Open Access

3‑Bromopyruvate sensitizes human breast cancer cells to TRAIL‑induced apoptosis via the phosphorylated AMPK‑mediated upregulation of DR5

Corrigendum in: /10.3892/or.2022.8264

  • Authors:
    • Yuzhong Chen
    • Li Wei
    • Xiaojing Zhang
    • Xianfu Liu
    • Yansong Chen
    • Song Zhang
    • Lanzhu Zhou
    • Qixiang Li
    • Qiong Pan
    • Surong Zhao
    • Hao Liu
  • View Affiliations

  • Published online on: August 14, 2018     https://doi.org/10.3892/or.2018.6644
  • Pages: 2435-2444
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Previous studies have indicated that the sensitivity of breast cancer cells to tumor necrosis factor‑related apoptosis‑inducing ligand (TRAIL)‑induced apoptosis is associated with the expression of death receptors on the cell membrane. However, drug resistance limits the use of TRAIL in cancer therapy. Numerous studies have indicated that death receptors, which induce apoptosis, are upregulated by the endoplasmic reticulum (ER) stress response. 3‑Bromopyruvate (3‑BP), an anticancer agent, inhibits cell growth and induces apoptosis through interfering with glycolysis. In the present study, it was demonstrated that 3‑BP synergistically sensitized breast cancer cells to TRAIL‑induced apoptosis via the upregulation of death receptor 5 (DR5). Furthermore, we found that the protein levels of glucose‑related protein 78 (GRP78) and CCAAT‑enhancer‑binding protein homologous protein (CHOP) increased following treatment with 3‑BP. The expression of Bax (in MCF‑7 cells) and caspase‑3 (in MDA‑MB‑231 cells) increased following co‑treatment with 3‑BP and TRAIL, whereas the expression of the anti‑apoptotic protein Bcl‑2 decreased. In order to investigate the molecular mechanism regulating this effect, the expression of adenosine monophosphate‑activated protein kinase (AMPK), activated by 3‑BP, was determined. It was demonstrated that phosphorylated‑AMPK was upregulated following treatment with 3‑BP. Notably, Compound C, an AMPK inhibitor, reversed the effects of 3‑BP. Finally, a synergistic antitumor effect of 3‑BP and TRAIL was observed in MCF‑7 cell xenografts in nude mice. In conclusion, these results indicated that 3‑BP sensitized breast cancer cells to TRAIL via the AMPK‑mediated upregulation of DR5.

Introduction

Breast cancer is the leading cause of cancer-related deaths among females and is the most frequently diagnosed type of cancer in women. The incidence of breast cancer in China is increasing by 2.2% per year (1). The currently available treatments for patients with breast cancer includes chemotherapy, surgical resection and radiotherapy. Considering that drug resistance is often associated with targeted therapies, there is a requirement for novel therapeutic strategies for the treatment of cancer (2).

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has a crucial role in cancer therapy; by binding to death receptor (DR)4 and DR5, it selectively induces apoptosis in cancer cells. TRAIL has reduced toxicicity compared with other members of the tumor necrosis factor (TNF) protein family, such as TNFα and Fas ligand (CD95L) (3). Pre-clinical trials have determined that TRAIL effectively inhibits tumor growth without inducing toxicity in both mice and non-human primates (4,5). However, cancer cells may aquire resistence to TRAIL-induced apoptosis via mutation or deficiency of DR4 or DR5. A number of studies have investigated whether the use of recombinant TRAIL may reverse this effect, by enabling the interaction of TRAIL with mutated receptors, or whether the combination of treatment with TRAIL and Chinese herbology (6) may increase the therapeutic efficacy and decrease the toxicity of TRAIL (7).

The expression of TRAIL receptor 2 (TRAIL-R2 or DR5) is crucial in TRAIL-induced apoptosis in cancer cells (8), and is upregulated following the inhibition of ATP synthesis in cancer cells (9). Cancer cells and healthy cells may be characterized according to their different energy metabolisms (10). In 1956, Warburg demonstrated that cancer cells produce energy predominantly via fermentation rather than through oxidative respiration as exhibited by healthy cells, even when sufficient oxygen is present (11). However, fermentation is associated with the higher consumption of glucose relative to oxidative respiration, as well as the greater production of lactate.

3-Bromopyruvate (3-BP), a hexokinase II inhibitor, can induce apoptosis in hepatocellular carcinoma cells by inducing endoplasmic reticulum (ER) stress (12). ER stress occurs upon accumulation of misfolded proteins in the ER, and results in the activation of the ER stress chaperone protein glucose-related protein 78 (GRP78) and the pro-apoptotic transcription factor CCAAT-enhancer-binding protein homologous protein (CHOP). Previous studies have indicated that DR5 is upregulated during the ER stress response (1315). Adenosine monophosphate-activated protein kinase (AMPK) is a nutrient and energy marker in the cells, and can also induce apoptosis (1618). Notably, numerous studies have suggested that c-Jun N-terminal kinase (JNK), also known as stress-activated protein kinase (SAPK), is potentially a regulator of AMPK-induced apoptosis. JNK regulates apoptosis via the phosphorylation of apoptosis related-proteins, such as B-cell f lymphoma 2 (Bcl-2), or via the activation of activator protein-1 (AP-1) (19). In addition, 3-BP suppresses the levels of cellular adenosine tri-phosphate (ATP) and activates AMPK, as well as the AMPK-mediated upregulation of Bcl-2-associated X protein (Bax), in which subsequently induces cell death via the mitochondrial pathway (20).

