BML-275, an AMPK inhibitor, induces DNA damage, G2/M arrest and apoptosis in human pancreatic cancer cells

  • Authors:
    • Hong-Quan Duong
    • Jae Seok Hwang
    • Hee Jeong Kim
    • Yeon-Sun Seong
    • Insoo Bae
  • View Affiliations

  • Published online on: October 17, 2012     https://doi.org/10.3892/ijo.2012.1672
  • Pages: 2227-2236
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adenosine monophosphate-activated protein kinase (AMPK) is a principal intracellular energy sensor which regulates energy producing pathways and energy requiring pathways when the cellular AMP/ATP ratio is altered. BML-275 (compound C), a well-known inhibitor of AMPK, has been found to induce apoptosis in myeloma, glioma and prostate cancer cells. However, the mechanisms responsible for the selective apoptotic effect(s) by BML-275 in cancer cells remain unknown. In the present study, BML-275 was investigated for its antitumor effect(s) in human pancreatic cancer cell lines. BML-275 inhibited the cell proliferation of 4 human pancreatic cancer cell lines (MIA PaCa-2, Panc-1, Colo-357 and AsPC-1). In addition, BML-275 significantly increased the generation of intracellular reactive oxygen species (ROS), followed by induction of DNA damage signaling and apoptosis. Furthermore, BML-275 induced cell cycle arrest in the G2/M phase. The inhibition of ROS generation by N-acetyl cysteine (NAC) significantly prevented the induction of DNA damage and apoptosis, but failed to prevent the induction of G2/M arrest by BML-275. Small interfering RNA (siRNA)-mediated knockdown of AMPKα increased the generation of intracellular ROS, DNA damage signaling and apoptosis without cell cycle arrest at the G2/M phase. These findings suggest that BML-275 exerts its antitumor effects by inducing ROS generation, DNA damage and apoptosis via inhibition of the AMPK pathway and by inducing G2/M arrest via a pathway independent of AMPK, implicating its potential application as an antitumor agent for pancreatic cancer.

Introduction

Pancreatic adenocarcinoma is one of the most lethal and poorly understood human malignancies. Because of the lack of effective systemic therapies the 5-year survival rate for patients with pancreatic adenocarcinoma has remained at 1–3% without a change over the past 25 years (1,2). To date, the only potential curative means is surgical resection, of which only 20% of patients are eligible. Alternative therapies, such as radiotherapy and chemotherapy remain largely ineffective. Therefore, the development and evaluation of novel targeted therapeutic agents that reduce the intrinsic drug resistance of this disease poses one of the greatest challenges in pancreatic cancer research and other intractable cancers.

AMP-activated protein kinase (AMPK), a serine/threonine kinase, is a highly conserved sensor of cellular energy status in eukaryotes and is widely known as a regulator of cell metabolism (3). AMPK is a heterotrimeric protein consisting of a catalytic α-subunit and regulatory β-/γ-subunits (4,5). It is phosphorylated at Thr172 in response to an increase in the ratio of AMP-to-ATP within its activation domain of α-subunit by upstream kinases LKB1 (68) and calmodulin-dependent protein kinase kinase β (CaMKKβ) (911). Several previous studies show that excessive AMPK activation by treatment of AMPK activator (such as Metformin, 5-aminoimidazole-4-carboxamide riboside (AICAR) or A769662) inhibits the growth and/or survival of various cancer cell lines (1219). Moreover, BML-275 (compound C), a potent, selective, and reversible ATP-competitive inhibitor of AMPK induces cell death in various types of cancers including myeloma, glioma, prostate and breast carcinoma cells (2023). In addition, inhibition of AMPK pathway by compound C sensitizes apoptosis by co-treatment with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), doxorubicin or cisplatin in human renal, leukemia, gastric carcinoma, colon carcinoma, and cervix adenocarcinoma cell lines (2426). Therefore, pharmacological inhibition of AMPK activity might be potentially useful in therapy of human solid tumors. However, the effect of AMPK inhibition of pancreatic cancer cell proliferation or survival has not been investigated.

Cell cycle deregulation resulting in uncontrolled cell proliferation is the one of the most frequent alterations that occurs during tumor development (27) and targeting of cell cycle progression and/or machinery is effective strategy to control aberrant proliferation of cancer cell (28,29). There are two major checkpoints, G1/S and G2/M checkpoints, are known to regulate the cell cycle. The G2/M checkpoint plays a key role in the maintenance of chromosomal integrity by allowing cells to repair DNA damage before entering mitosis. A key regulator of the cell cycle at G2/M checkpoint is cyclin dependent kinase 1 (CDK1), especially cell division cycle 2 (Cdc2). Cdc2 activation depends on the dephosphorylation of Tyr15 by Cdc25C (30). In addition, Cdc2 can be further regulated by GADD45 and 14-3-3 by p53 pathway (31). Reactive oxygen species (ROS) generation causes oxidative stress and has been shown to significantly function to controlling cancer cell survival (32). Oxidation of DNA bases and breakage of DNA strand may occurs as results of oxidative DNA damage and parts of these lesions are converted to DNA double-strand breaks (3335). BML-275 was reported to induce cell cycle arrest at G2/M-phase and ROS generation in U251 glioma cells (22). Therefore, understanding the molecular mechanisms of BML-275 to sensitize these cells to undergo BML-275-mediated G2/M arrest and apoptosis is an important issue for effective cancer therapy.

