AMPK signaling to promote radioresistance in nasopharyngeal carcinoma

AMPK signaling to promote radioresistance in nasopharyngeal carcinoma

ARTICLE IN PRESS Cancer Letters ■■ (2016) ■■–■■ Contents lists available at ScienceDirect Cancer Letters j o u r n a l h o m e p a g e : w w w. e l ...

3MB Sizes 4 Downloads 45 Views

ARTICLE IN PRESS Cancer Letters ■■ (2016) ■■–■■

Contents lists available at ScienceDirect

Cancer Letters j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / c a n l e t

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46

Q2 Original Articles

EBV-LMP1 suppresses the DNA damage response through DNA-PK/ AMPK signaling to promote radioresistance in nasopharyngeal carcinoma Q1 Jingchen Lu a,b,c, Hongde Li b,c, Zhijie Xu b,c, Min Tang b,c, Xinxian Weng b,c, Jiangjiang Li b,c,

Xinfang Yu d, Luqing Zhao e, Hongwei Liu f, Yongbin Hu e, Lifang Yang b,c,g, Meizuo Zhong a, Jian Zhou h, Jia Fan h, Ann M. Bode i, Wei Yi b,c, Jinghe Gao b,c, Lunquan Sun g, Ya Cao b,c,g,* a

Department of Medical Oncology, Xiangya Hospital, Central South University, Changsha, China Key Laboratory of Carcinogenesis of Chinese Ministry of Public Health, Xiangya School of Medicine, Central South University, Changsha, China c Key Laboratory of Chinese Ministry of Education, Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China d Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China e Department of Pathology, Xiangya Hospital, Central South University, Changsha, China f Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China g Molecular Imaging Center, Central South University, Changsha, China h Key Laboratory of Chinese Ministry of Education, Zhongshan Hospital, Zhongshan Hospital, Shanghai, China i The Hormel Institute, University of Minnesota, Austin, MN, USA b

A R T I C L E

I N F O

Article history: Received 24 March 2016 Received in revised form 25 May 2016 Accepted 26 May 2016 Keywords: LMP1 DNA damage response DNA-PK AMPK Nasopharyngeal carcinoma

A B S T R A C T

We conducted this research to explore the role of latent membrane protein 1 (LMP1) encoded by the Epstein–Barr virus (EBV) in modulating the DNA damage response (DDR) and its regulatory mechanisms in radioresistance. Our results revealed that LMP1 repressed the repair of DNA double strand breaks (DSBs) by inhibiting DNA-dependent protein kinase (DNA-PK) phosphorylation and activity. Moreover, LMP1 reduced the phosphorylation of AMP-activated protein kinase (AMPK) and changed its subcellular location after irradiation, which appeared to occur through a disruption of the physical interaction between AMPK and DNA-PK. The decrease in AMPK activity was associated with LMP1-mediated glycolysis and resistance to apoptosis induced by irradiation. The reactivation of AMPK significantly promoted radiosensitivity both in vivo and in vitro. The AMPKα (Thr172) reduction was associated with a poorer clinical outcome of radiation therapy in NPC patients. Our data revealed a new mechanism of LMP1-mediated radioresistance and provided a mechanistic rationale in support of the use of AMPK activators for facilitating NPC radiotherapy. © 2016 Published by Elsevier Ireland Ltd.

47 48 49 50 51 52 53 54 55 56 57 58 59 60

Introduction The Epstein–Barr virus (EBV) is an oncogenic virus implicated in the pathogenesis of a number of human malignancies of both lymphoid and epithelial origins and is associated with nearly 200,000 new malignancies each year worldwide [1–3]. Latent membrane protein 1 (LMP1) is a primary oncoprotein encoded by EBV that plays a key role in both the initiation and progression of nasopharyngeal carcinoma (NPC) [4]. Using newly developed support vector machine (SVM)-based methods, LMP1 was identified as a prognostic biomarker for NPC [5]. LMP1 activates several important oncogenic signaling pathways such as NF-κB, JNK, PI3-K/Akt, MAPK and JAK/ STAT and causes various downstream pathological changes in cell

61 62 63 64

* Corresponding author. Tel.: +86 731 84805448; fax: +86 731 84470589. E-mail address: [email protected] (Y. Cao).

proliferation, apoptosis and metastasis [6]. Recently, we found that suppression of LMP1 expression by the LMP1-targeted DNAzyme, DZ1, enhanced radiosensitivity both in vivo and in vitro [7], which suggested an important role for LMP1 in the regulation of the radiosensitivity of NPC cells. The DNA damage response (DDR) is a cellular mechanism that protects against DNA damage induced by endogenous and exogenous factors, it includes changes in cellular processes such as cell cycle regulation, DNA damage repair, apoptosis and chromatin remodeling. In recent years, the DDR has been recognized as an important innate tumor suppressor pathway [8–11]. DNA tumor virus infections activate and modulate DDR signaling pathways through multiple independent mechanisms [12–14]. Recent evidence suggests that EBV oncoproteins target multiple aspects of DDR signaling, such as heightening DNA damage by increasing the cellular levels of reactive oxygen species (ROS), disrupting the mitotic checkpoint and suppressing DNA repair mechanisms [15].

http://dx.doi.org/10.1016/j.canlet.2016.05.032 0304-3835/© 2016 Published by Elsevier Ireland Ltd.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

2

82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122

DNA double strand breaks (DSBs) are considered to be the most lethal form of DNA damage induced by DNA-damaging agents, such as ionizing irradiation. The ability to repair DSBs is related not only to cancer susceptibility but also to sensitivity of cells to radiotherapy and chemotherapy [16]. The DNA-dependent protein kinase (DNA-PK) is a critical component of the DSB repair machinery [17–20]. It is a serine/threonine kinase complex composed of a heterodimer of the catalytic subunit DNA-PKcs and the regulator subunit Ku proteins (Ku70/Ku86). In response to DSB formation, DNAPKcs is recruited to DSBs by the DNA end-binding Ku70/86 heterodimer and is rapidly phosphorylated at multiple serine and threonine residues. DNA-PKcs phosphorylation at the Thr2609 cluster region is particularly significant because it is critical for DSB repair [21,22]. In addition to its classical role in DSB repair, recent findings demonstrate damage-independent functions of DNA-PK that affect a variety of critical cellular processes associated with malignancy [23,24]. Specifically, a recent report defines a general role for DNA-PK in metabolic regulation [25–27]. AMP-activated protein kinase (AMPK) is a crucial energy sensor that helps maintain cellular energy homeostasis and especially as a negative regulator of glycolysis [28,29]. It is a serine/threonine protein kinase composed of a heterotrimeric complex, including α, β and γ subunits. The α-subunit of AMPK has catalytic activity and phosphorylation of AMPKα at Thr172 is necessary for full enzyme activity [30,31]. Besides acting as an energy sensor, recent work describes a novel function for AMPK as a sensor of genomic stress and a participant in the DDR pathway [32–34]. In this study, we report that in NPC cells, EBV-encoded LMP1 repressed DSB repair by inhibiting DNA-PK phosphorylation and activity. Moreover, LMP1 disrupted the physical interaction between DNA-PK and AMPK and reduced the phosphorylation of AMPK by disturbing the subcellular location of the protein after DNA damage. Reactivation of AMPK markedly inhibited glycolysis and promoted apoptosis induced by DNA damage, and enhanced radiosensitivity both in vivo and in vitro. This study revealed a new mechanism of LMP1-induced radioresistance occurring through the modulation of DDR signaling pathways and provided a mechanistic rationale supporting the use of AMPK activators for facilitating NPC radiotherapy.