Our previous study indicated that the hexokinase inhibitor 3-BP induced cell death in colon cancer cells via a number of different mechanisms (21). Therefore, we aimed to investigate whether 3-BP upregulated DR5, thus enhancing the anticancer effect of TRAIL. The results of the present study demonstrated that AMPK was induced by 3-BP to act as a regulator of apoptosis. Thus, we hypothesized that AMPK played a role in the ER stress response, and subsequent upregulation of CHOP and DR5. Furthermore, the results of the present study revealed that co-treatment with 3-BP and TRAIL induced apoptosis in breast cancer cells in a Bax- and caspase-dependent manner.

Materials and methods

Reagents and antibodies

Recombinant human TRAIL was purchased from Genentech Inc. (San Francisco, CA, USA). 3-BP, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and propidium iodide (PI) were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). The ATP assay kit was purchased from Merck KGaA. The AMPK inhibitor Compound C was purchased from Selleck Chemicals (Houston, TX, USA). A rabbit polyclonal antibody against DR5 (1:1,000 dilution; cat. no. ab199357) and a rabbit monoclonal antibody against CHOP (1:2,000 dilution; cat. no. ab179823) were purchased from Abcam (Cambridge, MA, USA). The rabbit polyclonal antibody against caspase-3 (1:1,000 dilution; cat. no. LBP72217) was obtained from Enzo Life Sciences, Inc. (Farmingdale, NY, USA). The GRP78 (1:1,000 dilution; cat. no. SC-3177) polyclonal antibody was purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). The rabbit polyclonal antibodies against AMPK-α1 (1:500 dilution; cat. no. 10929-2-AP), Bax (1:5,000 dilution; cat. no. 50599-2-Ig) and Bcl-2 (1:1,000 dilution; cat. no. 12789-1-AP) were supplied by ProteinTech Group, Inc. (Chicago, IL, USA). The rabbit polyclonal antibody against phosphorylated (p)-AMPKα (Thr172) (1:1,000 dilution; cat. no. 50081) was purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). All reagents were prepared to the recommended dilutions according to the manufacturer's instructions.

Cell culture

The human breast cancer cell lines MDA-MB-231 and MCF-7 were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), penicillin (10 U/ml), and streptomycin (100 U/ml). Cells were maintained at 37°C in a humidified atmosphere containing 5% CO2.

Cell viability assay

Cells were seeded in 96-well plates (6×103 cells/well) and treated with different concentrations of 3-BP (0, 40, 80, 160 and 320 µmol/l) or TRAIL (25, 50, 100, 200 and 400 ng/ml) for 24 h. Phosphate-buffered saline (PBS) containing 5 mg/ml MTT (15 µl) was added to each well, and the cells were incubated for a further 4 h. Subsequently, the medium was then replaced with dimethyl sulfoxide (DMSO; Biosharp, Inc., Hefei, China; 150 µl/well) in order to solubilize the formazan crystals. Finally, absorbance was determined at 490 nm using a plate reader (Synergy HT, Inc. Winooski, VT, USA).

PI staining

Cells were seeded in 12-well plates (1.5×105 cells/well) and incubated for 24 h, until the cells reached exponential phase. Subsequently, cells were treated for 24 h with various concentrations of 3-BP, TRAIL or both. Cells were subsequently stained using propidium iodide (PI; 600 µl/well) for 2 h and then analyzed using flow cytometry (Accuri™ C6 system; BD Biosciences, Franklin Lakes, NJ, USA).

ATP quantification

CellTiter-Glo Luminescent Cell Viability Assay kit (Promega, Madison, WI, USA) was used to investigate ATP levels, according to the manufacturer's instructions. Cells (1.5×105/well) were seeded in 12-well plates for 24 h and then treated with different concentrations of 3-BP for 4 h at 37°C. Following this, cells were collected and then lysed using radioimmunoprecipitation assay (RIPA) lysis buffer for 10 min on ice. Cell lysates were subsequently centrifuged at 13,225 × g for 5 min at 4°C. A nucleotide-releasing buffer (100 µl/well), ATP-monitoring enzyme (10 µl/well), and cell lysate (30 µl/well) were added to 96-well plates. Developed signals were then detected using a Luminoskan luminometer and a Varioskan™ Flash spectral scanning multimode reader (Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Cell surface staining