In this study, we performed experiments to determine anti-tumor effect(s) by BML-275 in human pancreatic cancer cell lines. Our results suggest that BML-275 regulates cell survival via targeting AMPK and generating ROS in multiple human pancreatic cancer cells.

Materials and methods

Cell culture and reagents

MIA PaCa-2, Panc-1, CFPAC-1 and BxPC-3 cells were purchased from American Type Culture Collection (ATCC, Manassas, VA, USA) and AsPC-1, Capan-1 and Colo-357 cells were obtained from Tissue Culture Shared Resource of Georgetown University Lombardi Comprehensive Cancer Center (Washington, DC, USA). Immortal human pancreatic ductal epithelial cells, HPDE6-C7 were acquired from Dr M.S. Tsao (36). AsPC-1, BxPC-3, Capan-1 and Colo-357 cells were cultured in RPMI-1640 media supplemented with fetal bovine serum (FBS; 20% for AsPC-1, 10% for Colo-357, Capan-1 and BxPC-3 cells), 100 U/ml penicillin, 100 μg/ml streptomycin and 1% sodium pyruvate. MIA PaCa-2 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) containing 10% FBS, 2.5% horse serum (HS), 100 U/ml penicillin and 100 μg/ml streptomycin. Panc-1 and CFPAC-1 cells were cultured in DMEM containing 10% FBS, 10 U/ml penicillin and 10 μg/ml streptomycin. HPDE6-C7 cells were cultured in keratinocyte serum-free (KSF) medium supplemented by an epidermal growth factor and bovine pituitary extract and 1X antibiotic-antimycotic. Cell culture reagents were purchased from BioWhittaker (Walkersville, MD, USA) and Invitrogen (Carlsbad, CA, USA). BML-275 was purchased from Tocris Bioscience (Ellisville, MO, USA), and A769662 was obtained from LC Laboratories (Woburn, MA, USA).

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

A total of 2,000 human pancreatic cancer cells, counted by the Luna Cell Counter (Logos Biosystems, Gyeonggi-Do, Korea) were plated in 96-well flat-bottom plates and then exposed to test the effects of BML-275 in various concentrations. At the indicated times, 10 μl of 1 mg/ml MTT (Sigma, St. Louis, MO, USA) in PBS was added to each well for 4 h. After centrifugation and removal of the medium, 150 μl of DMSO (Sigma) was added to each well to dissolve the formazan crystals. The absorbance was measured at 560 nm using an ELx808 Absorbance Microplate Reader (BioTek Instruments Inc., Winooski, VT, USA). Absorbance of untreated cells was designated as 100% and cell survival was expressed as a percentage of this value. Triplicate wells were assayed for each condition and standard deviation (SD) was determined.

Western blot (WB) analysis

Cells were grown to ∼70% confluence and reagents were added at the indicated concentrations. After exposure to BML-275 alone or in combination with NAC, cells were lysed in cell lysis buffer containing 20 mM Tris-HCl, 0.5 M NaCl, 0.25% Triton X-100, 1 mM EDTA, 1 mM EGTA, 10 mM β-glycophosphate, 10 mM NaF, 300 μM Na3VO4, 1 mM benzamidine, 2 μM PMSF and 1 mM DTT. Protein concentrations were determined by a BCA protein assay kit (Thermo Scientific, Rockford, IL, USA). Proteins were separated by SDS-PAGE, transferred on to PVDF membranes, blocked in 1X blocking buffer (Sigma) and probed with the following primary antibodies: phospho-ACC (S79), ACC, phospho-AMPKα (T172), AMPK, phospho-ATM (S1981), phospho-CHK2 (T68), phospho-Histone H2A.X (S139), XIAP and Survivin (Cell Signaling Technology, Boston, MA), Bcl2 and Poly-ADP-Ribose-Polymerase (PARP; BD Biosciences, Franklin, NJ, USA) and α-tubulin (Sigma). Then, the membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (Sigma) and visualized with a chemiluminescence kit (Santa Cruz Biotechnology, Santa Cruz, CA, USA) according to the manufacturer’s recommended protocol and exposed with X-ray film (American X-ray and Medical Supply, Jackson, CA, USA).

Clonogenic assay

Human pancreatic cancer cells (4x105 cells) were seeded in 60-mm dishes. Twenty-four hours after plating, varying concentrations of the drugs, either as a single agent or in combination, were added to the dishes. After treatment, cells (2,000 cells) were re-seeded in 60-mm dishes (triplicate). Each culture dish was incubated for 14 days and photographed after staining with 0.5% crystal violet in 1X PBS including 25% methanol. Colonies were examined under a light microscope and counted after capturing images by scanner. Colony numbers were calculated according to the percentage of the untreated cells (37).

Flow cytometry

Human pancreatic cancer cell lines were collected after treatment of BML-275 by trypsinization, washed with PBS and fixed overnight in 70% ethanol at −20°C. Cells were incubated with 20 μg/ml propidium iodide and 40 μg/ml RNase A in 1X PBS. Cells were analyzed on a FACSCalibur flow cytometer (Becton Dickinson, San Jose, CA, USA) at the Flow Cytometry and Cell Sorting Shared Resource, Georgetown University Lombardi Comprehensive Cancer Center. The acquired data were analyzed by Cell Quest Pro Analysis software (Becton Dickinson).