123

Cell lines and culture

124 125 126 127 128 129 130

CNE1-LMP1 and HNE2-LMP1 cells are NPC cell lines that were constructed from LMP1-negative NPC cell lines, CNE1 and HNE2, respectively, which were stably transfected with an LMP1 plasmid. Cells were grown in RPMI 1640 (Gibco BRL, Gaithersburg, MD, USA) supplemented with 10% fetal bovine serum (FBS; Hyclone, Logan, UT, USA).

131 132 133 134 135 136 137 138 139 140 141 142 143

NU7026, metformin, DAPI (4′,6′-diamidino-2-phenylindole) and the antibody against β-actin were purchased from Sigma-Aldrich (St. Louis, MO, USA). The antibody against LMP1 was from DAKO (Glostrup, Denmark). The antibodies against DNAPKcs, α-tubulin, Cy3-conjugated anti-rabbit IgG, Alexa 488-conjugated anti-mouse IgG and Alexa 488-conjugated anti-goat IgG for immunofluorescence were from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antibodies against phosphorylated DNAPKcs (Thr2609) and histone H3 were from Abcam (Cambridge, UK). Antibodies against AMPKα, phosphorylated AMPKα (Thr172) and phosphorylated histone H2AX (Ser139) were from Cell Signaling Technologies (Danvers, MA, USA). The Annexin-V FITC Apoptosis Kit was purchased from KeyGEN Biotech (Nanjing, Jiangsu, P.R. China) and Lipofectamine 2000 was purchased from Invitrogen (Carlsbad, CA, USA).

144 145 146 147

Protein samples (50–100 μg) were separated by 6–12% SDS-PAGE, transferred onto a nylon membrane and immunoblotted with primary antibodies. Binding of primary antibodies was detected using peroxidase-conjugated secondary antibodies and developed with an enhanced chemiluminescence reagent. Visualization of

Materials and methods

Reagents and antibodies

Western blot analysis

proteins was performed using the ChemiDoc XRS system with Image Lab software (Bio-Rad, Hercules, CA).

148 149

Co-immunoprecipitation (Co-IP)

150

Cells were disrupted with IP lysis buffer containing protease inhibitor cocktails (Roche Diagnostics, Mannheim, Germany). Aliquots of 2000 μg of proteins from each sample were pre-cleared by incubation with 20 μl of Dynabeads protein G (Invitrogen) for 1 h at 4 °C. Pre-cleared samples were incubated with a DNA-PKcs antibody (2 μg/ sample) overnight at 4 °C. Then 20 μl Dynabeads protein G was added into each sample and incubated for 2 h at 4 °C. The beads were then washed 3 times with cold lysis buffer, boiled, separated by 6–12% SDS–PAGE, and transferred onto a nylon membrane followed by Western blot analysis.

151 152 153 154 155 156 157 158

Immunofluorescence analysis

159

The cells were fixed and permeabilized with −20 °C cold methyl alcohol for 10 min, then blocked in 5% donkey serum in PBS for 1 h and then incubated with a primary antibody in PBS containing 1% BSA at 4 °C overnight. The cells were washed 3 times with PBS, and incubated for 30 min with a secondary antibody for 30 min. The cells were washed with PBS and stained with DAPI for 10 min and viewed by a confocal laser scanning microscope (LSM 510 META, Carl Zeiss, Germany).

160 161 162 163 164 165

Immunohistochemistry

166

Immunohistochemistry (IHC) was performed using a Histomouse SP Broad Spectrum DAB kit (Invitrogen-Zymed, Carlsbad, CA, USA). Paraffin sections were immunostained using a streptavidin–peroxidase procedure after microwave antigen retrieval. The signal was detected using a diaminobenzidine solution. The stained sections were independently examined by two pathologists and the staining intensity of each protein was scored.

167 168 169 170 171 172

Clonogenic survival assay

173 1 × 102,

Cells were counted and seeded in six-well plates in triplicate at densities of 1 × 103, 1 × 104 and 1 × 105 per well and exposed to 0, 4, 6 or 8 Gy doses of irradiation according to the density of the cells. After 7–14 days, cells were fixed with methanol, stained with crystal violet, and colonies containing more than 50 cells were counted using Image J. Results are plotted as the mean surviving fraction ± S.E.M. The survival curves were drawn using the GraphPad Prism 5 software program (GraphPad Software, La Jolla, CA, USA).

174 175 176 177 178 179 180

Comet assay

181

A neutral comet assay was performed using the comet assay kit (Trevigen, Gaithersburg, MD, USA) according to the manufacturer’s protocol.

182 183

DNA-PK activity assay

184

A kinase activity assay for DNA-PK was performed using the SignaTECT® DNADependent Protein Kinase Assay System kit (Promega) according to the manufacturer’s protocol. The radioactivity was determined in a liquid scintillation counter (TriCarb® 2810, PerkinElmer, Waltham, MA, USA).

185 186 187 188

Irradiation of cells

189

Cells were irradiated at a dose rate of 2 Gy/min at room temperature using an electron linear accelerator (Siemens Primus, E. Germany) as an X-ray source and were allowed to recover at 37 °C under 5% CO2 for the times indicated.

190 191 192

Animal experiments

193

All mice were maintained and manipulated according to strict guidelines established by the Medical Research Animal Ethics Committee, Central South University, China. Six-week-old female athymic nude mice (BALB/C) were injected with 5 × 106 CNE1-LMP1 cells. Tumor volumes were calculated using the formula: volume = length × width2/2. When the tumor volume reached 60–100 mm3, the animals were treated with intraperitoneal injection of metformin (250 mg/kg daily) or water every day [35]. In groups of mice subjected to irradiation treatment, local irradiation of 5 Gy was administered once a week for three weeks. Ten days after the last irradiation, the animals were sacrificed and the tumors were removed.

194 195 196 197 198 199 200 201 202

Statistical analysis

203

Data analysis was conducted by the SPSS software (version 21.0; SPSS). Statistical differences were determined using Student’s t-test or one-way ANOVA. The Kaplan–Meier method was used to estimate overall survival and the log-rank test was used to evaluate differences between survival curves. All values are expressed as mean values ± S.E. of 3 individual experiments. A value of p < 0.05 was considered statistically significant.

