EFLDO sensitizes liver cancer cells to TNFSF10‑induced apoptosis in a p53‑dependent manner

  • Authors:
    • Yanbo Qu
    • Zhixin Liao
    • Xinzhu Wang
    • Jing Zhang
    • Chao Liu
  • View Affiliations

  • Published online on: March 15, 2019     https://doi.org/10.3892/mmr.2019.10046
  • Pages: 3799-3806
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ent‑3α‑formylabieta‑8(14),13(15)‑dien‑16,12β‑olide (EFLDO) is a compound extracted from Euphorbia lunulata Bge exhibiting anti‑proliferative activity in vitro. In the present study, EFLDO was identified to sensitize HepG2 cells to tumor necrosis factor (TNF) superfamily member 10 (TNFSF10)‑induced apoptosis. Liver cancer cells were resistant to TNFSF10; however, EFLDO increased TNFSF10‑induced cancer cell viability inhibition and cell apoptosis induction as assessed by MTT assay and Annexin V‑fluorescein isothiocyanate (FITC)/propidium iodide assay, respectively. The western blotting results suggested that treatment with EFLDO increased TNFSF10‑induced upregulation of the protein expression levels of pro‑apoptotic proteins, including BCL2 associated agonist of cell death, BCL2 associated X, apoptosis regulator, caspase‑3 (CASP3) and CASP8. Furthermore, treatment with EFLDO increased TNFSF10‑mediated downregulation of the protein expression level of the anti‑apoptotic protein BCL2 apoptosis regulator. Notably, the increase in the activity of CASP3 was consistent with the western blotting results. Treatment with EFLDO sensitized liver cancer cells to TNFSF10, and apoptosis was induced via the upregulation of TNF receptor superfamily member 10a (TNFRSF10A) and TNFRSF10B in a tumor protein p53 (p53)‑dependent manner, as detected by reverse transcription‑quantitative polymerase chain reaction and western blot analyses. In addition, p53 was identified to be necessary for EFLDO‑induced sensitivity to TNFSF10, as assessed by western blotting and Annexin V‑FITC assay. Collectively, the present results suggested a novel mechanism underlying EFLDO function in liver cancer. Treatment with EFLDO was able to increase the antitumor effect of TNFSF10 in liver cancer cells in a p53‑dependent manner.

Introduction

Liver cancer is the fifth most common malignant tumor, and exhibits the second highest mortality rate among malignant tumors (13). Liver cancer has a high incidence rate, and accounts for ~55% of the cancer incidence worldwide (4). Additionally, the majority of patients with liver cancer at an advanced stage have a poor prognosis (5). Therefore, it is necessary to identify novel drugs to overcome the chemotherapeutic resistance of liver cancer cells and to develop novel treatments for patients with liver cancer.

Tumor necrosis factor (TNF) superfamily member 10 (TNFSF10) belongs to the TNF family, and is able to induce cell apoptosis in various types of tumor cells; however, its pro-apoptotic activity has not been detected in normal healthy cells (69). Cell apoptosis may be initiated by the interaction between TNFSF10 and TNF receptor superfamily member 10a (TNFRSF10A)/TNFRSF10B (10,11). Subsequently, caspase-8 (CASP8) is recruited to the death-inducing signaling complex and may cause the activation of CASP3, inducing cell apoptosis (1214). TNFSF10 resistance may be acquired via multiple molecular mechanisms, including the differential expression of death receptors and the increased expression of anti-apoptotic factors (15). Notably, treatments combining TNFSF10 with various anticancer drugs have been demonstrated to improve the anticancerogenic effects of a number of compounds (16).

Due to the antiproliferative activity exhibited by ent-3α-formylabieta-8(14),13(15)-dien-16,12β-olide (EFLDO) in MCF-7 cells and NCI-H460 cells (17), EFLDO has attracted the interest of various research studies (18). The aim of the present study was to investigate the antitumor activity of EFLDO and the potential molecular mechanisms underlying EFLDO function in promoting the inhibitory effects of TNFSF10 in liver cancer cells.

