Glycogen synthase kinase‑3β regulates tumor necrosis factor‑related apoptosis inducing ligand (TRAIL)‑induced apoptosis via the NF‑κB pathway in hepatocellular carcinoma

  • Authors:
    • Kai Fu
    • Huazheng Pan
    • Shihai Liu
    • Jing Lv
    • Zhaojun Wan
    • Jiao Li
    • Qing Sun
    • Jun Liang
  • View Affiliations

  • Published online on: October 13, 2015     https://doi.org/10.3892/ol.2015.3803
  • Pages: 3557-3564
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor necrosis factor (TNF)‑related apoptosis‑inducing ligand (TRAIL) is known for its ability to selectively induce apoptosis in malignant cells. However, human hepatocellular carcinoma (HCC) cells display resistance to TRAIL‑induced cell death. The present study investigated whether TRAIL‑induced apoptosis in HCC cells was enhanced by the administration of an inhibitor of glycogen synthase kinase‑3β (GSK‑3β) or by short hairpin RNA‑mediated inhibition of GSK‑3β. The results of the current study demonstrated that inhibition of GSK‑3β significantly impairs the expression of the nuclear factor‑κB (NF‑κB) target genes Bcl‑xL and clAP2 in HCC cells (P<0.05). This indicates that GSK‑3β may regulate NF‑κB target genes involved in cell survival. Furthermore, knockdown of Bcl‑xL significantly enhanced the sensitizing effect of GSK‑3β inhibitor on TRAIL‑induced apoptosis (P<0.05). Overall, the present study provides a rationale for further exploration of GSK‑3β inhibition combined with TRAIL as a novel treatment for HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common types of malignant primary liver cancer and is the third leading cause of cancer-related mortality worldwide (1). Although surgery remains the preferred therapeutic strategy for HCC, tumor size, hepatic functional reserve and/or portal hypertension may all limit the extent of surgical resection (2). Chemotherapy and internal radiation therapy are also used to treat liver cancer (3), however, both can lead to the damage of tissues and organs unaffected by cancer. The prognosis of HCC is poor due to the development of resistance to current chemotherapy regimens through the downregulation of various signaling pathways; in particular, those that control cell proliferation and survival, such as nuclear factor κB (NF-κB) (4). Thus, the development of novel, effective therapeutic strategies for HCC are required to improve the prognosis of this disease.

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) belongs to the TNF superfamily (5) and possesses a number of anti-cancer properties (6,7). For example, TRAIL can induce apoptosis in tumor cells by binding to the plasma membrane death receptors (DRs) TRAIL-R1 (DR4) and TRAIL-R2 (DR5) (8). Therefore, TRAIL is a potential candidate for cancer treatment (9). However, HCC cells are intrinsically resistant to TRAIL-induced cell death (10). This resistance to TRAIL is a major clinical challenge that leads to failure of treatment, poor prognosis and reduced survival of patients with HCC (10). Previous studies have demonstrated that TRAIL can activate the NF-κB signaling pathway, which activates genes that encode various key anti-apoptotic proteins, such as B-cell lymphoma-extra large (Bcl-xL) and inhibitor of apoptosis proteins (IAPs). These proteins contribute to TRAIL resistance (10,11). Thus, overcoming NF-κB-associated survival signals may enhance the antitumor effect of TRAIL in HCC cells.

Glycogen synthase kinase-3 (GSK-3) is a multifunctional serine/threonine protein kinase that participates in numerous cellular processes, including protein synthesis, glycogen metabolism, mitosis and apoptosis. Additionally, GSK-3 is involved in various signaling pathways, such as the Wnt/β-catenin signaling pathway (12). Two major GSK-3 isoforms (GSK-α and GSK-3β) have been identified in mammals; they are encoded by distinct genes and perform different functions (13,14). Mice with a homozygous deletion of the GSK-3β gene experience massive hepatocyte apoptosis during embryogenesis, leading to premature death (15). Furthermore, previous studies have demonstrated that GSK-3β is important in cell survival through its ability to regulate the NF-κB signaling pathway in hepatocytes (16).

In consideration of the role of NF-κB target genes on TRAIL-induced apoptosis, the present study aims to evaluate the effect of GSK-3β on TRAIL-induced cell death and examine the mechanism by which GSK-3β inhibition sensitizes HCC cells to TRAIL-induced apoptosis.

