Purvalanol induces endoplasmic reticulum stress-mediated apoptosis and autophagy in a time-dependent manner in HCT116 colon cancer cells

  • Authors:
    • Ajda Coker-Gürkan
    • Elif Damla Arisan
    • Pınar Obakan
    • Kübra Akalın
    • Utku Özbey
    • Narcin Palavan-Unsal
  • View Affiliations

  • Published online on: April 21, 2015     https://doi.org/10.3892/or.2015.3918
  • Pages: 2761-2770
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Purvalanol, a novel cyclin-dependent kinase inhibitor, is referred to as a strong apoptotic inducer which causes cell cycle arrest in various cancer cells such as prostate, breast and colon cancer cell lines. Various physiological and pathological conditions such as glucose starvation, inhibition of protein glycosylation and oxidative stress may cause an accumulation of unfolded proteins in the endoplasmic reticulum (ER), leading to the unfolded protein response (UPR) and autophagy. Lacking proteosomal function on aggregates of unfolded proteins, ER stress may induce autophagic machinery. Autophagy, an evolutionarily conserved process, is characterized by massive degradation of cytosolic contents. In the present study, our aim was to determine the time-dependent, ER-mediated apoptotic and autophagy induction of purvalanol in HCT 116 colon cancer cells. Fifteen micromoles of purvalanol induced a reduction in cell viability by 20 and 35% within 24 and 48 h, respectively. HCT 116 colon cancer cells were exposed to purvalanol, which activated ER stress via upregulation of PERK, IRE1α gene expression, eIF-2α phosphorylation and ATF-6 cleavage at early time-points in the HCT 116 colon cancer cells. Moreover, we determined that during purvalanol-mediated ER stress, autophagic machinery was also activated prior to apoptotic cell death finalization. Beclin-1 and Atg-5 expression levels were upregulated and LC3 was cleaved after a 6 h purvalanol treatment. Purvalanol induced mitochondrial membrane potential loss, caspase-7 and caspase-3 activation and PARP cleavage following a 48 h treatment. Thus, we conclude that the anticancer effect of purvalanol in HCT 116 cells was due to ER stress-mediated apoptosis; however, purvalanol triggered autophagy, which functions as a cell survival mechanism at early time-points.

Introduction

Cyclin-dependent kinases (CDKs), are members of the serine-threonine protein kinase family and are responsible for taking control of cell cycle regulation in eukaryotic cells. CDKs show their action by interacting with cyclins and different CDK-cyclin complexes regulate the cell cycle in the G1, S and G2/M phases (1). New anticancer therapy strategies refer to the inhibition of CDK-cyclin complexes as an important target to prevent uncontrolled proliferation and induce apoptosis in cancer cells (2). Olomoucine, roscovitine and purvalanol are examples of CDK inhibitors (CDKIs) designed and investigated for their apoptotic potential on cancer cells (3). Purvalanol is a purine-derived CDKI that binds with a high selectivity and competitively to the ATP binding side of CDK1/2 and leads to G2/M cell cycle arrest (4). Τheir impact on the apoptotic cell death mechanism requires further elucidation. Natural polyamines, putrescine, spermidine and spermine, play essential roles in the regulation of cell growth and proliferation. Increased levels of polyamines in cells are considered to be involved in cancer progression. Intracellular polyamine levels are under the control of several catabolic enzymes, such as spermidine/spermine-N-acetyl transferase (SSAT). Although purvalanol-induced cell cycle arrest and apoptotic cell death were demonstrated in prostate (5), breast (6) and colon cancer cells (7), the exact molecular mechanism of purvanol-induced apoptosis has not been elucidated yet.

Endoplasmic reticulum (ER) is an essential organelle responsible for protein synthesis, folding, post-translational modification of proteins and protein trafficking in eukaryotes (8). ER alerts a self-protective mechanism that is called ER stress during nutrient deprivation, pathogen infection, alterations in redox status, intraluminal Ca2+ levels and folding defective protein conditions (9). ER stress response is initiated by activation of three types of ER membrane receptors; inositol-requiring enzyme 1 (IRE1α), PRKR-like ER kinase (PERK) and activating transcription factor-6 (ATF-6). Activated and released IRE1α acts as an RNase to initiate transcription of XBP1 mRNA and it becomes a transcriptional activator for unfolded protein response (UPR) gene targets, such as BiP and calreticulin (10). Activation of ER membrane receptor PERK, during prolonged UPR, phosphorylates eIF2α to attenuate mRNA transcription within the cell (11). Recently, autophagy, a cytoprotective intracellular degradation system, is assumed to be activated by both IRE1α and PERK as an alternative way to degrade accumulated misfolded proteins (12). Concominantly, during UPR stress, ATF-6 is released from BiP and translocates from the ER to the Golgi. Cleaved ATF-6 migrates to the nucleus and transactivates various chaperones and major ER stress markers such as the CAAT-enhancer binding protein (CHOP) gene. CHOP is a transcription factor that regulates the expression of the Bcl-2 family members. Overexpression of CHOP induces apoptosis and its deficiency has been shown to protect cells from apoptotic cell death. Prolonged severe ER stress could eventually activate caspase-12 and caspase-dependent apoptosis (13). Thus, our aim in the present study was to understand the molecular basis of purvalanol-induced ER stress which leads to cell death in HCT 116 colon cancer cells.

