Open Access

Imatinib‑induced apoptosis of gastric cancer cells is mediated by endoplasmic reticulum stress

  • Authors:
    • Jung Lim Kim
    • Dae‑Hee Lee
    • Soyeon Jeong
    • Bo Ram Kim
    • Yoo Jin Na
    • Seong Hye Park
    • Min Jee Jo
    • Yoon A. Jeong
    • Sang Cheul Oh
  • View Affiliations

  • Published online on: December 19, 2018     https://doi.org/10.3892/or.2018.6945
  • Pages: 1616-1626
  • Copyright: © Kim 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

Imatinib is a powerful tyrosine kinase inhibitor that specifically targets BCR‑ABL, c‑KIT, and PDGFR kinases, and is used in the treatment of chronic myelogenous leukemia, gastrointestinal stromal tumors, and other types of cancers. However, the possible anticancer effects of imatinib in gastric cancer have not yet been explored. The present study evaluated the in vitro effects of imatinib on gastric cancer cells and determined the molecular mechanism underlying these effects. We determined that imatinib induced mitochondria‑mediated apoptosis of gastric cancer cells by involving endoplasmic reticulum (ER) stress‑associated activation of c‑Jun NH2‑terminal kinase (JNK). We also found that imatinib suppressed cell proliferation in a time‑ and dose‑dependent manner. Cell cycle analysis revealed that imatinib‑treated AGS cells were arrested in the G2/M phase of the cell cycle. Moreover, imatinib‑treated cells exhibited increased levels of phosphorylated JNK, and of the transcription factor C/EBP homologous protein, an ER stress‑associated apoptotic molecule. Results of cell viability assays revealed that treatment with a combination of imatinib and chemotherapy agents irinotecan or 5‑Fu synergistically inhibited cell growth, compared with treatment with any of these drugs alone. These data indicated that imatinib exerted cytotoxic effects on gastric cancer cells by inducing apoptosis mediated by reactive oxygen species generation and ER stress‑associated JNK activation. Furthermore, we revealed that imatinib induced the apoptosis of gastric cancer cells by inhibiting platelet‑derived growth factor receptor signaling. Collectively, our results strongly support the use of imatinib in the treatment of treating gastric cancer.

Introduction

Gastric cancer is a leading cause of cancer-related deaths worldwide (1). Recent studies have revealed that immune-targeting therapy improves the survival of patients with gastric cancer. Numerous chemotherapy regimens have been clinically examined for treating gastric cancer; however, there is an urgent need for novel therapeutic agents for the treatment of gastric cancer.

Imatinib mesylate (imatinib) is a powerful tyrosine kinase inhibitor that specifically targets BCR-ABL, KIT, and platelet-derived growth factor receptor (PDGFR) kinases and is used for treating chronic myelogenous leukemia (CML), gastrointestinal stromal tumors (GISTs), and other types of cancers (2,3). However, the cellular and molecular mechanisms underlying the antitumor effects of imatinib are unknown.

PDGFR-α and PDGFR-β are transmembrane tyrosine kinase receptors whose ligands play critical roles in cancer cell migration and proliferation (46). The PI3K/AKT pathway is an important downstream signaling pathway of PDGFR that plays important roles in promoting cell proliferation, suppressing cell motility, and inhibiting cell apoptosis. Increased PDGFR expression is also detected in various solid cancers (7).

The endoplasmic reticulum (ER) plays a major role in protein synthesis and maturation, lipid synthesis, calcium homeostasis, and protein folding (8). Misfolded and unfolded proteins, particularly secretory and transmembrane proteins produced in the ER, induce an evolutionarily conserved unfolded protein response (UPR), an ER stress pathway, which in turn induces various pathological events such as inflammation, aging, and neurodegenerative disorders. The UPR, which improves folding, is a survival response triggered by cells to restore ER homeostasis. Chronic accumulation of unfolded proteins in the ER may lead to death receptor-independent and mitochondria-mediated apoptotic pathways (912).

At low concentrations, intracellular oxidants function as signal transducers to induce growth factors, hypoxia, and other receptor-ligand systems (13,14). However, at concentrations above threshold levels, these oxidants induce damage of lipids, proteins, RNA, and DNA, thus triggering cell death through apoptosis and/or necrosis (1517). Reactive oxygen species (ROS) such as oxygen ions and peroxides are chemically reactive molecules containing oxygen that are formed as natural by-products of normal oxygen metabolism and play important roles in cell signaling and homeostasis (18). Biological functions of ROS and their potential roles in cancer development and disease progression have been investigated over the past several decades. ROS mediate cancer cell apoptosis induced by various anticancer agents and other stimuli (19). However, various studies have also shown that anticancer agents increase apoptosis of malignant cells by decreasing ROS production (20).