204 205 206 207 208 209

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275

Results LMP1 represses DSB repair in NPC cells Once DSBs are induced in cells, serine 139 of histone H2AX is quickly phosphorylated around the DSB site and is then referred to as γH2AX, which is a sensitive marker of DSBs [36]. To determine the effect of LMP1 expression on the ability of NPC cells to repair DSBs, we first measured γH2AX foci formation by immunofluorescence staining intensity after irradiation. Cells included LMP1-positive cells, CNE1-LMP1 and HNE2-LMP1, and LMP1-negative cells, CNE1 and HNE2. In non-irradiated NPC cells, the percentage of γH2AX-positive cells was very low. No difference was observed in either LMP1-positive or -negative cells. Irradiation induced a robust formation of γH2AX foci and the percentage of γH2AX-positive NPC cells increased by 8-fold (p < 0.05) at 0.5 h post-irradiation. At 24 h post-irradiation, a time point that most DSBs should be repaired, the percentage of γH2AX-positive cells was 12.7% in CNE1 cells compared to 29.7% in CNE1-LMP1 cells (p < 0.05; Fig. 1A) and was 11.2% and 28.6% in HNE2 and HNE2-LMP1 cells, respectively (p < 0.01; Fig. 1B). Then we used Western blot analysis to detect the protein level of γH2AX in NPC cells. In non-irradiated cells, the protein level of γH2AX was not different in CNE1 or CNE1-LMP1 cells, but was higher in HNE2-LMP1 cells compared to HNE2 cells (Fig. 1C, D). After exposure to irradiation, γH2AX levels in both CNE1-LMP1 and HNE2LMP1 cells were higher compared with those observed in CNE1 or HNE2 cells at different time points (Fig. 1C, D). We also used a neutral comet assay to investigate DSB repair capacity. Irradiation significantly increased comet formation in NPC cells and the comet tails were gradually eliminated as the DSBs were repaired (Fig. 1E, F). The length of the comet tails in LMP1-positive cells was significantly longer than LMP1-negative cells at either 1 or 4 h after irradiation. Specifically, CNE1-LMP1 cells exhibited tails of 200.945 nm vs. 151.699 nm in CNE1 cells at 1 h (p < 0.05) and 253.625 nm vs. 100.294 nm at 4 h (p < 0.01; Fig. 1G). Compared to HNE2 cells, HNE2LMP1 cells also retained longer comet tails of 167.852 nm vs. 116.297 nm at 1 h (p < 0.001) and 89.318 nm vs. 49.655 nm (p < 0.001) at 4 h after irradiation (Fig. 1H). These data indicated that LMP1 repressed the ability of NPC cells to perform DSB repair. LMP1 inhibits DSB repair by attenuating DNA-PK phosphorylation and activity Because DNA-PK plays a dominant role in DSB repair, we performed Western blot analysis to detect DNA-PKcs in LMP1-positive and -negative NPC cell lines. Results indicated that total DNAPKcs protein levels were not significantly different between CNE1LMP1 cells and CNE1 cells (Fig. 2A), but decreased in HNE2-LMP1 cells compared to levels observed in HNE2 cells (Fig. 2B). We then measured the mRNA level of DNA-PKcs and found no difference between LMP1-positive or -negative NPC cell lines (Fig. 2C). DNAPKcs phosphorylation at the Thr2609 cluster region is essential for its activity, we found that exposure to irradiation induced a robust phosphorylation of DNA-PKcs at Thr2609, and LMP1 substantially inhibited this phosphorylation in NPC cells (Fig. 2D, E). Accordingly, the DNA-PK activity decreased in LMP1-positive NPC cell lines (p < 0.001; Fig. 2F, G). After knockdown of LMP1 (Fig. 2H), the phosphorylation level of DNA-PKcs at Thr2609 increased (Fig. 2I, J). These data indicated that LMP1 inhibited the phosphorylation and activity of DNA-PK, but had an unsettling effect on the total protein level of DNA-PKcs and no effect on the mRNA level of DNA-PKcs. LMP1 inhibits AMPK phosphorylation through disrupting its subcellular location and the interaction between AMPK and DNA-PK DSB repair is accompanied by increased energy consumption. As AMPK is not just an energy sensor, but also a potential genomic

3

stress sensor participating in the DDR, we examined the impact of LMP1 on AMPK expression and phosphorylation. The total protein level of AMPKα was not different between LMP1-positive and -negative NPC cells. However, the phosphorylation of AMPKα at Thr172 was substantially decreased in LMP1-positive cells compared to LMP1-negative cells (Fig. 3A), suggesting that LMP1 influences AMPK phosphorylation without affecting the total levels of protein expression. In CNE1 cells, AMPKα was localized in both cytoplasm and nucleus (Fig. 3B), but after irradiation, the nuclear AMPKα shifted to the cytoplasm, resulting in an increase in cytoplasmic AMPKα protein expression. However, in CNE1-LMP1 cells, AMPKα was mainly distributed in the cytoplasm, and no substantial change in cytoplasmic AMPKα protein level was observed after irradiation. Consistent with the results of total protein expression, phosphorylated AMPKα (Thr172) localized in the cytoplasm was higher in CNE1 cells compared to CNE1-LMP1 cells (Fig. 3C). These results suggest that LMP1 might inhibit the phosphorylation of AMPKα by regulating its subcellular localization. A recent study suggests that DNA-PK is an important regulator of AMPK activation in response to energy depletion [25]. We hypothesize that LMP1 might regulate the phosphorylation of AMPK by modulating DNA-PK. In LMP1-positive cells, phosphorylation of AMPKα at Thr172 was suppressed by NU7026, a specific DNA-PK inhibitor. However, the phosphorylation was not affected in LMP1-negative cells (Fig. 3D). We next determined whether DNA-PK could physically interact with AMPK after irradiation and whether this association was affected by LMP1. DNA-PKcs was immunoprecipitated from NPC cells and the presence of the AMPKα subunit was detected by immunoblotting. DNA-PKcs coimmunoprecipitated with AMPKα in both LMP1-negative and -positive cells. However, greater amounts of the AMPKα protein co-immunoprecipitated with DNA-PKcs in LMP1-negative cells (Fig. 3E, F), indicating that the interaction of DNA-PKcs with AMPKα was disrupted by LMP1. To further expand these results, we performed an immunofluorescence assay. Irradiation induced the co-localization of DNA-PKcs and AMPKα in the cytoplasm of LMP1-negative cells; however, co-localization was almost undetectable in LMP1-positive cells (Fig. 3G, H). These data indicated that LMP1 could possibly inhibit the phosphorylation of AMPKα (Thr172) directly through DNA-PK or indirectly by disrupting the interaction of AMPKα with DNA-PKcs.