Materials and methods

Cell culture

HL7702, SMMC7721, HuH-7, HuH-6 and HepG2 Human HCC cell lines were purchased from the American Type Culture Collection (Manassas, VA, USA). Cells were grown at 37°C in a 5% CO2 humidified atmosphere, and cultured as a monolayer in RPMI-1640 medium (HyClone, Logan, UT, USA) supplemented with 100 U/ml penicillin, 100 µg/ml streptomycin, 2 mmol/l glutamine and 10% fetal bovine serum. HCC cell growth was observed and recorded regularly.

Reagents and antibodies

Annexin V-R-phycoerythrin (PE) and propidium iodide (PI) were purchased from Invitrogen Life Technologies (Carlsbad, CA, USA), and GSK-3β inhibitor (SB216763) was purchased from Sigma-Aldrich (St. Louis, MO, USA). Rabbit anti-human monoclonal antibodies raised against GSK-3β (cat no. 12456; 1:1,000), TRAIL (cat. no. 3219; 1:500), GAPDH (cat. no. 2118; 1:2,000) and β-catenin (cat no. 8480; 1:1,000) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Rabbit anti-human monoclonal antibodies raised against Bcl-xL (cat no. ab2568; 1:1,000) and cellular IAP2 (cIAP2; cat no. ab32059; 1:1,000) were purchased from Abcam (Cambridge, UK). Recombinant Ad5.TRAIL, short hairpin (sh) GSK-3β, shBcl-xL and short shcIAP2 constructs were obtained from the Central Laboratory, The Affiliated Hospital, Qingdao University (Qingdao, China).

GSK-3β inhibitor (SB216763)

SB216763 is potent and selective ATP-competitive GSK-3 inhibitor. It is equally effective at inhibiting human GSK-3α and GSK-3β (17). HepG2 and HuH-7 cells (1×105 cells/well) were seeded in 96-well plates and incubated with Dulbecco's modified Eagle's medium (DMEM; HyClone) for 24 h. The cells were then pretreated with 10 µM SB216763 in 10% fetal bovine serum and 1 µmol/l DMEM for 24 h at 37°C in an atmosphere of 5% CO2.

Construction of the pGenesil-GSK-3β short interfering (si)RNA

Three pairs of shRNAs were used for screening to obtain the most effective downregulation of the gene fragment. Non-targeting siRNA (Wuhan Genesil Biotechnology Co., Ltd., Wuhan, China) was used as the negative control. Using siRNA design software (http://sirna.wi.mit.edu/), the following three coding regions corresponding to the target GSK-3β were selected as siRNA target sequences in the enzyme site of the GFP-tagged pGenesil-1 vector (pGenesil-1-GFP; Wuhan Genesil Biotechnology Co., Ltd.): 5′-ACTGGTCGCCAT-CAAGAAA-3′ (471–489 bp); 5′-GAAAGCTA-GATCACTGTAA-3′ (536–554 bp); and 5′-GCCACT-GATTATACCTCTA-3′ (922–940 bp). In addition, an unrelated sequence was designed for the negative control: 5′-TTCTCCGAACGTCTCACGT-3′. Two oligonucleotides encoding the target shRNA and its complementary sequence were annealed and ligated, and the pGenesil-1-GFP vector was cleaved by BamHI and HindIII (Wuhan Genesil Biotechnology Co., Ltd.). Then the products of both reactions were recovered and purified. The shRNA oligonucleotide fragment and the pGenesil-1-GFP vector were ligated using T4 ligase (Takara Bio, Inc., Shiga, Japan), and the recombinant plasmids were termed pGenesil-GFP GSK-3β shRNA 1–3. Next, pGenesil-GFP-GSK-3β shRNAs 1–3 (8 µg) were transfected into HepG2 cells, respectively, using 20 µl Lipofectamine 2000 reagent (Invitrogen Life Technologies), according to the manufacturer's instructions. The most effective shRNA expression cassette, p-Genesil-GSK-3β shRNA 1, was selected as described previously (18), and excised from the pGenesil-1-GFP vector by BamHI and MluI, and ligated into a pLV-mCMV-ZsGreen-PGK-puro shuttle vector (Wuhan Genesil Biotechnology Co., Ltd.), termed pLV-GSK-3β shRNA. Plasmids were purified with a MaxPrep kit (Wuhan Genesil Biotechnology Co., Ltd.) and successful ligations were verified by sequencing (Sangon Biotech Co., Ltd., Shanghai, China). Recombinant lentiviral vectors were produced by co-transfecting HepG2 cells with the lentiviral expression plasmid and packaging plasmids using the calcium phosphate method. Briefly, 8 µg shRNA plasmid DNA, 5 µg lentiviral helper-1, 6 µg lentiviral helper-2 plasmids were mixed with sterile ddH2O to a final volume of 450 µl and mixed with 50 µl of 2.5 M CaCl2. Following transfection, infectious media containing shRNA lentiviral vectors was harvested at 48 and 72 h.