Materials and methods

Cell culture

HepG2 cells (19) and non-cancerous normal liver LO2 cells were purchased from The American Type Culture Collection (Manassas, VA, USA). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM; HyClone; GE Healthcare Life Sciences, Logan, UT, USA), supplemented with 10% (v/v) fetal bovine serum (HyClone; GE Healthcare Life Sciences) and 1% penicillin-streptomycin (HyClone; GE Healthcare Life Sciences) in an incubator with 5% CO2 at 37°C.

Drug treatments

Non-cancerous normal liver LO2 cells were treated with EFLDO (3 µM) and/or TNFSF10 (25 ng/ml, Thermo Fisher Scientific, Inc., Waltham, MA, USA), and cultured in an incubator with 5% CO2 at 37°C for 24 h. EFLDO was extracted from Euphorbia lunulata Bge as previously described (17). Hepatoblastoma cells were treated with EFLDO (3 µM), TNFSF10 (25 ng/ml), caspase inhibitor z-DEVD-FMK (working concentration, 40 µM; R&D Systems, Inc., Minneapolis, MN, USA; cat. no. FMK004) and/or the p53 inhibitor pifithrin-α (PFT-α; working concentrations, 20 and 40 µM; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and cultured in an incubator with 5% CO2 at 37°C for 24 h prior to further experimentation.

MTT assay

Hepatoblastoma HepG2 cells and non-cancerous normal liver LO2 cells were seeded into a 96-well plate at a density of 3×103 cells/well. Following addition of the indicated drug into the culture medium, the cells were incubated in an incubator with 5% CO2 at 37°C for 24 h. Subsequently, the cell viability was measured using the MTT assay at 570 nm. The MTT assay was performed using DMSO (Sigma-Aldrich; Merck KGaA) as the solvent to dissolve the formazan as previously described (20).

Annexin V-fluorescein isothiocyanate (FITC) assay

The Annexin V-FITC assay (Becton, Dickinson and Company, Franklin Lakes, NJ, USA) was performed to quantify the percentage of apoptotic cells, according to the manufacturer's protocol. HepG2 cells were incubated for 24 h, collected, washed twice with PBS and incubated in 1X binding buffer (provided in the Annexin V-FITC kit). Subsequently, Annexin V-FITC and propidium iodide (PI) were added to the cells and cells were incubated for 20 min at room temperature in the dark. Following incubation with Annexin V-FITC and PI, the cells were analyzed using a flow cytometer (Becton, Dickinson and Company) and the results were analyzed using the CellQuest Pro software (version 5.1; Becton, Dickinson and Company).

CASP3 Activity assay

CASP3 activity was measured using the CASP3 activity assay kit (Chemicon International; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. HepG2 cells were lysed using lysis buffer provided in the kit on ice for 10 min and centrifuged for 5 min at 12,000 × g at 4°C. Caspase substrate solution was added to the supernatant and incubated for 2 h at 37°C, subsequently, the activity was measured using an ELISA plate reader at 405 nm.

Western blot

HepG2 cells were incubated with DMEM for 48 h. Cells at a confluency of 70% were lysed using radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Haimen, China) containing a protease inhibitor cocktail (Roche Applied Science, Penzberg, Germany). The protein concentration was determined using Bradford Protein Assay kit (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Each lane was loaded with 0.05 µg total protein. Proteins were separated by 10% SDS-PAGE and transferred onto polyvinylidene fluoride (PVDF) membranes. The PVDF membranes were blocked with 5% non-fat milk for 1 h at room temperature, and incubated with the following primary antibodies: Anti-cleaved CASP3 (cat. no. 9661), cleaved CASP8 (cat. no. 9496), p53 (cat. no. 2527) (all 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA), GAPDH (cat. no. 59540), TNFRSF10A (cat. no. 32255), TNFRSF10B (cat. no. 166624), BCL2 associated agonist of cell death (BAD; cat. no. 8044), BCL2 associated X, apoptosis regulator apoptosis regulator (BAX; cat. no. 20067), BCL2 apoptosis regulator (BCL2; cat. no. 509) (all 1:1,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) overnight at 4°C. Horseradish peroxidase-conjugated goat anti-rabbit (cat. no. 7407) and goat anti-mouse (cat. no. 7076) secondary antibodies (all 1:10,000; Cell Signaling Technology, Inc.) were incubated with the membranes at room temperature for 2 h. Bands were developed with the electrochemiluminescence detection reagent (GE Healthcare, Chicago, IL, USA). GAPDH was used as the loading control and Image J software (version 1.8.0, National Institutes of Health, Bethesda, MD, USA) was used to compare the relative band intensities.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