Small interfering RNA (siRNA)

For the RNA interfering experiment, AMPKα-siRNA, 5′-CUGAGUUGCAUAUACUGUA-3′ and control-siRNA, 5′-GACGAGCGGCACGUGCACA-3′ were purchased from Bioneer (Daejeon, Korea). AMPKα-siRNA or control-siRNA were transfected into MIA PaCa-2 and Panc-1 cells using Lipofectamine 2000 (Invitrogen) according to the manufacturer’s procedure. After 48 h transfected cells were processed for cell cycle analysis, WB analysis and measurement of ROS generation.

ROS generation

For measurement of ROS generation, human pancreatic cancer cell lines were treated with BML-275 with or without N-acetyl cysteine (NAC) for the indicated times and then loaded with 50 μM 2′, 7′-dichlorofluorescin diacetate (DCFDA; Molecular Probes, Eugene, OR, USA) and 0.5 μg/ml Hoechst 33342 (HO; Sigma) for 30 min. After rinsing, fluorescent images were taken with fluorescence intensities were obtained with a Fluorocount at excitation/emission wavelengths of 490/530 nm (DCFDA) and 340/425 (HO), and values of ROS generation were obtained by determining the ratio of DCFDA/HO signals per well.

Statistical methods

Statistical comparisons were made using the two-tailed Student’s t-test where appropriate. Results were considered significant in at means *P<0.05, **P<0.01 and ***P<0.005. Data were expressed as the mean ± SD.

Results

Human pancreatic cancer cells and immortal human pancreatic duct epithelial cells express AMPKα

We first examined the total and phosphorylated form of AMPKα in AsPC-1, Panc-1, MIA PaCa-2, Capan-1, CFPAC-1, Colo-357, BxPC-3 and HPDE6-C7 cells. The WB result reveals that all pancreatic cell lines used for this study expressed the levels of both phosphorylated-AMPK and total-AMPK (Fig. 1). Next, we investigated the expression level of AMPK target protein, Acetyl-CoA Carboxylase (ACC). We found that there is relatively good correlation between the levels of phosphorylated-AMPK and phosphorylated-ACC among pancreatic cell lines (Fig. 1). To study the antitumor effect(s) by BML-275, AMPK inhibitor, in pancreatic cancer cells, we chose four pancreatic cancer cell lines: MIA PaCa-2, Panc-1, Colo-357 and AsPC-1 for further studies.

BML-275 induces apoptotic cell death

BML-275 is a potent ATP-mimetic competitive inhibitor of AMPK. In order to explore the antitumor effect(s) by BML-275, MIA PaCa-2, Panc-1, Colon-357 and AsPC-1 cells were treated with different concentrations of BML-275 (0, 1, 3, 5 and/or 10 μM) for 48 h and cell viability were measured by MTT assay. BML-275 inhibited cell survival in dose-dependent manner (Fig. 2A). Next, we performed clonogenic assay to determine the long-term growth inhibitory effect of BML-275. Cells were treated with various concentrations of BML-275 (0, 1, 3, 5 and/or 10 μM) for 1 day and continuously cultured in fresh media for 14 days and colony formation was measured by clonogenic assay. BML-275 significantly inhibited colony formation in dose-dependent manner and at 10 μM BML-275 all of the tested pancreatic cancer cells showed susceptibility to the AMPK inhibitor (Fig. 2B). MIA PaCa-2 cells showed increased sensitivity to BML-275 and Panc-1 cells showed relatively less sensitive to BML-275 among four pancreatic cancer cell lines tested (Fig. 2A and B).

To investigate mechanism of apoptosis by BML-275 treatment, MIA PaCa-2 and Panc-1 cells were treated with various concentrations of BML-275 or 10 μM A769662 for 24 h. Apoptotic cell death was detected by WB analysis of a molecular biomarker of apoptosis, PARP cleavage. On the contrary to cells treated with A769662, cells treated with BML-275 showed an increase of cleaved PARP in MIA PaCa-2 cells but not in Panc-1 cells (Fig. 3). However, BML-275 treatment decreased the expression of anti-apoptotic proteins such as Survivin, Bcl2 and XIAP in both cell lines (Fig. 3).

BML-275 induces G2/M arrest and sub-G1

We next investigated if the pharmacological inhibition of AMPK by BML-275 can affect the cell cycle progression in pancreatic cancer cell lines. MIA PaCa-2 and Panc-1 cells were treated with 10 μM BML-275 for 24 h and their cell cycle profiles were assessed by FACS analysis. BML-275 treatment significantly increased the cell population at G2/M-phase (from 15.9 to 58.7% in MIA PaCa-2 and from 19.4 to 42% in Panc-1) and significantly decreased the cell population at G0/G1-phase (from 52.8 to 26.4% in MIA PaCa-2 and from 44.9 to 32.4% in Panc-1) and S-phase (from 31.3 to 14.9% in MIA PaCa-2 cells and from 35.7 to 25.6% in Panc-1) (Fig. 4A). Moreover, we also observed increase of sub-G1 populations. BML-275 increased the sub-G1 population in Panc-1 (from 3 to 7.5%) and more significantly in MIA PaCa-2 (from 6.2 to 45.9%) (Fig. 4A).

