1 The anti-tumor efficiency of pterostilbene is promoted with a combined 2 treatment of Fas signaling or autophagy inhibitors in triple negative breast 3 cancer cells 4 Wei-Chih Chen1, Kuei-Yang Hsu2, Chao-Ming Hung3, Ying-Chao Lin4, Ning-Sun 5 Yang5, Chi-Tang Ho6, Sheng-Chu Kuo7, and Tzong-Der Way2,8,9* 6 1 7 8 China Medical University, Taichung, Taiwan 2 9 10 Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan 3 11 12 The Ph.D. Program for Cancer Biology and Drug Discovery, College of Medicine, Department of General Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan 4 13 Division of Neurosurgery, Buddhist Tzu Chi General Hospital, Taichung Branch, Taiwan 14 5 Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan 15 6 Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA 16 7 Graduate Institute of Pharmaceutical Chemistry, College of Pharmacy, China 17 Medical University, Taichung, Taiwan 18 8 19 University, Taichung, Taiwan 20 21 22 9 Institute of Biochemistry, College of Life Science, National Chung Hsing Department of Health and Nutrition Biotechnology, College of Health Science, Asia University, Taichung, Taiwan *Correspondence author: 1 23 Tzong-Der Way, Ph.D. 24 Department of Biological Science and Technology, College of Life Sciences, China 25 Medical University, Taichung, Taiwan 26 No.91 Hsueh-Shih Road, Taichung, Taiwan 40402 27 Tel: +886-4-2205-3366 ext: 2509 28 Fax: +886-4-2203-1075 29 E-mail: tdway@mail.cmu.edu.tw 30 31 32 33 34 35 36 37 2 38 Abstract 39 High expression of vimentin (canonical mesenchymal marker) links with poor 40 prognosis in triple negative breast cancer (TNBC), implying that vimentin may be a 41 potential biomarker in the application of TNBC therapy. Pterostilbene (PTE) shows 42 anti-invasion 43 epithelial-mesenchymal transition (EMT) in TNBC. Here, we showed that PTE 44 decreased vimentin level, but the effect was transient. PTE stimulated Fas signaling 45 which drove EMT by ERK1/2 and GSK3β/β-catenin pathways, supporting Fas 46 signaling induction involved in EMT regulation. PTE also triggered autophagy in 47 TNBC. The treatment of TNBC with 3-MA (autophagy inhibitor) not only sustained 48 PTE-inhibited EMT but also significantly promoted anti-proliferation, supporting that 49 the autophagy played a cyto-protective role and was associated with EMT. Taken 50 together, these data showed that Fas signaling and autophagy accelerated the 51 aggressiveness of TNBC. Inhibition of autophagy or Fas signaling may provide novel 52 targets for TNBC therapy. activity, hence, we investigated whether PTE inhibited 53 54 Keywords: Triple-negative breast cancer; Pterostilbene; Vimentin; Fas signaling; 55 Autophagy 56 3 57 1 Introduction 58 Triple negative breast cancer (TNBC) is approximately 15-20% of breast cancers 59 which lacks estrogen receptor (ER), progesterone receptor (PR), and human epidermal 60 growth factor receptor 2 (HER2). Due to high propensity for metastasis and poor 61 prognosis, TNBC is considered the most aggressive of all breast cancers. 1-3 Current 62 treatment of patients with TNBC limited to chemotherapy because of the absence of 63 specific molecular targets.4 However, TNBC standard therapy has been challenging 64 due to the data on TNBC treatment is insufficient. Hence, there is important to 65 establish new approaches to improve the poor prognosis of TNBC therapy. 66 Epithelial-mesenchymal transition (EMT), so called the loss of epithelial 67 phenotype and the acquisition of a mesenchymal characteristic, is associated with 68 tumor invasion and metastasis.5-7 Fas signaling, transforming growth factor (TGF-β), 69 hepatocyte growth factor (HGF), epidermal growth factor (EGF), and fibroblast 70 growth factor (FGF) are demonstrated to induce EMT.8-10 A recent study showed that 71 high expression of vimentin (canonical mesenchymal marker) linked with a high 72 histological grade and poor prognosis in TNBC. Vimentin may be a potential 73 biomarker in the application of TNBC prognosis.11,12 74 Fas (APO-1/CD95) belongs to the death receptor (DR) superfamily and is well 