Materials and methods

Drugs and antibodies

Purvalanol was purchased from Tocris Bioscience (Bristol, UK), dissolved in DMSO to make a 10 mM stock solution and stored at −20°C. Phospho-Rb, p53, p21, Bad, Bid, Bik, PUMA, Mcl-1, Bcl-xl, pro-caspase-3, pro-caspase-7, PARP, Beclin-1, p62, LC3, Atg-5, Atg-12, PERK, XBP1, caspase-12, JNK, ATF-6, CHOP, BiP, IRE1α, calnexin, calreticulin, PDI, c-jun, GAPDH and β-actin (each diluted 1:1,000) rabbit antibodies were purchased from Cell Signaling Technology (CST, Danvers, MA, USA). HRP-conjugated secondary anti-rabbit antibodies (diluted 1:3,000) were from CST.

Cell culture

HCT 116 colon cancer cells (CCL 247) were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). Cells were maintained in McCoy’s 5A medium (Gibco, Invitrogen, Carlsbad, CA, USA) with 10% fetal bovine serum (Pan Biotech, Aidenbach, Germany) and 100 U/100 mg/ml penicilin/streptomycin and grown in the presence of 5% CO2 in humidified air at 37°C (Heracell 150; Thermo Electron Corporation, Waltham, MA, USA).

Cell viability assay

The effects of purvalanol on cell viability were determined by colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT; Roche, Indianapolis, IN, USA) assay. Cells were seeded at a density of 1×104 cells/well in 96-well plates, allowed to attach overnight and treated for 24 and 48 h with various concentrations of purvalanol (0–30 μM). After purvalanol treatment, 10 μl of MTT reagent (5 mg/ml) was added to the cell culture medium for 4 h. Following removal of media, 200 μl DMSO was added to dissolve the formazan crystals, which are produced due to activated mitochondria. The absorbance of the suspensions was determined at 570 nm with a microplate reader (Bio-Rad, Hercules, CA, USA).

Trypan blue dye exclusion assay

Cells were seeded on 6-well plates (5×104 cells/well) and treated with purvalanol (15 μM) for 96 h. Every 24 h, the cells were trypsinized and stained with trypan blue and viable and dead cells were counted under light microscopy. Data were platted on a graph indicating the number of cells (y-axis) vs. time (x-axis).

Fluorescence microscopy
PI staining

HCT 116 cells (1×105) were seeded into 12-well plates and treated with purvalanol (0–30 μM) for 24 h. Following a dose-dependent purvalanol treatment, the cells were washed once with 1X PBS and stained with propidium iodide (PI) (50 mg/ml stock concentration in 1X PBS) fluorescent probe and were incubated for 10 min in the dark. Purvalanol-induced cell death was detected with fluorescence microscopy (Olympus, Tokyo, Japan).

DAPI staining

The cells were seeded in 12-well plates at a density of 1×105 cells/well and treated with purvalanol (0–30 μM) for 24 h. After treatment, the cells were washed once with 1X PBS. The cells were stained with 1 μl/ml 4′,6-diamidino-2-phenylindole (dAPI) (1 mg/ml stock concentration in 1X PBS) fluorescent probe and were incubated for 10 min in the dark. Dose-dependent purvalanol-induced nuclear DNA fragmentation was visualized using fluorescence microscopy.

DiOC6 staining

HCT 116 (1×105) cells were seeded into 12-well plates. Following exposure of cells to purvalanol (0–30 μM), they were washed once with 1X PBS, and then stained with 4 nM 3,3′-dihexyloxacarbocyanine iodide [DiOC6(3)] (40 nM stock concentration in DMSO; Calbiochem, La Jolla, CA, USA) fluorescent probe. Mitochondrial membrane potential (MMP) disruption was visualized by fluorescence microscopy.

Cell cycle analysis by PI staining

HCT 116 colon carcinoma cells at a density of 2×105 cells/well were seeded in 6-well plates, and then subsequently treated with purvalanol (15 μM) for 12, 24 and 48 h. Both floating and adherent cells were collected and fixed with 70% ethanol. Following incubation on ice for 30 min, the cells were diluted with 1X PBS. Samples were then centrifuged at 1,200 rpm for 5 min. Pellets were resuspended in 1X PBS, RNase (100 μg/ml) and PI solution (40 μg/ml). Samples were kept for 30 min at 37°C in the dark. Cell cycle distribution was analyzed by Accuri C6 (BD Biosciences, Oxford, UK). 10,000 events/sample were acquired and evaluated using BD Accuri C6 software (BD Biosciences).

CHOP activation by FACS flow analysis

HCT 116 colon carcinoma cells were seeded in 6-well plates and transfected with CHOP promoter (−649/+136) pmCherry-1 plasmid (0.5 mg/ml) using the Fugene6 (Promega, Sunnyvale, CA, USA). After a 24 h transfection, the cells were exposed to purvalanol (15 μM) for 12, 24 and 48 h. Following purvalanol treatment, mCherry-CHOP plasmid-transfected 10,000 cells/sample were determined by flow cytometry using FL-3 (red fluorescence) (FACScan, Attune; Applied Biosciences, Foster City, CA, USA).