HepG2 cells were incubated with DMEM for 48 h, and total RNA was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. mRNA was reverse-transcribed to cDNA using the PrimeScript™ RT reagent kit (Takara Bio, Inc., Otsu, Japan), according to the manufacturer's protocol. For cDNA synthesis, the RT reactions were incubated at 37°C for 1 h and at 70°C for 15 min. The sequences of the PCR primers used were as follows: TNFRSF10A forward: 5′-ACCTTCAAGTTTGTCGTCGTC-3′; TNFRSF10A reverse: 5′-CCAAAGGGCTATGTTCCCATT-3′; TNFRSF10B forward: 5′-GCCCCACAACAAAAGAGGTC-3′; TNFRSF10B reverse: 5′-AGGTCATTCCAGTGAGTGCTA-3′; GAPDH forward: 5′-ACAACTTTGGTATCGTGGAAGG-3′; GAPDH reverse: 5′-GCCATCACGCCACAGTTTC-3′. GAPDH gene was used as the reference gene. QuantiTect SYBR® Green RT-PCR kit (cat. no. 204245; Qiagen GmbH, Hilden, Germany) was used to analyze relative gene expression. The thermocycling conditions were as follows: Initial denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 60 sec, and a final elongation at 72°C for 15 sec.

Small interfering (siRNA) transfection

p53 siRNA (cat. no. sc-29435) and control siRNA (cat. no. sc-44238) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). For the siRNA transfection, HepG2 cells were seeded in 6-well plates at 3×105 cells/well and cultured in an incubator at 37°C overnight. On the following day, 0.67 µg siRNAs were mixed with Lipofectamine® RNAiMAX (Invitrogen; Thermo Fisher Scientific, Inc.) in Opti-MEM™ (Gibco; Thermo Fisher Scientific, Inc.) and incubated at room temperature for 15 min; subsequently, the mixtures were added into the culture medium and incubated at 37°C for 24 h prior to further experimentation.

Statistical analysis

All data were analyzed using GraphPad Prism 6.0 (GraphPad Software, Inc., La Jolla, CA, USA). Data are presented as the mean ± standard deviation. Comparisons between two groups and multiple groups were performed using Student's t-test and one-way analysis of variance followed by Newman-Keuls test, respectively. P<0.05 was considered to indicate a statistically significant difference. All the experiments were performed ≥3 times.

Results

Synergistic effects of EFLDO and TNFSF10 on HepG2 cell viability

The chemical structure of EFLDO is presented in Fig. 1A. The cytotoxicity of TNFSF10 at various concentrations (10, 20, 30, 40, 50 and 60 ng/ml) on LO2 and HepG2 cells was assessed using the MTT assay. The present MTT results suggested that TNFSF10 exhibited no obvious cytotoxicity in cells at concentrations ≤60 ng/ml (Fig. 1B).

Subsequently, the effects of various concentrations of EFLDO (2, 4, 6, 8 and 10 µM) and EFLDO (2, 4, 6, 8 and 10 µM) + TNFSF10 (25 ng/ml) on cell viability were detected. The present results suggested that the cell viability of HepG2 cells significantly decreased following treatment with EFLDO, in a concentration-dependent manner. Notably, the cell viability of normal liver LO2 cells was unaffected by treatment with EFLDO (Fig. 1C). Additionally, the cell viability of HepG2 cells was significantly decreased following the administration of EFLDO + TNFSF10 compared with administration of EFLDO alone. Furthermore, treatment with EFLDO (3 µM) + TNFSF10 (25 ng/ml) exhibited a significant antiproliferative activity (data not shown). Therefore, 25 ng/ml was selected at the working concentration of TNFSF10 and 3 µM was selected as the working concentration of EFLDO for further experiments.

EFLDO increases TNFSF10-induced cell apoptosis in HepG2 cells

The potential of EFLDO in sensitizing HepG2 cells to TNFSF10-induced apoptosis was investigated. Flow cytometry results suggested that the apoptotic rate of HepG2 cells following treatment with EFLDO + TNFSF10 increased compared with the control, TNFSF10 and EFLDO groups (Fig. 2A). The apoptotic rates for HepG2 cells in the TNFSF10 and EFLDO + TNFSF10 group were 9.57 and 60.7%, respectively (Fig. 2B). The present results suggested that the combination of EFLDO and TNFSF10 increased the effect of TNFSF10 in inducing the apoptosis of HepG2 cells.