75 known as an apoptosis-inducing receptor. Once Fas-ligand (FasL) binding with Fas 4 76 receptor, Fas activates caspase-dependent apoptosis by recruiting the Fas-associated 77 death domain (FADD), caspase 8 and caspase 10. In addition, Fas induces 78 non-apoptotic events in various cell types, such as the proliferation of human T 79 lymphocytes and human fibroblasts, the regulation of proinflammatory cytokines and 80 chemokines, tumor growth, and motility.13-15 81 Autophagy has been recognized to provide cells in adapting to the surroundings 82 via recycling cellular constituents in various physiopathology conditions, including 83 nutrient and growth factor deprivation, stress, microbial infection and diseases.16 84 Using pharmacological or genetic approaches to inhibit autophagy would enhance the 85 shift of cancer cells in apoptosis.17-19 It means that autophagy promoted both cell 86 survival and cell death. Autophagy has also been indicated to mediate tumor 87 progression including the aggressive characteristics of cancer cells, invasion and 88 metastasis. 89 Pterostilbene (trans-3,5-dimethoxy-4 ʹ -hydroxystilbene, PTE) is a natural 90 dimethylated analog of resveratrol from blueberries. The pharmacological properties 91 of PTE are similar to resveratrol such as anti-cancer, anti-inflammation, anti-oxidant, 92 anti-proliferative and analgesic properties in several cancer cells including breast, 93 melanoma, colon, liver, gastric and bladder.20-22 Compared to resveratrol, 94 pterostilbene has higher oral bioavailability (80% versus 20%), higher potential for 5 95 cellular uptake, longer half-life (105 minutes versus 14 minutes).23 In addition, dietary 96 administration of high doses of PTE (up to 3 g/Kg bw/day for 28 days) to mice has 97 been shown to be nontoxic.24, 98 HepG2 cells.26 However, the effects of PTE on TNBC and the underlying mechanisms 99 of EMT remain unclear. In the present study we investigated whether PTE could 100 inhibit EMT in TNBC and the possible regulatory mechanisms. Interestingly, our 101 results demonstrated that EMT inhibition by PTE is associated with decreased in 102 vimentin protein level, but the effect is transient. The novelty of this study is that Fas 103 signaling and autophagy contributed to EMT promotion in TNBC. Hence, a combined 104 treatment of PTE with inhibitors of Fas signaling or autophagy may be a beneficial 105 therapy for TNBC patients. 25 PTE reduced tumor invasion and metastasis of 106 107 2 Materials and methods 108 2.1 Cell culture 109 The human breast cancer cell lines used in this study were MDA-MB-231 and 110 BT-549. MDA-MB-231 and BT-549 cells were mesenchymal phenotype with high 111 vimentin expression but undetectable E-cadherin expression. BT-549 cells were 112 grown in RPMI 1640 (Invitrogen Corporation, Carlsbad, CA, USA); MDA-MB-231 113 cells were grown in DMEM/F12 (Invitrogen Corporation, Carlsbad, CA, USA). 6 114 Medium was supplemented with 10% fetal bovine serum (FBS), 2 mM L-glutamine, 115 100 U penicillin and 100 g streptomycin (Invitrogen Corporation, Carlsbad, CA, 116 USA). All cell lines were grown in a humidified incubator at 37 °C under 5% CO2 in 117 air. 118 2.2 Reagents and Antibodies 119 PTE was provided by Chi-Tang Ho (Department of Food Science, Rutgers 120 University). The purity of PTE is 96%. PTE was resuspended in DMSO. 121 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl 122 3-methyladenine [3-MA (autophagy inhibitor)] and primary antibody -actin were 123 purchased from Sigma Chemical Co. (St. Louis, Mo, USA). PD98059 [an 124 extracellular signal-regulated kinase1/2 (ERK1/2) inhibitor] was purchased from 125 Calbiochem (EMD Chemical, San Diego, CA, USA). NOK-1 (FasL inhibitor) and 126 primary antibody Fas were purchased from Santa Cruz Biotechnology (Santa Cruz, 127 CA, USA). All inhibitors were added into the culture medium 2 h before treatment. 