GFP-LC3 localization by FACS flow analysis

HCT 116 colon carcinoma cells were seeded in 6-well plates and transfected with a green fluorescent protein (GFP)-tagged LC3-expressing vector (0.5 μ/ml) using Fugene6. After a 24 h transfection, the cells were exposed to purvalanol (15 μM) for 12, 24 and 48 h. Following purvalanol treatment, GFP-LC3 plasmid-transfected 10,000 cells/sample were determined by flow cytometry using FL-1 (green fluorescence) (FACScan, Attune; Applied Biosciences).

Cell death ELISA assay

HCT 116 cells (1×104) were seeded in 96-well plates and treated with 15 μM purvalanol for 12, 24 and 48 h. Cytoplasmic histone-associated-DNA fragments (mono- and oligonucleosomes) were determined using Cell Death detection ELISA Plus assay, according to the manufacturer’s instructions (Roche). Briefly, cell lysates were placed in a streptavidin-coated microplate. A mixture of anti-histone-biotin and anti-DNA-POD was added and incubated for 2 h at 15–25°C. Following the removal of unbound antibodies by a washing procedure, POD was determined photometrically at 405 nm with 2,2′-azino-di-[3-ethylbenzthiazoline sulfonate (6)] diammonium salt (ABTS) as a substrate. In order to determine the DNA fragments following drug treatment in colon carcinoma cells, total DNA content was isolated.

Protein extraction and immunoblotting

HCT 116 colon carcinoma cells were treated with the appropriate concentrations of purvalanol in a time-dependent manner. First, all the samples were washed with ice-cold 1X PBs and lysed on ice in a solution containing 20 mM Tris-HCl (pH 7.5), 150 mM NaCl, Nonidet P-40 0.5%, (v/v), 1 mM EDTA, 0.5 mM PMSF, 1 mM DTT and protease inhibitor cocktail (Complete, Roche). After cell lysis, the cell debris were removed by centrifugation for 15 min at 13,200 rpm, and protein concentrations were determined by the Bradford protein assay (BioRad). Total protein lysates (30 μg) were separated on a 12% SDS-PAGE and transferred onto PVDF membranes (Roche). The membranes were then blocked with 5% milk blocking solution in Tris buffer saline (TBS)-Tween 20 (Sigma) and incubated with appropriate primary and horseradish peroxidase (HRP)-conjugated secondary antibodies (CST) in antibody buffer containing 5% (v/v) milk blocking solution. Following a gentle washing step with 1X TBS-Tween 20, the proteins were analyzed using an enhanced chemiluminescence detection system (ECL). Bands were exposed to Lumi-Film Chemiluminescent Detection (Roche).

Statistical analysis

All the experiments were statistically analyzed by two-way ANOVA using GraphPad Prism 6 (GraphPad Software, La Jolla, CA, USA). Statistically significant results by ANOVA were further analyzed by Bonferroni post-hoc analysis (where indicated). A p<0.05 was considered to indicate a statistically significant result. Error bars in the graphs were generated using ± standard deviation (SD) values. Band intensities were quantified using imageJ software and normalized to β-actin or GAPDH.

Results

Purvalanol inhibits cell viability and proliferation in a dose-dependent manner

In order to understand the time- and dose-dependent effects of purvalanol treatment on HCT 116 cell viability, we performed an MTT assay. According to the MTT assay, 15 μM of purvalanol decreased the cell viability by 25% in HCT 116 cells within 24 h. Exposure to purvalanol for 48 h decreased cell viability by 40% (Fig. 1A). Moreover, PI and DAPI staining of cells after dose-dependent purvalanol treatment for 24 h showed that cell death and nuclear condensation were increased in the HCT 116 colon cancer cells. In addition, we also determined that MMP disruption was increased in dose-dependent manner in the HCT 116 colon cancer cells (Fig. 1B). Cell survival analysis with trypan blue dye exclusion assay showed that 15 μM purvalanol inhibited cell proliferation after 24 h and this cytostatic effect was maintained until 72 h (Fig. 1C).

Purvalanol induces cell cycle arrest in a time-dependent manner

The effect of purvalanol on the cell cycle profile of HCT 116 cells was examined following PI staining by FACS flow analysis. 15 μM of purvalanol increased the subG1 population in a time-dependent manner (12, 24 and 48 h), to 1.7, 2 and 8.9%, respectively (Fig. 2A). In order to confirm FACS flow analysis, we analyzed major cell cycle coordinating proteins such as phosphorylated Rb, p53 and p21 by immunob-lotting. According to the immunoblotting results, purvalanol treatment increased the expression of p53 in a time-dependent manner, but significant upregulation was determined after a 48 h purvalanol treatment in the HCT 116 colon cancer cells. However, expression of p21 was increased only after a 24 h purvalanol treatment. Moreover, protein expression of the phosphorylated form of Rb (807/811) was sharply downregulated only after 12 h in HCT 116 colon cancer cells (Fig. 2B).