Cell viability assay

The effect of shGSK-3β and shRNA on HCC cell viability was measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The HepG2 cell line, which is derived from well-differentiated hepatocellular carcinoma, can be grown successfully on a large scale, secretes numerous plasma proteins, and is composed of adherent epithelial-like cells that grow as monolayers and in small aggregates; therefore, these cells were used to investigate cell viability. HepG2 cells (5×103 cells/well) were seeded in 96-well plates in triplicate, and infected with shGSK-3β and negative control shRNA at a multiplicity of infection (MOI) of 10. Control cells were treated with DMEM. After growing for 24, 48 and 72 h at 37°C in a 5% CO2 atmosphere, 20 µl of 5 mg/ml MTT [in phosphate-buffered saline (PBS)] was added to each well and continually incubated for 4 h at 37°C in a CO2 incubator. The formazan granules obtained from the cells were dissolved in 150 µl dimethylsulfoxide for 10 min. Cell viability was measured in terms of the optical density using an enzyme-linked immune detector (Multiskan GO; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at a wavelength of 570 nm. Each cell viability assay was performed in triplicate.

Flow cytometric analysis

HuH-7 cells were treated with 10 µM GSK-3β inhibitor, Ad5.TRAIL, shBcl-xL and shcIAP2 alone or in combination at an MOI of 10 for 48 h at 37°C in a 5% CO2 atmosphere. Adherent and suspended cells were collected, washed in PBS and then suspended in Annexin binding buffer (Immunotech, Marseille, France). Subsequently, cells were stained with Annexin V-PE or PI to distinguish between apoptotic and dead cells. All steps were conducted in accordance with the Annexin binding buffer manufacturer's instructions. Finally, the stained cells were analyzed by flow cytometry using a FC 500 MPL Flow Cytometer (Becton Dickinson, San Jose, CA, USA).

Western blot analysis

Cell samples were lysed in ice-cold lysis buffer (Beyotime Institute of Biotechnology) with 1% phenylmethylsulfonyl fluoride for 30 min and then centrifuged at 10,000 × g for 20 min at 4°C. The protein concentration of the resulting supernatant was determined using a bicinchoninic acid protein assay kit (Beyotime Institute of Biotechnology). Proteins (50 µg) were separated by 12% SDS-PAGE electrophoresis (Beyotime Institute of Biotechnology) and subsequently transferred to polyvinylidene difluoride membranes. Membranes were blocked with 5% non-fat dry milk in Tris-buffered saline/Tween-20 (0.05%, v/v) for 2 h at room temperature and incubated overnight at 4°C with the rabbit TRAIL, GSK-3β, β-catenin, Bcl-xL, cIAP2 and GAPDH primary antibodies. The blots were washed and incubated with a horseradish peroxidase-conjugated secondary antibody (Agilent Technologies, Santa Clara, CA, USA) and developed with the chemiluminescent substrate ECL Plus (Pierce Biotechnology, Inc., Rockford, IL, USA). An autoradiograph was obtained, and protein levels were measured using a Fluor-S scanner for grayscale determination and Quantity One software for analysis (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Quantification by reverse transcription quantitative-polymerase chain reaction (RT-qPCR)