276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318

Reactivation of AMPK reverses LMP1-mediated NPC cell radioresistance Recent findings suggest that activation of AMPK could radiosensitize tumor cells. To determine whether LMP1-mediated AMPK inactivation confers radioresistance to NPC cells, we treated cells with metformin (5 μM), which is a pharmacological drug that can activate AMPK [28], and then examined the colony formation. Reactivation of AMPK significantly reduced colony formation of LMP1-positive NPC cells (Fig. 4A, B) and resulted in a lower survival rate after irradiation compared with LMP1-negative NPC cells (p < 0.01; Fig. 4C, D). To further investigate whether reactivation of AMPK could reverse LMP1-induced radioresistance in vivo, we treated athymic nude mice bearing CNE1-LMP1 xenografts with metformin alone, irradiation alone, or both metformin and irradiation. Treatment with metformin alone had no effect on tumor growth and irradiation alone significantly suppressed tumor growth, whereas metformin combined with irradiation therapy further inhibited tumor growth (p < 0.01; Fig. 4E, F). These data indicated that reactivation of AMPK could reverse LMP1-induced cellular radioresistance in NPC cells.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340

ARTICLE IN PRESS 4

341 342 343 344 345 346 347 348 349 350

J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

Fig. 1. LMP1 represses double-strand break repair of nasopharyngeal carcinoma cells. (A) Analysis of H2AX foci at different times after 4 Gy 6 MV X-ray irradiation. Immunofluorescence staining for γ-H2AX followed by confocal microscopy was performed. Cells displaying 10 or more foci were counted as positive and 300–500 cells were counted. Representative images of γH2AX foci and the percentage of CNE1 and CNE1-LMP1 cells displaying γH2AX foci are shown. (B) Representative images of γH2AX foci and the percentage of HNE2 and HNE2-LMP1 cells displaying γH2AX foci are shown. (C, D) Detection of γH2AX in CNE1/CNE1-LMP1 or HNE2/HNE2-LMP1 cells at different times after irradiation. Total proteins were or were not exposed to 4 Gy irradiation and prepared at 0.5, 1, 4 or 24 h after irradiation. The γH2AX protein level was determined by Western blot analysis using specific antibodies to detect γH2AX and β-actin. (E and F) Neutral comet assay of CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells. Comet images of DSBs detected by neutral single cell gel electrophoresis at 0.5, 1, 4 and 24 h after 4 Gy X-ray irradiation are shown. (G and H) The repair kinetics of 4 Gy-induced DNA DSBs were detected by comet assay. The tail moment was used as the endpoint of DSBs. One hundred individual comets were counted per time point for each experiment. The tail moments were measured and data are represented as the mean tail moment from 3 independent experiments. Data are shown as mean values ± S.D. from 3 independent experiments. For A, B, G, and H, the asterisks indicate a significant (*p < 0.05, **p < 0.01 and ***p < 0.001) difference.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

351 352 353 354 355 356 357 358 359 360

5

Fig. 2. LMP1 inhibits double-strand break repair by inhibiting DNA-PK phosphorylation and activity. (A, B) CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells were or were not exposed to 4 Gy X-ray irradiation. After 24 h, cells were harvested and analyzed for DNA-PKcs protein levels. (C) Evaluation of DNA-PKcs mRNA levels by real-time RT-PCR analysis in CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells at 24 h after irradiation at a dose of 4 Gy. (D, E) CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells were exposed to 4 Gy irradiation. After 30 min, cells were harvested and analyzed for DNA-PKcs phosphorylation at Thr2609. (F, G) CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells were exposed to 4 Gy irradiation. After 30 min, whole-cell extracts (25–50 μg) prepared from NPC cells were assayed for DNA-PK activity as described in Materials and Methods. (H) The knockdown effect of shLMP1 in CNE1-LMP1 cells. CNE1-LMP1 cells were transfected with shLMP1 and at 48 h after transfection, cells were harvested and analyzed for LMP1 protein levels. (I) CNE1-LMP1 cells were transfected with shLMP1 and at 48 h after transfection, CNE1-LMP1 and CNE1 cells were exposed to 4 Gy irradiation. After 30 min, cells were harvested and analyzed for DNA-PKcs phosphorylation at Thr2609. (J) HNE2-LMP1 cells were transfected with shLMP1 and at 48 h after transfection, HNE2LMP1 and HNE2 cells were exposed to 4 Gy irradiation. After 30 min, cells were harvested and analyzed for DNA-PKcs phosphorylation at Thr2609.Data are shown as mean values ± S.D. from 3 independent experiments and for F, the asterisks indicate a significant (***p < 0.001) difference.

361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377

Reactivation of AMPK inhibits LMP1-induced glycolysis and promotes apoptosis of NPC cells after DNA damage Next we investigated the possible mechanisms of activation of AMPK in promoting radiosensitivity. Emerging evidence indicates that AMPK is a negative regulator of aerobic glycolysis (i.e., the Warburg effect) in cancer cells and suppresses tumor growth in vivo. Thus, we examined the level of glucose consumption and lactate production in NPC cells after irradiation. Compared with LMP1negative cells, LMP1-positive NPC cells displayed increased glucose consumption and lactate production. After treatment with metformin (Fig. 5A), glucose consumption and lactate production decreased dramatically in LMP1-positive NPC cells (Fig. 5B-E). These findings indicated that LMP1 might promote glycolysis by inhibiting AMPK. Apoptosis is a major form of cell death induced by irradiation. To gain insight into the role of AMPK in apoptosis induced by DNA

damage in NPC cells, we performed fluorescence-activated cell sorting (FACS) to assess apoptosis in LMP1-positive and -negative NPC cells. Results indicated that LMP1-negative NPC cells underwent substantial apoptosis induced by irradiation, whereas LMP1positive cells were relatively resistant (Fig. 5F, G). However, treatment with metformin significantly increased apoptosis in LMP1-positive cells. These results suggested that LMP1 might inhibit DNA damageinduced apoptosis by inhibiting AMPK. Decreased phosphorylation of DNA-PK and AMPK corresponds with overall survival of NPC patients Because decreased phosphorylation of AMPKα (Thr172) plays an important role in LMP1-mediated radioresistance, we determined whether reduction of DNA-PK and AMPK phosphorylation was associated with the prognosis of NPC patients. We used IHC staining

