Downregulation of glucose-regulated protein 78 enhances the cytotoxic effects of curcumin on human nasopharyngeal carcinoma cells

  • Authors:
    • Xiaoming Yu
    • Long Shi
    • Li Yan
    • Hui Wang
    • Yan Wen
    • Xiuping Zhang
  • View Affiliations

  • Published online on: August 23, 2018     https://doi.org/10.3892/ijmm.2018.3837
  • Pages: 2943-2951
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Curcumin is a conventional Chinese medicine, which exerts a marked effect on various tumor types and suppresses tumor invasion. The present study analyzed the antineoplastic effects of curcumin on human nasopharyngeal carcinoma (NPC) cells and determined the effects of endoplasmic reticulum (ER) stress on curcumin‑induced cytotoxicity. The Cell Counting Kit‑8 assay examined the viability of SUNE1 and SUNE2 NPC cells. The Annexin V/propidium iodide staining technique was used to detect cell apoptosis and flow cytometry was used to examine cell cycle distribution. Western blotting and immunofluorescence were used to detect ER stress‑associated molecules. Furthermore, the toxic effects of curcumin treatment alongside glucose‑regulated protein 78 (GRP78) knockdown using small interfering (si)RNA, and treatment with the pan‑caspase inhibitor Z‑VAD‑FMK and the protein kinase B (AKT) inhibitor MK‑2206 were detected. The results demonstrated that curcumin markedly reduced cell viability, blocked cell cycle progression and induced apoptosis of human NPC cells. In addition, curcumin activated ER stress‑associated proteins to participate in the apoptosis of human NPC cells. siRNA‑induced knockdown of GRP78 may be able to strengthen the toxic effects of curcumin through mediating the AKT signaling pathway. These findings indicated that downregulation of GRP78 promoted the therapeutic effects of curcumin on NPC cells. The present study identified a potential, novel therapeutic method for the treatment of NPC.

Introduction

Nasopharyngeal carcinoma (NPC) is the most common primary malignancy of the nasopharynx, which frequently occurs in South China and Southeast Asia (1). The main treatment strategy for NPC is a combination of radiotherapy and chemotherapy; however, due to distant metastasis and resistance to treatment, the clinical prognosis of NPC is poor (2). In addition, numerous therapeutic measures can cause serious side effects and lead to multidrug resistance (3). Therefore, it is necessary to identify a novel, safe and effective method to treat NPC. At present, for the prevention, inhibition or delay of carcinogenesis, treatment with natural, synthetic or biological chemicals is regarded as being able to induce relatively effective chemoprevention (4).

Curcumin, 1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione, is present in turmeric (Curcuma longa), and has been used to treat various diseases, including allergies, coryza, cough, anorexia, rhinitis and hepatic conditions, in Asia for centuries (5). In addition, curcumin is able to suppress proliferation and promote apoptosis in various malignant diseases (6-9). However, the functions and effects of curcumin on NPC cells require further research. Some specific functions that have been detected in other tumor models include induction of apoptosis via c-Jun N-terminal kinase and p38 mitogen-activated protein kinase signaling pathway regulation (10), endoplasmic reticulum (ER) stress (11) and autophagy (12).

The ER is an organelle, which can fold and synthesize transmembrane, intraorganellar and secretory proteins, and accumulate intracellular calcium. Disturbance of ER homeostasis leads to ER stress and the subsequent accumulation of unfolded or misfolded proteins; this is known as the unfolded protein response (UPR). The UPR induces a series of signaling pathways to maintain ER homeostasis via the upregulation of molecular chaperones that can accelerate immunoglobulin folding and protein synthesis attenuation (13). Three ER transmembrane receptors inhibit the three signaling pathways that comprise the UPR; inositol-requiring enzyme 1α (IRE1α), activating transcription factor 6 and eukaryotic translation initiation factor 2α kinase 3 (14). Initially, UPR signaling serves a crucial role in restoring ER homeostasis; however, continuous or prolonged UPR results in cell apoptosis via caspase-12, caspase-4 and B-cell lymphoma 2 (Bcl-2). The present study aimed to investigate the effects of the UPR on the toxic activity of curcumin on NPC cells. The effects of interference in the UPR pathway on the toxic effects of curcumin on human NPC cells were also analyzed.