Cells (2.5×105/ml) were seeded in 6-well plates, treated with PBS or 3-BP (80 µmol/l), and then incubated for 24 h. Subsequently, non-specific antibody binding sites were blocked using PBS containing 10% FBS for 20 min. Cells were then washed with PBS, re-suspended in 200 µl PBS, aliquoted into two tubes and incubated with 20 µl of an antibody-containing solution (rabbit anti-DR5 in PBS with 1% FBS; dilution 1:1,000) for 30 min on ice. Subsequently, the cells were washed twice with PBS, pelleted and incubated with 100 µl FITC-conjugated goat anti-rabbit IgG (1:100 dilution; cat. no. BL033A; Biosharp) for 30 min on ice. Cell surface staining was investigated via flow cytometry using the Accuri™ C6 flow cytometer (BD Biosciences).

Western blot analysis

Harvested cells were washed with PBS and then lysed using RIPA buffer for 30 min on ice. Subsequently, cell lysates were centrifuged at 13,225 × g for 30 min at 4°C. The proteins were determined by BCA assay. Subsequently, proteins (50 µg) were loaded per lane, and then separated on a 10 or 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred onto a nitrocellulose membrane (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Membranes were then blocked using 5% skimmed milk in PBS for 4 h at 20–25°C and incubated overnight at 4°C with the primary antibodies. After washing with Tris-buffered saline containing 1% Tween®−20 (Beyotime Institute of Biotechnology, Haimen, China), membranes were incubated with the corresponding secondary antibodies. β-actin was used as the loading control.

In vivo experiments

In order to investigate the antitumor effect of 3-BP and TRAIL, female nude mice (BALB/c; 4–5-weeks old and 18–20 g) were used. Mice were purchased from the Animal Experimental Center of Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China) and maintained under specific pathogen-free conditions (26–28°C, air pressure difference was 10–20 kPa, 10-h light/14-h dark cycle, food and water were taken ad libitum). Mice were injected subcutaneously with MCF-7 cells (4×106 cells/mouse) to induce tumor formation. A total of 20 mice with tumors of >100 mm3 were randomly divided into four groups (five per group), and injected intraperitoneally with either PBS (0.2 ml), 3-BP (8 mg/kg), TRAIL (0.1 mg/kg) or both 3-BP (8 mg/kg) and TRAIL (8 mg/kg) every 4 days. Body weight was monitored prior to each injection. Tumor volume was calculated using the following formula: Length × width2/2. Mice were sacrificed by cervical dislocation after 28 days of treatment. Following treatment for 28 days, the solid tumors were resected from mice, stored in 4% formalin solution, cut into sections and subsequently subjected to with hematoxylin and eosin (H&E) or TUNEL staining. All procedures performed in this study involving animals were in accordance with the ethical standards of the Institutional Animal Care and Use Committee of Bengbu Medical College.

Statistical analysis

Data are expressed as the mean ± the standard error of the mean (SEM). Statistical analyses were performed using SPSS 16.0 software (SPSS, Inc., Chicago, IL, USA). The difference among groups was calculated by one-way analysis of variance (ANOVA) followed by Least Significant Difference (LSD) test. P<0.05 was considered to indicate a statistically significant difference.

Results

3-BP enhances TRAIL-induced apoptosis in breast cancer cells

MCF-7 and MDA-MB-231 cells, were selected as they are estrogen/progesterone receptor positive and negative, respectively, which frequently exhibit different drug sensitivities. To investigate the effects of 3-BP and TRAIL on cell viability, MCF-7 and MDA-MB-231 cells were treated with various concentrations of 3-BP and TRAIL for 24 h. The results of the MTT assay and PI staining analyses demonstrated that 3-BP (80 and 160 µmol/l) and TRAIL (400 ng/ml) significantly inhibited cell viability (Fig. 1A). Notably, concomitant treatment with 3-BP (80 µmol/l) and TRAIL (200 ng/ml) inhibited cell viability to a greater extent compared with treatment with 3-BP or TRAIL alone (P<0.05, Fig. 1B and C). Therefore, the results indicated that co-treatment with 3-BP and TRAIL synergistically induced apoptosis in breast cancer cells.

3-BP inhibits ATP generation and upregulates the expression of DR5

Taking the above-mentioned results into consideration, we investigated whether 3-BP affected the activity and/or expression of one or more mediators in the enhancement of the anticancer activity of TRAIL in breast cancer cells. To investigate this, we determined whether 3-BP affected ATP levels and/or the expression of DR5. It was demonstrated that treatment with 3-BP induced ATP depletion in breast cancer cells (Fig. 2A). Similarly, DR5 staining revealed that the expression of DR5 was enhanced in breast cancer cells treated with 3-BP compared with control cells (Fig. 2B).

