Epidermal Growth Factor-induced Cell Death and Radiosensitization in Epidermal Growth Factor Receptor-overexpressing Cancer Cell Lines

Similar documents
Differential Effects of Recombinant Human EGF on Proliferation and Radiation Survival of Normal Fibroblast and Cancer Cell Lines

Supplementary Information POLO-LIKE KINASE 1 FACILITATES LOSS OF PTEN-INDUCED PROSTATE CANCER FORMATION

Supporting Information

Supplementary data Supplementary Figure 1 Supplementary Figure 2

SUPPLEMENTARY INFORMATION

Supplementary Figure 1: si-craf but not si-braf sensitizes tumor cells to radiation.

Supplementary Table 1. Characterization of HNSCC PDX models established at MSKCC

Proteomic profiling of small-molecule inhibitors reveals dispensability of MTH1 for cancer cell survival

RayBio KinaseSTAR TM Akt Activity Assay Kit

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot

A549 and A549-fLuc cells were maintained in high glucose Dulbecco modified

HCC1937 is the HCC1937-pcDNA3 cell line, which was derived from a breast cancer with a mutation

A Hepatocyte Growth Factor Receptor (Met) Insulin Receptor hybrid governs hepatic glucose metabolism SUPPLEMENTARY FIGURES, LEGENDS AND METHODS

ab E3 Ligase Auto- Ubiquitilylation Assay Kit

Supporting Information

hexahistidine tagged GRP78 devoid of the KDEL motif (GRP78-His) on SDS-PAGE. This

Fang et al. NMuMG. PyVmT unstained Anti-CCR2-PE MDA-MB MCF MCF10A

Supplementary Information

SUPPLEMENT. Materials and methods

Protocol for Gene Transfection & Western Blotting

Instructions for Use. APO-AB Annexin V-Biotin Apoptosis Detection Kit 100 tests

MTC-TT and TPC-1 cell lines were cultured in RPMI medium (Gibco, Breda, The Netherlands)

SUPPLEMENTAL MATERIAL. Supplementary Methods

The Schedule and the Manual of Basic Techniques for Cell Culture

TECHNICAL BULLETIN. Catalog Number RAB0447 Storage Temperature 20 C

Supplemental Experimental Procedures

Supplemental Information

A protocol for enhancement of the AAV-mediated expression of transgenes

A dual PI3 kinase/mtor inhibitor reveals emergent efficacy in glioma

CytoSelect Tumor- Endothelium Adhesion Assay

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

Impact of hyper-o-glcnacylation on apoptosis and NF-κB activity SUPPLEMENTARY METHODS

2,6,9-Triazabicyclo[3.3.1]nonanes as overlooked. amino-modification products by acrolein

Online Data Supplement. Anti-aging Gene Klotho Enhances Glucose-induced Insulin Secretion by Upregulating Plasma Membrane Retention of TRPV2

Supplementary Information Supplementary Fig. 1. Elevated Usp9x in melanoma and NRAS mutant melanoma cells are dependent on NRAS for 3D growth.

Molecular Mechanism of EGFR Signaling Pathway Mediating Proliferation and Migration of U251 Glioma Cell Line

TSH Receptor Monoclonal Antibody (49) Catalog Number MA3-218 Product data sheet

Figure S1. Generation of inducible PTEN deficient mice and the BMMCs (A) B6.129 Pten loxp/loxp mice were mated with B6.

LDL Uptake Cell-Based Assay Kit

Imaging of glycolytic metabolism in primary glioblastoma cells with

B16-F10 (Mus musculus skin melanoma), NCI-H460 (human non-small cell lung cancer

NF-κB p65 (Phospho-Thr254)

Chapter 2. Aims & Objectives

SUPPLEMENTAL INFORMATION

TFEB-mediated increase in peripheral lysosomes regulates. Store Operated Calcium Entry

University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; e Boone Pickens

Validation & Assay Performance Summary

SUPPLEMENTARY INFORMATION. Supplementary Figures S1-S9. Supplementary Methods

HIV-1 Virus-like Particle Budding Assay Nathan H Vande Burgt, Luis J Cocka * and Paul Bates