However, the role of ROS, and mechanisms underlying their antitumorigenic function in solid tumors, particularly gastric cancer, are unknown. In the present study, we evaluated whether imatinib exerted antitumorigenic effects on gastric cancer cells by inducing apoptosis. Our results indicated that IRE1α-c-Jun NH2-terminal kinase (JNK)- and C/EBP homologous protein (CHOP)-associated ER stress was involved in imatinib-induced apoptosis of gastric cancer cells, suggesting that imatinib is a potential chemotherapeutic agent for treating gastric cancer.

Materials and methods

Reagents and antibodies

Imatinib was purchased from Novartis International AG (Basel, Switzerland). JNK inhibitor SP600125 was obtained from EMD/Merck KGaA (Darmstadt, Germany). Rabbit antibodies against PARP (1:1,000 dilution; cat. no. 9542), caspase-3 (1:1,000 dilution; cat. no. 9662), Bid (1:1,000 dilution; cat. no. 2002), Bim (1:1,000 dilution; cat. no. 2819), Puma (1:1,000 dilution; cat. no. 4976), Noxa (1:1,000 dilution; cat. no. 14766), p-JNK (1:1,000 dilution; cat. no. 9251), JNK (1:1,000 dilution; cat. no. 9252), PDGFR-α (1:1,000 dilution; cat. no. 3164), PDGFR-β (1:1,000 dilution; cat. no. 3169), p-PDGFR-α (1:1,000 dilution; cat. no. 4547), p- PDGFR-β (1:1,000 dilution; cat. no. 3161), AKT (1:1,000 dilution; cat. no. 9272), p-AKT (1:1,000 dilution; cat. no. 4060), 4EBP1 (1:1,000 dilution; cat. no. 9644), p-4EBP1 (1:1,000 dilution; cat. no. 2855), p70 S6 kinase (1:1,000 dilution; cat. no. 2708), p-p70 S6 kinase (Ser371) (1:1,000 dilution; cat. no. 9208), p-p70 S6 kinase (Thr 389) (1:1,000 dilution; cat. no. 9205), mTOR (1:1,000 dilution; cat. no. 2983), p38 MAPK (1:1,000 dilution; cat. no. 9212), p-p38 MAPK (1:1,000 dilution; cat. no. 9211), p44/42 MAPK (ERK1/2) (137F5) (1:1,000 dilution; cat. no. 4695), p-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (1:1,000 dilution; cat. no. 4370), eIF2α (D7D3) (1:1,000 dilution; cat. no. 5324), p-eIF2α (Ser51) (1:1,000 dilution; cat. no. 3597) and GRP94 (1:1,000 dilution; cat. no. 2104) and IRE1α (1:1,000 dilution; cat. no. 3294) were obtained from Cell Signaling Technology, Inc. (Beverly, MA, USA). Rabbit antibody against p-IRE1 α (1:1,000 dilution; cat. no. ab48187) were obtained from Abcam (Cambridge, UK). Mouse antibodies against BCL2-associated X protein (Bax) ATF6 (1:1,000 dilution; cat. no. sc-20067), Bcl-2 (1:1,000 dilution; cat. no. sc-509), Survivin (1:1,000 dilution; cat. no. sc-17779) and CHOP (GADD 153) (1:1,000 dilution; cat. no. sc-7351) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Mouse antibodies against ATF6 (1:1,000 dilution; cat. no. NBP1-40256) were obtained from Novus Biologicals, LLC (Littleton, CO, USA). Anti-actin (1:2,000 dilution; cat. no. A2228) and DCFH-DA (cat. no. D6883) were purchased from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany).

Cell culture

Human gastric cancer cell line AGS was obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA), and SNU-638 and MKN45 cell lines were purchased from the Korean Cell Line Bank (Seoul, Korea). CHOP−/− and corresponding wild-type MEF cell lines were provided by Dr Randal J. Kaufman (Sanford Burnham Medical Research Institute, CA, USA). Gastric cancer cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS) and 1% antibiotic-antimycotic solution (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified atmosphere of 5% CO2.

Cell viability assay

Cell growth rate and viability were determined by performing 3-(4,5-dimethylthiazol-2-ly)-2,5-diphenyl tetrazolium bromide (MTT)-based colorimetric assays (Sigma-Aldrich; Merck KGaA). Cells were grown in 96-well plates containing medium supplemented with 10% fetal bovine serum at a density of 1×104 cells/well. The medium in each well contained different concentrations of imatinib. After 48 h, 50 µl MTT solution was added to each well, and the cells were incubated at 37°C for 4 h. Next, the supernatant was aspirated, and the dye was solubilized using 200 µl dimethyl sulfoxide (DMSO). Cell viability was determined according to the manufacturer's instructions by measuring the absorbance in individual wells at 595 nm using a microplate reader.

