom to promote the restraint of cyclophosphamide on Lewis lung cancer. Zhongguo Zhong Xi Yi Jie He Za Zhi. 32:537–542. 2012.(In Chinese).

9 

Song JK, Jo MR, Park MH, et al: Cell growth inhibition and induction of apoptosis by snake venom toxin in ovarian cancer cell via inactivation of nuclear factor κB and signal transducer and activator of transcription 3. Arch Pharm Res. 35:867–876. 2012.PubMed/NCBI

10 

Liu Z, Deng M, Xiang J, et al: A novel spider peptide toxin suppresses tumor growth through dual signaling pathways. Curr Mol Med. 12:1350–1360. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Wang M, Liu Q, Luo H, et al: Jingzhaotoxin-II, a novel tarantula toxin preferentially targets rat cardiac sodium channel. Biochem Pharmacol. 76:1716–1727. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Deng M, Kuang F, Sun Z, et al: Jingzhaotoxin-IX, a novel gating modifier of both sodium and potassium channels from Chinese tarantula Chilobrachys jingzhao. Neuropharmacology. 57:77–87. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Zeng X, Deng M, Lin Y, Yuan C, Pi J and Liang S: Isolation and characterization of Jingzhaotoxin-V, a novel neurotoxin from the venom of the spider Chilobrachys jingzhao. Toxicon. 49:388–399. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Liao Z, Yuan C, Deng M, et al: Solution structure and functional characterization of jingzhaotoxin-XI: a novel gating modifier of both potassium and sodium channels. Biochemistry. 45:15591–15600. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Wang M, Diao J, Li J, et al: JZTX-IV, a unique acidic sodium channel toxin isolated from the spider Chilobrachys jingzhao. Toxicon. 52:871–880. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Yuan C, Liu Z, Hu W, Gao T and Liang S: JZTX-XIII, a Kv channel gating modifier toxin from Chinese tarantula Chilobrachys jingzhao. Toxicon. 59:265–271. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Xiao Y, Tang J, Yang Y, et al: Jingzhaotoxin-III, a novel spider toxin inhibiting activation of voltage-gated sodium channel in rat cardiac myocytes. J Biol Chem. 279:26220–26226. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Liu Z, Zhao Y, Li J, et al: The venom of the spider Macrothele raveni induces apoptosis in the myelogenous leukemia K562 cell line. Leuk Res. 36:1063–1066. 2012.PubMed/NCBI

19 

Gao L, Yu S, Wu Y and Shan B: Effect of spider venom on cell apoptosis and necrosis rates in MCF-7 cells. DNA Cell Biol. 26:485–489. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Gao L, Shan BE, Chen J, Liu JH, Song DX and Zhu BC: Effects of spider Macrothele raven venom on cell proliferation and cytotoxicity in HeLa cells. Acta Pharmacol Sin. 26:369–376. 2005.

21 

Gao L, Shen JB, Sun J and Shan BE: Effect of the venom of the spider Macrothele raveni on the expression of p21 gene in HepG2 cells. Sheng Li Xue Bao. 59:58–62. 2007.PubMed/NCBI

22 

Gao L, Feng W, Shan BE and Zhu BC: Inhibitory effect of the venom of spider Macrothele raveni on proliferation of human hepatocellular carcinoma cell line BEL-7402 and its mechanism. Ai Zheng. 24:812–816. 2005.(In Chinese).

Related Articles

Journal Cover

June 2013
Volume 7 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun M, Zhao X, Zhao H, Zhang W, Li F, Chen M, Li H and Li G: Recombinant Escherichia coli Trx-JZTX-III represses the proliferation of mouse hepatocellular carcinoma cells through induction of cell cycle arrest. Mol Med Rep 7: 1800-1804, 2013
APA
Sun, M., Zhao, X., Zhao, H., Zhang, W., Li, F., Chen, M. ... Li, G. (2013). Recombinant Escherichia coli Trx-JZTX-III represses the proliferation of mouse hepatocellular carcinoma cells through induction of cell cycle arrest. Molecular Medicine Reports, 7, 1800-1804. https://doi.org/10.3892/mmr.2013.1432
MLA
Sun, M., Zhao, X., Zhao, H., Zhang, W., Li, F., Chen, M., Li, H., Li, G."Recombinant Escherichia coli Trx-JZTX-III represses the proliferation of mouse hepatocellular carcinoma cells through induction of cell cycle arrest". Molecular Medicine Reports 7.6 (2013): 1800-1804.
Chicago
Sun, M., Zhao, X., Zhao, H., Zhang, W., Li, F., Chen, M., Li, H., Li, G."Recombinant Escherichia coli Trx-JZTX-III represses the proliferation of mouse hepatocellular carcinoma cells through induction of cell cycle arrest". Molecular Medicine Reports 7, no. 6 (2013): 1800-1804. https://doi.org/10.3892/mmr.2013.1432