3-BP upregulates CHOP, GRP78 and the phosphorylation of AMPK and augments TRAIL-induced Bax and caspase-3 levels

Previous studies have demonstrated that 3-BP induced apoptosis in hepatocellular carcinoma cells via ER stress (12). Therefore, the present study aimed to investigate whether 3-BP induced ER stress in breast cancer cells by determining the expression of ER stress-associated proteins in cells treated with 3-BP. The results of western blot analysis revealed that CHOP and GRP78 levels were enhanced in breast cancer cells following treatment with 3-BP (Fig. 3A). In addition, previous studies have demonstrated that 3-BP induced apoptosis via the disruption of energy metabolism (20). Therefore, the present study aimed to investigate the level of p-AMPK in cells treated with 3-BP. The results of western blot analysis revealed that treatment with 3-BP increased levels of p-AMPK in a dose-dependent manner, as well as the AMPK-α levels overall (Fig. 3A). Furthermore, the expression levels of Bax in MCF-7 cells and caspase-3 in MDA-MB-231 cells were increased in breast cancer cells following co-treatment with 3-BP and TRAIL. However, co-treatment with 3-BP and TRAIL decreased the expression of Bcl-2 in breast cancer cells (Fig. 3B).

The AMPK inhibitor Compound C reduces the TRAIL-synergizing effect of 3-BP on cell growth inhibition and apoptosis

Subsequently, we investigated whether 3-BP sensitized breast cancer cells to growth inhibition and apoptosis by TRAIL via the AMPK activation. MCF-7 and MDA-MB-231 cells were treated with Compound C (an AMPK inhibitor), and 3-BP, TRAIL or both 3-BP and TRAIL. The results of the MTT assays demonstrated that the viability of cells treated with either 3-BP, TRAIL or 3-BP and TRAIL and Compound C did not exhibit a significant difference compared with the control group (Fig. 4A). Similar results were obtained using PI staining, which indicated the number of apoptotic cells upon each treatment (Fig. 4B). Therefore, the results suggested that Compound C attenuated the TRAIL-synergistic effect of 3-BP.

Compound C inhibits 3-BP-induced upregulation of DR5, GRP78, CHOP and p-AMPK apoptosis-associated proteins

To investigate the regulatory role of AMPK in mediating the effects of 3-BP associated with the upregulation of ER stress- and apoptosis-associated proteins, MCF-7 and MDA-MB-231 cells were treated with Compound C. Membrane receptor staining revealed that the fluorescence intensity of DR5 did not increase in breast cancer cells following treatment with Compound C (Fig. 5A). In addition, the results demonstrated that DR5, CHOP, GRP78 and p-AMPK levels did not significantly increase in cells treated with various concentrations of 3-BP and Compound C compared with the control cells (Fig. 5B). Similarly, the expression of Bax and Bcl-2 in MCF-7 cells and caspase-3 levels in MDA-MB-231 cells treated with Compound C, 3-BP, TRAIL, or 3-BP and TRAIL did not demonstrate a significant increase compared to with the control cells (Fig. 5C). These results suggested that AMPK activation may be involved in the ER stress response induced by 3-BP in breast cancer cells.

Antitumor efficacy of 3-BP and TRAIL in tumor xenografts

MCF-7 cells were inoculated hypodermically into the forelimb of nude mice to induce the formation of xenograft tumors. Once the tumors reached a mean volume of ~100 mm3, mice matched for tumor volumes were divided into 4 groups and treated with either PBS, 3-BP, TRAIL or 3-BP and TRAIL. Following 28 days of treatment, the tumor volumes of mice treated with either PBS, 3-BP, TRAIL, or 3-BP and TRAIL were ~1,200±100, ~850±71, ~700±77 and ~232±40 mm3, respectively (Fig. 6A). To investigate the hepatotoxicity and nephrotoxicity of the aforementioned treatments, the levels of serological markers including aspartate aminotransferase (AST), alanine-aminotransferase (ALT), blood urea nitrogen (BUN) and creatinine (Cr) were determined. The results revealed that mice did not exhibit marked levels of toxicity following treatment with either PBS, 3-BP, TRAIL or 3-BP and TRAIL, as the expression of these markers was unchanged (Fig. 6B). The results of H&E staining demonstrated that necrosis occurred in the tumors of groups treated with 3-BP, TRAIL or both 3-BP and TRAIL, and the necrotic area in the group treated with both 3-BP and TRAIL was larger compared with the other treatment groups (Fig. 6C). H&E staining of the liver and kidney revealed that no evident damage was induced following the treatments (Fig. 6C). Finally, TUNEL staining demonstrated that the number of apoptotic cells in the group treated with 3-BP and TRAIL was increased compared with the other groups (Fig. 6D). Therefore, the results suggested that the antitumorigenic effect of 3-BP and TRAIL was associated with low hepatotoxicity and nephrotoxicity in vivo.