Purvalanol activates ER stress

In order to evalute the time-dependent effect of purvalanol on ER stress via activated Ca2+ metabolism, we performed immunoblotting. According to the immunblotting results, purvalanol upregulated both PDI and calnexin expression levels within 1 h of purvalanol exposure in the HCT 116 colon cancer cells. However, calre-ticulin, an ER resident protein with Ca2+ buffering effect, was downregulated in a time-dependent manner following 15 μM of purvalanol treatment (Fig. 3A). We next examined the expression profiles of ER membrane proteins and their downstream target key molecules by immunoblotting. BiP expression was upregulated after a 3 h purvalanol treatment and this effect was increased in a time-dependent manner. Moreover, a 1 h purvalanol treatment induced PERK expression and phosphorylation of eIF-2α within a 3 h drug exposure in the HCT 116 cells. Another ER membrane receptor, IRE1α, was also upregulated following time-dependent purvalanol treatment. Late response of cells to purvalanol treatment related with ER stress was determined by the cleavage of ATF-6 after 12 h. CHOP, a transcriptional target of both cleaved ATF-6 and ATF-4, was found to be significantly upregulated following a 12 h purvalanol treatment (Fig. 3B). In order to examine the purvalanol-induced CHOP activation, we performed FACS flow analysis after transfection of the CHOP promoter (−649/+136) pmCherry-1 plasmid (Fig. 3C). According to FACS flow analysis, purvalanol-induced CHOP activation via ATF-4 transcriptional activity was determined after 48 h.

Purvalanol-induced ER stress activates autophagy in HCT 116 colon cancer cells

To evaluate the effect of purvalanol on autophagic regulation related with its influence on ER membrane receptor activation, we examined the involvement of LC3 via GFP-LC3 plasmid transfection in the HCt 116 cells. According to FACS flow analysis, purvalanol induced GFP-LC3 intensity within a 12 h drug exposure and this effect was maintained for 24 h (Fig. 4A). Moreover, Beclin-1, Atg-5 and Atg-12 expression levels were upregulated; p62 degradation and LC3 cleavage were observed at early time-points following purvalanol treatment in the HCT 116 colon cancer cells (Fig. 4B).

Prolonged purvalanol treatment causes apoptotic cell death by modulating the Bcl-2 family members in HCT 116 colon cancer cells

The apoptotic induction due to purvalanol was determined by Cell Death ELISA assay. As shown in Fig. 5A, exposure of cells to 15 μM purvalanol for 12, 24 and 48 h induced apoptosis by 2.5-, 3.5- and 5.5-fold compared to the untreated control cells (p<0.05 and p<0.001), respectively. The apoptotic induction of purvalanol was found to be caspase-dependent. As shown in Fig. 5B, caspase-7 and caspase-3 were activated by causing PARP cleavage. As shown in Fig. 5C, 15 μM purvalanol for 12, 24 and 48 h induced MMP loss by 75, 55 and 40%, respectively (p<0.05 and p<0.001). To analyze the functional role of Bcl-2 family members in mitochondria-mediated apoptosis due to purvalanol treatment, the modulation of the expression levels of anti- and pro-apoptotic proteins was examined in the HCT 116 cells by western blot analysis. Purvalanol down-regulated Bcl-xl and Mcl-1 and upregulated Bik, Bad, Bid, Bad and PUMA protein expression levels (Fig. D). Therefore, we conclude that purvalanol induced mitochondria-mediated apoptosis by activating caspases and modulating Bcl-2 family members.

Discussion

Dysregulation of molecular machinery of the cell cycle process plays an important role in tumor progression and malignancy (1). Thus, control of the cell cycle by pharmacological CDKIs, is believed to be an important therapeutic target for cancer therapy. CDKIs have been shown to be promising chemotherapeutic agents in various cancer types such as colon (7), prostate (5) and breast cancer (6). Purvalanol, a new generation CDKI, has been shown to cause cell cycle arrest at the G2/M checkpoint of the cell cycle by inhibiting the ATP binding site of CDK1 and CDK2 (14) and to induce apoptosis through activating caspases and cytochrome-c release in gastric cancer cells (15). In our previous study, we determined that 30 μM purvalanol induced apoptotic cell death by modulating the expression of the Bcl-2 family members via activating polyamine catabolic metabolism in HCT 116 colon cancer cells (7). Another previous study from our laboratory showed that 25 μM purvalanol induced apoptosis via activation of caspases, which led to dysfunction of MMP in MCF-7 breast cancer cells (6). A lower dose of purvalanol (15 μM) used in this study also inhibited cell proliferation via cell cycle arrest and induced cell death through MMP loss (Figs. 1 and 2). Although a number of reports have demonstrated the molecular action of roscovitine, there are few research studies concerning purvalanol-induced apoptotic cell death. Thus, clarification of molecular targets of purvalanol treatment on apoptosis and autophagic mechanisms might increase the therapeutic efficiency of the drug.