Transfected and untreated cells were collected and washed with PBS. Total RNA was extracted from the cells using TRIzol® reagent (Invitrogen Life Technologies) and complementary (c)DNA (1 µl) was generated using a PrimeScript® RT reagent kit (Takara, Chiga, Japan) at a total volume of 20 µl, according to the manufacturer's instructions. This cDNA was then used in each amplification reaction. The primers used were as follows: Forward, 5′-CTGGGCTACACTGAGCACC-3′ and reverse, 5′-AAGTGGTCGTTGAGGGCAATG-3′ for GSK-3β; and forward, 5′-AGACGCTCCCTGTGATTTATGT-3′ and reverse, 5′-CCGATGGCAGATTCCAAAGG-3′ for GAPDH. Reactions were performed using SYBR® Premix Ex Taq™ (Takara) under the following PCR conditions: 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 30 sec. Amplification specificity was also confirmed by generating a melting curve for each sample. The expression of proteins was assessed by normalization of the cycle threshold (Ct) of these genes to that of the housekeeping gene GAPDH. A Ct value was obtained from each amplification curve using the software provided by the manufacturer (Roche Diagnostics GmbH, Mannheim, Germany).

Statistical analysis

Student's t-test and one-way analysis of variance were performed for continuous variables. The χ2 or Fisher's exact test were used for categorical variables. Error bars in all cases represent the standard error of the mean. All statistical analyses were performed using SAS 9.0 software (SAS Institute, Inc., Cary, NC, USA) and, using two-sided tests, P<0.05 indicated a statistically significant difference.

Results

GSK-3β downregulation inhibits the proliferation of HCC cells

Previous studies have reported that direct downregulation of GSK-3 protein expression inhibits glioma cell growth (19). GSK-3β is one of the two GSK-3 isoforms and its expression was determined in five cell lines using western blot analysis in the present study (Fig. 1). To examine the role of GSK-3β in HCC cells, the effect of downregulation of GSK-3β protein expression on HCC cell growth was investigated. HepG2 cells were divided into three groups: Blank HepG2 cells, shGSK-3β-treated cells and shRNA-treated control cells. Western blotting and RT-qPCR identified that GSK-3β protein and mRNA expression levels, respectively, were significantly downregulated by shGSK-3β in HepG2 cells (P<0.05; Fig. 2A–C). Furthermore, shRNA-mediated downregulation of GSK-3β resulted in growth inhibition, as revealed by the MTT assay. Compared with the shRNA control cells, there was a significant growth delay in shGSK-3β-treated cells by 72 h (P<0.05; Fig. 2D). These results demonstrate that GSK-3β downregulation inhibited the proliferation of HCC cells.

GSK-3β inhibitor sensitizes HCC cells to TRAIL-induced apoptosis

The protein expression level of GSK-3β was detected by western blotting following treatment of HepG2 cells with SB216763 (a specific of inhibitor of GSK-3β) and GSK-3β shRNA. As GSK-3 phosphorylates β-catenin for proteasomal degradation, the level of phosphorylated β-catenin reflects the activity of GSK-3. It follows that pretreatment with a GSK-3 inhibitor completely inhibits phosphorylation of β-catenin, thus, increasing the levels of β-catenin (20). To determine the effects of GSK-3β inhibitor on GSK-3β activity in HepG2 cells, western blot was used to analyze the protein expression levels of β-catenin. Western blotting confirmed that the GSK-3β inhibitor significantly inhibited GSK-3β expression compared with the blank control (P<0.05; Fig. 3A and B). The results also demonstrated that GSK-3β inhibition significantly enhanced β-catenin expression (P<0.05), indirectly indicating effective inhibition of GSK-3β activity in HepG2 cells. This enhancement of β-catenin expression occurred in a dose-dependent manner (Fig. 3C and D). Our previous study verified that infection of non-small cell lung cancer cells with Ad5.TRAIL resulted in significant cytotoxicity (21). To investigate the potential role of GSK-3β in TRAIL-induced apoptosis, the effect of a GSK-3β inhibitor on TRAIL-induced cytotoxicity was analyzed using fluorescence-activated cell sorting. Due to the high expression of GSK-3β compared with the other HCC cells investigated, HuH-7 cells were used for this experiment. HuH-7 cells were treated with GSK-3β inhibitor and Ad5.TRAIL alone or in combination. The results indicated that combined treatment with GSK-3β inhibitor and Ad5.TRAIL led to a significant increase in apoptosis compared with untreated cells (P<0.01). By contrast, GSK-3β inhibitor/Ad5.TRAIL combined treatment resulted in a significant induction of cell apoptosis (P<0.05). Thus, the present study demonstrated that TRAIL-induced apoptosis is enhanced following GSK-3β inhibition in HuH-7 cells (Fig. 4A and B).