Cell cycle and apoptosis analyses

Cell cycle was analyzed by performing propidium iodide (PI) staining. Adherent cells were harvested by treatment with trypsin and were fixed with 5 mM EDTA and 85% ethanol. Fixed cells were incubated with 50 µg/ml PI and 20 µg/ml RNase at 37°C for 30 min and were analyzed by performing flow cytometry. Apoptotic cells were stained using an Annexin V-fluorescein isothiocyanate (FITC) kit (Beckman Coulter, Inc., Brea, CA, USA), according to the manufacturer's instructions, and were then analyzed.

Western blot analysis

Cells harvested 48 h after imatinib treatment were centrifuged at 14,000 × g and 4°C for 5 min and were washed twice with phosphate-buffered saline (PBS). Next, the cells were lysed in 30 µl ice-cold RIPA buffer and were centrifuged again at 14,000 × g and 4°C for 30 min. The obtained pellet was stored at −70°C for further analysis. Protein concentrations were determined using the Bradford method (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Lysates corresponding to equal protein amounts were boiled in sample buffer for 5 min. Next, the samples were resolved by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) on 10 or 12% gels, with 50 µg total protein loaded in each lane of the gel. The resolved proteins were transferred onto a 0.45-µm nitrocellulose membranes. The membranes were blocked with 5% non-fat dry milk in Tris-buffered saline with Tween-20 (TBST) buffer for 2 h to prevent non-specific binding, and was then washed three times with TBST for 10 min each. Next, the membrane was incubated with specific primary antibodies, followed by incubation at 4°C overnight with horseradish peroxidase-conjugated secondary antibody and then incubated with anti-mouse (1:2,500 dilution; cat. no. 170-6516; Bio-Rad Laboratories) or anti-rabbit (1:2,500 dilution; cat. no. 7074; Cell Signaling Technology, Inc.) specific polyclonal secondary antibodies for 2 h at 4°C. Then, the membrane was washed three times with TBST for 10 min each. Signals were detected by exposure to X-ray film using an EZ-Western Lumi Pico kit (DoGEN, Seoul, Korea; cat. no. DG-WP-250).

ROS generation

ROS generation was assessed in cells treated with imatinib for 30 or 60 min. Intracellular ROS levels were determined using 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA), which oxidizes within cells to fluorescent dichlorofluorescein. Control and imatinib-treated cells were incubated with 100 µM DCFH-DA for 30 min at 37°C.

Next, the cells were fixed in 3.7% paraformaldehyde for 10 min at room temperature. Subsequently, the cells were co-stained with 2 µM 4′,6-diamidino-2-phenylindole (Molecular Probes; Thermo Fisher Scientific, Inc.) at 37°C. After washing three times with PBS, the cells were mounted using VECTASHIELD mounting medium (Vector Laboratories, Inc., Burlingame, CA, USA), and immunofluorescence was detected using a confocal microscope (Carl Zeiss AG, Oberkochen, Germany). Then, MEF of MEF CHOP−/− cells were stained with 100 µM DCFH-DA for 30 min at 37°C. The cells were then transferred to FACS tubes and analyzed using the FL1 channel on a FACScan cytometer (BD Biosciences, Franklin Lakes, NJ, USA).

Statistical analysis

Statistical analysis was performed using GraphPad InStat 5 software (GraphPad Software, Inc., San Diego, CA, USA). Statistical significance was determined by one-way analysis of variance (ANOVA) followed by Bonferroni post hoc test for multiple comparisons or Student's t-test. For all tests, P<0.05 or P<0.01 were considered to indicate statistically significant differences.

Results

Imatinib decreases the viability of gastric cancer cells

To investigate the antitumor activity of imatinib against gastric cancer cells (AGS, MKN45 and SNU638 cells), the cells were incubated at different concentrations of imatinib for 48 h and their viability was determined by performing MTT assays. Results of the MTT assays revealed that imatinib treatment significantly decreased cell viability in a dose-dependent manner. Statistically significant cytotoxic effects were observed after treatment with >50 µM imatinib (Fig. 1A). Next, we determined the effect of imatinib on cell cycle progression. For this, the cells were treated with various concentrations of imatinib, and cell cycle progression was assessed after 48 h by performing flow cytometry with PI staining. Imatinib treatment for 48 h significantly increased the percentage of cells in the sub-G1 and G2/M phases of the cell cycle, in a dose-dependent manner. These data indicated that imatinib treatment arrested gastric cancer cells in the sub-G1 phase of the cell cycle (Fig. 1B), and also indicated that imatinib exerted anti-proliferative effects on gastric cancer cells.