Phospho-AKT Sampler Kit

Downregulation of angiotensin type 1 receptor and nuclear factor-κb. by sirtuin 1 contributes to renoprotection in unilateral ureteral

p47 negatively regulates IKK activation by inducing the lysosomal degradation of polyubiquitinated NEMO

ab LDL Uptake Assay Kit (Cell-Based)

Essential Medium, containing 10% fetal bovine serum, 100 U/ml penicillin and 100 µg/ml streptomycin. Huvec were cultured in

Impact factor: Reporter:4A1H0019 Chen Zi Hao 4A1H0023 Huang Wan ting 4A1H0039 Sue Yi Zhu 4A1H0070 Lin Guan cheng 4A1H0077 Chen Bo xuan

human Total Cathepsin B Catalog Number: DY2176

TECHNICAL BULLETIN. Phospho-Akt (pser 473 ) ELISA Kit for detection of human, mouse, or rat phospho-akt (pser 473 ) in cell and tissue lysates

Effect of FADD expression during UVC carcinogenesis

Targeted mass spectrometry (LC/MS/MS) for Olaparib pharmacokinetics. For LC/MS/MS of Olaparib pharmacokinetics metabolites were extracted from

SUPPLEMENTARY INFORMATION

Anti-ceramide Antibody Prevents the Radiation GI Syndrome in Mice

LDL Uptake Cell-Based Assay Kit

STAT3 (py705)/ Pan STAT3 (Human/Mouse/Rat) ELISA Kit

Supplementary Materials and Methods

Chemical Chaperones Mitigate Experimental Asthma By Attenuating Endoplasmic

Supplementary Figure 1

Supplemental material for Hernandez et al. Dicoumarol downregulates human PTTG1/Securin mrna expression. through inhibition of Hsp90

Supplementary Figure 1 Lymphocytes can be tracked for at least 4 weeks after

Supplementary Figure 1 Role of Raf-1 in TLR2-Dectin-1-mediated cytokine expression

Analyses of Intravesicular Exosomal Proteins Using a Nano-Plasmonic System

IMMP8-1. Different Mechanisms of Androg and IPAD on Apoptosis Induction in Cervical Cancer Cells

The Preventive Effect of Recombinant Human Growth Factor (rhegf) on the Recurrence of Radiodermatitis

Amersham ECL Prime Western blotting reagent

Supplementary Materials and Methods

Advances in Computer Science Research, volume 59 7th International Conference on Education, Management, Computer and Medicine (EMCM 2016)

Effec<ve Use of PI3K and MEK Inhibitors to Treat Mutant K Ras G12D and PIK3CA H1047R Murine Lung Cancers

Chloroquine inhibits cell growth and induces cell death in A549 lung cancer cells

STAT3 (py705) (Human/Mouse/Rat) ELISA Kit

a b G75 G60 Sw-2 Sw-1 Supplementary Figure 1. Structure predictions by I-TASSER Server.

SUPPORTING MATREALS. Methods and Materials

CD14 + S100A9 + Monocytic Myeloid-Derived Suppressor Cells and Their Clinical Relevance in Non-Small Cell Lung Cancer

KILLER CARBS: EFFECTS OF 50% DEXTROSE TREATMENT ON POSTNATAL HYPOGLYCEMIC RATS

Protocol for Western Blo

SOMAPLEX REVERSE PHASE PROTEIN MICROARRAY HUMAN KIDNEY TUMOR & NORMAL TISSUE

The Annexin V Apoptosis Assay

William C. Comb, Jessica E. Hutti, Patricia Cogswell, Lewis C. Cantley, and Albert S. Baldwin

Programmed necrosis, not apoptosis, is a key mediator of cell loss and DAMP-mediated inflammation in dsrna-induced retinal degeneration

Discovery and Optimization of Inhibitors of STAT3 Activation for the Treatment of Squamous Cell Carcinoma of the Head and Neck

The Need for a PARP in vivo Pharmacodynamic Assay

SUPPLEMENTAL EXPERIMENTAL PROCEDURES

Research on the inhibitory effect of metformin on human oral squamous cell carcinoma SCC-4 and CAL-27 cells and the relevant molecular mechanism.