EFLDO promotes TNFSF10-induced apoptosis of HepG2 cells in a caspase-dependent manner

Previous studies demonstrated that activation of CASP8 induced by TNFSF10 may activate CASP3 and CASP9 (1214). To investigate the molecular mechanisms underlying cell apoptosis induced by treatment with EFLDO + TNFSF10, the activity levels of CASP8 and 3 in HepG2 cells treated with TNFSF10 and/or EFLDO were detected. The present results suggested that proteolytic cleavage of proCASP8 and proCASP3 increased following treatment with EFLDO + TNFSF10 compared with the control, TNFSF10 and EFLDO groups (Fig. 3A). CASP3 activity in the EFLDO + TNFSF10 group increased to 7.0-fold compared with the control group, which was increased compared with TNFSF10 (0.7-fold) and EFLDO (1.4-fold) groups, and was significantly decreased following treatment with the caspase inhibitor z-DEVD-FMK (Fig. 3B). Subsequently, treatment with EFLDO + TNFSF10 significantly increased the protein expression levels of BCL2 associated agonist of cell death (BAD) and BAX, and decreased the protein expression level of BCL2 in HepG2 cells compared with the control, TNFSF10 and EFLDO groups (Fig. 3C). The present results suggested that EFLDO significantly sensitized HepG2 cells to TNFSF10-induced apoptosis in a caspase-dependent manner.

EFLDO sensitizes HepG2 cells to TNFSF10-induced cell apoptosis by increasing the expression levels of TNFRSF10A and TNFRSF10B

TNFRSF10A and TNFRSF10B (TNFSF10 receptors) were identified to be able to initiate the apoptotic cascade by binding to TNFSF10 (21). The present RT-qPCR and western blotting results suggested that treatment with EFLDO or EFLDO + TNFSF10 increased the mRNA (Fig. 4A) and protein expression levels (Fig. 4B) of TNFRSF10A and TNFRSF10B compared with the control group. A previous study observed that p53 may serve a role in regulating the functions of TNFRSF10A and TNFRSF10B (22). The present western blotting results suggested that, compared with the control group, the protein expression level of p53 was significantly increased following treatment with EFLDO or EFLDO + TNFSF10 (Fig. 4C). The effects of EFLDO on the protein expression levels of p53 and TNFRSF10B were inhibited by treatment with PFT-α (Fig. 4D). The present results suggested that treatment with EFLDO + TNFSF10 increased the protein expression levels of TNFRSF10A and TNFRSF10B by increasing the protein expression level of p53 in HepG2 cells.

EFLDO increases TNFSF10-induced cell apoptosis in a p53-dependent manner in HepG2 cells

To investigate the role of p53 on the effects of EFLDO in increasing TNFSF10-induced cell apoptosis, p53 was silenced by transfecting a siRNA targeting p53 (si-p53) in HepG2 cells (Fig. 5A). Knockdown of p53 significantly increased the activity of CASP3 in liver cancer cells (Fig. 5B). si-p53 significantly increased the apoptosis of HepG2 cells following treatment of TNFSF10 + EFLDO (Fig. 5C). The present results suggested that inhibition of p53 may increase EFLDO-induced TNFSF10 sensitivity in HepG2 cells.

Discussion

Previous studies in liver cancer demonstrated that the upregulation of antiapoptotic genes, and the increase in the protein expression levels of dysadherin and various cytokines, may serve roles in the negative outcome of certain therapies and in cancer metastasis (2325). Combinatorial treatments were identified to exhibit an increased effectiveness compared with single-agent chemotherapy in various types of cancer, including liver cancer (26). Nevertheless, the efficacy of chemotherapy to treat cancer may be decreased by the occurrence of drug resistance. Therefore, the identification of novel therapies is required to improve the treatment of patients with liver cancer.

Due to its strong antiproliferative activity in MCF-7 and NCI-H460 cells (17), EFLDO has attracted the interest of various research studies (18). However, the effects of the combination of EFLDO with additional anticancer drugs in liver cancer remain unknown.