Materials and methods

Cell culture

SUNE1 and SUNE2 NPC cells were purchased from the American Type Culture Collection (Manassas, VA, USA). These cells were cultured in Dulbecco's modified Eagle's medium (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) or RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin, and 100 µg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in an atmosphere containing 95% air and 5% CO2.

Reagents and antibodies

Curcumin, Z-VAD-FMK (Z-VAD) and MK-2206 were purchased from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). Primary antibodies against GRP78 (cat. no. ab21685), GAPDH (cat. no. ab37168), cleaved poly (ADP-ribose) polymerase (PARP; cat. no. ab32561), caspase-3 (cat. no. ab90437), caspase-9 (cat. no. ab25758) and caspase-12 (cat. no. ab62484) were obtained from Abcam (Cambridge, UK). Antibodies against CCAAT-enhancer-binding protein homologous protein (CHOP; cat. no. 2895), IRE1 (cat. no. 3294), phosphorylated (p)-eukaryotic initiation factor 2α (eIF2α; cat. no. 9721), Bcl-2 (cat. no. 2872), protein kinase B (AKT; cat. no. 9272) and p-AKT (Ser473) (cat. no. 9271) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA).

Measurement of cell viability

Cell viability was analyzed using the Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan). Initially, cells (5×103/well) were cultured in culture medium and were inoculated into 96-well plates. After 24 h, 0, 10, 20, 40 and 80 µM curcumin was added to each well and the cells were cultured at 37°C for a further 24 h. The original culture medium was replaced with a mixture of fresh medium and CCK-8 solution (10 µl). The optical density (OD) index of the cells was measured at 450 nm after culturing at 37°C for 2 h. The percentage of inhibition of cell activity was calculated according to the following formula: (control OD value-experiment OD value)/control group OD value ×100%.

Cell cycle analysis by flow cytometry

Human NPC cells were cultured in the aforementioned culture medium. Subsequently, once the cells reached 50% confluence, they were treated with 80 µM curcumin at 37°C for 24 h and were collected in order to analyze the cell cycle. Briefly, 5×104 cells were incubated with DAPI and RNase A (Thermo Fisher Scientific, Inc.) for 30 min at room temperature, and the cells were then suspended in 0.5 ml propidium iodide (PI; BD Biosciences, San Jose, CA, USA) solution and incubated for 30 min at room temperature in the dark, according to the manufacturer's protocol. Subsequently, cell cycle distribution was analyzed using flow cytometry [fluorescence-activated cell sorting (FACS) analysis; BD FACSDiva software v8.0.1; BD Biosciences].

Apoptotic analysis by flow cytometry

Cells (4×105/well) were seeded in the aforementioned culture medium in 6-well plates and were incubated at 37°C for 24 h. Subsequently, cells were treated with fresh medium containing the required concentrations of curcumin (0, 20, 40 and 80 µM), or the cells were pretreated with 50 µM Z-VAD, followed by treatment with or without 80 µM curcumin at 37°C for 24 h. Cells were then stained using the Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (BD Biosciences). According to the manufacturer's protocol, cells were treated for 15 min with a mixture of Annexin V (5 µl) and PI (5 µl) at room temperature. Subsequently, the stained cells were observed by flow cytometry (FACS analysis; BD FACSDiva software v8.0.1, BD Biosciences).