GSK-3β modulates a specific set of NF-κB target genes

NF-κB is a critical factor in protecting hepatocytes from apoptosis (22). TRAIL has been reported to activate the transcription factor NF-κB, which transactivates genes encoding the key anti-apoptotic proteins Bcl-xL and IAPs, both of which have been implicated in TRAIL resistance (10,11). The current findings demonstrated that GSK-3β inhibitor sensitizes HCC cells to TRAIL-induced apoptosis, suggesting that GSK-3β may be involved in the regulation of NF-κB activation. To prove this hypothesis, the effect of GSK-3β on NF-κB target genes, which encode the key anti-apoptotic proteins Bcl-xL and cIAP2, was evaluated. Western blotting demonstrated that downregulation of GSK-3β by shGSK-3β or GSK-3β inhibitor in HepG2 HCC cells significantly decreased the expression of Bcl-xL and cIAP2 (P<0.05; Fig. 5A and B). The results indicated that GSK-3β has the potential to regulate NF-κB target genes involved in cell survival.

Knockdown of Bcl-xL enhances the effect of GSK-3β inhibitor on TRAIL-induced apoptosis

A recent study revealed that Bcl-xL downregulation contributes to GSK-3β inhibitor-induced sensitization to TRAIL in pancreatic ductal adenocarcinoma cells (11). To evaluate the role of Bcl-xL and cIAP2 on TRAIL-induced apoptosis in HCC cells, shRNA was used to generate HuH-7 cells with stable knockdown of each gene. Subsequently, the response of shBcl-xL- and shcIAP2-treated HuH-7 cells to TRAIL-induced apoptosis was determined. The results indicated that Bcl-xL and cIAP2 knockdown each markedly enhanced TRAIL-induced apoptosis (Fig. 6). To further examine whether Bcl-xL actively participates in GSK-3β inhibitor-mediated TRAIL sensitization, apoptosis was measured following treatment with GSK-3β inhibitor (Fig. 6). The results indicated that knockdown of Bcl-xL significantly enhances the effect of GSK-3β inhibitor on TRAIL-induced apoptosis (P<0.05; Fig. 6).

Discussion

GSK-3β modulates cell survival and apoptosis through multiple intracellular signaling pathways (23,24). In the present study, downregulation of GSK-3β resulted in growth inhibition in HCC cells, which is consistent with previous reports (12). The aim of the current study was to investigate whether inhibition of GSK-3β could induce TRAIL-reduced apoptosis in HCC cells. The results herein demonstrated that chemical inhibition or genetic silencing of GSK-3β sensitizes HCC cells to TRAIL-induced apoptosis. Furthermore, it was found that GSK-3β inhibitor-induced TRAIL sensitization depends upon the activity of NF-κB. These results provide a novel mechanism for GSK-3β modulation of TRAIL sensitivity in HCC cells.

TRAIL resistance has been observed in a variety of cell types (25,26) and is caused by the differential expression of TRAIL receptors, as well as by intracellular molecules, such as Akt and Bcl-2, which modulate the downstream effect of TRAIL signaling (27,28). GSK-3β has been implicated in apoptosis under a variety conditions, including DNA damage, endoplasmic reticulum stress and hypoxia (13,29,30). GSK-3β inhibitors are used in the treatment of Alzheimer's disease, diabetes and inflammatory disorders (31,32). In an oncology setting, previous studies have shown that GSK-3β inhibitors sensitize prostate cancer cells to TRAIL-induced apoptosis (33). The present study demonstrates the mechanism by which GSK-3β inhibits TRAIL-induced apoptosis of sensitized HCC cells. The rationale was based on the premise that GSK-3β inhibition in HCC cells downregulates the expression of key anti-apoptotic proteins that mediate TRAIL resistance. For example, previous studies have demonstrated that TRAIL stimulates NF-κB activation, while inhibition of NF-κB sensitizes cells to TRAIL-mediated apoptosis in various cell lines (10,34). Since the first report that genetic ablation of the murine GSK-3β gene perturbs NF-κB activation (35), the role of GSK-3β in NF-κB signaling has been investigated in a number of cell types. However, conflicting results have been reported regarding the role of GSK-3β in promoting NF-κB activity, ranging from major defects in TNFα-induced inhibitor of κBα (IκBα) phosphorylation to minor effects on cytosolic signaling pathways and p65 nuclear translocation (16,36). In addition, Bcl-2 was reported to inhibit TRAIL-mediated cell death (27). IAPs constitute a protein family that regulates programmed cell death (37), and a recent study demonstrated that GSK-3β induces Bcl-xL and cIAP2 expression in pancreatic cancer cells (11). Consistent with this, the results of the present study indicate that GSK-3β may also regulate the expression of the BCL2L1 and BIRC3 genes, which encode anti-apoptosis proteins Bcl-xL and cIAP2 in HCC cells, respectively, and are known downstream targets of NF-κB. Notably, suppression of Bcl-xL and cIAP2 sensitized HCC cells to TRAIL-induced apoptosis. Furthermore, knockdown of Bcl-xL signaling enhanced the effect of GSK-3β inhibitor on TRAIL-induced apoptosis. These results are consistent with a previous study that demonstrated a role for Bcl-xL in mediating TRAIL resistance (25). A limitation of the current study is that only two NF-κB target genes were examined. Thus, it is unknown whether the NF-κB inhibitor IκBα and a number of other NF-κB target genes are not involved in mediating TRAIL resistance. The detailed mechanisms underlying GSK-3β regulation of the NF-κB signaling pathway have yet to be fully elucidated.