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393

ARTICLE IN PRESS 6

394 395 396 397 398 399 400 401 402 403 404 405 406

J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

Fig. 3. LMP1 inhibits AMPK phosphorylation through disrupting its subcellular localization and the interaction between AMPK and DNA-PK. (A) CNE1/CNE1-LMP1 and HNE2/ HNE2-LMP1 cells were exposed to 4 Gy irradiation. After 30 min, cells were harvested and analyzed for total AMPKα protein levels and phosphorylation at Thr172. (B) CNE1 and CNE1-LMP1 cells were either left untreated or treated with 4Gy irradiation. After 30 min, cells were fractionated into cytoplasmic or nuclear extracts as described in Materials and Methods. Protein (50 μg) was separated by SDS-PAGE and probed for AMPKα, histone H3 and tubulin as indicated. (C) CNE1 and CNE1-LMP1 cells were either left untreated or treated with 4 Gy irradiation. After 30 min, cells were fractionated into cytoplasmic or nuclear extracts. Protein (50 μg) was separated by SDS-PAGE and probed for AMPKα (Thr172), histone H3 and tubulin as indicated. (D) CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells were irradiated in the presence or absence of NU7026 (10 μM, 1 h before irradiation). After 30 min, cells were harvested and analyzed for AMPKα phosphorylation at Thr172. (E, F) CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells were exposed to 4 Gy of irradiation and then whole-cell lysates were prepared. Proteins were immunoprecipitated from the lysates with a DNA-PKcs antibody and the immunoprecipitates were subjected to immunoblot analysis with an antibody specific for AMPKα or control immunoglobulin (normal rabbit IgG). Representative blots from 3 independent experiments are shown. (G, H) CNE1/CNE1-LMP1 and HNE2/HNE2-LMP1 cells were mock-treated or exposed to 4 Gy of irradiation and allowed to recover for 30 min before fixation and staining with monoclonal antibodies to detect DNA-PKcs (green) and AMPKα (red). The images were acquired using confocal microscopy. Representative images from 3 independent experiments are shown. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

407 408 to examine the expression status of LMP1, DNA-PKcs (Thr2609), 409 AMPKα (Thr172) and γH2AX in a commercial NPC tissue array. 410 Clinical–pathological characteristics of 60 patients are listed in the 411 Supplementary data (Table S1). Results showed that the expres412 sion of LMP1 was significantly higher in NPC tumors (p = 0.001). 413 Strong or moderate expression of phosphorylated DNA-PKcs 414 (Thr2609) and AMPKα (Thr172) was found in normal nasopharyn415 geal epithelium, but a low level of these 2 phosphorylated proteins 416 was common in primary NPC specimens (p < 0.05) and correlated 417 significantly with the expression of LMP1 (Fig. 6A). The correla418 tion coefficient between LMP1 and phosphorylated DNA-PKcs 419 Q4 (Thr2609) was 0.389 (p < 0.01) or phosphorylated AMPKα (Thr172) 420 was 0.353 (p < 0.05; Fig. 6B). In addition, the correlation coeffi421 cient between phosphorylated DNA-PKcs (Thr2609) and 422 phosphorylated AMPKα (Thr172) was 0.458 (p < 0.001; Fig. 6B). These 423 findings indicated that DNA-PKcs and AMPKα inactivation corre424 lates with LMP1 expression in NPC. 425 In contrast, the expression of γH2AX was significantly higher in 426 NPC tumors compared to expression in normal nasopharyngeal ep427 ithelium (p < 0.001). Strong or moderate expression was observed 428 in NPC tumors, particularly in tumors with positive LMP1 stain-

ing. The correlation coefficient between LMP1 and γH2AX was 0.566 (p < 0.01) and the correlation between phosphorylated DNA-PKcs (Thr2609) and γH2AX was 0.476 (p < 0.01). In contrast, no significant correlation was observed between phosphorylated AMPKα (Thr172) and γH2AX (0.064, p > 0.05). These data indicated that LMP1 inhibits DSB repair in NPC cells at least partly through DNA-PK inactivation. We divided NPC patients into two groups based on the expression level of phosphorylated DNA-PKcs (Thr2609) and phosphorylated AMPKα (Thr172) and analyzed the correlation between their expression level and the 5-year overall survival (OS) rate. Results of a univariate analysis indicated that the expression of DNA-PKcs (Thr2609) was not associated with the 5-year OS rate, which was 66.67% in the DNA-PKcs (Thr2609)negative group compared to 70.37% in the DNA-PKcs (Thr2609)positive group (p = 0.757; Fig. 6C). However, the 5-year OS rate was significantly lower in AMPKα (Thr172)-negative patients compared to AMPKα (Thr172)-positive patients (53.57% vs. 78.26%, p = 0.037; Fig. 6D). These results suggested that AMPKα (Thr172) reduction was associated with a poorer clinical outcome of radiation therapy in NPC patients.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

450 451 452 453 454 455 456 457 458 459 460 461

7

Fig. 4. Reactivation of AMPK reverses LMP1-induced NPC cell radioresistance. (A) Sensitization of radioresistant LMP1-positive NPC cells by activation of AMPK. Colony formation of CNE1 and CNE1-LMP1 cells treated or not treated with metformin (5 μM) for 1 h before irradiation was evaluated 10 days after treatment with a single dose of 0, 4, 6 or 8 Gy irradiation. (B) Colony formation of HNE2 and HNE2-LMP1 cells treated or not treated with metformin (5 μM 1 h before irradiation) was evaluated 7 days after treatment with a single dose of 0, 4, 6 or 8 Gy irradiation. (C) Survival curve of CNE1 and CNE1-LMP1 cells treated or not treated with metformin. Surviving fractions were calculated by comparing the colony number of each treatment group with untreated groups (0 Gy). Results are plotted as the mean surviving fraction ± S.E.M. The survival curves were drawn using the GraphPad Prism 5 software program. (D) Survival curve of HNE2 and HNE2-LMP1 cells treated or not treated with metformin. Surviving fractions were calculated by comparing the colony number of each treatment group with untreated groups (0 Gy). Results are plotted as the mean surviving fraction ± S.E.M. The survival curves were drawn using the GraphPad Prism 5 software program, the asterisks indicate a significant (**p < 0.01 and ***p < 0.001) difference. (E) Representative images of xenografts from different treatment groups. The nasopharyngeal carcinoma xenograft model was established using CNE1-LMP1 cells. When the tumor volume reached 60–100 mm3, the animals were irradiated with a single dose of 5 Gy per week for 3 weeks alone or combined with intraperitoneal injection of metformin (250 mg/ kg daily). The tumor volume was measured every 2 days. Ten days after the last irradiation, the animals were sacrificed and the tumors were removed. (F) The tumor growth curve of different treatment groups in a CNE1-LMP1 cell xenograft model, the asterisks indicate a significant (**p < 0.01 and ***p < 0.001) difference.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

8

462 463 464 465 466 467 468 469 470

Fig. 5. Repression of AMPK activity by LMP1 promotes glycolysis and inhibits apoptosis of NPC cells after DNA damage. (A) CNE1 and CNE1-LMP1 cells were treated with metformin (5 μM) for 1 h. Then the cells were harvested and analyzed for phosphorylation at AMPKα (Thr172). (B, C) CNE1 and CNE1-LMP1 cells were treated with 4 Gy of irradiation in the presence or absence of metformin (5 μM, 1 h before irradiation). The relative levels of glucose consumption and lactate production rate at the indicated times were examined in these cell lines using the Automatic Biochemical Analyzer. (D, E) HNE2 and HNE2-LMP1 cells were irradiated with 4 Gy of irradiation in the presence or absence of metformin (5 μM, 1 h before irradiation). The relative levels of glucose consumption and lactate production rate at the indicated times were examined. (F, G) FACS analyses of apoptosis in CNE1 and CNE1-LMP1 cells and HNE2 and HNE2-LMP1 cells. Cells were irradiated in the presence or absence of metformin (5 μM, 1 h before irradiation). After 24 h cells were harvested and stained with Annexin-V and propidium iodide, and then analyzed using a FACScan. The percentages of Annexin-Vpositive apoptotic cells are shown as mean values ± S.D. from 3 independent experiments. For B–G, the asterisks indicate a significant (* p < 0.05; **p < 0.01; ***p < 0.001) difference.