Imatinib induces apoptosis of gastric cancer cells

We performed Annexin V/PI double staining to evaluate whether imatinib induced apoptosis of the three gastric cancer cell lines. Results of Annexin V/PI double staining revealed that the percentage of Annexin V/PI-positive cells significantly increased, indicating that imatinib treatment increased the early apoptosis of gastric cancer cells (Fig. 2A). Next, we performed western blot analysis to confirm imatinib-induced apoptosis of gastric cancer cells, and to identify the underlying mechanism. Imatinib treatment for 48 h increased poly(ADP-ribose) polymerase (PARP) cleavage, and cleaved caspase-3 and −9 expression in AGS cells (Fig. 2B). Bcl-2 is an anti-apoptotic protein, and Bax is a pro-apoptotic protein. We observed that Bax and Bcl-2 levels were unaffected in imatinib-treated AGS cells. Pro-apoptotic Bcl-2 family proteins (Noxa and Bid) were induced in imatinib-treated cells. Similar results were obtained for imatinib-treated MKN45 and SNU638 gastric cancer cells (Fig. 2C and D). These results indicated that imatinib treatment induced apoptosis of gastric cancer cells.

Imatinib decreases PDGFR signaling in gastric cancer cells

Imatinib is a powerful tyrosine kinase inhibitor that specifically targets BCR-ABL, KIT, and PDGFR kinases, and is used for treating CML, GISTs, and other types of cancers (3,2124). In the present study, c-KIT and BCR-ABL expression was not detected in the three gastric cancer cell lines (data not shown). Next, we examined the effects of imatinib on PDGFR signaling in AGS cells and found that imatinib efficiently suppressed PDGFR-induced downstream signaling (Fig. 3A). Imatinib inhibited the phosphorylation of PDGFR-α, PDGFR-β, AKT and p70S6K (Ser371). Notably, the phosphorylation of PDGFR-β decreased more markedly than that of PDGFR-α in imatinib-treated cells. However, no significant changes were observed in total protein levels in imatinib-treated cells.

Imatinib induces apoptosis by phosphorylating JNK in gastric cancer cells

Various cellular stresses and stimuli induce mitogen-activated protein kinase (MAPK) signaling that contributes to apoptotic induction (25). To determine the effect of imatinib on the expression and activity of molecules involved in MAPK signaling, we treated AGS cells with 30, 50 and 100 µM imatinib for 48 h and performed western blot analysis. Imatinib treatment increased JNK phosphorylation in a dose-dependent manner, without affecting total JNK levels, but did not affect p38 and ERK phosphorylation (Fig. 3B). To verify the involvement of JNK in imatinib-induced apoptosis, the cells were co-treated with a JNK pathway inhibitor (SP600125) and imatinib (50 µM). Treatment of gastric cancer cells with SP600125 significantly decreased their imatinib-induced apoptosis due to JNK inhibition (Fig. 3C). Moreover, treatment with SP600125 completely abolished imatinib-induced PARP activation (Fig. 3D). These results indicated that imatinib-induced apoptosis was mediated by the JNK-MAPK pathway.

Imatinib-induced apoptosis of gastric cancer cells is mediated by ER stress induction and ROS generation

AGS and SNU638 gastric cancer cells were used to investigate mechanisms underlying imatinib-induced apoptosis. Recent research has indicated that ER stress plays a crucial role in the regulation of apoptosis. To confirm that ER stress was involved in imatinib-induced apoptosis, we examined the expression of ER stress-associated proteins, namely, activating transcription factor 6 (ATF6), glucose-regulated protein 94 (GRP-74), eIF2α, p-eIF2α, IRE1α, p-IRE1α, and CHOP in imatinib-treated AGS and SNU638 cells. Notably, imatinib induced the expression of GRP94, p-eIF2α, p-IRE1 α and CHOP in a concentration- and time-dependent manner in both AGS and SNU638 cells (Fig. 4A-C), indicating that imatinib-induced apoptosis was mediated by the ER stress pathway. A recent study indicated that several anticancer agents induced oxidative stress by promoting ROS generation, thus inducing cytotoxicity and apoptosis in cancer cells (19). To investigate the potential role of ROS in imatinib-induced apoptosis, we determined ROS production in imatinib-treated gastric cancer cells using DCFH-DA. Imatinib-treated cells produced significant fluorescence signals. In addition, imatinib induced ROS production in a time-dependent manner (Fig. 4D). CHOP−/− MEFs were resistant to the effects of PARP-1 cleavage compared with wild-type CHOP MEFs (Fig. 4E). Imatinib reduced the elevated ROS levels in CHOP−/− MEFs, providing evidence that imatinib reduced endogenous ROS levels in a CHOP-dependent manner (Fig. 4F). To examine the relationship between JNK and ER-stress activation during imatinib exposure, AGS cells were pretreated with an inhibitor of JNK (SP600125), then treated with imatinib. Pretreatment with SP600125 significantly decreased imatinib-induced CHOP (Fig. 4G). These results collectively indicated that imatinib-induced apoptosis of gastric cancer cells was mediated by ROS generation and ER stress.