The present results suggested that TNFSF10 did not induce cytotoxicity. By contrast, the combination of EFLDO and TNFSF10 presented a synergistic effect in suppressing cell viability of HepG2 cells; however, this combinatorial treatment did not affect the viability of non-cancerous normal liver LO2 cells. Subsequently, EFLDO was identified to sensitize HepG2 cells to TNFSF10-induced apoptosis. The present results suggested that apoptotic rates of HepG2 cells increased significantly following the cotreatment with TNFSF10 and EFLDO compared with single-treatment with TNSFS10 or EFLDO, indicating a potential synergistic effect between TNFSF10 and EFLDO in inducing apoptosis in liver cancer cells.

Furthermore, the activities of CASP8 and 3 following treatment with TNFSF10 and/or EFLDO were detected to investigate the molecular mechanisms underlying cell apoptosis induced by the combined treatment with EFLDO and TNFSF10. The present results suggested that liver cancer cell apoptosis was induced by EFLDO + TNFSF10 via the activation of CASP3 and CASP8. Treatment with EFLDO and TNSFS10 induced the upregulation of pro-apoptotic molecules, including BAD and BAX, and the downregulation of the anti-apoptotic factor BCL2. The present results suggested that EFLDO may promote TNFSF10-induced liver cancer cell apoptosis in a caspase-dependent manner.

p53 was previously identified to serve a role in regulating the function of TNFRSF10A and TNFRSF10B (22). p53 is able to inhibit cell proliferation by inducing cell apoptosis and cell cycle arrest in response to cellular stresses (2729). The present results suggested that the synergistic effects between EFLDO and TNFSF10 in increasing the protein expression level of TNFRSF10B may be due to the upregulation of p53, since the protein expression level of TNFRSF10B was decreased following treatment with the p53 inhibitor PFT-α. In addition, knockdown of p53 increased the activity of CASP3 and increased cell apoptosis in HepG2 cells, suggesting that p53 may regulate the synergistic effects between EFLDO and TNFSF10 in liver cancer cells. However, treatment with EFLDO and TNFSF10 led to an increase in the expression level of p53, in contrast with the results of p53 knockdown. p53 may be involved in various pathways regulating HepG2 cells, and further studies are required to confirm the present results.

In conclusion, combinatorial treatment with TNFSF10 and EFLDO induced cell apoptosis in HepG2 cells by promoting the caspase pathway, suggesting a potential therapeutic effect of the combination of EFLDO and TNFSF10 in treating liver cancer.

Acknowledgements

The authors would like to thank The Pharmaceutical Engineering Department of Southeast University for technical support of this study.

Funding

The project was funded by the priority academic program development of Jiangsu Higher Education Institutions (grant no. 1107047002), and the Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Guangxi Normal University; grant nos. CMEMR2016-B06 to ZXL).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YQ acquired the data, analyzed and interpreted the data, drafted the manuscript and revised it critically. ZL designed and conceived the study and revised the manuscript. XW, JZ and CL performed the experiments. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Takada Y, Tohyama T and Watanabe J: Biological markers of hepatocellular carcinoma for use as selection criteria in liver transplantation. J Hepatobiliary Pancreat Sci. 22:279–286. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Ling D, Xia H, Park W, Hackett MJ, Song C, Na K, Hui KM and Hyeon T: pH-sensitive nanoformulated triptolide as a targeted therapeutic strategy for hepatocellular carcinoma. ACS Nano. 8:8027–8039. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Kassebaum NJ, Bertozzi-Villa A, Coggeshall MS, Shackelford KA, Steiner C, Heuton KR, Gonzalez-Medina D, Barber R, Huynh C, Dicker D, et al: Global, regional, and national levels and causes of maternal mortality during 1990–2013: A systematic analysis for the Global burden of disease study 2013. Lancet. 384:980–1004. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Alsaied OA, Sangwan V, Banerjee S, Krosch TC, Chugh R, Saluja A, Vickers SM and Jensen EH: Sorafenib and triptolide as combination therapy for hepatocellular carcinoma. Surgery. 156:270–279. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Chen W, Zhang Y, Li Y, Zhang J, Zhang T, Fu B, Zhang Q and Jiang N: Constitutive expression of Wnt/β-catenin target genes promotes proliferation and invasion of liver cancer stem cells. Mol Med Rep. 13:3466–3474. 2016. View Article : Google Scholar : PubMed/NCBI