Western blot analysis

Cells were treated with either curcumin (0-80 µM) or MK-2206 (5 µM) alone or in combination for 24 h. Alternatively, cells underwent GRP78 knockdown by transient transfection with small interfering (si)RNA (20 nM) for 24 h, after which, the transfected NPC cells were treated with or without curcumin (80 µM) in complete medium at 37°C for 24 h. The cells were then lysed in cold radioimmuno-precipitation assay lysis buffer (Sigma-Aldrich; Merck KGaA) supplemented with 1 nM phenylmethylsufonyl fluoride, and were centrifuged at 12,000 × g for 10 min at 4°C. The concentration of proteins extracted from cells was determined using a bicinchoninic acid assay (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Total proteins (20-25 µg) were separated by 8-15% SDS-PAGE and transferred onto a polyvinylidene fluoride membrane (EMD Millipore, Billerica, MA, USA). The membrane was then blocked in a mixture of 5% skimmed milk and PBS containing 0.1% Tween-20 for 2 h at room temperature, and was incubated with primary antibodies at 4°C overnight [1:1,000 dilutions for GRP78, cleaved PARP, caspase-3, caspase-9, caspase-12, CHOP, IRE1, p-eIF2α, Bcl-2, AKT, p-AKT (Ser473) and GAPDH]. Membranes were then incubated with horseradish peroxidase-conjugated secondary antibodies (cat. nos. 7074 and 7076; 1:2,000; Cell Signaling Technology, Inc.) for 1 h at room temperature, followed by visualization with enhanced chemiluminescence immunoblotting detection reagents (EMD Millipore). Protein band intensities were semi-quantified by densitometric analysis using ImageJ software 1.6.0_20 (National Institutes of Health, Bethesda, MD, USA).

Immunofluorescence assay

A total of 1×105 human NPC cells were placed in each well of 6-well chamber slides and were treated with curcumin (80 µM) at 37°C for 24 h. PBS was used to wash the cells, and they were then immobilized and permeabilized with 4% paraformaldehyde and 0.1% Triton X-100 for 15 min at room temperature. Immunofluorescence staining was then conducted. Initially, the processed human NPC cells were stained with primary antibodies (1:1,000 for GRP78; 1:3,000 for CHOP) for 2 h at room temperature, and were then cultured with secondary antibodies conjugated to Alexa Fluor 488 (green) (1:200) or Alexa Fluor 546 (red) (1:200) (cat. nos. A28175 and A-11071; Invitrogen; Thermo Fisher Scientific, Inc.) for detection of CHOP and GRP78 for 2 h at room temperature, respectively. Subsequently, the cells were stained with DAPI (Abcam). Fluorescent images of the NPC cells were captured and analyzed under a light microscope (Olympus Corporation, Tokyo, Japan).

RNA interference

GRP78-specific (cat. no. sc-29338) and non-silencing scrambled siRNA (cat. no. sc-37007) were obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). According to the manufacturer's protocol, 4×105 human NPC cells/well were plated in 6-well plates and transfected with siRNAs using Lipofectamine® 2000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.) for 24 h. Subsequently, the transfected NPC cells were exposed to curcumin (80 µM) in complete medium at 37°C for 24 h.

Statistical analysis

All experiments were repeated in triplicate. Data are presented as the means ± standard deviation. The differences among multiple groups were analyzed by one-way analysis of variance followed by Dunnett's test using SPSS software (version 17.0; SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statically significant difference.

Results

Curcumin reduces the viability of human NPC cells

In order to investigate the role of curcumin in the survival of NPC cells in vitro, SUNE1 and SUNE2 cells were exposed to various concentrations of curcumin (0-80 µM) for 1 day, and inhibition of cell proliferation was evaluated by CCK-8. As shown in Fig. 1, inhibition of NPC cell viability increased with increasing concentrations of curcumin, from 13.58% inhibition following treatment with 10 µM curcumin to 40.60% inhibition following treatment with 80 µM curcumin in SUNE1 cells, and from 15.16% inhibition following treatment with 10 µM curcumin to 43.27% inhibition following treatment with 80 µM curcumin in SUNE2 cells.