DNA damage sensor CHK1/CHK2 plays a role in G2/M checkpoint via the ataxia-telangiectasia mutated (ATM)/ATM-RAD3-related (ATR) pathway. In order to further elucidate the molecular mechanism leading to BML-275-mediated G2/M arrest, we determined the activation of DNA damage signaling pathway. Treatment of MIA PaCa-2 and Panc-1 cells with BML-275 for 24 h increased the phosphorylation of ATM at Ser1981 and CHK2 at Thr68 in dose-dependent manner (Fig. 4B). These results coincide with cell cycle arrest in both cell lines. However, the phosphorylation of Histone H2A.X at Ser139, which is the molecular marker of DNA double-strand breaks, more significantly increased in MIA PaCa-2 than in Panc-1 cells (Fig. 4B). Increased levels of CHK2 and H2A.X phosphorylation were more obvious in MIA PaCa-2 cells (Fig. 4B). On the contrary, cells treated by 10 μM A769662 for 24 h did not induce the phosphorylation levels of ATM, CHK2 or Histone H2A.X in either cell line.

BML-275 decreases AMPKα activity in human pancreatic cancer cells

In order to determine the decrease in cell survival and increase in apoptotic cell death closely correlates with the level of inhibition of AMPK activity, cells were pretreated with 10 μM A769662 for 6 h and administered with various concentrations of BML-275 for 24 h. The treatment of 10 μM A769662 for 6 h without BML-275 significantly activated accumulation of phosphorylated levels of AMPKα and ACC in both cell lines (Fig. 5). However, BML-275 treatment reduced the phosphorylation of AMPKα and ACC exerted by A769662 in a dose-dependent manner (Fig. 5), suggesting that antitumor effect(s) by BML-275 closely correlates with the level of inhibition of AMPK activity in human pancreatic cancer cell lines.

The generation of ROS by BML-275 is critically required for the induction of cell death but not G2/M arrest

Since oxidative stress is a potent inducer of apoptosis, we next investigated if BML-275 could cause a generation of ROS in pancreatic cancer cell lines. We determined ROS generation by measuring the fluorescence of DCF which is formed by the oxidation of DCFDA by peroxides. Our results demonstrated early ROS generation by BML-275 in both cell lines (Fig. 6A). BML-275-induced ROS generation was significantly diminished by incubation with the antioxidant agent, NAC (Fig. 6B). NAC also rescued BML-275-mediated inhibition of cell survival by MTT assay (data not shown) and clonogenic assay (Fig. 6C). It also relieved the cleavage of PARP by BML-275 treatment in MIA PaCa-2 cells (Fig. 6D). BML-275-mediated phosphorylation of H2A.X at Ser139 also inhibited by NAC pretreatment (Fig. 6E). However, NAC administration did not alleviate G2/M arrest induced by BML-275 treatment (Fig. 6F), suggesting that BML-275-mediated G2/M arrest is ROS-independent at least in pancreatic cancer cell lines used for this study.

Figure 6

ROS generation is critically required for apoptotic cell death by BML-275. (A) The result of ROS generation analysis of MIA PaCa-2 and Panc-1 cells treated with 10 μM BML-275 in different time intervals (0, 6, 12 and/or 24 h) and treated with DCFDA for a further 0.5 h to measure ROS generation. Experiments were repeated 3 times and similar results were obtained. Error bars represent the standard deviation. ***Represents statistically significant difference with p-value <0.005 between 10 μM BML-275 12 h treated group and control group. (B) The result of ROS generation analysis of MIA PaCa-2 and Panc-1 cells pretreated with 10 mM NAC for 1 h and further treated with 10 μM BML-275 for 12 h and then treated with DCFDA for further 0.5 h to measure ROS generation. Experiments were repeated 3 times and similar results were obtained. Error bars represent the standard deviation. **Represents statistically significant difference with p-value <0.01 between 10 μM BML-275 treated group and 10 mM NAC plus 10 μM BML-275 group. (C) A clonogenic assay of MIA PaCa-2 and Panc-1 cells pretreated with 10 mM NAC for 1 h and further treated with 3 μM BML-275 for 24 h was used to determine the long-term response. Colony numbers were counted and calculated as a relative percentage (%) of the untreated control cells (left) and representative photograph of colony formation assay results are shown (right). Experiments were repeated 3 times and similar results were obtained. Error bars represent the standard deviation. **Represents statistically significant difference with p-value <0.01 between 10 mM NAC plus 3 μM BML-275 and 3 μM BML-275 group. (D) A WB analysis of MIA PaCa-2 and Panc-1 cells pretreated with 10 mM NAC for 1 h and further treated with 10 μM BML-275 for 24 h was used to determine apoptotic cell death by anti-cleaved PARP antibody. (E) A WB analysis of MIA PaCa-2 and Panc-1 cells pretreated with 10 mM NAC for 1 h and further treated with 10 μM BML-275 for 24 h was used to determine the phosphorylated forms of Histone H2A.X (S139). The specific phosphorylation site(s) of each kinase is indicated in parentheses. (F) Analysis by FACS of MIA PaCa-2 and Panc-1 cells pretreated with 10 mM NAC for 1 h and treated with 10 μM BML-275 for 24 h was used to determine cell cycle arrest. Experiments were repeated 3 times and similar results were obtained. Error bars represent the standard deviation.