128 Primary antibodies E-cadherin, Zeb1, FasL, Twist, MMP2, MMP9, phospho-GSK3β 129 (S2448), β-catenin, Beclin-1, LC3-Ⅰ/Ⅱ and -actin were purchased from Cell 130 Signaling Technology (Beverly, MA, USA). Primary vimentin was purchased from 131 Abcam Inc. (Cambridge, MA, USA). Secondary antibodies, HRP-conjugated Goat tetrazolium 7 bromide (MTT), 132 anti-Mouse IgG and Goat anti-Rabbit IgG, were obtained from Millipore (Billerica, 133 MA, USA). 134 2.3 Cell viability assay 135 MDA-MB-231 and BT-549 cells were seeded in a 24-well plate (2104 cells/well) 136 overnight, and then were treated with indicated times of PTE with or without 137 NOK-1or NOK-1 only. Cell viability was examined by the MTT assay. Briefly, 80 L 138 MTT solution (2 mg/mL) was added to each well to make a final volume of 500 L 139 and incubated for 1.5 h at 37 °C. The supernatant was aspirated, and the 140 MTT-formazan crystals formed by metabolically viable cells were dissolved in 500 141 L of DMSO. Finally, the absorbance at O.D. 570 nm was detected by enzyme-linked 142 immunosorbent assay (ELISA) reader. 143 2.4 Morphology observation 144 MDA-MB-231 and BT-549 cells (2104) were seeded in each well of a 24-well plate 145 and incubated in a 37°C incubator with 5% CO2 overnight. MDA-MB-231 and 146 BT-549 cells were treated with indicated times of PTE with or without NOK-1or 147 NOK-1 only and then cells incubated at 37°C for 48 h. Representative photographs 148 were taken at 200x magnification using a Nikon TE2000-U inverted microscope. 149 2.5 Quantitative real-time PCR 150 Total RNA was isolated from cultured cells using the TRIzol reagent (Life 8 151 Technologies, Inc., Grand Island, NY, USA) according to the manufacturer’s 152 instructions. Real-time quantitative PCR was performed using the Real-time PCR 153 system 7900 (Applied Biosystems). The primer sequences for E-cadherin, Vimentin 154 and glyceraldehyde-6-phosphoate dehydrogenase (GAPDH; used as internal control) 155 are as follows: GAPDH (5’-AAGGTCGGAGTCAACGGATTTG-3’, 5’-CCATGGGT 156 GGAATCATATTGGAA-3’), E-cadherin (5’-GACCGGTGCAATCTTCAAA-3’, 5′-T 157 TGACGCCGAGAGCTACAC-3’), Vimentin (5’-GACAATGCGTCTCTGGCACGT 158 CTT-3’, 5’-TCCTCCGCCTCCTGCAGGTTCTT-3’). The thermal cycle condition 159 included maintaining the reactions at 50℃for 2min and at 95℃ for 10 min, and then 160 alternating for 40 cycles between 95℃ for 15 s and 60℃ for 1 min. The relative 161 gene expression for each sample was determined using the formula 2 ( 162 (GAPDH)- Ct (target)) 163 levels. 164 2.6 Western blot analysis 165 Cells were seeded onto a 100-mm culture dishes (1106/dish) containing 10% FBS. 166 Cells were than treated with various agents as indicated in the figure captions. After 167 treatment, the total proteins were extracted by adding 50 L of gold lysis buffer (50 168 mM Tris–HCl, pH 7.4; 1 mM NaF; 150 mM NaCl; 1 mM EGTA; 1 mM 169 phenylmethylsulfonyl floride; 1% NP-40; and 10 mg/ml leupeptin) to the cell pellets. Ct ) = 2(Ct , which relected the target gene expression normalized to GAPDH 9 170 Lysate proteins were determined by the Lowry protein assay (Bio-Rad Laboratories). 171 The samples (50 g of proteins) of total cell lysates were resolved by sodium dodecyl 172 sulfate-polyacrylamide gel electrophoresis and (SDS-PAGE), transferred to 173 nitrocellulose membranes. Membranes were blocked with 5% BSA (Sigma, St. Louis, 174 MO, USA) for 1 h at room temperature, and probed with primary antibody for 1.5 h at 175 room temperature or overnight at 4 °C followed by HRP-conjugated appropriated 176 secondary antibodies. 177 2.7 Statistical analysis 178 One-way analysis of variance (ANOVA) was used for the comparison of more than 179 two mean values. Results represent at least two to three independent experiments. 180 Results with a P value less than 0.05 were considered statistically significant. *, p 181 0.05. 182 183 3 Results 184 3.1 Proliferation-inhibitory effect of PTE on TNBC cells 185 To evaluate the cell proliferation of PTE against mesenchymal phenotype TNBC cells, 186 MDA-MB-231 and BT-549 cells were treated with various concentrations for 48 h 187 and determined using the MTT assay. PTE exhibited the cell growth inhibition in a 188 dose-dependent manner (Figure 1B). This data showed that PTE decreased cell 10 189 proliferation in TNBC cells. 