Curcumin induces NPC cell apoptosis and cell cycle arrest

In order to determine the role of curcumin in apoptotic cell death, SUNE1 and SUNE2 cells were exposed to various concentrations of curcumin (20-80 µM) for 24 h. Apoptotic cell death was analyzed using the Annexin V/PI analysis. As shown in Fig. 2A-D, early apoptosis of SUNE1 cells was markedly increased from 1.49 to 17.45% and late apoptosis of SUNE1 cells was increased from 0.72 to 21.38% following exposure to 80 µM curcumin for 24 h. In addition, early and late apoptosis increased from 1.16 to 25.12% and 2.23 to 15.93% in 80 µM curcumin-treated SUNE2 cells, respectively. As shown in Fig. 2E, cell cycle distribution of curcumin-treated SUNE1 and SUNE2 cells was determined. The majority of curcumin (80 µM)-treated NPC cells were arrested at G0/G1 phase; the percentage of cells in G0/G1 phase was increased to 68.85 and 70.88% in SUNE1 cells and SUNE2 cells, respectively. These data indicated that curcumin can induce cell death via apoptosis.

Curcumin induces ER stress in human NPC cells

To determine the effects of curcumin on ER stress in human NPC cells, specific ER-associated proteins were detected. Western blot analysis demonstrated that IRE1, p-eIF2α, CHOP and GRP78 were increased following curcumin treatment (Fig. 3A-D). To ascertain whether curcumin can increase the expression of markers that are associated with ER stress in NPC cells, CHOP and GRP78 were analyzed by immunofluorescence staining. As shown in Fig. 3E, immunofluorescence staining of CHOP and GRP78 was markedly increased following curcumin (80 µM) treatment in SUNE1 and SUNE2 cells. Previous studies have reported that prolonged ER stress can active caspase-12, and caspase-12 has an important influence on inducing cell death (15,16). In order to analyze the effects of curcumin on ER stress and toxicity, and to investigate the underlying mechanisms, western blotting was used to detect the expression of the caspase-12 protein in NPC cells. As shown in Fig. 4A-D, compared with in the control group, activation of caspase-12 was increased in curcumin-treated SUNE1 and SUNE2 cells. Furthermore, the expression levels of the anti-apoptotic protein Bcl-2 were significantly decreased (Fig. 4A-D). These results indicated that ER stress may induce apoptosis.

GRP78 knockdown enhances curcumin-induced apoptosis of human NPC cells

GRP78, which is the most important marker of the UPR, has been reported to be associated with chemoresistance (17,18). siRNA was used to determine the effects of GRP78 knockdown on curcumin-induced apoptosis of SUNE1 and SUNE2 cells. As shown in Fig. 5A, following GRP78 siRNA transfection, in cells that were treated with curcumin, cell viability was significantly reduced. In addition, GRP78 siRNA transfection significantly inhibited the protein expression levels of GRP78, improved PARP, caspase-3 and caspase-9 cleavage (Fig. 5B and C), and increased cell apoptosis (Fig. 6A and B) of curcumin-treated SUNE1 and SUNE2 cells. Subsequently, the pan-caspase inhibitor Z-VAD was used to investigate the role of curcumin in cell death. As shown in Fig. 6C and D, a marked reduction in cell death was detected in SUNE1 and SUNE2 cells pretreated with Z-VAD. This finding indicated that curcumin-mediated apoptosis may be associated with caspase activation.