During homeostatic conditions in eukaryotic cells, ER is essential in biosynthesis and signaling functions, and several resident chaperones, Ca2+ binding proteins and folding enzymes such as calnexin, calreticulin and protein disulfide isomerase (PDI) are known to monitor ER biological functions (16). It was shown that a lack of calnexin within the cell induced a resistant profile against ER stress-mediated apoptosis as it acts with caspase-8 for Bap31 protein cleavage (17). In addition, although overexpression of calreticulin in HeLa cells was shown to enhance the sensitivity against ceramide-induced apoptosis (18), knockout of calreticulin increased the resistance phenotype (19). Moreover, silencing of calreticulin in bladder cancer cells suppressed cell proliferation, migration and attachment, whereas overexpression of calreticulin enhanced cell migration and attachment (20). Thus, in the present study, we determined that purvalanol induced UPR as a first response. Both PDI and calnexin expression profiles were upregulated following a 1 h purvalanol treatment and this effect was observed until 12 h. In addition, calreticulin expression was downregulated at early time-points following purvalanol treatment which modulated Ca2+ levels and cell adhesion (Fig. 3A). These results suggest that purvalanol might first trigger ER stress via ER chaperone activation in HCT 116 colon cancer cells.

Accumulation of unfolded or misfolded proteins in the ER lumen during hypoxia, oxidative injury, a high-fat diet or viral infections was found to induce various intracellular signaling pathways via activation of receptors localized on the ER membrane (9). During ER stress, disassociation of ER membrane receptors from their ligand BiP caused receptor-mediated nuclear activation of various transcription factors such as ATF-6, ATF-4 and CHOP (12). ER stress- activated ER membrane receptor, ATF-6 p90, is activated following BiP disassociation and cleavage to form ATF-6 p50, a regulatory protein of X-box binding protein 1 (XBP-1) mRNA (21). Cleaved and activated ATF-6 p50 protein translocates to the nucleus and triggers UPR target genes such as BiP and XBP1 (22). Similar to this finding, N-butylidenephthalide was shown to induce ER-stress by triggering ATF-6 cleavage and BiP upregulation within 6 h after drug exposure in LNCaP and PC3 prostate cancer cell lines (23). Both ATF-6 and IRE1α trigger the transcription of ER chaperones during ER stress conditions via acting on XBP1 mRNA (24). Moreover, IRE1α interaction with TRAF-2 triggers apoptotic cell death via JNK activation (25). Although 6-shogaol induced apoptosis without any significant effect on IRE1α in SMMC-7721 hepatocellular carcinoma cells (26), 20-O-(β-D-glucopyranosyl)-20(S)-protopanaxadiol activated the IRE1α phosphorylation and splicing of XBP1 in Ht29 human colon cancer cells (27). In addition, upregulation of IRE-1α was determined after a 6-h N-butylidenephthalide treatment in prostate cancer cell lines (23). In the present study, although we demonstrated upregulation of IRE-1α expression and an increase in XBP1 splicing, a significant upregulation of JNK expression was observed after a 24-h purvalanol treatment (Fig. 3B). Concominantly, purvalanol induced apoptotic cell death via activation of caspase-3 and caspase-7 and PARP cleavage (Fig. 5B). Thus, we conclude that short-term purvalanol treatment induces ATF-6-mediated ER stress and long-term purvalanol induces apoptotic cell death through activation of JNK in HCT 116 colon cancer cells.

The third ER membrane receptor, PERK, is dimerized, and auto-phosphorylation of itself activates the phosphorylation of eIF-2α that attenuates protein translation (11). PERK activation and eIF-2α phosphorylation were determined after a 6-h 20-O-(β-D-glucopyranosyl)-20(S)-protopanaxadiol treatment in HT29 human colon cancer cells (27). 6-shogaol induced apoptosis via activation of PERK and its downstream target eIF-2α in SMMC-7721 hepatocellular carcinoma cells (26). In addition, luteolin-induced eIF-2α phosphorylation within a 2 h drug treatment triggered the ER stress-mediated apoptosis in NCI-H460 lung carcinoma cells (28). We determined that PERK expression was upregulated only after a 1 h purvalanol treatment and phosphorylation of eIF-2α was also observed during 1–6 h of purvalanol treatment (Fig. 3B). Purvalanol might induce UPR and attenuate protein translation at early time-points in HCT 116 cells by activating the PERK/eIF-2α/ATF-4 signaling axis.

CHOP, is a key ER stress-induced transcription factor and transcription activation is through ATF-6 and ATF-4 (29). Deficiency of CHOP protected cells from ER-induced apoptotic cell death (30) and overexpression of CHOP stimulated cell cycle arrest and apoptosis (31). Activated CHOP expression has been implicated to repress anti-apoptotic Bcl-2 protein (32) and induce apoptotic cell death by activating Bim (33). We showed that CHOP expression was significantly upregulated after a 6 h purvalanol treatment and this effect was increased in a time-dependent manner (Fig. 3B). Moreover, when we analyzed the activation of CHOP via the CHOP promoter (−649/+136) inserted mCherry expression plasmid, mCherry expression was found to be elevated following a 48 h purvalanol treatment (Fig. 3C). CHOP-mediated ER stress-induced apoptotic cell death occurred after a 48 h purvalanol treatment in the HCT 116 colon cancer cells.