Imatinib and chemotherapy agents synergistically decrease growth of gastric cancer cells

We examined the effect of combination treatment with imatinib and chemotherapy agents on the viability of gastric cancer cells. Treatment with imatinib or irinotecan alone resulted in limited suppression of the viability of gastric cancer cells (<15%) at 24 h (Fig. 5A and C). In contrast, combination treatment with a fixed concentration of irinotecan and varying concentrations of imatinib significantly decreased the viability of gastric cancer cells. We used 50 µM imatinib for performing subsequent experiments since this concentration of imatinib significantly reduced cell viability during combination treatment with irinotecan, with only a slight decrease in the percentage of viable cells. Next, we examined whether the combined effect of irinotecan and imatinib treatment on the viability of gastric cancer cells was mediated by apoptotic induction. Co-treatment of gastric cancer cells with irinotecan and imatinib significantly increased the percentage of apoptotic cells, whereas treatment with irinotecan or imatinib alone only slightly increased the percentage of apoptotic cells (Fig. 5D) compared with that among untreated cells. Next, we evaluated the effect of combination treatment with 0.5–2 µM 5-Fu and 30–50 µM imatinib on the viability of AGS cells by performing MTT assays. Combination treatment with 50 µM imatinib and 2 µM 5-Fu significantly inhibited the growth of AGS cells compared with treatment with imatinib or 5-Fu alone (Fig. 5B and E). Similarly, imatinib also increased irinotecan or 5-Fu-mediated inhibition of MKN45 and SNU638 cell growth (Fig. 5F-I). These findings indicated that combined treatment with imatinib and chemotherapy agents (irinotecan or 5-Fu) effectively induced the apoptosis of gastric cancer cells.

Discussion

This is the first study, to the best of our knowledge, to show that imatinib induces the apoptosis of gastric cancer cells through the JNK/ROS/ER stress pathway (Fig. 6). It is important to determine mechanisms underlying imatinib-induced death of gastric cancer cells. Imatinib is a tyrosine kinase inhibitor with activity against the BCR-ABL fusion oncoprotein, PDGFR, and c-KIT in GISTs, prostate cancer, malignant gliomas, and ovarian cancer (2629). In the present study, c-KIT and BCR-ABL were not expressed in the gastric cancer cell lines (data not shown). Mireskandari et al reported that expression of c-KIT in gastric cancer appears to be a very unlikely event (30). Imatinib was revealed to induce apoptosis in, and may modulate the metastasis of, gastric cancer cells by upregulating KAI1 expression (31). Biswas et al reported that imatinib induced programmed cell death in retinal ganglion cells by inhibiting PDGFR-mediated PI3K/AKT signaling (32).

Another study suggested that the effect of imatinib on the migration of medulloblastoma cells was not mediated by early induction of apoptosis (33). A recent study indicated that treatment with low and high concentrations of imatinib induced cell growth arrest and apoptosis, respectively, in glioblastoma cells. Consistently, results of the present study revealed that imatinib induced apoptosis at relatively high concentrations (20–100 µM), and inhibited cell metastasis at lower concentrations (1–10 µM) (data not shown). However, the mechanism underlying imatinib-induced cell death is not completely understood. To clearly determine the mechanism underlying imatinib-induced apoptosis, we identified the possible involvement of a MAPK subfamily protein, since accumulating evidence suggests important regulatory roles of MAPKs in different physiological and pathological processes (34). It was observed that imatinib treatment activated JNK in the late stage, but did not activate ERK. Imatinib-induced activation of JNK/MAPK in the present study indicated that these proteins perform distinct physiological functions in determining the fate of gastric cancer cells. Similarly, Chang et al reported that treatment with high-dose imatinib induced JNK phosphorylation by elevating ROS production in melanoma cells (34). A study by Yu et al revealed that treatment with 5 mM STI571 interrupted cytoprotective 42/44 MAPK activation response in human myeloid leukemia cells (35). These results indicated that iron chelators activate different target MAPKs in different cell types.

ER stress is suggested to be a significant contributor to cell death. JNK activation plays a significant role in UPR (36,37). Induction of the UPR in the ER, which causes ER stress, induces several pathological and physiological alterations such as glucose depletion, hypoxia, and oxidative stress. Han et al reported that imatinib decreased JNK activation and ER stress in the liver of a diabetic mouse model (38). However, imatinib induced ER stress in gastric cancer cells. Moreover, we found that imatinib induced the apoptosis of gastric cancer cells by modulating ER stress. This is the first study to report that imatinib induced significant apoptosis of gastric cancer cells, which is mediated by ER stress. Imatinib was also revealed to trigger ER stress in CML cells expressing BCR-ABL (39). In contrast, Zhang et al reported that imatinib did not induce ER stress in Ph1-positive leukemia cells (40). These results indicated that imatinib induced ER stress in a cell-specific manner. IRE1α-mediated JNK activation in the ER induced apoptosis. Notably, we found that imatinib-induced apoptosis of gastric cancer cells was mediated by the JNK/ROS/ER stress pathway.