Electrical Stimulation Control Nerve Regeneration via the p38 Mitogen-activated Protein Kinase and CREB

Comparison of Young and Old Cardiac Telocytes Using Atomic Force Microscopy

Cell Viability, DNA Damage And Relative Mitotic Arrest Dependence On Radiation Dose

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

1. Materials and Methods 1.1 Animals experiments process The experiments were approved by the Institution Animal Ethics Committee of Jilin University

Effect of starvation-induced autophagy on cell cycle of tumor cells

(a) Significant biological processes (upper panel) and disease biomarkers (lower panel)

Grass pollen immunotherapy induces Foxp3 expressing CD4 + CD25 + cells. in the nasal mucosa. Suzana Radulovic MD, Mikila R Jacobson PhD,

Transcription:

Epidermal Growth Factor-induced Cell Death and Radiosensitization in Epidermal Growth Factor Receptor-overexpressing Cancer Cell Lines KYUBO KIM 1, HONG-GYUN WU 1,2,3 and SANG-ROK JEON 1 1 Department of Radiation Oncology, and 2 Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea; 3 Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea Abstract. Background/Aim: The aim of the present study was to suggest potential mechanisms of epidermal growth factor (EGF)-induced cell death and radiosensitization in EGF receptor (EGFR)-overexpressing cancer cell lines. Materials and Methods: Two EGFR-overexpressing cancer cell lines (AMC-HN3, and A431), one EGFR-null cancer cell line (H520) and normal fibroblasts were cultured with 0.01-1000 nm of recombinant human EGF (rhegf), and a clonogenic assay was performed. After culturing serum-starved cells with 10 nm rhegf, the expression patterns of two apoptosis-associated proteins (cleaved caspase-3 and cleaved poly(adp-ribose) polymerase (PARP)) and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway were measured using immunoblotting. The radiosensitizing effect of EGF was also evaluated with a clonogenic assay and phosphorylated H2AX (γh2ax) immunofluorescence. Results: In the clonogenic assay, the number of colonies was decreased in a dose-dependent manner in EGFR-overexpressing cancer cell lines. The expression of cleaved caspase-3 and cleaved PARP was significantly induced in EGFR-overexpressing cancer cell lines. As for the PI3K/AKT/mTOR signaling pathway, EGF paradoxically suppressed the expression of PI3K, AKT and mtor in a timedependent manner. As for the radiosensitizing effect, EGF enhanced the radiosensitivity of AMC-HN3 and A431 cells. Conclusion: rhegf treatment induced cell death in the two EGFR-overexpressing cancer cell lines, and the mode of cell death was apoptosis. Moreover, cell death might be associated Correspondence to: Hong-Gyun Wu, MD, Ph.D., Department of Radiation Oncology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, Republic of Korea. Tel: +82 220723177, Fax: +82 27653317, e-mail: wuhg@snu.ac.kr Key Words: EGF, EGFR-overexpressing cancer cell line, apoptosis, PI3K/AKT/mTOR signaling pathway, radiosensitization. with the paradoxical suppression of PI3K/AKT/mTOR signaling pathway. rhegf in combination with radiation augmented the radiation effect in EGFR-overexpressing cancer cell lines via inhibition of DNA damage repair. Epidermal growth factor (EGF) was isolated in the early 1960s, and was named as such because it stimulates the proliferation of epithelial cells (1). It is produced by platelets, macrophages, and monocytes (2), and is detected in various normal tissues and body fluids such as skin, mucosa, tears, and saliva. EGF is well-known to interact with EGF receptor (EGFR) on epidermal cells and fibroblast, and to induce epithelial proliferation, modification of migration and differentiation processes (3, 4). EGF-induced cancer cell death was reported in several EGFR-overexpressing cancer cell lines as early as the 1980s (5, 6); the mode of cell death was known to be apoptosis. We also confirmed EGF-induced apoptosis in EGFRoverexpressing cancer cell lines. When combined with radiation, EGF does not inhibit the cell-killing effect of radiation and even enhances radiation-induced cell death in EGFR-overexpressing cancer cell lines (7, 8). However, the exact mechanism of EGF-induced cancer cell death and radiosensitization has not yet been elucidated. The main pathways downstream of EGFR activation include the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR), Ras/mitogen-activated protein kinase (RAS/MAPK), phospholipase C, gamma/protein kinase C (PLCγ/PKC), and signal transducers and activators of transcription (STAT) pathways (9). Among these, the STAT pathway is the most intensively investigated mechanism involving EGF-induced cancer cell death (10). However, the consequences of other signal pathway activations are still unclear. Moreover, PI3K/AKT/mTOR pathway activation is generally considered to enhance tumor growth, therefore, the EGF-induced cancer cell death is difficult to explain in relation to this pathway. 0250-7005/2015 $2.00+.40 245