ER stress-induced autophagy is assumed to be activated as an alternative way to degradate accumulated unfolded/misfolded protein aggregates instead of the proteasome degradation system (12). Both IRE1α and PERK pathways were demonstrated to be associated with ER stress-induced autophagy regulation in cancer cells (34). In contrary, other studies reported that only PERK-eIF-2α signaling was involved in autophagy induction by ER stress (35). Autophagy, an evolutionarily conserved process, characterized by massive degradation of cytosolic contents initiates the fusion of autophagosomes to endosomes and lysosomes, which engulf cytoplasmic contents within a double-membrane vacuole (36). This physiological process is carried out through interaction of various molecules such as Bcl-2, Beclin-1, Atg-5 and Atg-12 (37). During autophagic induction, LC3-I (16 kDa) is cleaved to form LC3-II (14 kDa) that is involved in autophagic vacuole membrane formation by interacting with cytoplasmic protein p62 (38). As autophagy is assumed to be a cell survival mechanism against chemotherapeutic drugs in cancer cells, inhibition of autophagy by specific inhibitors induced drug-mediated apoptotic cell death in prostate (39), breast (40) and colon (41) cancer cells. Moreover, in NCI-H460 lung carcinoma cells, although luteolin induced ER stress activated apoptosis, autophagy was shown to be activated and inhibition of autophagy induced apoptotic cell death (28). Thus, in our present study we determined that another CDKI, purvalanol, induced autophagy induction by upregulation of Beclin-1, Atg-5, Atg-12 protein expression and LC3-II formation during a 6–12 h purvalanol treatment (Fig. 4B) only after Perk/eIF-2α induced ER stress.

The inactive state of ER membrane receptors can be stimulated by slight ER stress conditions leading to translation inhibition. Under this situation, autophagy induction could occur as a cell protection mechanism. However, prolonged ER stress can also result in apoptotic cell death through various molecular key players such as caspase-12, JNK and CHOP (42). Our previous studies found that long-term exposure of purvalanol triggered mitochondrial-mediated and caspase-dependent apoptotic cell death in MCF-7 cells following treatment with 25 μM purvalanol (6) and in HCT 116 colon cancer cells after of 30 μM purvalanol (7). In the present study, a lower dose of purvalanol for a long-term exposure triggered caspase-dependent apoptotic cell death via modulation of Bcl-2 family members and this effect was significant after a 24 h purvalanol treatment in the HCT 116 colon cancer cells (Fig. 5A–D). Finally, we conclude that purvalanol induced ER stress-mediated autophagy as a first response in HCT 116 colon cancer cells against purvalanol. Prolonged ER stress activated both CHOP and JNK- mediated apoptotic cell death.

Acknowledgments

The present study was supported by the Istanbul Kultur University Scientific Projects Support Center and the TUBITAK Scientific Projects Support Center (2209 program).

Abbreviations:

ABTS

2,2′-azino-di-[3-ethylbenzthiazoline sulfonate (6)] diammonium salt

ATF-6

activating transcription factor-6

CDK

cyclin-dependent kinase

CDKI

cyclin-dependent kinase inhibitor

CHOP

CAAT/enhancer-binding protein

DiOC6

3,3′-dihexyloxacarbocyanine iodide

DMSO

dimethyl sulfoxide

ER

endoplasmic reticulum

GFP

green fluorescent protein

HRP

horseradish peroxidase

IRE1

inositol-requiring enzyme 1

JNK

c-Jun N-terminal kinase

LC3

microtubule-associated protein 1A/1B-light chain 3

MMP

mitochondrial membrane potential

MTT

3-4,5-dimethyl-2-thiazolyl-2,5-diphenyl-2H-tetrazolium bromide

UPR

unfolded protein response

PERK

PRKR-like ER kinase

PBS

phosphate- buffered saline

PI

propidium iodide

POD

peroxidase

PVDF

polyvinyldifluoride

Rb

retinoblastoma

SDS-PAGE

sodium dodecyl sulphate polyacrylamide gel electrophoresis

TBS

Tris-buffered saline

XBP-1

X-box binding protein 1

References

1 

Harper JW and Adams PD: Cyclin-dependent kinases. Chem Rev. 101:2511–2526. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Knockaert M, Greengard P and Meijer L: Pharmacological inhibitors of cyclin-dependent kinases. Trends Pharmacol Sci. 23:417–425. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Gray N, Détivaud L, Doerig C and Meijer L: ATP-site directed inhibitors of cyclin-dependent kinases. Curr Med Chem. 6:859–875. 1999.PubMed/NCBI

4 

Gray NS, Wodicka L, Thunnissen AM, Norman TC, Kwon S, Espinoza FH, Morgan DO, Barnes G, Le Clerc S, Meijer L, et al: Exploiting chemical libraries, structure, and genomics in the search for kinase inhibitors. Science. 281:533–538. 1998. View Article : Google Scholar : PubMed/NCBI

5 

Arisan ED, Obakan P, Coker-Gurkan A, Calcabrini A, Agostinelli E and Unsal NP: CDK inhibitors induce mitochondria-mediated apoptosis through the activation of polyamine catabolic pathway in LNCaP, DU145 and PC3 prostate cancer cells. Curr Pharm Des. 20:180–188. 2014. View Article : Google Scholar