Discussion

Breast cancer is an aggressive malignancy. It is the most frequently diagnosed cancer and its incidence has been increasing in both economically developed and developing countries (22). Numerous therapeutic strategies are available for the treatment of patients with breast cancer, including chemotherapy, surgery and radiotherapy (23). However, the mortality rate for patients with breast cancer remains high. Therefore, novel and effective therapies for the treatment of patients with breast cancer are required. It has been demonstrated that TRAIL is an effective anticancer agent that inhibits the proliferation of cancer cells and tumor growth in xenograft models (2426). However, loss or mutation of the death receptors targeted by TRAIL may lead to drug resistance (27). Therefore, it is important to determine the molecular mechanisms underlying TRAIL-induced apoptosis in order to develop novel therapeutic agents that circumvent resistance (28,29).

The ER stress response pathway regulates cancer cell fate, and may induce autophagy, senescence, or apoptosis (30). Protein sensors such as PKR-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1) and activator transcription factor 6 (ATF6) initiate the ER stress response and regulate the unfolded protein response (UPR) under both pathological and physiological conditions (31). Specifically, the PERK/eukaryotic initiation factor 2 (eIF2α)/activator transcription factor 4 (ATF4) axis upregulates CHOP, which subsequently induces apoptosis (32,33). The expression of DR5 is regulated by CHOP, the predominant regulator of the ER stress response. Several previous studies point in this direction: Lu et al revealed that ER stressors regulated the transcription of DR5 via the UPR mediator, CHOP (15). In addition, Guo et al demonstrated that tunicamycin sensitized human colon cancer cells to TRAIL-induced apoptosis via the JNK-CHOP-mediated upregulation of DR5 expression (13). Chen et al determined that the expression of DR5 in human esophageal cancer cells is regulated by the ATF4-CHOP-DR5 axis (14). In addition, treatment with caffeic acid phenethyl ester upregulated the protein level of DR5 and promoted apoptosis in hepatocarcinoma Hep3B cells through CHOP (34). While ER stress sensors and mediators have been revealed to be involved in the regulation of death receptors, the molecular mechanisms underlying 3-BP and TRAIL-induced apoptosis in human breast cancer cells have not yet been clarified. In the present study, it was demonstrated that treatment with 3-BP and TRAIL in MCF-7 and MDA-MB-231 breast cancer cells is associated with the suppression of cell viability in a dose-dependent manner. Furthermore, the results revealed that co-treatment with 3-BP and TRAIL significantly increased apoptosis compared with 3-BP or TRAIL alone, thus suggesting that 3-BP and TRAIL exhibit a synergistic anticancer effect both in vitro and in a tumor xenograft model. Furthermore, 3-BP was demonstrated to induce the ER stress response and subsequently to upregulate GRP78 and CHOP levels in MCF-7 and MDA-MB-231 cells. Simultaneously, 3-BP was revealed to increase the protein expression level of DR5, which subsequently enhanced the sensitivity of cells to TRAIL. These data suggested that the anticancer efficacy of 3-BP in human breast cancer cells is regulated via the activation of ER stress and the upregulation of DR5, which, in turn, sensitizes cells to TRAIL treatment.

There are three types of cell death, namely apoptosis, autophagic cell death and necrosis, largely defined by the morphology of the dying cells (35). The death receptor and mitochondrial pathways play major roles in apoptotic cell death despite the existence of additional regulatory pathways associated with apoptosis. DR5 is upregulated in ER stress-induced apoptosis and is an important factor in this mechanism (35). DR5 binds to its ligands and then induces apoptosis via recruitment of the caspase-activation platform. UPR sensitizes cancer cells to TRAIL-induced apoptosis by enhancing the enzymatic activity of caspase-8 and caspase-3/7. Conversely, mitochondrial outer membrane permeabilization (MMOP) has an important role in the mitochondrial pathway of apoptosis (36). MMOP is regulated by members of the Bcl-2 family. Bcl-2 proteins are classified into three groups: The pro-apoptotic effector proteins (e.g., Bax and Bak), anti-apoptotic Bcl-2 proteins (e.g., Bcl-2, Bcl-xL and Mcl-1), and BH3-only proteins (e.g., Bad, Bim, Bid and Noxa) (37). The results of the present study suggested that co-treatment with 3-BP and TRAIL decreased the expression of Bcl-2 in MCF-7 cells and upregulated the expression of Bax in MCF-7 cells and caspase-3 in MDA-MB-231 cells. Bax expression was investigated in MCF-7 cells, as these cells do not express caspase-3 (38). The results of the present study indicated that the co-treatment with 3-BP and TRAIL may be involved in caspase-3- and Bcl-2 family-mediated apoptosis in breast cancer cells.