6 

He S, Chen Y, Chen XP, Zhang WG, Wang HP, Zhao YZ and Wang SF: Antitumor effects of soluble TRAIL in human hepatocellular carcinoma. J Huazhong Univ Sci Technolog Med Sci. 25:51–54. 2005. View Article : Google Scholar : PubMed/NCBI

7 

He SQ, Rehman H, Gong MG, Zhao YZ, Huang ZY, Li CH, Zhang WG and Chen XP: Inhibiting survivin expression enhances TRAIL-induced tumoricidal activity in human hepatocellular carcinoma via cell cycle arrest. Cancer Biol Ther. 6:1247–1257. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Zhang Y, Ma H, Zhang J, Liu S, Liu Y and Zheng D: AAV-mediated TRAIL gene expression driven by hTERT promoter suppressed human hepatocellular carcinoma growth in mice. Life Sci. 82:1154–1161. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Zhang Y, Qu ZH, Cui M, Guo C, Zhang XM, Ma CH and Sun WS: Combined endostatin and TRAIL gene transfer suppresses human hepatocellular carcinoma growth and angiogenesis in nude mice. Cancer Biol Ther. 8:466–473. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Zhang B, Shan H, Li D, Zhu KS, Jiang ZB and Huang MS: Cisplatin sensitizes human hepatocellular carcinoma cells, but not hepatocytes and mesenchymal stem cells, to TRAIL within a therapeutic window partially depending on the upregulation of DR5. Oncol Rep. 25:461–468. 2011.PubMed/NCBI

11 

Lee JY, Jung KH, Morgan MJ, Kang YR, Lee HS, Koo GB, Hong SS, Kwon SW and Kim YS: Sensitization of TRAIL-induced cell death by 20(S)-ginsenoside Rg3 via CHOP-mediated DR5 upregulation in human hepatocellular carcinoma cells. Mol Cancer Ther. 12:274–285. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Okano H, Shiraki K, Inoue H, Kawakita T, Yamanaka T, Deguchi M, Sugimoto K, Sakai T, Ohmori S, Fujikawa K, et al: Cellular FLICE/caspase-8-inhibitory protein as a principal regulator of cell death and survival in human hepatocellular carcinoma. Lab Invest. 83:1033–1043. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Ganten TM, Haas TL, Sykoraet J, Stahl H, Sprick MR, Fas SC, Krueger A, Weigand MA, Grosse-Wilde A, Stremmel W, et al: Enhanced caspase-8 recruitment to and activation at the DISC is critical for sensitisation of human hepatocellular carcinoma cells to TRAIL-induced apoptosis by chemotherapeutic drugs. Cell Death Differ. 11 (Suppl 1):S86–S96. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Su CL, Wu CJ, Chen FN, Wang BJ, Sheu SR and Won SJ: Supernatant of bacterial fermented soybean induces apoptosis of human hepatocellular carcinoma Hep 3B cells via activation of caspase 8 and mitochondria. Food Chem Toxicol. 45:303–314. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Youness ER, El Nemr M, Oraby FS, Ahmed NM, Moghni MA, Aly HF and Ahmed HH: Evaluation of apoptotic marker Bcl2, CD4+, human hepatocyte growth factor and metalloproteinase-9 as tumor markers for patients with hepatocellular carcinoma. Indian J Clin Biochem. 29:351–356. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Zhang L and Fang B: Mechanisms of resistance to TRAIL-induced apoptosis in cancer. Cancer Gene Ther. 12:228–237. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Liu C, Liao ZX, Liu SJ, Qu YB and Wang HS: Two new derivatives from Euphorbia lunulata bge and their anti-proliferation activities. Fitoterapia. 96:33–38. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Qu YB, Liao ZX, Liu C, Wang XZ and Zhang J: EFLDO induces apoptosis in hepatic cancer cells by caspase activation in vitro and suppresses tumor growth in vivo. Biomed Pharmacother. 100:407–416. 2018. View Article : Google Scholar : PubMed/NCBI