GSK-3β inhibition can overcome TRAIL resistance through the extrinsic apoptotic pathway by promoting the initial step of death-inducing signaling complex formation, leading to caspase-8 and caspase-3 activation in breast cancer cells (38). The results of the current study revealed that GSK-3β can regulate the TRAIL-dependent inhibition of apoptosis at the mRNA and protein levels in HCC cells. Thus, the NF-κB signaling pathways are promising targets for cancer therapy (39). The canonical and noncanonical pathways can be inhibited to modulate NF-κB activity, and possibly treat various types of disease. The present study indicates that GSK-3β inhibitor enhances TRAIL-induced apoptosis in HCC cells. Furthermore, GSK-3β contributes to TRAIL resistance by modulating the expression of a set of specific anti-apoptotic NF-κB target genes. The subcellular location (nucleus or cytoplasm) in which GSK-3β regulates NF-κB function was not explored in the present study. Future studies may determine whether GSK-3β contributes to the expression of Bcl-xL and cIAP2 via nucleic or cytoplasmic targeting and, thus, indicate whether the signaling cascades differ.

In conclusion, the present study demonstrates that GSK-3β suppression sensitizes HCC cells to TRAIL-induced apoptosis, and is dependent on the NF-κB signaling pathway. An understanding of the role of GSK-3β in TRAIL resistance may offer improved therapeutic strategies for patients with HCC.

Acknowledgements

The present study was supported by a grant from the National Natural Science Foundation of China (grant no. 81372632).

References

1 

El-Serag HB and Rudolph KL: Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastroenterology. 132:2557–2576. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Llovet JM, Burroughs A and Bruix J: Hepatocellular carcinoma. Lancet. 362:1907–1917. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Cheng CF, Lu IH, Tseng HW, Sun CY, Lin LT, Kuo ZK, Pan IH and Ko CH: Antitumor Effect of Periplocin in TRAIL-Resistant Human Hepatocellular Carcinoma Cells through Downregulation of IAPs. Evid Based Complement Alternat Med. 2013:Article ID 958025. 2013.

4 

Chiao PJ, Na R, Niu J, Sclabas GM, Dong Q and Curley SA: Role of Rel/NF-kappaB transcription factors in apoptosis of human hepatocellular carcinoma cells. Cancer. 95:1696–1705. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Wiley SR, Schooley K, Smolak PJ, et al: Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity. 3:673–682. 1995. View Article : Google Scholar : PubMed/NCBI

6 

Wajant H, Pfizenmaier K and Scheurich P: TNF-related apoptosis inducing ligand (TRAIL) and its receptors in tumor surveillance and cancer therapy. Apoptosis. 7:449–459. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Srivastava RK: TRAIL/Apo-2L: Mechanisms and clinical applications in cancer. Neoplasia. 3:535–546. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Spierings DC, de Vries EG, Vellenga E, et al: Tissue distribution of the death ligand TRAIL and its receptors. J Histochem Cytochem. 52:821–831. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Smyth MJ, Takeda K, Hayakawa Y, et al: Nature's TRAIL - on a path to cancer immunotherapy. Immunity. 18:1–6. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Omar HA, Arafa SA, Maghrabi IA and Weng JR: Sensitization of hepatocellular carcinoma cells to Apo2L/TRAIL by a novel Akt/NF-κB signalling inhibitor. Basic Clin Pharmacol Toxicol. 114:464–471. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Zhang JS, Herreros-Villanueva M, Koenig A, et al: Differential activity of GSK-3 isoforms regulates NF-κB and TRAIL- or TNFα induced apoptosis in pancreatic cancer cells. Cell Death Dis. 5:e11422014. View Article : Google Scholar : PubMed/NCBI