In this study, we evaluated the changes of expression of the EGF EGFR signaling pathway after EGF treatment in EGFR-overexpressing cancer cell lines to suggest the potential mechanisms of EGF-induced cancer cell death. In addition, we also evaluated the radiosensitizing effect of EGF and its mechanism. Materials and Methods Study drug. recombinant human EGF (rhegf) used in this study was provided by Daewoong Parmaceutical Co. Ltd. (Seoul, Korea). A spray form of this product is commercially available and indicated for diabetic foot in Korea. Cell lines. A431 (human epidermoid carcinoma) and H520 (human lung squamous cell carcinoma) were purchased from the American Type Culture Collection (Manassas, VA, USA). AMC-HN3 (human head and neck squamous cell carcinoma) was a generous gift from Professor Sang-wook Lee at University of Ulsan, Seoul, Korea. We selected AMC-HN3 and A431 as EGFR-overexpressing cancer cell lines because AMC-HN3 is the cell line with the highest expression of EGFR among tested in our previous experiment (7), and A431 is well-known as a cell line highly expressing EGFR (5). Normal primary human cervix fibroblast was also used, and this was a kind gift from Professor Yong-Sang Song at Seoul National University, Seoul, Korea. Fibroblast was selected as a normal cell control, and we also confirmed the EGF-induced proliferation of fibroblast in previous work (7). Cells were maintained at 37 C and 5% CO 2 and cultured in RPMI-1640 (Gibco BRL, Gaithersburg, MD, USA), Dulbecco s Modified Eagle s Medium (DMEM) (Gibco BRL, Grand Island, NY, USA), and Minimum Essential Medium (MEM) (Gibco BRL, Grand Island, NY, USA). All media were supplemented with 10% fetal bovine serum (FBS) (Sigma, St. Louis, MO, USA). Serum-free media (0.05% FBS) were also used in order to exclude the effects of other growth factors present in the FBS. In vitro clonogenic assay. Mid log-phase cells from monolayer cultures were trypsinized and plated in six-well culture plates at cell densities that were optimized for the experimental conditions indicated: 2 10 2 cells were plated for AMC-HN3 cells, and 1 10 2 cells for other cell lines according to treatment conditions. The cells were then incubated for 24 h prior to treatment. To measure the effect of rhegf, 0, 0.01, 0.1, 1, 10, 100, and 1000 nm rhegf were added to the medium. Cells were incubated for additional 10 days at 37 C without changing the medium, resulting in the formation of colonies. The cells were then fixed with 100% methanol and stained with 0.5% crystal violet in methanol. The numbers of colonies (defined as more than 50 cells) were counted. The surviving fraction (SF) was defined as the ratio of the numbers of colonies to the numbers of initially plated cells corrected by plating efficiency (PE), where PE was the SF of untreated cells. Each experiment was independently repeated three times. Western blot analysis. Cells were harvested using cold lysis buffer (intron Biotechnology, Seoul, Korea). The lysates were sonicated, clarified by centrifugation at 30,285 g for 20 min at 4 C, and the supernatants were utilized for analysis. Protein concentrations of the samples were determined with a BCA protein assay kit (PIERCE, Rockford, IL, USA). The protein was mixed with 5X sample buffer (2% SDS, 5% β-mercaptoethanol, 20% glycerol, 0.001% bromophenol-blue, 0.1 M Tris-HCl, ph 6.8) and boiled for 10 minutes. Equal amounts of protein were separated by 8-12% and transferred to membranes (Millipore Corp., Bedford, MA, USA). Membranes were blocked for 1 h at room temperature with TBST [10 nm Tris-HCl (ph 7.5), 150 mm NaCl, 0.1% Tween 20] containing 5% dry milk, and then washed three times with TBST. Membranes were probed at 4 C overnight with polyclonal rabbit anti-egfr (Cell Signaling Technology, Beverly, MA, USA), pegfr (Cell Signaling Technology), cleaved caspase-3 (Cell Signaling Technology), cleaved poly(adp-ribose) polymerase (PARP) (Cell Signaling Technology), AKT (Cell Signaling Technology), PI3K (Cell Signaling Technology), mtor (Cell Signaling Technology), and β-actin (Santa Cruz Biotechnology, Santa Cruz, CA, USA). The dilutions were 1:500 for anti-egfr, and 1:1000 for other antibodies. The blots were then incubated with peroxidase-conjugated goat anti-rabbit IgG (Jackson ImmunoResearch Laboratories, West Grove, PA, USA) at a 1:1000 dilution for 2 h at room temperature and washed three times with TBST. As a loading control, the same membranes were stripped and probed with monoclonal anti-α-tubulin (Sigma) at a 1:3,000 dilution at 4 C overnight, followed by an incubation with the goat antimouse IgG used above at a 1:1000 dilution for 1 hour at room temperature and three washes with TBST. Immunoreactive proteins were visualized with WEST-ONE chemiluminescent substrate (intron Biotechnology) and signals were detected with LAS-3000 (FujiFilm, Tokyo, Japan). In vitro clonogenic assay: combination of rhegf and radiation. In the experiments using radiation with and without rhegf, 1 10 2 cells were plated for dose of 2 Gy, 3 10 2 cells for 5 Gy, and 1 10 3 cells for 10 Gy, for all cancer cell lines (AMC-HN3, A431, and H520) as well as fibroblasts. The cells were then incubated for 24 hours prior to treatment. To compare the combined effect of rhegf and radiation with that of radiation alone, cells were irradiated with 6 MV X-rays generated by a linear accelerator (Clinac 6EX; Varian, Palo Alto, CA, USA), at doses of 0, 2, 5, and 10 Gy. Where indicated, rhegf was added to the medium at the concentration of 10 nm for 24 h, after which cells were irradiated in the presence of rhegf. After 24 h, the medium was removed, and cells were incubated in drug-free media for 10 days to form colonies. Each experiment was independently repeated three times. Immunofluorescent detection of γh2ax. Cells were grown and treated in tissue culture chamber slides (Nalge Nunc International, Naperville, IL, USA). At specified times, medium was aspirated and cells were fixed in 4% paraformaldehyde for 10 minutes at room temperature. Paraformaldehyde was aspirated, and the cells were treated with a 0.2% NP40/PBS solution for 15 min. Cells were then washed in PBS twice, and antibody to phosphorylated H2AX (γh2ax) (Cell Signaling Technology) was added at a dilution of 1:200 in 1% bovine serum albumin and incubated overnight at 4 C. Cells were again washed twice in PBS before incubating in the dark with a fluorescein isothiocyanate (FITC)-labeled secondary antibody (Invitrogen, Camarillo, CA, USA) at a dilution of 1:50 in 1% bovine serum albumin for 1 h. The secondary antibody solution was then aspirated and the cells were washed twice in PBS. Cells were then incubated in the dark with 4,6-diamindino-2-phenylindole (1 μg/ml) 246