471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493

Discussion The global burden of mortality from EBV-attributed malignancies accounts for 1.8% of all cancer deaths in 2010, in which NPC and gastric cancer accounted for 92% of all EBV-attributed cancer deaths [37,38]. Human DNA tumor viruses often impinge on the DDR in order to establish the necessary cellular milieu to replicate viral DNA. Studies have shown that EBV-encoded LMP1 can modulate the DDR by inhibiting DNA repair through different mechanisms, such as repressing the PI3-K/Akt/FOXO3a pathway or p53-mediated DNA repair and transcriptional activity [39,40]. In this study, we demonstrated that LMP1 could repress DSB repair in NPC cells, at least partly by inhibiting the phosphorylation and activity of DNA-PK, a central component of DSB repair. Corresponding with this, γH2AX, a marker for DSBs, was significantly higher in NPC tumors, especially in LMP1-positive tumors, suggesting that LMP1 impaired the ability to repair DSBs in NPC and might produce genetic instability, which is a cause of cancer. AMPK is a critical regulator of cellular energy homeostasis. Recent work describes a novel function for AMPK as a sensor of genomic stress and as a participant in the DDR pathway. Herein, we found that phosphorylated AMPKα (Thr172) mainly localized in the cy-

toplasm after DNA damage and was substantially decreased in CNE1LMP1 cells compared to CNE1 cells. After exposure to irradiation, AMPKα localization in the nucleus shifted to the cytoplasm in CNE1 cells, but not in CNE1-LMP1 cells. These findings suggested that the decrease in phosphorylation of AMPKα (Thr172) in LMP1-positive NPC cells was partly due to the decrease in total protein levels of AMPKα in the cytoplasm. Thus, LMP1 might participate in regulating the subcellular location of AMPKα after DNA damage. A recent study revealed that in human glioma cells, DNA-PKcs interacted with the regulatory γ subunit of AMPK and positively regulated AMPK phosphorylation and activation under glucose-deprived conditions. This suggested that DNA-PK is an important regulator of AMPK activation in response to energy depletion [25]. To elucidate the role of DNA-PK in regulating AMPK activation in response to irradiation, we treated LMP1-positive cells with NU7026, a specific DNA-PK inhibitor, and found that the phosphorylation of AMPKα (Thr172) was further decreased, suggesting that DNA-PK is a positive regulator of AMPK activation in response to DNA damage. We also observed that the catalytic subunit of DNA-PK physically interacted with the α-subunit of AMPK under DNA damage conditions, whereas LMP1 disrupted this interaction. These data indicated that the interaction between AMPK and DNA-PK promotes the phos-

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

516 517 518 519 520 521

9

Fig. 6. Decreased phosphorylation of DNA-PK and AMPK corresponds with overall survival of NPC patients. (A) Immunohistochemistry analysis was used to examine the expression of LMP1, DNA-PKcs (Thr2609), AMPKα (Thr172) and γH2AX in an NPC tissue array. Case 1 was a biopsy from a normal nasopharyngeal epithelium and Cases 2 and 3 were both biopsies from NPC tumors. (B) Correlation between LMP1 and phosphorylated DNA-PKcs (Thr2609), AMPKα (Thr172) and γH2AX expression in the NPC tissue array. (C) Five-year overall survival rates of NPC patients with DNA-PKcs (Thr2609)-negative or -positive expression were estimated with the Kaplan–Meier method by log-rank test (p = 0.757). (D) Five-year overall survival rates of NPC patients with AMPKα (Thr172)-negative or -positive expression were estimated with the Kaplan– Meier method by log-rank test (p = 0.037).

522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552

phorylation of AMPKα at Thr172 and LMP1 might affect the phosphorylation by disrupting this interaction. Recently, a series of our studies confirmed that LMP1 contributes to the radioresistance of NPC. LMP1 might promote radioresistance by regulating tumor angiogenesis through the JNKs/ HIF-1 pathway or glycolysis through the up-regulation of hexokinase 2 (HK2), a rate-limiting enzyme of glycolysis, or by inhibiting telomerase activity in NPC cells [41–43]. Other studies showed that LMP1-induced cancer stem cell (CSC)-like properties might contribute to radioresistance in NPC cells [44]. Here we found that reactivation of AMPK by metformin could substantially reverse radioresistance of LMP1-positive NPC cells both in vitro and in vivo, suggesting that AMPK plays an important role in regulating LMP1mediated radioresistance. AMPK is considered a negative regulator of aerobic glycolysis and recent findings revealed that the lactate concentration was significantly correlated with tumor response to fractionated irradiation, probably due to its anti-oxidative capacity [45–47]. We found that LMP1-positive NPC cells displayed dramatically increased glucose consumption and lactate production after irradiation, a metabolic signature consistent with the Warburg effect. When AMPK was activated with metformin, glucose consumption and lactate production in LMP1-positive NPC cells dropped back to a similar level as observed in LMP1-negative NPC cells. Apoptosis is a primary means of death after DNA damage. We found that LMP1 profoundly inhibited irradiation-induced apoptosis, but apoptosis increased dramatically in LMP1-positive cells after reactivation of AMPK. Together these findings suggested that high concentrations of lactate and resistance to apoptosis may contribute to radioresistance in LMP1-positive NPC cells, whereas AMPK reactivation reversed ra-

dioresistance by negatively regulating aerobic glycolysis and promoting apoptosis. A low level of phosphorylated AMPK is common in primary NPC specimens and significantly correlates with the expression of LMP1 [48]. Our data revealed that a lower level of phosphorylated AMPKα (Thr172) predicted a poorer 5-year overall survival rate in NPC patients. A previous study showed that DNA-PKcs overexpression was observed in 36.8% of NPC tumor specimens and was highly correlated to advanced clinical stages and poor survival [49]. Another study revealed that negative expression of DNA-PKcs was detected in 35 of 87 (40.2%) NPC patients and was significantly associated with poor patient survival [50]. A separate study by Lee et al., however, showed no association between DNA-PKcs overexpression and the clinical outcome of NPC [51]. Herein, we found no significant relationship between phosphorylated DNA-PKcs (Thr2609) expression and the 5-year overall survival rate of NPC patients, which might possibly be due to the small sample size. Overall, this study demonstrated that EBV-encoded LMP1 impaired DSB repair induced by irradiation, possibly by inhibiting DNAPK phosphorylation and activity. Moreover, LMP1 reduced the phosphorylation of AMPK and disturbed its subcellular localization after irradiation, possibly by disrupting the physical interaction between AMPKα and DNA-PKcs. The decrease in AMPK activity was involved in LMP1-mediated glycolysis and resistance to apoptosis induced by irradiation, which might confer radioresistance to NPC cells. Our data revealed a novel mechanism for LMP1-mediated radioresistance by suppressing the DDR through DNA-PK/AMPK signaling. This could provide a mechanistic rationale supporting the use of AMPK activators, such as metformin, for enhancing NPC radiotherapy.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582

ARTICLE IN PRESS J. Lu et al. / Cancer Letters ■■ (2016) ■■–■■

10

583 584 585 586 587 588

Acknowledgments This work was supported by the National High Technology Research and Development Program of China (2009AA02Z403, 2012AA02A501) and the National Key Basic Research Program of China (2011CB504300, 2011CB504305).