19 

López-Terrada D, Cheung SW, Finegold MJ and Knowles BB: HepG2 is a hepatoblastoma-derived cell line. Hum Pathol. 40:1512–1515. 2009. View Article : Google Scholar

20 

Twentyman P and Luscombe M: A study of some variables in a tetrazolium dye (MTT) based assay for cell growth and chemosensitivity. Br J Cancer. 56:279–285. 1987. View Article : Google Scholar : PubMed/NCBI

21 

Fossati S, Ghiso J and Rostagno A: TRAIL death receptors DR4 and DR5 mediate cerebral microvascular endothelial cell apoptosis induced by oligomeric Alzheimer's Aβ. Cell Death Dis. 3:e3212012. View Article : Google Scholar : PubMed/NCBI

22 

Tomasetti M, Andera L, Alleva R, Borghi B, Neuzil J and Procopio A: Alpha-tocopheryl succinate induces DR4 and DR5 expression by a p53-dependent route: Implication for sensitisation of resistant cancer cells to TRAIL apoptosis. FEBS Lett. 580:1925–1931. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, Hoey T, Gurney A, Huang EH, Simeone DM, et al: Phenotypic character-ization of human colorectal cancer stem cells. Proc Natl Acad Sci USA. 104:10158–10163. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Ino Y, Gotoh M, Sakamoto M, Tsukagoshi K and Hirohashi S: Dysadherin, a cancer-associated cell membrane glycoprotein, down-regulates E-cadherin and promotes metastasis. Proc Natl Acad Sci USA. 99:365–370. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Nam JS, Hirohashi S and Wakefield LM: Dysadherin: A new player in cancer progression. Cancer Lett. 255:161–169. 2007. View Article : Google Scholar : PubMed/NCBI

26 

El-Shemi AG, Ashshi AM, Na Y, Li YM, Al-Allaf FA, Oh E, Jung BK and Yun CO: Combined therapy with oncolytic adenoviruses encoding TRAIL and IL-12 genes markedly suppressed human hepatocellular carcinoma both in vitro and in an orthotopic transplanted mouse model. J Exp Clin Cancer Res. 35:742016. View Article : Google Scholar : PubMed/NCBI

27 

Chiu CT, Yeh TS, Hsu JC and Chen M: Expression of Bcl-2 family modulated through p53-dependent pathway in human hepatocellular carcinoma. Dig Dis Sci. 48:670–676. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Franklin DA, He Y, Leslie PL, Tikunov AP, Fenger N, Macdonal JM and Zhang Y: p53 coordinates DNA repair with nucleotide synthesis by suppressing PFKFB3 expression and promoting the pentose phosphate pathway. Sci Rep. 6:380672016. View Article : Google Scholar : PubMed/NCBI

29 

Romano FJ, Rossetti S, Conteduca V, Schepisi G, Cavaliere C, Di Franco R, La Mantia E, Castaldo L, Nocerino F, Ametrano G, et al: Role of DNA repair machinery and p53 in the testicular germ cell cancer: A review. Oncotarget. 7:85641–85649. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 19 Issue 5

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
Qu Y, Liao Z, Wang X, Zhang J and Liu C: EFLDO sensitizes liver cancer cells to TNFSF10‑induced apoptosis in a p53‑dependent manner. Mol Med Rep 19: 3799-3806, 2019
APA
Qu, Y., Liao, Z., Wang, X., Zhang, J., & Liu, C. (2019). EFLDO sensitizes liver cancer cells to TNFSF10‑induced apoptosis in a p53‑dependent manner. Molecular Medicine Reports, 19, 3799-3806. https://doi.org/10.3892/mmr.2019.10046
MLA
Qu, Y., Liao, Z., Wang, X., Zhang, J., Liu, C."EFLDO sensitizes liver cancer cells to TNFSF10‑induced apoptosis in a p53‑dependent manner". Molecular Medicine Reports 19.5 (2019): 3799-3806.
Chicago
Qu, Y., Liao, Z., Wang, X., Zhang, J., Liu, C."EFLDO sensitizes liver cancer cells to TNFSF10‑induced apoptosis in a p53‑dependent manner". Molecular Medicine Reports 19, no. 5 (2019): 3799-3806. https://doi.org/10.3892/mmr.2019.10046