GRP78 knockdown reduces AKT activity in human NPC cells

The AKT pathway is important in the modification of cell growth and proliferation (19). Previous studies have reported that the GRP78 signaling pathway can regulate the AKT signaling pathway (20,21). Therefore, the present study aimed to determine whether AKT was involved in GRP78-mediated cell death during curcumin treatment. As shown in Fig. 7A and B, treatment of SUNE1 and SUNE2 cells with curcumin (80 µM) significantly suppressed AKT phosphorylation, whereas total AKT was only slightly altered. Knockdown of GRP78 via siRNA alongside curcumin treatment led to a marked reduction in AKT phosphorylation compared with in cells treated with curcumin only. To further investigate the effects of GRP78 expression and AKT phosphorylation, MK-2206, a chemical inhibitor of AKT, was used to treat cells prior to treatment with curcumin. As shown in Fig. 7C and D, treating SUNE1 and SUNE2 cells with MK-2206 reduced activation of AKT. Furthermore, MK-2206 markedly decreased the expression levels of GRP78 (Fig. 7C and D). These results indicated that GRP78 knockdown may reduce AKT activity.

Discussion

NPC is a common cancer of the nasopharynx, with a five year survival rate of 40-70% (22). Numerous patients with NPC experience recurrence or metastasis due to resistance to standard therapy with radiation (23). Therefore, it is very important to identify novel treatment modalities. Curcumin, which is a polyphenolic dienone, has been demonstrated to induce apoptosis in several human malignancies (24-27); however, the mechanisms underlying the suppressive effects of curcumin on NPC remain to be completely explored. The present study demonstrated that curcumin included ER stress, cell cycle arrest and apoptosis of human NPC cells. Knockdown of GRP78 by siRNA was able to significantly enhance curcumin-induced NPC cell apoptosis.

Previous studies have reported that ER stress is not only conducive to cell survival, but also induces apoptosis in various cell types (28,29). The aim of the UPR is to reinstate appropriate ER homeostasis; however, if ER stress perseveres, the UPR signaling pathways can trigger apoptosis. Therefore, the balance between the survival signal and the apoptosis signal determines the survival and apoptosis of cells. GRP78 is an ER molecular chaperone, which is important in folding and assembling proteins (30). Previous research has indicated that constitutive over expression of GRP78 is associated with chemoresistance in cancer treatment (31), whereas knockdown of GRP78 can enhance chemosensitivity in several tumor cells (18,32,33). According to the present study, treatment of human NPC cells with curcumin led to ER stress, and curcumin-induced ER stress in SUNE1 and SUNE2 cells was associated with the up regulation of GRP78. In addition, silencing GRP78 using siRNA significantly enhanced the cytotoxic and apoptotic effects of curcumin in SUNE1 and SUNE2 cells. These data revealed that GRP78 may be crucial in protecting human NPC cells from curcumin-induced apoptosis. Therefore, downregulation of GRP78 may markedly improve the sensitivity of human NPC cells to curcumin.

Activation of the AKT signaling pathway is crucial in modifying cell proliferation and motility in various types of cancer; therefore, inhibition of AKT phosphorylation could suppress tumor cell growth and proliferation (34,35). In addition, it has been demonstrated that the AKT signaling pathway is associated with chemoresistance and maintaining GRP78 expression (36,37). The present results revealed that GRP78 knockdown by siRNA significantly inhibited the phosphorylation of AKT in curcumin-treated cells. Furthermore, inhibiting AKT activity using the chemical inhibitor MK-2206 prevented curcumin-mediated GRP78 induction, and suppressed AKT activity, thus suggesting that GRP78 may function in NPC to restrict chemotherapeutic-induced cytotoxicity via regulating the AKT signaling pathway.

In conclusion, this study suggested that curcumin may inhibit the viability and promote the apoptosis of human NPC cells. Downregulation of GRP78 may further enhance curcumin-induced SUNE1 and SUNE2 cell apoptosis. These findings are promising; however, further research is required to develop novel therapeutic strategies for human NPC.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81400376) and thee Shandong Province Natural Science Foundation of China (grant no. BS2014YY017).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

XY performed experiments and analyzed data; XZ designed the research, interpreted data and edited the manuscript; LS wrote the manuscript and analyzed the data; LY analyzed the data and reviewed the manuscript; HW interpreted data and reviewed the manuscript; YW interpreted data and edited the manuscript. 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.