Generally, for patients with gastric cancer, therapy is combined with cytotoxic chemotherapy and targeted therapy (41). Therefore, it is very important to find a target agent that has synergistic effects while reducing toxicity of cytotoxic agents. Clinical studies on the combination of imatinib, cisplatin and 5-fluoruracil or capecitabine have been reported (42). In one of these clinical trials, the safety and tolerability of combination of imatinib plus 5-fluoruracil was confirmed.

In summary, it was revealed that imatinib is a potent antitumor agent that induces ER stress-mediated apoptosis of gastric cancer cells. We observed that imatinib induced ER stress by activating IRE1α, p-JNK, and CHOP. To the best of our knowledge, this is the first study to determine mechanisms underlying imatinib-induced apoptosis of gastric cancer cells. However, further studies are required to determine the antitumorigenic effects of imatinib in animal models. According to pathological features, it is necessary to further study the anticancer effect of imatinib in gastric cancer cells. Thus, our results indicated that imatinib-induced activation of ER stress is a novel therapeutic strategy for treating gastric cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the Korea University Guro Hospital (O1600121) and supported by a grant from the National Research Foundation (NRF) of Korea funded by the Korean government (MSIP) (NRF-2017R1A2B2011684).

Availability of data and materials

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

Authors' contributions

SCO and JLK conceived and designed the study and critically revised the manuscript. JLK and DHL designed and performed the experiments, analyzed the data and were major contributors in writing the manuscript. SJ and BRK supervised the western blot experiments and also performed the statistical analysis. YJN, SHP, MJJ and YAJ performed the Annexin V/PI apoptosis assay experiments. SJ, BRK, YJN, SHP, MJJ and YAJ provided advice on the experiments and technical assistance. SCO supervised the study. 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

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

BAX

BCL2-associated X protein

CHOP

C/EBP-homologous protein

eIF2α

eukaryotic translation initiation factor 2α

ER

endoplasmic reticulum

FITC

fluorescein isothiocyanate

JNK

c-Jun NH2-terminal kinase

MAPK

mitogen-activated protein kinase

MTT

3-(4,5-dimethylthiazol-2-ly)-2,5-diphenyl tetrazolium bromide

PARP

poly(ADP-ribose) polymerase

PBS

phosphate-buffered saline

PI

propidium iodide

ROS

reactive oxygen species

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Nakatani H, Araki K, Jin T, Kobayashi M, Sugimoto T, Akimori T, Namikawa T, Okamoto K, Nakano T, Okabayashi T, et al: STI571 (Glivec) induces cell death in the gastrointestinal stromal tumor cell line, GIST-T1, via endoplasmic reticulum stress response. Int J Mol Med. 17:893–897. 2006.PubMed/NCBI

3 

Kadivar A, Kamalidehghan B, Akbari Javar H, Karimi B, Sedghi R and Noordin MI: Antiproliferation effect of imatinib mesylate on MCF7, T-47D tumorigenic and MCF 10A nontumorigenic breast cell lines via PDGFR-β, PDGF-BB, c-Kit and SCF genes. Drug Des Dev Ther. 11:469–481. 2017. View Article : Google Scholar

4 

Donovan J, Abraham D and Norman J: Platelet-derived growth factor signaling in mesenchymal cells. Front Biosci. 18:106–119. 2013. View Article : Google Scholar

5 

Cumpănas AA, Cimpean AM, Ferician O, Ceausu RA, Sarb S, Barbos V, Dema A and Raica M: The involvement of PDGF-B/PDGFRβ axis in the resistance to antiangiogenic and antivascular therapy in renal cancer. Anticancer Res. 36:2291–2295. 2016.PubMed/NCBI