AMPK is a sensor of cellular energy status that regulates metabolism in cellular processes. AMPK is activated by changes in the adenosine mono-phosphate (AMP)/ATP or adenosine di-phosphate (ADP)/ATP ratio (16,17). Numerous studies indicated that activated AMPK induced apoptosis in pancreatic cancer cells, renal carcinoma Caki cells, neuroblastoma and colorectal cancer cells (3942). Furthermore, previous studies demonstrated that the inhibition of glycolysis, or caloric restriction may promote AMPK-induced apoptosis in cancer cells (43,44), which indicated that the activation and phosphorylation of AMPK could be used as a therapeutic strategy for the treatment of cancer. We hypothesized that 3-BP, an inhibitor of glycolysis, activated AMPK through an effect on cellular energy status. Western blot assays demonstrated that levels of p-AMPK were increased following treatment with 3-BP in a concentration-dependent manner. However, the expression of AMPK-α did not show a significant difference post-treatment. To investigate the association between p-AMPK and ER stress response, MCF-7 and MDA-MB-231 cells were treated with Compound C, an inhibitor of AMPK. Western blot analysis revealed that the increased expression levels of ER stress response associated proteins following treatment with 3-BP and TRAIL was attenuated following treatment with Compound C. In addition, the viability of cells treated with Compound C and 3-BP and/or TRAIL significantly decreased, while the expression of Bcl-2 family proteins (Bcl-2 and Bax) and caspase-3 did not significantly differ compared with the control group. These results indicated that the 3-BP induced ER stress response may be associated with the activation and phosphorylation of AMPK.

In conclusion, the results of the present study demonstrated that 3-BP, a hexokinase II inhibitor that interferes with glycolysis, upregulated GRP78, CHOP and DR5 expression levels and enhanced TRAIL-induced apoptosis in MCF-7 and MDA-MB-231 breast cancer cells. These results revealed that the ER stress response had a crucial role in sensitizing human breast cancer cells to TRAIL-induced apoptosis, potentially via the activation and phosphorylation of AMPK. Further studies are required to determine whether the simultaneous treatment with 3-BP and TRAIL may represent a novel therapeutic strategy for patients with cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Science Foundation of China (no. 81372899), the Natural Science Foundation of Anhui Province (no. 1508085MH166), the Provincial Science and Technology Cooperation Project of Anhui Province (no. 1503062024), the Foundation of Bengbu Medical College (BYKY1692) and the Key Project of Natural Science Research for College and University of Anhui Province (no. KJ2016A486).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

HL and SZ conceived and designed the experiments. YC and LW performed the experiments. XZ, XL, YC, SZ, LZ, SZ and HL analyzed the data. Image processing was conducted by QL and QP. YC wrote and proofread the paper. SZ and HL revised the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All procedures performed in this study involving animals were in accordance with the ethical standards of the Bengbu Medical College Experimental Animal Ethics Committee.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

TRAIL

tumor necrosis factor-related apoptosis-inducing ligand

DR5

death receptor 5

3-BP

3-bomopyruvate

CHOP

CCAAT/enhancer-binding protein-homologous protein

ER

endoplasmic reticulum

GRP78

glucose-regulated protein 78

DMSO

dimethyl sulfoxide

AMPK

AMP activated protein kinase

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide

PI

propidium iodide

PBS

phosphate-buffered saline

DMEM

Dulbecco's modified Eagle's medium

PVDF

polyvinyl difluoride

H&E

hematoxylin and eosin

ATP

adenosine tri-phosphate

References

1 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Niraula S and Ocana A: Mechanism of drug resistance in relation to site of metastasis: Meta-analyses of randomized controlled trials in advanced breast cancer according to anticancer strategy. Cancer Treat Rev. 50:168–174. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Hellwig CT and Rehm M: TRAIL signaling and synergy mechanisms used in TRAIL-based combination therapies. Mol Cancer Ther. 11:3–13. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Ashkenazi A and Herbst RS: To kill a tumor cell: The potential of proapoptotic receptor agonists. J Clin Invest. 118:1979–1990. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, Chin W, Jones J, Woodward A, Le T, et al: Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med. 5:157–163. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Ho TF and Chang CC: A promising ‘TRAIL’ of tanshinones for cancer therapy. Biomedicine. 5:232015. View Article : Google Scholar : PubMed/NCBI

7 

Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA, Marsters SA, Blackie C, Chang L, McMurtrey AE, Hebert A, et al: Safety and antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest. 104:155–162. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Kim EH, Yoon MJ, Kim SU, Kwon TK, Sohn S and Choi KS: Arsenic trioxide sensitizes human glioma cells, but not normal astrocytes, to TRAIL-induced apoptosis via CCAAT/enhancer-binding protein homologous protein-dependent DR5 upregulation. Cancer Res. 68:266–275. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Liu H, Jiang CC, Lavis CJ, Croft A, Dong L, Tseng HY, Yang F, Tay KH, Hersey P and Zhang XD: 2-Deoxy-D-glucose enhances TRAIL-induced apoptosis in human melanoma cells through XBP-1-mediated upregulation of TRAIL-R2. Mol Cancer. 8:1222009. View Article : Google Scholar : PubMed/NCBI

10 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Warburg O: On the origin of cancer cells. Science. 123:309–314. 1956. View Article : Google Scholar : PubMed/NCBI