Acknowledgments

The authors would like to thank Dr. Edward C. Mignot (Shandong University, Jinan, China) for linguistic advice.

References

1 

Lee AW, Fee WE Jr, Ng WT and Chan LK: Nasopharyngeal carcinoma: Salvage of local recurrence. Oral Oncol. 48:768–774. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Xu Y, Zhang J, Shi W and Liu Y: Anticancer effects of 3,3′-diindolylmethane are associated with G1 arrest and mitochondria-dependent apoptosis in human nasopharyngeal carcinoma cells. Oncol Lett. 5:655–662. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Xiao WW, Huang SM, Han F, Wu SX, Lu LX, Lin CG, Deng XW, Lu TX, Cui NJ and Zhao C: Local control, survival, and late toxicities of locally advanced nasopharyngeal carcinoma treated by simultaneous modulated accelerated radiotherapy combined with cisplatin concurrent chemotherapy: Long-term results of a phase 2 study. Cancer. 117:1874–1883. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Huang J, Liang ZD, Wu TT, Hoque A, Chen H, Jiang Y, Zhang H and Xu XC: Tumor-suppressive effect of retinoid receptor-induced gene-1 (RRIG1) in esophageal cancer. Cancer Res. 67:1589–1593. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Ammon HP and Wahl MA: Pharmacology of Curcuma longa. Planta Med. 57:1–7. 1991. View Article : Google Scholar : PubMed/NCBI

6 

He M, Li Y, Zhang L, Li L, Shen Y, Lin L, Zheng W, Chen L, Bian X, Ng HK and Tang L: Curcumin suppresses cell proliferation through inhibition of the Wnt/β-catenin signaling pathway in medulloblastoma. Oncol Rep. 32:173–180. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Qiao Q, Jiang Y and Li G: Curcumin enhances the response of non-Hodgkin's lymphoma cells to ionizing radiation through further induction of cell cycle arrest at the G2/M phase and inhibition of mTOR phosphorylation. Oncol Rep. 29:380–386. 2013. View Article : Google Scholar

8 

Du Q, Hu B, An HM, Shen KP, Xu L, Deng S and Wei MM: Synergistic anticancer effects of curcumin and resveratrol in Hepa1-6 hepatocellular carcinoma cells. Oncol Rep. 29:1851–1858. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Pan Y, Xiao J, Liang G, Wang M, Wang D, Wang S and Yang H: A new curcumin analogue exhibits enhanced antitumor activity in nasopharyngeal carcinoma. Oncol Rep. 30:239–245. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Yu X, Zhong J, Yan L, Li J, Wang H, Wen Y and Zhao Y: Curcumin exerts antitumor effects in retinoblastoma cells by regulating the JNK and p38 MAPK pathways. Int J Mol Med. 38:861–868. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Cao A, Li Q, Yin P, Dong Y, Shi H, Wang L, Ji G, Xie J and Wu D: Curcumin induces apoptosis in human gastric carcinoma AGS cells and colon carcinoma HT-29 cells through mitochondrial dysfunction and endoplasmic reticulum stress. Apoptosis. 18:1391–1402. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Zanotto-Filho A, Braganhol E, Klafke K, Figueiró F, Terra SR, Paludo FJ, Morrone M, Bristot IJ, Battastini AM, Forcelini CM, et al: Autophagy inhibition improves the efficacy of curcumin/temozolomide combination therapy in glioblastomas. Cancer Lett. 358:220–231. 2015. View Article : Google Scholar