12 

Forde JE and Dale TC: Glycogen synthase kinase 3: A key regulator of cellular fate. Cell Mol Life Sci. 64:1930–1944. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Song L, De Sarno P and Jope RS: Central role of glycogen synthase kinase-3beta in endoplasmic reticulum stress-induced caspase-3 activation. J Biol Chem. 277:44701–44708. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Pak C and Miyamoto S: A new alpha in line between KRAS and NF-κB activation? Cancer Discov. 3:613–615. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Yao HB, Shaw PC, Wong CC and Wan DCC: Expression of glycogen synthase kinase-3 isoforms in mouse tissuesand their transcription in the brain. J Chem Neuroanat. 23:291–297. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Schwabe RF and Brenner DA: Role of glycogen synthase kinase-3 in TNF-alpha-induced NF-kappaB activation and apoptosis in hepatocytes. Am J Physiol Gastrointest Liver Physiol. 283:G204–G211. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Coghlan MP, Culbert AA, Cross DA, et al: Selective small molecule inhibitors of glycogen synthase kinase-3 modulate glycogen metabolism and gene transcription. Chem Biol. 7:793–803. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Liu J, Jiang G, Liu S, et al: Lentivirus-delivered short hairpin RNA targeting SNAIL inhibits HepG2 cell growth. Oncol Rep. 30:1483–1487. 2013.PubMed/NCBI

19 

Kotliarova S, Pastorino S, Kovell LC, Kotliarov Y, Song H, Zhang W, Bailey R, Maric D, Zenklusen JC, Lee J, et al: Glycogen synthase kinase-3 inhibition induces glioma cell death through c-MYC, nuclear factor-kappaB, and glucose regulation. Cancer Res. 68:6643–6651. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Kim SJ, Lim JY, Lee JN, Choe SK, Kim YI, Song SR, Cho M, So HS and Park R: Activation of β-catenin by inhibitors of glycogen synthase kinase-3 ameliorates cisplatin-induced cytotoxicity and pro-inflammatory cytokine expression in HEI-OC1 cells. Toxicology. 320:74–82. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Zhang H, Sui A, Wang Z, Liu S and Yao R: Adenovirus-mediated TRAIL expression and downregulation of Bcl-2 expression suppresses non-small cell lung cancer growth in vitro and in vivo. Int J Mol Med. 30:358–364. 2012.PubMed/NCBI