12 

Yu SJ, Yoon JH, Yang JI, Cho EJ, Kwak MS, Jang ES, Lee JH, Kim YJ, Lee HS and Kim CY: Enhancement of hexokinase II inhibitor-induced apoptosis in hepatocellular carcinoma cells via augmenting ER stress and anti-angiogenesis by protein disulfide isomerase inhibition. J Bioenerg Biomembr. 44:101–115. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Guo X, Meng Y, Sheng X, Guan Y, Zhang F, Han Z, Kang Y, Tai G, Zhou Y and Cheng H: Tunicamycin enhances human colon cancer cells to TRAIL-induced apoptosis by JNK-CHOP-mediated DR5 upregulation and the inhibition of the EGFR pathway. Anticancer Drugs. 28:66–74. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Chen P, Hu T, Liang Y, Li P, Chen X, Zhang J, Ma Y, Hao Q, Wang J, Zhang P, et al: Neddylation inhibition activates the extrinsic apoptosis pathway through ATF4-CHOP-DR5 axis in human esophageal cancer cells. Clin Cancer Res. 22:4145–4157. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Lu M, Lawrence DA, Marsters S, Acosta-Alvear D, Kimmig P, Mendez AS, Paton AW, Paton JC, Walter P and Ashkenazi A: Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science. 345:98–101. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Lopez M, Nogueiras R, Tena-Sempere M and Diéguez C: Hypothalamic AMPK: A canonical regulator of whole-body energy balance. Nat Rev Endocrinol. 12:421–432. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Carling D and Viollet B: Beyond energy homeostasis: The expanding role of AMP-activated protein kinase in regulating metabolism. Cell Metab. 21:799–804. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Graf D, Reinehr R, Kurz AK, Fischer R and Häussinger D: Inhibition of taurolithocholate 3-sulfate-induced apoptosis by cyclic AMP in rat hepatocytes involves protein kinase A-dependent and -independent mechanisms. Arch Biochem Biophys. 415:34–42. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Dhanasekaran DN and Reddy EP: JNK signaling in apoptosis. Oncogene. 27:6245–6251. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Bodur C, Karakas B, Timucin AC, Tezil T and Basaga H: AMP-activated protein kinase couples 3-bromopyruvate-induced energy depletion to apoptosis via activation of FoxO3a and upregulation of proapoptotic Bcl-2 proteins. Mol Carcinog. 55:1584–1597. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Sun Y, Liu Z, Zou X, Lan Y, Sun X, Wang X, Zhao S, Jiang C and Liu H: Mechanisms underlying 3-bromopyruvate-induced cell death in colon cancer. J Bioenerg Biomembr. 47:319–329. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Francis A, Thomas J, Fallowfield L, Wallis M, Bartlett JM, Brookes C, Roberts T, Pirrie S, Gaunt C, Young J, et al: Addressing overtreatment of screen detected DCIS; the LORIS trial. Eur J Cancer. 51:2296–2303. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Huang K, Zhang J, O'Neill KL, Gurumurthy CB, Quadros RM, Tu Y and Luo X: Cleavage by Caspase 8 and mitochondrial membrane association activate the BH3-only protein bid during TRAIL-induced apoptosis. J Biol Chem. 291:11843–11851. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Wilson NS, Yang A, Yang B, Couto S, Stern H, Gogineni A, Pitti R, Marsters S, Weimer RM, Singh M, et al: Proapoptotic activation of death receptor 5 on tumor endothelial cells disrupts the vasculature and reduces tumor growth. Cancer Cell. 22:80–90. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Akazawa Y, Mott JL, Bronk SF, Werneburg NW, Kahraman A, Guicciardi ME, Meng XW, Kohno S, Shah VH, Kaufmann SH, et al: Death receptor 5 internalization is required for lysosomal permeabilization by TRAIL in malignant liver cell lines. Gastroenterology. 136(2365–2376): e1–e7. 2009.

27 

Pennarun B, Meijer A, de Vries EG, Kleibeuker JH, Kruyt F and de Jong S: Playing the DISC: Turning on TRAIL death receptor-mediated apoptosis in cancer. Biochim Biophys Acta. 1805:123–140. 2010.PubMed/NCBI

28 

Knoll G, Bittner S, Kurz M, Jantsch J and Ehrenschwender M: Hypoxia regulates TRAIL sensitivity of colorectal cancer cells through mitochondrial autophagy. Oncotarget. 7:41488–41504. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Iaboni M, Russo V, Fontanella R, Roscigno G, Fiore D, Donnarumma E, Esposito CL, Quintavalle C, Giangrande PH, de Franciscis V, et al: Aptamer-miRNA-212 conjugate sensitizes NSCLC cells to TRAIL. Molecular therapy. Mol Ther Nucleic Acids. 5:e2892016. View Article : Google Scholar : PubMed/NCBI