Knockdown of AMPKα induces ROS generation and apoptosis but not G2/M arrest

Since the inhibition of AMPK by BML-275 induced DNA damage, G2/M arrest and apoptosis in human pancreatic cancer cells, MIA PaCa-2 and Panc-1 cells were transfected with control-siRNA or AMPKα-siRNA to compare the effect(s) of BML-275 and knockdown of AMPKα. Knockdown of AMPKα with concentration of 0.1 or 0.2 μM AMPKα-siRNA suppressed the level of total and phosphorylated form of AMPKα and phosphorylated form of ACC in MIA PaCa-2 and Panc-1 cells (Fig. 7A). In addition, knockdown of AMPKα also induced apoptotic cell death as evidenced by induction of PARP cleavage in MIA-PaCa-2 cells (Fig. 7B) and accumulation of sub-G1 cells in FACS analysis in MIA PaCa-2 cells (from 19.4% by control to 34.1% by 0.1 μM AMPKα siRNA) but to a lesser extent in Panc-1 cells (from 7.0% by control to 11.3% by 0.1 μM AMPKα-siRNA (Fig. 7D). Knockdown of AMPKα in MIA PaCa-2 cells also induced phosphorylation of H2A.X at Ser139 indicating DNA damage (Fig. 7C). The Panc-1 cells show resistance to phosphorylation of H2A.X similarly to BML-275 treatment (Fig. 4B). However, knockdown of AMPKα activity fails to display a cell cycle arrest in G2/M-phase in MIA PaCa-2 and Panc-1 cells (Fig. 7D). Finally, AMPKα knockdown induced ROS generation with increasing concentration of AMPKα-siRNA (Fig. 7E). Taken together, in pancreatic cancer cell lines, targeting of AMPKα is able to induce DNA damage, ROS generation and apoptotic cell death but not G2/M arrest.

Discussion

In this study, we investigated the molecular mechanism of antitumor effect(s) of BML-275, an AMPK inhibitor, in human pancreatic adenocarcinoma. We found that: i) the levels of total and phosphorylated form of AMPKα and ACC vary in several different human pancreatic cancer cell lines; ii) BML-275 inhibits cell proliferation in MIA PaCa-2, Panc-1, Colo-357 and AsPC-1 cells; iii) BML-275 induces DNA damage, apoptosis and G2/M arrest; iv) the ROS generation by BML-275 is critically required for the DNA damage and apoptosis but not G2/M arrest and v) knockdown of AMPKα induces ROS generation, DNA damage and apoptosis but not G2/M arrest. This is the first report showing that BML-275 induces DNA damage, G2/M arrest and apoptosis in pancreatic cancer cell lines.

AMPK is a survival factor for cancer cells. It is involved in the augmentation of energy production through the activation of glucose uptake, glycolysis and fatty acid oxidation in response to ATP-depleting stresses (38). Solid tumors outgrowing the existing vasculature are continuously exposed to a microenvironment in which the supply of both oxygen and nutrition is limited. Previous studies showed that AMPK is critical for cancer cell adaptation in response to hypoxia or glucose deprivation (3942). The protective role of AMPK is not restricted to nutrient stress, as this enzyme seems to play an important role in protecting tumor cells from apoptosis induced by chemotherapeutic agents such as doxorubicin, cisplatin and TRAIL (2426). In addition, pharmacological inhibition of AMPK by BML-275 induced apoptotic cell death in myeloma, glioma, prostate cancer and breast carcinoma cells (2023). Moreover, transfection with dominant-negative AMPK or AMPKα-siRNA was also sufficient to reduce cell proliferation of BHK, HeLa and PC12 pheochromocytoma cells or CWR22Rv1 and LNcaP prostate cancer cells (21,43). Comparing with effective apoptosis inducing dose of BML-275 treated in other cancer cell lines reported previously (2023), most of the pancreatic cancer cell lines responded to BML-275 with different levels of responsiveness. Pancreatic cancer cell lines with relatively high level of phosphorylated AMPK showed more susceptibility to BML-275 treatment (MIA PaCa-2 and Colo-357), and those with low phosphorylated AMPK showed relatively decreased sensitivity (Panc-1 and AsPc-1).

Cancer cells usually exhibit increased levels on intracellular ROS, which in turn can initiate various cycles leading to further metabolic malfunction and ROS generation (44,45). ROS cause oxidative damage to DNA, proteins, lipids and other cellular components and therefore also significant cellular stress (45). A proposed therapeutic strategy against cancer is to treat cancer cells with pharmacological agents that have pro-oxidant properties which increase the intracellular ROS generation to a toxic threshold that triggers cell death in the cancer cells without harming normal cells (44). Vuvicevic et al showed that BML-275 induces ROS generation in glioma cell line, but AMPKα-siRNA treatment fails to induce ROS generation and apoptosis (22). In this study, an increased generation of ROS upon either BML-275 or AMPKα-siRNA treatment was observed and the intracellular accumulation of ROS seems to be one of critical factors in BML-275-induced apoptosis. To verify this speculation, NAC, scavenger of oxygen-free radicals, was challenged with BML-275. NAC relieved BML-275 or AMPK-siRNA mediated ROS production and improved cell viability based on the clonogenic assay, which suggested that both chemical and genetic inhibitor regulate cell viability via repressing AMPK activity.