Kim et al: EGF-induced Cell Death and Radiosensitization Figure 1. Clonogenic assays of recombinant human epidermal growth factor (rhegf)-treated cells. Cells were cultured with different concentrations of rhegf (0, 0.01, 0.1, 1, 10, 100, and 1000 nm) for 10 days, and clonogenic assays were performed. Data shown are mean values in three independent experiments. in PBS for 30 minutes and washed twice, and coverslips were mounted with an antifade solution (Vector Laboratories, Burlingame, CA, USA). Slides were examined on a Leica DMRXA fluorescent microscope (Leica, Wetzlar, Germany). Images were captured by a Photometrics Sensys CCD camera (Leica, Wetzlar, Germany). Results Cell proliferation. AMC-HN3, A431, H520 and normal fibroblasts were cultured under different concentrations of rhegf (0.01-1000nM) for 10 days, and clonogenic assays were performed. For AMC-HN3 and A431, the number of colonies decreased in a dose-dependent manner, whereas the number of colonies of normal fibroblasts increased. For H520, which is the EGFR-null cell line, there was no change in the number of colonies (Figure 1). Apoptosis-associated proteins. After culturing serum-starved cells with 10 nm rhegf for different time periods (5 min, and 4, 8, 24, 48, and 72 h), we measured expression levels of two apoptosis-associated proteins, cleaved caspase-3 and cleaved PARP using immunoblotting. Treatment with rhegf induced a significant temporal increase of cleaved caspase-3 and cleaved PARP expression in the EGFR-overexpressing cell lines (AMC-HN3 and A431), whereas there was no expression in normal fibroblasts. For H520, there was no expression of cleaved caspase-3 and no significant temporal increase in the expression of cleaved PARP (Figure 2). PI3K/AKT/mTOR signaling pathway. PI3K/AKT/mTOR pathway proteins including AKT, PI3K, and mtor were evaluated in a similar manner to the above. rhegf was not associated with a significant increase in the expression of PI3K, AKT, and mtor in EGFRoverexpressing cell lines (AMC-HN3 and A431), whereas AKT and mtor expression was substantially enhanced in H520 and normal fibroblasts (Figure 3). 247