22 

Kim HS, Loughran PA, Rao J, Billiar TR and Zuckerbraun BS: Carbon monoxide activates NF-kappaB via ROS generation and Akt pathways to protect against cell death of hepatocytes. Am J Physiol Gastrointest Liver Physiol. 295:G146–G152. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Jope RS and Johnson GV: The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 29:95–102. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Beurel E and Jope RS: The paradoxical pro- and anti-apoptotic actions of GSK3 in the intrinsic and extrinsic apoptosis signaling pathways. Prog Neurobiol. 79:173–189. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Chen KF, Yeh PY, Hsu C, Hsu CH, Lu YS, Hsieh HP, Chen PJ and Cheng AL: Bortezomib overcomes tumor necrosis factor-related apoptosis-inducing ligand resistance in hepatocellular carcinoma cells in part through the inhibition of the phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem. 284:11121–11133. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Newsom-Davis T, Prieske S and Walczak H: Is TRAIL the holy grail of cancer therapy? Apoptosis. 14:607–623. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Munshi A, Pappas G, Honda T, McDonnell TJ, Younes A, Li Y and Meyn RE: TRAIL (APO-2L) induces apoptosis in human prostate cancer cells that is inhibitable by Bcl-2. Oncogene. 20:3757–3765. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Chen X, Thakkar H, Tyan F, Gim S, Robinson H, Lee C, Pandey SK, Nwokorie C, Onwudiwe N and Srivastava RK: Constitutively active Akt is an important regulator of TRAIL sensitivity in prostate cancer. Oncogene. 20:6073–6083. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Loberg RD, Vesely E and Brosius FC III: Enhanced glycogen synthase kinase-3beta activity mediates hypoxia-induced apoptosis of vascular smooth muscle cells and is prevented by glucose transport and metabolism. J Biol Chem. 277:41667–41673. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Watcharasit P, Bijur GN, Zmijewski JW, Song L, Zmijewska A, Chen X, Johnson GV and Jope RS: Direct, activating interaction between glycogen synthase kinase-3beta and p53 after DNA damage. Proc Natl Acad Sci USA. 99:7951–7955. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Klamer G, Song E, Ko KH, O'Brien TA and Dolnikov A: Using small molecule GSK3β inhibitors to treat inflammation. Curr Med Chem. 17:2873–2881. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Medina M and Avila J: Glycogen synthase kinase-3 (GSK-3) inhibitors for the treatment of Alzheimer's disease. Curr Pharm Des. 16:2790–2798. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Liao X, Zhang L, Thrasher JB, Du J and Li B: Glycogen synthase kinase-3β suppression eliminates tumor necrosis factor-related apoptosis-inducing ligand resistance in prostate cancer. Mol Cancer Ther. 2:1215–1222. 2003.PubMed/NCBI

34 

Huerta-Yepez S, Vega M, Jazirehi A, Garban H, Hongo F, Cheng G and Bonavida B: Nitric oxide sensitizes prostate carcinoma cell lines to TRAIL-mediated apoptosis via inactivation of NF-kappa B and inhibition of Bcl-xl expression. Oncogene. 23:4993–5003. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Hoeflich KP: Requirement for glycogen synthase kinase-3β cell survival and NF-κB activation. Nature. 406:86–90. 2001. View Article : Google Scholar

36 

Wilson W III and Baldwin AS: Maintenance of constitutive IkappaB kinase activity by glycogen synthase kinase-3alpha/beta in pancreatic cancer. Cancer Res. 68:8156–8163. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Deveraux QL and Reed JC: IAP family proteins - suppressors of apoptosis. Genes Dev. 13:239–252. 1999. View Article : Google Scholar : PubMed/NCBI

38 

Sun M, Zhou T, Jonasch E and Jope RS: DDX3 regulates DNA damage-induced apoptosis and p53 stabilization. Biochim Biophys Acta. 1833:1489–1497. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Carbone C and Melisi D: NF-kB as a target for pancreatic cancer therapy. Expert Opin Ther Targets. 16:1–10. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 10 Issue 6

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
Fu K, Pan H, Liu S, Lv J, Wan Z, Li J, Sun Q and Liang J: Glycogen synthase kinase‑3β regulates tumor necrosis factor‑related apoptosis inducing ligand (TRAIL)‑induced apoptosis via the NF‑κB pathway in hepatocellular carcinoma. Oncol Lett 10: 3557-3564, 2015
APA
Fu, K., Pan, H., Liu, S., Lv, J., Wan, Z., Li, J. ... Liang, J. (2015). Glycogen synthase kinase‑3β regulates tumor necrosis factor‑related apoptosis inducing ligand (TRAIL)‑induced apoptosis via the NF‑κB pathway in hepatocellular carcinoma. Oncology Letters, 10, 3557-3564. https://doi.org/10.3892/ol.2015.3803
MLA
Fu, K., Pan, H., Liu, S., Lv, J., Wan, Z., Li, J., Sun, Q., Liang, J."Glycogen synthase kinase‑3β regulates tumor necrosis factor‑related apoptosis inducing ligand (TRAIL)‑induced apoptosis via the NF‑κB pathway in hepatocellular carcinoma". Oncology Letters 10.6 (2015): 3557-3564.
Chicago
Fu, K., Pan, H., Liu, S., Lv, J., Wan, Z., Li, J., Sun, Q., Liang, J."Glycogen synthase kinase‑3β regulates tumor necrosis factor‑related apoptosis inducing ligand (TRAIL)‑induced apoptosis via the NF‑κB pathway in hepatocellular carcinoma". Oncology Letters 10, no. 6 (2015): 3557-3564. https://doi.org/10.3892/ol.2015.3803