30 

Tameire F, Verginadis II and Koumenis C: Cell intrinsic and extrinsic activators of the unfolded protein response in cancer: Mechanisms and targets for therapy. Semin Cancer Biol. 33:3–15. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Senft D and Ronai ZA: UPR, autophagy, and mitochondria crosstalk underlies the ER stress response. Trends Biochem Sci. 40:141–148. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Hetz C: The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 13:89–102. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Oyadomari S and Mori M: Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ. 11:381–389. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Dilshara MG, Jayasooriya RG, Park SR, Choi YH, Choi IW and Kim GY: Caffeic acid phenethyl ester enhances TRAIL-mediated apoptosis via CHOP-induced death receptor 5 upregulation in hepatocarcinoma Hep3B cells. Mol Cell Biochem. 418:13–20. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L and Kroemer G: Cell death modalities: Classification and pathophysiological implications. Cell Death Differ. 14:1237–1243. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Brahmbhatt H, Oppermann S, Osterlund EJ, Leber B and Andrews DW: Molecular pathways: Leveraging the BCL-2 interactome to kill cancer cells-mitochondrial outer membrane permeabilization and beyond. Clin Cancer Res. 21:2671–2676. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Green DR and Llambi F: Cell death signaling. Cold Spring Harb Perspect Biol. 7:pii: a006080. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Jänicke RU: MCF-7 breast carcinoma cells do not express caspase-3. Breast Cancer Res Treat. 117:219–221. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Lu PH, Chen MB, Ji C, Li WT, Wei MX and Wu MH: Aqueous Oldenlandia diffusa extracts inhibits colorectal cancer cells via activating AMP-activated protein kinase signalings. Oncotarget. 7:45889–45900. 2016.PubMed/NCBI

40 

Lennon JC, Butini S, Campiani G, O'Meara A, Williams DC and Zisterer DM: Involvement of AMP-activated protein kinase in mediating pyrrolo-1,5-benzoxazepine-induced apoptosis in neuroblastoma cells. Invest New Drugs. 34:663–676. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Jiang Z, Chen X, Chen K, Sun L, Gao L, Zhou C, Lei M, Duan W, Wang Z, Ma Q and Ma J: YAP inhibition by resveratrol via activation of AMPK enhances the sensitivity of pancreatic cancer cells to gemcitabine. Nutrients. 8:pii: E546. 2016. View Article : Google Scholar

42 

Han MA, Min KJ, Woo SM, Seo BR and Kwon TK: Eupafolin enhances TRAIL-mediated apoptosis through cathepsin S-induced down-regulation of Mcl-1 expression and AMPK-mediated Bim upregulation in renal carcinoma Caki cells. Oncotarget. 7:65707–65720. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Meynet O, Zunino B, Happo L, Pradelli LA, Chiche J, Jacquin MA, Mondragón L, Tanti JF, Taillan B, Garnier G, et al: Caloric restriction modulates Mcl-1 expression and sensitizes lymphomas to BH3 mimetic in mice. Blood. 122:2402–2411. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Pradelli LA, Bénéteau M, Chauvin C, Jacquin MA, Marchetti S, Muñoz-Pinedo C, Auberger P, Pende M and Ricci JE: Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation. Oncogene. 29:1641–1652. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 40 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Wei L, Zhang X, Liu X, Chen Y, Zhang S, Zhou L, Li Q, Pan Q, Zhao S, Zhao S, et al: 3‑Bromopyruvate sensitizes human breast cancer cells to TRAIL‑induced apoptosis via the phosphorylated AMPK‑mediated upregulation of DR5 Corrigendum in /10.3892/or.2022.8264. Oncol Rep 40: 2435-2444, 2018
APA
Chen, Y., Wei, L., Zhang, X., Liu, X., Chen, Y., Zhang, S. ... Liu, H. (2018). 3‑Bromopyruvate sensitizes human breast cancer cells to TRAIL‑induced apoptosis via the phosphorylated AMPK‑mediated upregulation of DR5 Corrigendum in /10.3892/or.2022.8264. Oncology Reports, 40, 2435-2444. https://doi.org/10.3892/or.2018.6644
MLA
Chen, Y., Wei, L., Zhang, X., Liu, X., Chen, Y., Zhang, S., Zhou, L., Li, Q., Pan, Q., Zhao, S., Liu, H."3‑Bromopyruvate sensitizes human breast cancer cells to TRAIL‑induced apoptosis via the phosphorylated AMPK‑mediated upregulation of DR5 Corrigendum in /10.3892/or.2022.8264". Oncology Reports 40.5 (2018): 2435-2444.
Chicago
Chen, Y., Wei, L., Zhang, X., Liu, X., Chen, Y., Zhang, S., Zhou, L., Li, Q., Pan, Q., Zhao, S., Liu, H."3‑Bromopyruvate sensitizes human breast cancer cells to TRAIL‑induced apoptosis via the phosphorylated AMPK‑mediated upregulation of DR5 Corrigendum in /10.3892/or.2022.8264". Oncology Reports 40, no. 5 (2018): 2435-2444. https://doi.org/10.3892/or.2018.6644