Figure 2. Recombinant human epidermal growth factor (rhegf)-induced changes in expression of epidermal growth factor receptor (EGFR), poly(adp-ribose) polymerase (PARP), and caspase-3 with respect to duration of treatment. After cells were treated with 10 nm rhegf for different time periods (5 min and, 4, 8, 24, 48 and 72 h), expression levels of cleaved caspase-3 and cleaved PARP were measured by immunoblotting. Effect on EGFR phosphorylation. To further elucidate the mechanism of EGF-induced cancer cell death, we measured EGFR phosphorylation after culturing serum-starved cells with 10 nm rhegf for different time periods (5, 10, 30 and 60 minutes) in the two EGFR-overexpressing cancer cell lines (AMC-HN3 and A431) and normal fibroblasts. rhegf treatment induced phosphorylation of EGFR residue Tyr845 in normal fibroblasts, whereas EGFR was phosphorylated at Tyr845, Tyr992, Tyr1045 and Tyr1068 in AMC-HN3 and A431 cell lines treated with rhegf (Figure 4). Combination of rhegf and radiation. We subjected two EGFR-overexpressing cell lines (AMC-HN3 and A431) to ionizing radiation, followed by a 10-day culture with either an unsupplemented medium or a medium containing 10 nm of rhegf. In the clonogenic assays, we observed that rhegf had a radio-sensitizing effect on both cell lines (Figure 5). PI3K/AKT/mTOR pathways were also evaluated in the experiment combining rhegf and radiation. There was no significant increase in the expression of PI3K, AKT, or mtor in either of the cell lines (Figure 6). Immunofluorescent detection of γh2ax. The radiosensitizing effect of EGF was assessed by γh2ax immunofluorescence (Figure 7). Radiation-induced γh2ax foci formation in AMC- HN3, A431, and normal fibroblast cells was observed. For comparison, cells treated with 5-Gy alone, and cells exposed to 10 nm rhegf prior to 5-Gy irradiation are shown. Cells were fixed and stained for γh2ax analysis 2 h after irradiation. In both AMC-HN3 and A431 cells, immunofluorescence visualization of nuclei and γh2ax foci were detected. Cells treated with 5-Gy alone and cells exposed to 10 nm rhegf prior to 5 Gy irradiation were compared and an increased radiation-induced γh2ax foci formation was found. 248

Kim et al: EGF-induced Cell Death and Radiosensitization Figure 3. Recombinant human epidermal growth factor (rhegf)-induced changes in expression of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway with respect to duration of treatment. After cells were treated with 10 nm rhegf for different time periods (5 min and, 4, 8, 24, 48 and 72 h), expression levels of PI3K, AKT, and mtor were measured using immunoblotting. Figure 4. Patterns of epidermal growth factor receptor (EGFR) phosphorylation. After cells were treated with 10 nm recombinant human epidermal growth factor (rhegf) for different time periods (5, 10, 30 and 60 min), EGFR phosphorylation was measured using immunoblotting. 249