190 3.2 PTE transiently regulated EMT in TNBC cells 191 Previously, PTE has been reported to exert an inhibition of tumor invasion,26 192 suggesting that PTE may inhibit EMT in TNBC cells. To test this hypothesis, we 193 examined PTE effects on morphological changes in MDA-MB-231 and BT-549 cells 194 and representative photographs were taken at 200x magnification using a Nikon 195 TE2000-U inverted microscope. MDA-MB-231 and BT-549 cells exposed to PTE for 196 6 and 12 h displayed morphology changes for towards a classic ‘cobblestone’ 197 epithelial morphology. However, both cells reversed to form a more fibroblast-like 198 morphology, a critical marker of EMT (Figure 2A and 2B). MDA-MB-231 and 199 BT-549 cells are mesenchymal phenotype TNBC which lack detectable E-cadherin 200 expression.27, 28 We next examined the effect of PTE on E-cadherin and vimentin 201 protein expression. After treatment with PTE for 6 and 12 h, the E-cadherin protein 202 level was temporary upregulated, but did not detect after 24 h treatment in 203 MDA-MB-231 cells. After treatment with PTE for 6 h, E-cadherin was slight detected 204 and undetected after 12 h and 24 h treatment in BT-549 cells. Inversely, the vimentin 205 protein level was temporary downregulated, however, after 24 h the downregulation 206 of vimentin was restored (Figure 2C and 2D). We also examined the effect of PTE on 207 E-cadherin and Vimentin transcription levels. After treatment with PTE, E-cadherin 11 208 mRNA was detected in MDA-MB-231 and BT-549 cells, respectively. Vimentin 209 mRNA was down-regulated after PTE-treatment in MDA-MB-231 and BT-549 cells, 210 respectively (Figure 2E and 2F). These data showed that PTE regulated transiently 211 E-cadherin and vimentin via transcription level in TNBC cells. We next determined 212 whether PTE regulated EMT through EMT-inducing transcription factors. In 213 MDA-MB-231 cells, Zeb1 protein level was slightly downregulated and recovered 214 after 6 h treatment, however, twist protein level did not alter after PTE treatment. In 215 BT-549 cells, PTE decreased Zeb1 and twist protein levels during 6 and 12 h 216 treatment, however, after 24 h PTE treatment, the expression of Zeb1 and twist were 217 recovered. We also determined whether PTE regulated MMP2 and MMP9 expressions 218 which are related to the invasion and metastasis. The results were similar to Figure 2C 219 and 2D, the expressions of MMP2 and MMP9 were decreased in 6-12 h post-PTE 220 incubation interval but the expressions were reversed after 24 h treatment in 221 MDA-MB-231 cells (Figure 2G) and BT-549 cells (Figure 2G and 2H). These results 222 showed that the inhibition of EMT by PTE is transient in TNBC cells. 223 3.3 Fas signaling contributed to PTE transiently regulated EMT 224 We next explored the mechanism of PTE transiently regulated EMT in TNBC cells. 225 PTE induced apoptosis through Fas/FasL-mediated pathway29 which related with 226 EMT induction,8 suggesting that PTE may regulate EMT through Fas signaling in 12 227 TNBC cells. To test this hypothesis, MDA-MB-231 and BT-549 cells were incubated 228 with PTE for 6, 12, and 24 h and determined Fas protein expression by western blot 229 assay. PTE stimulated the expression of Fas and FasL and the maximum expression 230 were detected at 24 h (Figure 3A) and 12 h (Figure 3B), respectively. To further 231 explore whether Fas signaling mediated in transient regulation of EMT by PTE, we 232 used NOK-1 (FasL inhibitor) to inhibit Fas signaling in TNBC cells. To examine 233 whether NOK-1 is able to block PTE-induced morphological alternation in 234 MDA-MB-231 and BT-549 cells, representative photographs were taken. The 235 morphology of NOK-1 treatment only was triggered from ‘fibroblast-like’ to 236 ‘epithelial-like’ in MDA-Mb-231 and BT-549 cells, respectively. Addition of NOK-1 237 to PTE treatment maintained PTE-induced ‘epithelial-like’ morphology (Figure 3C 238 and 3D). It is also interesting to note that cotreatment with NOK-1 and PTE displayed 239 an irregulated appearance suggesting another cellular transformation, such as cell 240 death program, it may be undergoing. We also determined the vimentin expression. 