The G2/M checkpoint plays an important role in cellular response to genotoxic stimuli. The G2/M checkpoint prevents cells from entering mitosis when DNA is damaged, providing an opportunity for repair and stopping the proliferation of damaged cells which help to maintain genomic stability (46). CHK1 and CHK2 kinases are activated at G2-phase checkpoint by DNA damage or unreplicated chromosomal DNA (47), and inactivate Cdc25C through its phosphorylation (48,49). Cdc25C was the protein phosphatase responsible for dephosphorylating and activating Cdc2, a crucial step in regulating the entry of all eukaryotic cells into the M-phase of the cell cycle. In this study, BML-275 induces cell cycle arrest at G2/M-phase possibly through the phosphorylation and activation of CHK2 kinase. The pretreatment of NAC restores the generation of ROS by BML-275 treatment in MIA PaCa-2 cell line, however, the cell cycle arrest at G2/M phase cannot be relieved, suggesting unknown effects of BML-275 or non-target effects may play a role in G2/M arrest. Previously AMPKα-siRNA treatment was reported to induce G2/M arrest in the absence of ROS generation and with no apparent cell death in U251 glioma cells (22). However, in pancreatic cancer cell line, the AMPKα-siRNA treatment induces generation of ROS and apoptotic cell death but no apparent G2/M arrest. Thus, our finding suggests that pancreatic cancer cells may be able to override the cell cycle arrest (G2/M) in response to AMPK knockdown by siRNA. On the other hand, the mechanism of DNA damage and cell death induced by BML-275 seems to be via inhibition of AMPK activity followed by stimulation of ROS production. Panc-1 is known as relatively more resistant to various antitumor agents among several pancreatic cancer cell lines (5052). Our study also show panc-1 as more resistant to apoptotic response (cell death and PARP cleavage) upon the treatment of BML-275 and AMPKα-siRNA. Although we could not demonstrate the mechanism of resistance of Panc-1 to BML-275 treatment, this may be due to its increased multidrug resistance (MDR) gene products and/or constitutively activated cell surviving signaling pathways that confer intrinsic drug resistance (5054).

In conclusion, our findings implicate that BML-275 induces DNA damage and apoptosis through AMPK-dependent mechanism and induces G2/M arrest through AMPK-independent mechanism (Fig. 8). Although the molecular mechanism of antitumor effect(s) by BML-275 requires further investigation, this compound seems to be a novel potential therapeutic agent to treat human pancreatic cancer.

Acknowledgements

IB was supported by National Institutes of Health (1R03CA152530), the National Research Foundation of Korea [R31-10069; World Class University (WCU) program] and the Georgetown University Lombardi Comprehensive Cancer Center (P30-CA051008).

References

1. 

A JemalR SiegelJ XuE WardCancer statistics, 2010CA Cancer J Clin60277300201010.3322/caac.20073

2. 

MR KeighleyGastrointestinal cancers in EuropeAliment Pharmacol Ther18730200310.1046/j.0953-0673.2003.01722.x

3. 

DG HardieAMP-activated/SNF1 protein kinases: conserved guardians of cellular energyNat Rev Mol Cell Biol8774785200710.1038/nrm224917712357

4. 

DG HardieD CarlingThe AMP-activated protein kinase - fuel gauge of the mammalian cells?Eur J Biochem246259273199710.1111/j.1432-1033.1997.00259.x9208914

5. 

DG HardieJW ScottDA PanER HudsonManagement of cellular energy by the AMP-activated protein kinase systemFEBS Lett546113120200310.1016/S0014-5793(03)00560-X12829246

6. 

D CarlingThe AMP-activated protein kinase cascade - a unifying system for energy controlTrends Biochem Sci291824200410.1016/j.tibs.2003.11.00514729328

7. 

DG HardieThe AMP-activated protein kinase pathway - new players upstream and downstreamJ Cell Sci11754795487200410.1242/jcs.0154015509864

8. 

BB KahnT AlquierD CarlingDG HardieAMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolismCell Metab11525200510.1016/j.cmet.2004.12.00316054041

9. 

SA HawleyDA PanKJ MustardL RossJ BainAM EdelmanBG FrenguelliDG HardieCalmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinaseCell Metab2919200510.1016/j.cmet.2005.05.00916054095

10. 

RL HurleyKA AndersonJM FranzoneBE KempAR MeansLA WittersThe Ca2+/calmodulin-dependent protein kinase kinases are AMP-activated protein kinase kinasesJ Biol Chem2802906029066200515980064

11. 

A WoodsK DickersonR HeathSP HongM MomcilovicSR JohnstoneM CarlsonD CarlingCa2+/calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cellsCell Metab221332005

12. 

C CampàsJM LopezAF SantidriánM BarragánB BellosilloD ColomerJ GilAcadesine activates AMPK and induces apoptosis in B-cell chronic lymphocytic leukemia cells but not in T lymphocytesBlood10136743680200312522004

13. 

BA KefasY CaiK KerchhofsZ LingG MartensH HeimbergD PipeleersM Van de CasteeleMetformin-induced stimulation of AMP-activated protein kinase in beta-cells impairs their glucose responsiveness and can lead to apoptosisBiochem Pharmacol68409416200410.1016/j.bcp.2004.04.003

14. 

M SaitohK NagaiK NakagawaT YamamuraS YamamotoT NishizakiAdenosine induces apoptosis in the human gastric cancer cells via an intrinsic pathway relawant to activation of AMP-activated protein kinaseBiochem Pharmacol6720052011200410.1016/j.bcp.2004.01.020

15. 

R RattanS GiriAK SinghI Singh5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside inhibits cancer cell proliferation in vitro and in vivo via AMP-activated protein kinaseJ Biol Chem2803958239593200510.1074/jbc.M50744320016176927

16. 

W ZhouWF HanLE LandreeJN ThupariML PinnT BililignEK KimA VadlamudiSM MedghlchiR El MeskiniGV RonnettCA TownsendFP KuhajdaFatty acid synthase inhibition activates AMP-activated protein kinase in SKOV3 human ovarian cancer cellsCancer Res6729646971200710.1158/0008-5472.CAN-06-343917409402

17. 