Figure 5. Effects of recombinant human epidermal growth factor (rhegf) on cell killing by ionizing radiation. Cells were irradiated at doses of 0, 2, 5, and 10 Gy. The cell numbers used were 1 10 2 for dose of 2 Gy, 3 10 2 cells for 5 Gy, and 1 10 3 cells for 10 Gy. Where indicated, 10 nm rhegf was added to the medium for 24 h, after which cells were irradiated in the presence of rhegf. After 24 h, the medium was removed, and cells were incubated in drug-free media for 10 days to form colonies. Discussion In the present study, we reaffirmed that EGF induces apoptosis in several EGFR-overexpressing cancer cell lines, but not in EGFR-null cell line or normal fibroblasts. Given that the growth-inhibitory effect of EGF has been known to be associated with the concentration of EGF, we tested different concentrations of rhegf, and demonstrated that concentrations as low as 0.01 nm can cause significant cell death, especially in AMC-HN3 cells. This finding was somewhat contradictory to that of Gulli et al.'s study, in which they demonstrated that 10 nm EGF inhibited proliferation of A431 cells, but 0.01 nm EGF increased cell proliferation compared to untreated cells (11). Given these observations, we used a 10-nM dose of rhegf in the subsequent experiments, which is a supra-physiological level because the physiologic concentration of EGF is known to be 0.1-1 nm (12). In immunoblotting, the expression of 250

Kim et al: EGF-induced Cell Death and Radiosensitization Figure 6. Recombinant human epidermal growth factor (rhegf) and radiation-induced changes in expression of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway with respect to duration of treatment. A: A431 cell line, B: HN3 cell line. After cells were irradiated at doses of 2 and 5 Gy in the presence of 10 nm rhegf for different time periods (5 min and, 4, 8, 24, 48 and 72 h), expression levels of PI3K, AKT, and mtor were measured using immunoblotting. cleaved caspase-3 and cleaved PARP was increased with time after rhegf treatment, suggesting that the mechanism of EGF-induced cell death is apoptosis. Furthermore, the radiosensitizing effect of EGF in EGFR-overexpressing cell lines was also demonstrated. As for the mechanism of EGF-induced cancer cell death, Chin et al. reported that STAT1 was activated by EGF, and then activation of STAT1 signaling pathway resulted in the expression of caspase 1 and apoptosis (13). Later, Grudinkin et al. also reported that EGF-induced apoptosis in A431 is dependent on STAT1, but not on STAT3 (14). More recently, Tikhomirov et al. suggested p38 MAPK-dependent apoptosis as the mechanism (15). However, it was the PI3K/AKT/mTOR signaling pathway that was associated with cancer cell survival among the major pathways downstream of EGFR activation (16). In the present study, we evaluated the changes of expression of the EGF EGFR signaling pathway after EGF treatment in EGFR-overexpressing cancer cell lines, and the paradoxical suppression of PI3K/AKT/mTOR signaling pathway was observed, which was contradictory to the expected observation with the activation of EGFR by EGF. This phenomenon was 251

Figure 7. Immunofluorescent detection of phosphorylated H2AX (γh2ax). Cells were irradiated at a dose of 5 Gy. Where indicated, cells were exposed to 10 nm recombinant human epidermal growth factor (rhegf) prior to 5-Gy irradiation. Cells were fixed and stained for γh2ax analysis 2 h after irradiation. also observed when the two EGFR-overexpressing cell lines (AMC-HN3 and A431) were treated with rhegf in combination with radiation. However, the exact mechanism of the paradoxical suppression of PI3K/AKT/mTOR signaling pathway was not evaluated in our experiment. The initial activation of the pathway and the subsequent suppression could be interpreted as feedback inhibition of the signaling pathway. However, the different patterns of EGFR phosphorylation between cancer cell lines and normal cervix fibroblasts might suggest that the initial step of the EGF EGFR signaling pathway is responsible for the deferential effect of EGF. One possible hypothesis is that specific phospho-tyrosines serve to recruit specific effector molecules to activate several signaling pathways and subsequent feedback inhibition. Further studies are required for the elucidation of the exact mechanisms. In conclusion, rhegf treatment induced cell death in the two EGFR-overexpressing cancer cell lines, and the mode of cell death was apoptosis. Moreover, this cell death might be associated with the paradoxical suppression of PI3K/ AKT/mTOR signaling pathway. rhegf in combination with radiation augmented the radiation effect in EGFRoverexpressing cancer cell lines via inhibition of DNA damage repair. Conflicts of Interest None. Acknowledgements This work was supported by a grant of the Korea Health technology R&D Project, Ministry of Health & Welfare, Republic of Korea (A120313), and National R&D Program through the Dong-nam Institute of Radiological & Medical Sciences(DIRAMS) funded by the Ministry of Education, Science and Technology (50595-2013). 252