6 

Kazlauskas A: PDGFs and their receptors. Gene. 614:1–7. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Pietras K, Sjöblom T, Rubin K, Heldin CH and Ostman A: PDGF receptors as cancer drug targets. Cancer Cell. 3:439–443. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Minamino T, Komuro I and Kitakaze M: Endoplasmic reticulum stress as a therapeutic target in cardiovascular disease. Circ Res. 107:1071–1082. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Tsai YC and Weissman AM: The unfolded protein response, degradation from endoplasmic reticulum and cancer. Genes Cancer. 1:764–778. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Malhotra JD, Miao H, Zhang K, Wolfson A, Pennathur S, Pipe SW and Kaufman RJ: Antioxidants reduce endoplasmic reticulum stress and improve protein secretion. Proc Natl Acad Sci USA. 105:18525–18530. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Thuerauf DJ, Marcinko M, Gude N, Rubio M, Sussman MA and Glembotski CC: Activation of the unfolded protein response in infarcted mouse heart and hypoxic cultured cardiac myocytes. Circ Res. 99:275–282. 2006. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Mansfield KD, Simon MC and Keith B: Hypoxic reduction in cellular glutathione levels requires mitochondrial reactive oxygen species. J Appl Physiol. 97:1358–1366. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Waypa GB, Guzy R, Mungai PT, Mack MM, Marks JD, Roe MW and Schumacker PT: Increases in mitochondrial reactive oxygen species trigger hypoxia-induced calcium responses in pulmonary artery smooth muscle cells. Circ Res. 99:970–978. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Ostadal P, Elmoselhi AB, Zdobnicka I, Lukas A, Elimban V and Dhalla NS: Role of oxidative stress in ischemia-reperfusion-induced changes in Na+,K+-ATPase isoform expression in rat heart. Antioxid Redox Signal. 6:914–923. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Nojiri H, Shimizu T, Funakoshi M, Yamaguchi O, Zhou H, Kawakami S, Ohta Y, Sami M, Tachibana T, Ishikawa H, et al: Oxidative stress causes heart failure with impaired mitochondrial respiration. J Biol Chem. 281:33789–33801. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Morgan WA: DNA single-strand breakage in mammalian cells induced by redox cycling quinones in the absence of oxidative stress. J Biochem Toxicol. 10:227–232. 1995. View Article : Google Scholar : PubMed/NCBI

18 

Devasagayam TP, Tilak JC, Boloor KK, Sane KS, Ghaskadbi SS and Lele RD: Free radicals and antioxidants in human health: Current status and future prospects. J Assoc Physicians India. 52:794–804. 2004.PubMed/NCBI

19 

Gong K, Xie J, Yi H and Li W: CS055 (Chidamide/HBI-8000), a novel histone deacetylase inhibitor, induces G1 arrest, ROS-dependent apoptosis and differentiation in human leukaemia cells. Biochem J. 443:735–746. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Chang YJ, Huang YP, Li ZL and Chen CH: GRP78 knockdown enhances apoptosis via the down-regulation of oxidative stress and Akt pathway after epirubicin treatment in colon cancer DLD-1 cells. PLoS One. 7:e351232012. View Article : Google Scholar : PubMed/NCBI

21 

Atari-Hajipirloo S, Nikanfar S, Heydari A and Kheradmand F: Imatinib and its combination with 2,5-dimethyl-celecoxibinduces apoptosis of human HT-29 colorectal cancer cells. Res Pharm Sci. 12:67–73. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Akram AM, Iqbal Z, Akhtar T, Khalid AM, Sabar MF, Qazi MH, Aziz Z, Sajid N, Aleem A, Rasool M, et al: Presence of novel compound BCR-ABL mutations in late chronic and advanced phase imatinib sensitive CML patients indicates their possible role in CML progression. Cancer Biol Ther. 18:214–221. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Mahon FX: Discontinuation of TKI therapy and ‘functional’ cure for CML. Best Prac Res Clin Haematol. 29:308–313. 2016. View Article : Google Scholar

24 

Abu-Amna M, Awadie H and Bar-Sela G: Imatinib-induced gastrointestinal vascular ectasia in a patient with advanced gIST: Case report and literature review. Anticancer Res. 36:6151–6154. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, He C and Pan H: p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 344:174–179. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Balachandran VP, Cavnar MJ, Zeng S, Bamboat ZM, Ocuin LM, Obaid H, Sorenson EC, Popow R, Ariyan C, Rossi F, et al: Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med. 17:1094–1100. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Nakatani H, Kobayashi M, Jin T, Taguchi T, Sugimoto T, Nakano T, Hamada S and Araki K: STI571 (Glivec) inhibits the interaction between c-KIT and heat shock protein 90 of the gastrointestinal stromal tumor cell line, GIST-T1. Cancer Sci. 96:116–119. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Matei D, Emerson RE, Lai YC, Baldridge LA, Rao J, Yiannoutsos C and Donner DD: Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer. Oncogene. 25:2060–2069. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Li J, Kleeff J, Guo J, Fischer L, Giese N, Büchler MW and Friess H: Effects of STI571 (gleevec) on pancreatic cancer cell growth. Mol Cancer. 2:322003. View Article : Google Scholar : PubMed/NCBI

30 

Mireskandari M, Shafaii AF, Kayser G and Kayser K: Lack of CD117 and rare bcl-2 expression in stomach cancer by immunohistochemistry. An immunohistochemical study with review of the literature. Diagn Pathol. 1:72006. View Article : Google Scholar : PubMed/NCBI