13 

Bortolozzi R, Viola G, Porcù E, Consolaro F, Marzano C, Pellei M, Gandin V and Bassoj G: A novel copper(I) complex induces ER-stress-mediated apoptosis and sensitizes B-acute lymphoblastic leukemia cells to chemotherapeutic agents. Oncotarget. 5:5978–5991. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Xu C, Bailly-Maitre B and Reed JC: Endoplasmic reticulum stress: Cell life and death decisions. J Clin Invest. 115:2656–2664. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Zhong J, Dong X, Xiu P, Wang F, Liu J, Wei H, Xu Z, Liu F, Li T and Li J: Blocking autophagy enhances meloxicam lethality to hepatocellular carcinoma by promotion of endoplasmic reticulum stress. Cell Prolif. 48:691–704. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Zhong J, Xiu P, Dong X, Wang F, Wei H, Wang X, Xu Z, Liu F, Li T, Wang Y and Li J: Meloxicam combined with sorafenib synergistically inhibits tumor growth of human hepatocellular carcinoma cells via ER stress-related apoptosis. Oncol Rep. 34:2142–2150. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Li W, Wang W, Dong H, Li Y, Li L, Han L, Han Z, Wang S, Ma D and Wang H: Cisplatin-induced senescence in ovarian cancer cells is mediated by GRP78. Oncol Rep. 31:2525–2534. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Pi L, Li X, Song Q, Shen Y, Lu X and Di B: Knockdown of glucose-regulated protein 78 abrogates chemoresistance of hypo-pharyngeal carcinoma cells to cisplatin induced by unfolded protein in response to severe hypoxia. Oncol Lett. 7:685–692. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Yip PY: Phosphatidylinositol 3-kinase-AKT-mammalian target of rapamycin (PI3K-Akt-mTOR) signaling pathway in non-small cell lung cancer. Transl Lung Cancer Res. 4:165–176. 2015.PubMed/NCBI

20 

Kelber JA, Panopoulos AD, Shani G, Booker EC, Belmonte JC, Vale WW and Gray PC: Blockade of Cripto binding to cell surface GRP78 inhibits oncogenic Cripto signaling via MAPK/PI3K and Smad2/3 pathways. Oncogene. 28:2324–2336. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Wey S, Luo B, Tseng CC, Ni M, Zhou H, Fu Y, Bhojwani D, Carroll WL and Lee AS: Inducible knockout of GRP78/BiP in the hematopoietic system suppresses Pten-null leukemogenesis and AKT oncogenic signaling. Blood. 119:817–825. 2012. View Article : Google Scholar :

22 

Zhang W, Yang P, Gao F, Yang J and Yao K: Effects of epigallocatechin gallate on the proliferation and apoptosis of the nasopharyngeal carcinoma cell line CNE2. Exp Ther Med. 8:1783–1788. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Ma J, Wen ZS, Lin P, Wang X and Xie FY: The results and prognosis of different treatment modalities for solitary metastatic lung tumor from nasopharyngeal carcinoma: A retrospective study of 105 cases. Chin J Cancer. 29:787–795. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Yin H, Zhou Y, Wen C, Zhou C, Zhang W, Hu X, Wang L, You C and Shao J: Curcumin sensitizes glioblastoma to temozolomide by simultaneously generating ROS and disrupting AKT/mTOR signaling. Oncol Rep. 32:1610–1616. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Peng SF, Lee CY, Hour MJ, Tsai SC, Kuo DH, Chen FA, Shieh PC and Yang JS: Curcumin-loaded nanoparticles enhance apoptotic cell death of U2OS human osteosarcoma cells through the Akt-Bad signaling pathway. Int J Oncol. 44:238–246. 2014. View Article : Google Scholar