Kim et al: EGF-induced Cell Death and Radiosensitization References 1 Cohen S: The stimulation of epidermal proliferation by a specific protein (EGF). Dev Biol 12: 394-407, 1965. 2 Servold SA: Growth factor impact on wound healing. Clin Pediatr Med Surg 8: 937-953, 1991. 3 Weaver LT, Gonnella PA, Israel EJ and Walker WA: Uptake and transport of epidermal growth factor by the small intestinal epithelium of the fetal rat. Gastroenterology 98: 828-837, 1990. 4 Steidler NE and Reade PC: Histomorphological effects of epidermal growth factor on skin and oral mucosa in neonatal mice. Arch Oral Biol 25: 37-43, 1980. 5 Gill GN and Lazar CS: Increased phosphotyrosine content and inhibition of proliferation in EGF-treated A431 cells. Nature 293: 305-307, 1981. 6 Filmus J, Pollak MN, Cailleau R and Buick RN: MDA-468, a human breast cancer cell line with a high number of epidermal growth factor (EGF) receptors, has an amplified EGF receptor gene and is growth inhibited by EGF. Biochem Biophys Res Commun 128: 898-905, 1985. 7 Kwon EK, Lee SH, Kim K, Wu HG and Lee SW: Differential effects of recombinant human EGF on proliferation and radiation survival of normal fibroblast and cancer cell lines. Open Transl Med J 1: 9-15, 2009. 8 Lee SH and Wu HG: Inhibitory effect of epidermal growth factor on the proliferation of lung cancer cell lines. J Lung Cancer 9: 64-71, 2010. 9 Brand TM, Iida M, Li C and Wheeler DL: The nuclear epidermal growth factor receptor signaling network and its role in cancer. Discov Med 12: 419-432, 2011. 10 Quesnelle KM, Boehm AL, and Grandis JR: STAT-mediated EGFR signaling in cancer. J Cell Biochem 102: 311-319, 2007. 11 Gulli LF, Palmer KC, Chen YQ and Reddy KB: Epidermal growth factor-induced apoptosis in A431 cells can be reversed by reducing the tyrosine kinase activity. Cell Growth Differ 7: 173-178, 1996. 12 Lembach KJ: Induction of human fibroblast proliferation by epidermal growth factor (EGF): enhancement by an EGFbinding arginine esterase and by ascorbate. Proc Natl Acad Sci USA 73: 183-187, 1976. 13 Chin YE, Kitagawa M, Kuida K, Flavell RA and Fu XY: Activation of the STAT signaling pathway can cause expression of caspase 1 and apoptosis. Mol Cell Biol 17: 5328-5337, 1997. 14 Grudinkin PS, Zenin VV, Kropotov AV, Dorosh VN and Nikolsky NN: EGF-induced apoptosis in A431 cells in dependent on STAT1, but not on STAT3. Eur J Cell Biol 86: 591-603, 2007. 15 Tikhomirov O and Carpenter G: Ligand-induced, p38-dependent apoptosis in cells expression high levels of epidermal growth factor receptor and ErbB-2. J Biol Chem 279: 12988-12996, 2004. 16 Han W and Lo HW: Landscape of EGFR signaling network in human cancers: biology and therapeutic response in relation to receptor subcellular locations. Cancer Lett 318: 124-134, 2012. Received August 3, 2014 Revised October 28, 2014 Accepted November 3, 2014 253