A IsakovicL HarhajiD StevanovicZ MarkovicM Sumarac-DumanovicV StarcevicD MicicV TrajkovicDual antiglioma action of metformin: cell cycle arrest and mitochondria-dependent apoptosisCell Mol Life Sci6412901302200710.1007/s00018-007-7080-417447005

18. 

R OkoshiT OzakiH YamamotoK AndoN KoidaS OnoT KodaT KamijoA NakagamaraH KizakiActivation of AMP-activated protein kinase induces p53-dependent apoptotic cell death in response to energetic stressJ Biol Chem28339793987200810.1074/jbc.M70523220018056705

19. 

TK SenquptaGM LeclercTT Hsieh-KinserGJ leclercI SinghJC BarredoCytotoxic effect of 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) on childhood acute lymphoblastic leukemia (ALL) cells: implication for targeted therapyMol Cancer646200710.1186/1476-4598-6-4617623090

20. 

P BaumannS Mandl-WeberB EmmerichC StrakaR SchmidmaierInhibition of adenosine monophosphate-activated protein kinase induces apoptosis in multiple myeloma cellsAnticancer Drugs18405410200710.1097/CAD.0b013e32801416b617351392

21. 

HU ParkS SuyM DannerV DaileyY ZhangH LiDR HydukeBT CollinsG GagnonB KallakuryD KumarML BrownA FornaceA DritschiloSP CollinsAMP-activated protein kinase promotes human prostate cancer cell growth and survivalMol Cancer Ther8733741200910.1158/1535-7163.MCT-08-063119372545

22. 

L VuvicevicM MisirkicK JanjetovicL Harhaji-TrajkovicM PricaD StevanovicE IsenovicE SudarM Sumarac-DumanovicD MicicV TrajkovicAMP-activated protein kinase-dependent and-independent mechanisms underlying in vitro antiglioma action of compound CBiochem Pharmacol7716841693200910.1016/j.bcp.2009.03.00519428322

23. 

J JinTD MullenQ HouJ BielawskiA BielawskaX ZhangLM ObeidYA HannunYT HsuAMPK inhibitor compound C stiumulates ceramine production and promotes BAx redistribution and apoptosis in MCF-7 breast carcinoma cellsJ Lipid Res5023892397200910.1194/jlr.M900119-JLR20019528633

24. 

JH JangTJ LeeES YangS Min doYH KimSH KimYH ChoiJW ParkKS ChoiTK KwonCompound C sensitizes caki renal cancer cells to TRAIL-induced apoptosis through reactive oxygen species-mediated down-regulation of c-FLIPL and Mcl-1Exp Cell Res31621942203201010.1016/j.yexcr.2010.04.02820451517

25. 

Q ZhuB ShenB ZhangW ZhangSH ChinJ JinDF LiaoInhibition of AMP-activated protein kinase pathway sensitizes human leukemia K562 cells to nontoxic concentration of doxorubicinMol Cell Biochem340275281201010.1007/s11010-010-0428-320339906

26. 

HS KimJT HwangH YunSG ChiSJ LeeI KangKS YoonWJ ChoeSS KimJ HaInhibition of AMP-activated protein kinase sensitizes cancer cells to cisplatin-induced apoptosis via hyper-induction of p53J Biol Chem28337313742200810.1074/jbc.M70443220018079115

27. 

K CollinsT JacksNP PavletichThe cell cycle and cancerProc Natl Acad Sci USA9427762778199710.1073/pnas.94.7.27769096291

28. 

JK BuolamwiniCell cycle molecular targets in novel anti-cancer drug discoveryCurr Pharm Des6379392200010.2174/138161200340094810788588

29. 

M HajduchL HavlieekJ VeselyR NovotnyV MihalM StrnadSynthetic cyclin dependent kinase inhibitors. New generation of potent anti-cancer drugsAdv Exp Med Biol457341353199910.1007/978-1-4615-4811-9_3710500810

30. 

J PinesFour-dimensional control of the cell cycleNat Cell Biol1E73E79199910.1038/1104110559915

31. 

WR TaylorGR StarkRegulation of the G2/M transition by p53Oncogene2018031815200110.1038/sj.onc.120425211313928

32. 

S UedaH NakamuraH MasutaniT SasadaA TakabayashiY YamaokaJ YodoiBaicalin induces apoptosis via mitochondrial pathway as prooxidantMol Immunol38781791200210.1016/S0161-5890(01)00115-811841838

33. 

KB BeckmanBN AmesOxidative decay of DNAJ Biol Chem2721963319636199710.1074/jbc.272.32.196339289489

34. 

J CadetT DelatourT DoukiD GasparuttoJP PougetJL RavanatS SauvaigoHydroxyl radicals and DNA base damageMutal Res424921199910.1016/S0027-5107(99)00004-4

35. 

MM VilenchikAG KnudsonEndogenous DNA double-strand breaks:production, fidelity of repair and induction of cancerProc Natl Acad Sci USA1001287112876200310.1073/pnas.213549810014566050

36. 

T FurukawaWP DuquidL RosenbergJ VialletDA GallowayMS TsaoLong-term culture and immortalization of epithelial cells from normal adult human pancreatic ducts transfected by the E6E7 gene of human papilloma virus 16Am J Pathol1481763177019968669463

37. 

HQ DuongHJ KimHJ KangYS SeongI BaeZSTK474, a PI3K inhibitor, suppresses proliferation and sensitizes human pancreatic adenocarcinoma cells to gemcitabineOncol Rep27182188201221993922

38. 