6 

Obakan P, Arisan ED, Özfiliz P, Çoker-Gürkan A and Palavan-Ünsal N: Purvalanol A is a strong apoptotic inducer via activating polyamine catabolic pathway in MCF-7 estrogen receptor positive breast cancer cells. Mol Biol Rep. 41:145–154. 2014. View Article : Google Scholar

7 

Gürkan AC, Arisan ED, Obakan P and Palavan-Ünsal N: Inhibition of polyamine oxidase prevented cyclin-dependent kinase inhibitor-induced apoptosis in HCT 116 colon carcinoma cells. Apoptosis. 18:1536–1547. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Kaufman RJ: Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev. 13:1211–1233. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Schroder M and Kaufman RJ: ER stress and the unfolded protein response. Mutat Res. 569:29–63. 2005. View Article : Google Scholar

10 

Yoshida H: Molecular biology of the ER stress response. Seikagaku. 76:617–630. 2004.In Japanese. PubMed/NCBI

11 

Harding HP, Zhang Y, Bertolotti A, Zeng H and Ron D: Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol Cell. 5:897–904. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Ogata M, Hino S, Saito A, Morikawa K, Kondo S, Kanemoto S, Murakami T, Taniguchi M, Tanii I, Yoshinaga K, et al: Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol. 26:9220–9231. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Morishima N, Nakanishi K, Takenouchi H, Shibata T and Yasuhiko Y: An endoplasmic reticulum stress-specific caspase cascade in apoptosis. Cytochrome c-independent activation of caspase-9 by caspase-12. J Biol Chem. 277:34287–34294. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Iizuka D, Inanami O, Kashiwakura I and Kuwabara M: Purvalanol A enhances cell killing by inhibiting up-regulation of CDC2 kinase activity in tumor cells irradiated with high doses of X rays. Radiat Res. 167:563–571. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Villerbu N, Gaben AM, Redeuilh G and Mester J: Cellular effects of purvalanol A: a specific inhibitor of cyclin-dependent kinase activities. Int J Cancer. 97:761–769. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Molinari M, Eriksson KK, Calanca V, Galli C, Cresswell P, Michalak M and Helenius A: Contrasting functions of calreticulin and calnexin in glycoprotein folding and ER quality control. Mol Cell. 13:125–135. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Zuppini A, Groenendyk J, Cormack LA, Shore G, Opas M, Bleackley RC and Michalak M: Calnexin deficiency and endoplasmic reticulum stress-induced apoptosis. Biochemistry. 41:2850–2858. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Pinton P, Ferrari D, Rapizzi E, Di Virgilio F, Pozzan T and Rizzuto R: The Ca2+ concentration of the endoplasmic reticulum is a key determinant of ceramide-induced apoptosis: significance for the molecular mechanism of Bcl-2 action. EMBO J. 20:2690–2701. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Nakamura K, Bossy-Wetzel E, Burns K, Fadel MP, Lozyk M, Goping IS, Opas M, Bleackley RC, Green DR and Michalak M: Changes in endoplasmic reticulum luminal environment affect cell sensitivity to apoptosis. J Cell Biol. 150:731–740. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Lu YC, Chen CN, Wang B, Hsu WM, Chen ST, Chang KJ, Chang CC and Lee H: Changes in tumor growth and metastatic capacities of J82 human bladder cancer cells suppressed by down-regulation of calreticulin expression. Am J Pathol. 179:1425–1433. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Yoshida H, Haze K, Yanagi H, Yura T and Mori K: Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J Biol Chem. 273:33741–33749. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Haze K, Yoshida H, Yanagi H, Yura T and Mori K: Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell. 10:3787–3799. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Chiu SC, Chen SP, Huang SY, Wang MJ, Lin SZ, Harn HJ and Pang CY: Induction of apoptosis coupled to endoplasmic reticulum stress in human prostate cancer cells by n-butylidenephthalide. PLoS One. 7:e337422012. View Article : Google Scholar : PubMed/NCBI

24 

Yoshida H, Okada T, Haze K, Yanagi H, Yura T, Negishi M and Mori K: Endoplasmic reticulum stress-induced formation of transcription factor complex ERSF including NF-Y (CBF) and activating transcription factors 6alpha and 6beta that activates the mammalian unfolded protein response. Mol Cell Biol. 21:1239–1248. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Urano F, Bertolotti A and Ron D: IRE1 and efferent signaling from the endoplasmic reticulum. J Cell Sci. 113:3697–3702. 2000.PubMed/NCBI

26 

Hu R, Zhou P, Peng YB, Xu X, Ma J, Liu Q, Zhang L, Wen XD, Qi LW, Gao N, et al: 6-Shogaol induces apoptosis in human hepatocellular carcinoma cells and exhibits anti-tumor activity in vivo through endoplasmic reticulum stress. PLoS One. 7:e396642012. View Article : Google Scholar : PubMed/NCBI

27 

Zhang R, Chung Y, Kim HS, Kim DH, Kim HS, Chang WY and Hyun JW: 20-O-(beta-D-glucopyranosyl)-20(S)-protopanaxadiol induces apoptosis via induction of endoplasmic reticulum stress in human colon cancer cells. Oncol Rep. 29:1365–1370. 2013.PubMed/NCBI