26 

Wu J, Tang Q, Zhao S, Zheng F, Wu Y, Tang G and Hahn SS: Extracellular signal-regulated kinase signaling-mediated induction and interaction of FOXO3a and p53 contribute to the inhibition of nasopharyngeal carcinoma cell growth by curcumin. Int J Oncol. 45:95–103. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Yang SJ, Lee SA, Park MG, Kim JS, Yu SK, Kim CS, Kim JS, Kim SG, Oh JS, Kim HJ, et al: Induction of apoptosis by diphenyldifluoroketone in osteogenic sarcoma cells is associated with activation of caspases. Oncol Rep. 31:2286–2292. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Schroder M and Kaufman RJ: The mammalian unfolded protein response. Annu Rev Biochem. 74:739–789. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Zhang K and Kaufman RJ: The unfolded protein response: A stress signaling pathway critical for health and disease. Neurology. 66:S102–S109. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Li J and Lee AS: Stress induction of GRP78/BiP and its role in cancer. Curr Mol Med. 6:45–54. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Lee AS: GRP78 induction in cancer: Therapeutic and prognostic implications. Cancer Res. 67:3496–3499. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Huang KH, Kuo KL, Chen SC, Weng TI, Chuang YT, Tsai YC, Pu YS, Chiang CK and Liu SH: Down-regulation of glucose-regulated protein (GRP) 78 potentiates cytotoxic effect of celecoxib in human urothelial carcinoma cells. PLoS One. 7:e336152012. View Article : Google Scholar : PubMed/NCBI

33 

Wang J, Yin Y, Hua H, Li M, Luo T, Xu L, Wang R, Liu D, Zhang Y and Jiang Y: Blockade of GRP78 sensitizes breast cancer cells to microtubules-interfering agents that induce the unfolded protein response. J Cell Mol Med. 13:3888–3897. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Singh S, Trevino J, Bora-Singhal N, Coppola D, Haura E, Altiok S and Chellappan SP: EGFR/Src/Akt signaling modulates Sox2 expression and self-renewal of stem-like side-population cells in non-small cell lung cancer. Mol Cancer. 11:732012. View Article : Google Scholar : PubMed/NCBI

35 

Qin J, Ji J, Deng R, Tang J, Yang F, Feng GK, Chen WD, Wu XQ, Qian XJ, Ding K and Zhu XF: DC120, a novel AKT inhibitor, preferentially suppresses nasopharyngeal carcinoma cancer stem-like cells by downregulating Sox2. Oncotarget. 6:6944–6958. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Li B, Li J, Xu WW, Guan XY, Qin YR, Zhang LY, Law S, Tsao SW and Cheung AL: Suppression of esophageal tumor growth and chemoresistance by directly targeting the PI3K/AKT pathway. Oncotarget. 5:11576–11587. 2014.PubMed/NCBI

37 

Gray MJ, Mhawech-Fauceglia P, Yoo E, Yang W, Wu E, Lee AS and Lin YG: AKT inhibition mitigates GRP78 (glucose-regulated protein) expression and contribution to chemoresistance in endometrial cancers. Int J Cancer. 133:21–30. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 42 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu X, Shi L, Yan L, Wang H, Wen Y and Zhang X: Downregulation of glucose-regulated protein 78 enhances the cytotoxic effects of curcumin on human nasopharyngeal carcinoma cells. Int J Mol Med 42: 2943-2951, 2018
APA
Yu, X., Shi, L., Yan, L., Wang, H., Wen, Y., & Zhang, X. (2018). Downregulation of glucose-regulated protein 78 enhances the cytotoxic effects of curcumin on human nasopharyngeal carcinoma cells. International Journal of Molecular Medicine, 42, 2943-2951. https://doi.org/10.3892/ijmm.2018.3837
MLA
Yu, X., Shi, L., Yan, L., Wang, H., Wen, Y., Zhang, X."Downregulation of glucose-regulated protein 78 enhances the cytotoxic effects of curcumin on human nasopharyngeal carcinoma cells". International Journal of Molecular Medicine 42.5 (2018): 2943-2951.
Chicago
Yu, X., Shi, L., Yan, L., Wang, H., Wen, Y., Zhang, X."Downregulation of glucose-regulated protein 78 enhances the cytotoxic effects of curcumin on human nasopharyngeal carcinoma cells". International Journal of Molecular Medicine 42, no. 5 (2018): 2943-2951. https://doi.org/10.3892/ijmm.2018.3837