589 590 591 592

Conflict of interest None.

593 594 595 596 597

Appendix: Supplementary material Supplementary data to this article can be found online at doi:10.1016/j.canlet.2016.05.032.

598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660

References [1] D. Holmes, The cancer-virus cures, Nat. Med. 20 (2014) 571–574. [2] S.H. Hutajulu, J. Kurnianda, I.B. Tan, J.M. Middeldorp, Therapeutic implications of Epstein-Barr virus infection for the treatment of nasopharyngeal carcinoma, Ther. Clin. Risk Manag. 10 (2014) 721–736. [3] M.P. Thompson, R. Kurzrock, Epstein-Barr virus and cancer, Clin. Cancer Res. 10 (2004) 803–821. [4] T. Yoshizaki, S. Kondo, N. Wakisaka, S. Murono, K. Endo, H. Sugimoto, et al., Pathogenic role of Epstein-Barr virus latent membrane protein-1 in the development of nasopharyngeal carcinoma, Cancer Lett. 337 (2013) 1–7. [5] H.Y. Wang, B.Y. Sun, Z.H. Zhu, E.T. Chang, K.F. To, J.S. Hwang, et al., Eight-signature classifier for prediction of nasopharyngeal [corrected] carcinoma survival, J. Clin. Oncol. 29 (2011) 4516–4525. [6] C.W. Dawson, R.J. Port, L.S. Young, The role of the EBV-encoded latent membrane proteins LMP1 and LMP2 in the pathogenesis of nasopharyngeal carcinoma (NPC), Semin. Cancer Biol. 22 (2012) 144–153. [7] Y. Cao, L. Yang, W. Jiang, X. Wang, W. Liao, G. Tan, et al., Therapeutic evaluation of Epstein-Barr virus-encoded latent membrane protein-1 targeted DNAzyme for treating of nasopharyngeal carcinomas, Mol. Ther. 22 (2014) 371– 377. [8] J. Bartek, J. Bartkova, J. Lukas, DNA damage signalling guards against activated oncogenes and tumour progression, Oncogene 26 (2007) 7773–7779. [9] J. Bartkova, Z. Horejsi, K. Koed, A. Kramer, F. Tort, K. Zieger, et al., DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis, Nature 434 (2005) 864–870. [10] A. Ciccia, S.J. Elledge, The DNA damage response: making it safe to play with knives, Mol. Cell 40 (2010) 179–204. [11] S.P. Jackson, J. Bartek, The DNA-damage response in human biology and disease, Nature 461 (2009) 1071–1078. [12] M.S. Chaurushiya, M.D. Weitzman, Viral manipulation of DNA repair and cell cycle checkpoints, DNA Repair (Amst) 8 (2009) 1166–1176. [13] P.A. Nikitin, M.A. Luftig, At a crossroads: human DNA tumor viruses and the host DNA damage response, Future Virol. 6 (2011) 813–830. [14] M.D. Weitzman, J.B. Weitzman, What’s the damage? The impact of pathogens on pathways that maintain host genome integrity, Cell Host Microbe 15 (2014) 283–294. [15] B. Gruhne, R. Sompallae, M.G. Masucci, Three Epstein-Barr virus latency proteins independently promote genomic instability by inducing DNA damage, inhibiting DNA repair and inactivating cell cycle checkpoints, Oncogene 28 (2009) 3997–4008. [16] K. Sakata, M. Someya, Y. Matsumoto, M. Hareyama, Ability to repair DNA double-strand breaks related to cancer susceptibility and radiosensitivity, Radiat. Med. 25 (2007) 433–438. [17] L. Lundholm, P. Haag, D. Zong, T. Juntti, B. Mork, R. Lewensohn, et al., Resistance to DNA-damaging treatment in non-small cell lung cancer tumor-initiating cells involves reduced DNA-PK/ATM activation and diminished cell cycle arrest, Cell Death Dis. 4 (2013) e478. [18] T. Noguchi, T. Shibata, S. Fumoto, Y. Uchida, W. Mueller, S. Takeno, DNA-PKcs expression in esophageal cancer as a predictor for chemoradiation therapeutic sensitivity, Ann. Surg. Oncol. 9 (2002) 1017–1022. [19] B. Rigas, S. Borgo, A. Elhosseiny, V. Balatsos, Z. Manika, H. Shinya, et al., Decreased expression of DNA-dependent protein kinase, a DNA repair protein, during human colon carcinogenesis, Cancer Res. 61 (2001) 8381–8384. [20] M. Someya, K. Sakata, Y. Matsumoto, H. Yamamoto, M. Monobe, H. Ikeda, et al., The association of DNA-dependent protein kinase activity with chromosomal instability and risk of cancer, Carcinogenesis 27 (2006) 117–122. [21] A.J. Davis, B.P. Chen, D.J. Chen, DNA-PK: a dynamic enzyme in a versatile DSB repair pathway, DNA Repair (Amst) 17 (2014) 21–29. [22] N. Jette, S.P. Lees-Miller, The DNA-dependent protein kinase: a multifunctional protein kinase with roles in DNA double strand break repair and mitosis, Prog. Biophys. Mol. Biol. 117 (2015) 194–205.