28 

Park SH, Park HS, Lee JH, Chi GY, Kim GY, Moon SK, Chang YC, Hyun JW, Kim WJ and Choi YH: Induction of endoplasmic reticulum stress-mediated apoptosis and non-canonical autophagy by luteolin in NCI-H460 lung carcinoma cells. Food Chem Toxicol. 56:100–109. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Ma Y, Brewer JW, Diehl JA and Hendershot LM: Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol. 318:1351–1365. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Zinszner H, Kuroda M, Wang X, Batchvarova N, Lightfoot RT, Remotti H, Stevens JL and Ron D: CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev. 12:982–995. 1998. View Article : Google Scholar : PubMed/NCBI

31 

Barone MV, Crozat A, Tabaee A, Philipson L and Ron D: CHOP (GADD153) and its oncogenic variant, TLS-CHOP, have opposing effects on the induction of G1/S arrest. Genes Dev. 8:453–464. 1994. View Article : Google Scholar : PubMed/NCBI

32 

McCullough KD, Martindale JL, Klotz LO, Aw TY and Holbrook NJ: Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol. 21:1249–1259. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Puthalakath H, O’Reilly LA, Gunn P, Lee L, Kelly PN, Huntington ND, Hughes PD, Michalak EM, Mckimm-Breschkin J, Motoyama N, et al: ER stress triggers apoptosis by activating BH3-only protein Bim. Cell. 129:1337–1349. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Vidal RL and Hetz C: Crosstalk between the UPR and autophagy pathway contributes to handling cellular stress in neurodegenerative disease. Autophagy. 8:970–972. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Carra S, Brunsting JF, Lambert H, Landry J and Kampinga HH: HspB8 participates in protein quality control by a non-chaperone-like mechanism that requires eIF2{alpha} phosphorylation. J Biol Chem. 284:5523–5532. 2009. View Article : Google Scholar

36 

Martinet W, Agostinis P, Vanhoecke B, Dewaele M and De Meyer GR: Autophagy in disease: a double-edged sword with therapeutic potential. Clin Sci (Lond). 116:697–712. 2009. View Article : Google Scholar

37 

Ku B, Woo JS, Liang C, Lee KH, Hong HS, E X, Kim KS, Jung JU and Oh BH: Structural and biochemical bases for the inhibition of autophagy and apoptosis by viral BCL-2 of murine gamma-herpesvirus 68. PLoS Pathog. 4:e252008. View Article : Google Scholar : PubMed/NCBI

38 

Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G and Johansen T: p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem. 282:24131–24145. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Kumar D, Shankar S and Srivastava RK: Rottlerin induces autophagy and apoptosis in prostate cancer stem cells via PI3K/Akt/mTOR signaling pathway. Cancer Lett. 343:179–189. 2014. View Article : Google Scholar

40 

Cui Q, Tashiro S, Onodera S, Minami M and Ikejima T: Autophagy preceded apoptosis in oridonin-treated human breast cancer MCF-7 cells. Biol Pharm Bull. 30:859–864. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Li J, Hou N, Faried A, Tsutsumi S, Takeuchi T and Kuwano H: Inhibition of autophagy by 3-MA enhances the effect of 5-FU- induced apoptosis in colon cancer cells. Ann Surg Oncol. 16:761–771. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Schönthal AH: Endoplasmic reticulum stress and autophagy as targets for cancer therapy. Cancer Lett. 275:163–169. 2009. View Article : Google Scholar

Related Articles

Journal Cover

June-2015
Volume 33 Issue 6

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
Coker-Gürkan A, Arisan ED, Obakan P, Akalın K, Özbey U and Palavan-Unsal N: Purvalanol induces endoplasmic reticulum stress-mediated apoptosis and autophagy in a time-dependent manner in HCT116 colon cancer cells. Oncol Rep 33: 2761-2770, 2015
APA
Coker-Gürkan, A., Arisan, E.D., Obakan, P., Akalın, K., Özbey, U., & Palavan-Unsal, N. (2015). Purvalanol induces endoplasmic reticulum stress-mediated apoptosis and autophagy in a time-dependent manner in HCT116 colon cancer cells. Oncology Reports, 33, 2761-2770. https://doi.org/10.3892/or.2015.3918
MLA
Coker-Gürkan, A., Arisan, E. D., Obakan, P., Akalın, K., Özbey, U., Palavan-Unsal, N."Purvalanol induces endoplasmic reticulum stress-mediated apoptosis and autophagy in a time-dependent manner in HCT116 colon cancer cells". Oncology Reports 33.6 (2015): 2761-2770.
Chicago
Coker-Gürkan, A., Arisan, E. D., Obakan, P., Akalın, K., Özbey, U., Palavan-Unsal, N."Purvalanol induces endoplasmic reticulum stress-mediated apoptosis and autophagy in a time-dependent manner in HCT116 colon cancer cells". Oncology Reports 33, no. 6 (2015): 2761-2770. https://doi.org/10.3892/or.2015.3918