31 

Shandiz SA, Farasati S, Saeedi B, Baghbani-Arani F, Asl EA, Keshavarz-Pakseresht B, Rahimi A, Assadi A, Noorbazargan H, RahimpourHesari M, et al: Up regulation of KAI1 gene expression and apoptosis effect of imatinib mesylate in gastric adenocarcinoma (AGS) cell line. Asian Pac J Trop Dis. 6:120–125. 2016. View Article : Google Scholar

32 

Biswas SK, Zhao Y and Sandirasegarane L: Imatinib induces apoptosis by inhibiting PDGF-but not insulin-induced PI 3-kinase/Akt survival signaling in RGC-5 retinal ganglion cells. Mol Vis. 15:1599–1610. 2009.PubMed/NCBI

33 

Abouantoun TJ and MacDonald TJ: Imatinib blocks migration and invasion of medulloblastoma cells by concurrently inhibiting activation of platelet-derived growth factor receptor and transactivation of epidermal growth factor receptor. Mol Cancer Ther. 8:1137–1147. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Chang SP, Shen SC, Lee WR, Yang LL and Chen YC: Imatinib mesylate induction of ROS-dependent apoptosis in melanoma B16F0 cells. J Dermatol Sci. 62:183–191. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Yu C, Krystal G, Varticovksi L, McKinstry R, Rahmani M, Dent P and Grant S: Pharmacologic mitogen-activated protein/extracellular signal-regulated kinase kinase/mitogen-activated protein kinase inhibitors interact synergistically with STI571 to induce apoptosis in Bcr/Abl-expressing human leukemia cells. Cancer Res. 62:188–199. 2002.PubMed/NCBI

36 

Lenna S, Han R and Trojanowska M: Endoplasmic reticulum stress and endothelial dysfunction. IUBMB Life. 66:530–537. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Kim JL, Lee DH, Na YJ, Kim BR, Jeong YA, Lee SI, Kang S, Joung SY, Lee SY, Oh SC and Min BW: Iron chelator-induced apoptosis via the ER stress pathway in gastric cancer cells. Tumour Biol. 37:9709–9719. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Han MS, Chung KW, Cheon HG, Rhee SD, Yoon CH, Lee MK, Kim KW and Lee MS: Imatinib mesylate reduces endoplasmic reticulum stress and induces remission of diabetes in db/db mice. Diabetes. 58:329–336. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Pattacini L, Mancini M, Mazzacurati L, Brusa G, Benvenuti M, Martinelli G, Baccarani M and Santucci MA: Endoplasmic reticulum stress initiates apoptotic death induced by STI571 inhibition of p210 bcr-abl tyrosine kinase. Leuk Res. 28:191–202. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Zhang X, Inukai T, Akahane K, Hirose K, Kuroda I, Honna H, Goi K, Kagami K, Tauchi T, Yagita H, et al: Endoplasmic reticulum stress inducers, but not imatinib, sensitize Philadelphia chromosome-positive leukemia cells to TRAIL-mediated apoptosis. Leuk Res. 35:940–949. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Lopez A, Harada K, Mizrak Kaya D and Ajani JA: Current therapeutic landscape for advanced gastroesophageal cancers. Ann Transl Med. 6:78–96. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Mayr M, Becker K, Schulte N, Belle S, Hofheinz R, Krause A, Schmid RM, Röcken C and Ebert MP: phase I study of imatinib, cisplatin and 5-fluoruracil or capecitabine in advanced esophageal and gastric adenocarcinoma. BMC Cancer. 12:587–593. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 41 Issue 3

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
Kim JL, Lee DH, Jeong S, Kim BR, Na YJ, Park SH, Jo MJ, Jeong YA and Oh SC: Imatinib‑induced apoptosis of gastric cancer cells is mediated by endoplasmic reticulum stress. Oncol Rep 41: 1616-1626, 2019
APA
Kim, J.L., Lee, D., Jeong, S., Kim, B.R., Na, Y.J., Park, S.H. ... Oh, S.C. (2019). Imatinib‑induced apoptosis of gastric cancer cells is mediated by endoplasmic reticulum stress. Oncology Reports, 41, 1616-1626. https://doi.org/10.3892/or.2018.6945
MLA
Kim, J. L., Lee, D., Jeong, S., Kim, B. R., Na, Y. J., Park, S. H., Jo, M. J., Jeong, Y. A., Oh, S. C."Imatinib‑induced apoptosis of gastric cancer cells is mediated by endoplasmic reticulum stress". Oncology Reports 41.3 (2019): 1616-1626.
Chicago
Kim, J. L., Lee, D., Jeong, S., Kim, B. R., Na, Y. J., Park, S. H., Jo, M. J., Jeong, Y. A., Oh, S. C."Imatinib‑induced apoptosis of gastric cancer cells is mediated by endoplasmic reticulum stress". Oncology Reports 41, no. 3 (2019): 1616-1626. https://doi.org/10.3892/or.2018.6945