[23] J.F. Goodwin, K.E. Knudsen, Beyond DNA repair: DNA-PK function in cancer, Cancer Discov. 4 (2014) 1126–1139. [24] X. Kong, Y. Shen, N. Jiang, X. Fei, J. Mi, Emerging roles of DNA-PK besides DNA repair, Cell. Signal. 23 (2011) 1273–1280. [25] P.N. Amatya, H.B. Kim, S.J. Park, C.K. Youn, J.W. Hyun, I.Y. Chang, et al., A role of DNA-dependent protein kinase for the activation of AMP-activated protein kinase in response to glucose deprivation, Biochim. Biophys. Acta 1823 (2012) 2099–2108. [26] R.H. Wong, I. Chang, C.S. Hudak, S. Hyun, H.Y. Kwan, H.S. Sul, A role of DNA-PK for the metabolic gene regulation in response to insulin, Cell 136 (2009) 1056–1072. [27] R.H. Wong, H.S. Sul, DNA-PK: relaying the insulin signal to USF in lipogenesis, Cell Cycle 8 (2009) 1977–1978. [28] B. Faubert, G. Boily, S. Izreig, T. Griss, B. Samborska, Z. Dong, et al., AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo, Cell Metab. 17 (2013) 113–124. [29] D.G. Hardie, D.R. Alessi, LKB1 and AMPK and the cancer-metabolism link – ten years after, BMC Biol. 11 (2013) 36. [30] S. Krishan, D.R. Richardson, S. Sahni, Gene of the month. AMP kinase (PRKAA1), J. Clin. Pathol. 67 (2014) 758–763. [31] S. Krishan, D.R. Richardson, S. Sahni, Adenosine monophosphate-activated kinase and its key role in catabolism: structure, regulation, biological activity, and pharmacological activation, Mol. Pharmacol. 87 (2015) 363–377. [32] T. Sanli, A. Rashid, C. Liu, S. Harding, R.G. Bristow, J.C. Cutz, et al., Ionizing radiation activates AMP-activated kinase (AMPK): a target for radiosensitization of human cancer cells, Int. J. Radiat. Oncol. Biol. Phys. 78 (2010) 221–229. [33] T. Sanli, G.R. Steinberg, G. Singh, T. Tsakiridis, AMP-activated protein kinase (AMPK) beyond metabolism: a novel genomic stress sensor participating in the DNA damage response pathway, Cancer Biol. Ther. 15 (2014) 156–169. [34] V.E. Zannella, D. Cojocari, S. Hilgendorf, R.N. Vellanki, S. Chung, B.G. Wouters, et al., AMPK regulates metabolism and survival in response to ionizing radiation, Radiother. Oncol. 99 (2011) 293–299. [35] E. Lengyel, L.M. Litchfield, A.K. Mitra, K.M. Nieman, A. Mukherjee, Y. Zhang, et al., Metformin inhibits ovarian cancer growth and increases sensitivity to paclitaxel in mouse models, Am. J. Obstet. Gynecol. 212 (2015) 479.e1–479.e10. [36] D. Firsanov, A. Vasilishina, A. Kropotov, V. Mikhailov, Dynamics of gammaH2AX formation and elimination in mammalian cells after X-irradiation, Biochimie 94 (2012) 2416–2422. [37] G. Khan, M.J. Hashim, Global burden of deaths from Epstein-Barr virus attributable malignancies 1990–2010, Infect. Agent. Cancer 9 (2014) 38. [38] J.K. Oh, E. Weiderpass, Infection and cancer: global distribution and burden of diseases, Ann. Glob. Health 80 (2014) 384–392. [39] Y.R. Chen, M.T. Liu, Y.T. Chang, C.C. Wu, C.Y. Hu, J.Y. Chen, Epstein-Barr virus latent membrane protein 1 represses DNA repair through the PI3K/Akt/FOXO3a pathway in human epithelial cells, J. Virol. 82 (2008) 8124–8137. [40] M.T. Liu, Y.T. Chang, S.C. Chen, Y.C. Chuang, Y.R. Chen, C.S. Lin, et al., Epstein-Barr virus latent membrane protein 1 represses p53-mediated DNA repair and transcriptional activity, Oncogene 24 (2005) 2635–2646. [41] L. Xiao, Z.Y. Hu, X. Dong, Z. Tan, W. Li, M. Tang, et al., Targeting Epstein-Barr virus oncoprotein LMP1-mediated glycolysis sensitizes nasopharyngeal carcinoma to radiation therapy, Oncogene 33 (2014) 4568–4578. [42] L. Yang, L. Liu, Z. Xu, W. Liao, D. Feng, X. Dong, et al., EBV-LMP1 targeted DNAzyme enhances radiosensitivity by inhibiting tumor angiogenesis via the JNKs/HIF-1 pathway in nasopharyngeal carcinoma, Oncotarget 6 (2015) 5804–5817. [43] L. Yang, Z. Xu, L. Liu, X. Luo, J. Lu, L. Sun, et al., Targeting EBV-LMP1 DNAzyme enhances radiosensitivity of nasopharyngeal carcinoma cells by inhibiting telomerase activity, Cancer Biol. Ther. 15 (2014) 61–68. [44] C.F. Yang, L.X. Peng, T.J. Huang, G.D. Yang, Q.Q. Chu, Y.Y. Liang, et al., Cancer stem-like cell characteristics induced by EB virus-encoded LMP1 contribute to radioresistance in nasopharyngeal carcinoma by suppressing the p53-mediated apoptosis pathway, Cancer Lett. 344 (2014) 260–271. [45] K. Goetze, S.S. Meyer, A. Yaromina, D. Zips, M. Baumann, W. Mueller-Klieser, Glycolysis-related gene induction and ATP reduction during fractionated irradiation. Markers for radiation responsiveness of human tumor xenografts, Strahlenther. Onkol. 189 (2013) 782–788. [46] V. Quennet, A. Yaromina, D. Zips, A. Rosner, S. Walenta, M. Baumann, et al., Tumor lactate content predicts for response to fractionated irradiation of human squamous cell carcinomas in nude mice, Radiother. Oncol. 81 (2006) 130–135. [47] U.G. Sattler, S.S. Meyer, V. Quennet, C. Hoerner, H. Knoerzer, C. Fabian, et al., Glycolytic metabolism and tumour response to fractionated irradiation, Radiother. Oncol. 94 (2010) 102–109. [48] A.K. Lo, K.W. Lo, C.W. Ko, L.S. Young, C.W. Dawson, Inhibition of the LKB1-AMPK pathway by the Epstein-Barr virus-encoded LMP1 promotes proliferation and transformation of human nasopharyngeal epithelial cells, J. Pathol. 230 (2013) 336–346. [49] S.S. Yan, L. Liu, Z.G. Liu, M.S. Zeng, L.B. Song, Y.F. Xia, [Expression and clinical significance of DNA-PKcs in nasopharyngeal carcinoma], Ai Zheng 27 (2008) 979–983. [50] J. Yang, X. Xu, Y. Hao, J. Chen, H. Lu, J. Qin, et al., Expression of DNA-PKcs and BRCA1 as prognostic indicators in nasopharyngeal carcinoma following intensity-modulated radiation therapy, Oncol. Lett. 5 (2013) 1199–1204. [51] S.W. Lee, K.J. Cho, J.H. Park, S.Y. Kim, S.Y. Nam, B.J. Lee, et al., Expressions of Ku70 and DNA-PKcs as prognostic indicators of local control in nasopharyngeal carcinoma, Int. J. Radiat. Oncol. Biol. Phys. 62 (2005) 1451–1457.

Please cite this article in press as: Jingchen Lu, et al., EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma, Cancer Letters (2016), doi: 10.1016/j.canlet.2016.05.032

661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745