DG HardieD CarlingM CarlsonThe AMP-activated/SNF1 protein kinase subfamily: metabolic sensors of the eukaryotic cells?Annu Rev Biochem67821855199810.1146/annurev.biochem.67.1.8219759505

39. 

M LeeJT HwangHJ LeeSN JungI KangSG ChiSS KimJ HaAMP-activated protein kinase activity is critical for hypoxia-inducible factor-1 transcriptional activity and its target gene expression under hypoxic conditions in DU145 cellsJ Biol Chem2783965339661200310.1074/jbc.M306104200

40. 

H YunM LeeSS KimJ HaGlucose deprivation increases mRNA stability of vascular endothelial growth factor through activation of AMP-activated protein kinase in DU145 prostate carcinomaJ Biol Chem28099639972200510.1074/jbc.M412994200

41. 

K KatoT OguraA KishimotoY MinegishiN NakajimaM MiyazakiH EsumiCritical roles of AMP-activated protein kinase in constitutive tolerance of cancer cells to nutrient deprivation and tumor formationOncogene2160826090200210.1038/sj.onc.120573712203120

42. 

H EsumiK IzuishiK KatoK HashimotoY KurashimaA KishimotoT OguraT OzawaHypoxia and nitric oxide treatment confer tolerance to glucose starvation in a 5′-AMP-activated protein kinase dependent mannerJ Biol Chem2773279132798200212091379

43. 

MM ShawWK GurrRJ McCrimmonDF SchordererRS Sherwin5′AMP-activated protein kinase alpha deficiency enhances stress-induced apoptosis in BHK and PC12 cellsJ Cell Mol Med112862982007

44. 

D TrachoothamJ AlexandreP HuangTargeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?Nat Rev Drug Discov8579591200910.1038/nrd280319478820

45. 

J LuoNL SoliminiSJ ElledqePrinciples of cancer therapy: oncogene and non-oncogene addictionCell136823837200910.1016/j.cell.2009.02.02419269363

46. 

M LobrichPA JeggoThe impact of a negligent G2/M checkpoint on genomic instability and cancer inductionNat Rev Cancer7861869200710.1038/nrc224817943134

47. 

VA SmitsRH MedemaChecking out the G(2)/M transitionBiochim Biophys Acta1519112200110.1016/S0167-4781(01)00204-411406266

48. 

T GotohK OhsumiT MatsuiH TakisawaT KishimotoInactivation of the checkpoint kinase Cds1 is dependent on cyclin B-Cdc2 kinase activation at the meiotic G2/M-phase transition in xenopus oocytesJ Cell Sci11433973406200111591827

49. 

SV SinghA Herman-AntosiewiczAV SinghKL LewSK SrivastavaR KamathKD BrownL ZhangR BaskaranSulforaphane-induced G2/M phase cell cycle arrest involves checkpoint kinase 2-mediated phosphorylation of cell division cycle 25CJ Biol Chem2792581325822200410.1074/jbc.M31353820015073169

50. 

W HuanwenL ZhiyongS XiaohuaR XinyuW KaiL TonghuaIntrinsic chemoresistance to gemcitabine is associated with constitutive and laminin-induced phosphorylation of FAK in pancreatic cancer cell linesMol Cancer8125200910.1186/1476-4598-8-12520021699

51. 

JN KreutzerM RuzzeneB GuerraEnhancing chemosensitivity to gemcitabine via RNA interfence targeting the catalytic subunits of protein kinase CK2 in human pancreatic cancer cellsBMC Cancer1040201010.1186/1471-2407-10-44020718998

52. 

AV DanilovD NeupaneAS NagarajaEV FeofanovaLA HumphriesJ DiRenzoM KorcDeltaNp63alpha-mediated induction of epidermal growth factor receptor promotes pancreatic cancer cell growth and chemoresistancePloS One6e26815201110.1371/journal.pone.002681522053213

53. 

W HagmannR JesnowskiJM LöhrInterdependence of gemcitabine treatment, transporter expression, and resistance in human pancreatic carcinoma cellsNeoplasia12740747201020824050

54. 

Z YaoY OkabayashiY YutsudoT KitamuraW OgawaM KasugaRole of Akt in growth and survival of Panc-1 pancreatic cancer cellsPancreas244246200210.1097/00006676-200201000-0000611741181

Related Articles

Journal Cover

December 2012
Volume 41 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Duong H, Hwang JS, Kim HJ, Seong Y and Bae I: BML-275, an AMPK inhibitor, induces DNA damage, G2/M arrest and apoptosis in human pancreatic cancer cells. Int J Oncol 41: 2227-2236, 2012
APA
Duong, H., Hwang, J.S., Kim, H.J., Seong, Y., & Bae, I. (2012). BML-275, an AMPK inhibitor, induces DNA damage, G2/M arrest and apoptosis in human pancreatic cancer cells. International Journal of Oncology, 41, 2227-2236. https://doi.org/10.3892/ijo.2012.1672
MLA
Duong, H., Hwang, J. S., Kim, H. J., Seong, Y., Bae, I."BML-275, an AMPK inhibitor, induces DNA damage, G2/M arrest and apoptosis in human pancreatic cancer cells". International Journal of Oncology 41.6 (2012): 2227-2236.
Chicago
Duong, H., Hwang, J. S., Kim, H. J., Seong, Y., Bae, I."BML-275, an AMPK inhibitor, induces DNA damage, G2/M arrest and apoptosis in human pancreatic cancer cells". International Journal of Oncology 41, no. 6 (2012): 2227-2236. https://doi.org/10.3892/ijo.2012.1672