241 After 24 h PTE treatment, the downregulation of vimentin was restored, however, 242 cotreatment with NOK-1 and PTE repressed the restored expression of vimentin in 243 MDA-MB-231 and BT-549 cells (Figure 3E and 3F). Moreover, we investigated 244 whether NOK-1 affect PTE-induced growth inhibition. Pretreated with NOK-1 for 2 h 245 in the absence or presence of PTE, NOK-1 alone had no significant effect on growth 13 246 inhibition but the cell proliferation was significantly inhibited after combined 247 treatment with NOK-1 and PTE (Figure 3G). NOK-1 enhanced PTE-induced growth 248 inhibition in both MDA-MB-231 and BT-549 cells, respectively. These results showed 249 that Fas signaling might contribute to the transient regulation of EMT by PTE and Fas 250 signaling inhibition increased the effect of PTE-induced growth inhibition. 251 3.4 Activation of ERK1/2 related with PTE transiently regulated EMT 252 ERK1/2 was activated during Fas-induced EMT in gastrointestinal cancer,8 suggesting 253 that the transient regulation of EMT by PTE might include ERK1/2 activation. To test 254 this hypothesis, MDA-MB-231 and BT-549 cells were incubated with PTE for 15, 30, 255 60 and 90 min and determined p-ERK1/2 and ERK1/2 protein expressions by western 256 blot assay. After PTE treatment, p-ERK1/2 was activated in MDA-MB-231 and 257 BT-549 cells (Figure 4A and 4B). Moreover, when both cells were pretreated with 258 PD98059 (10 M) for 2 h before PTE treatment, the cotreatment repressed the 259 restored expression of vimentin in MDA-MB-231 and BT-549 cells (Figure 4C and 260 4D). These results showed that ERK1/2 activation was acquired in PTE transiently 261 regulated EMT in TNBC cells. 262 3.5 Glycogen synthase kinase-3 beta (GSK3β) and -catenin involved in PTE 263 transiently regulated EMT 264 GSK3β inhibition was phosphorylated by ERK1/2.30 ERK1/2 activation was acquired 14 265 in PTE transiently regulated EMT, suggesting that GSK3β and β-catenin might be 266 included in PTE transiently regulated EMT. To test this hypothesis, MDA-MB-231 267 and BT-549 cells were incubated with PTE for 6, 12 and 24 h and determined the 268 protein levels of p-GSK3β and β-catenin. The protein levels of p-GSK3β and 269 β-catenin were upregulation in MDA-MB-231 and BT-549 cells (Figure 5A and 5B). 270 We pretreated with NOK-1 for 2 h in the absence or presence of PTE to confirm 271 whether GSK3β inactivation was regulated by Fas signaling. The accumulation of 272 pGSK3β and β-catenin were suppressed by NOK-1 in the presence of PTE for 24 h 273 (Figure 5C and 5D). These results showed that PTE transiently regulated EMT might 274 be through the GSK3β/β-catenin pathway in TNBC cells. 275 3.6 Autophagy induction involved in PTE transiently regulated EMT 276 PTE treatment induced autophagy at an earlier stage and then apoptosis at a later stage 277 in human bladder cancer cells.31 We next determined whether PTE treatment induced 278 autophagy in TNBC cells. The autophagy markers (LC3-Ⅰ/Ⅱ) and autophagy-related 279 protein (Beclin-1) were determined by western blotting. PTE caused the conversion of 280 LC3 from LC3-Ⅰ to LC3-Ⅱ and increased Beclin-1 protein level in MDA-MB-231 281 and BT-549 cells (Figure 6A and Figure 6B). These results indicated that PTE induced 282 growth inhibition through autophagy. Previous studies suggested that autophagy 283 induced cancer cell EMT and invasion,32,33 suggesting that autophagy induction might 15 284 be involved in PTE transiently regulated EMT. MDA-MB-231 and BT-549 cells were 285 pretreated with 3-MA (autophagy inhibitor) for 2 h in the absence or presence of PTE. 286 After 24 h PTE treatment, the downregulation of vimentin was restored, however, 287 cotreatment with 3-MA and PTE repressed the restored expression of vimentin in 288 MDA-MB-231 and BT-549 cells (Figure 6C and 6D). We further examined whether 289 NOK-1 affected PTE-induced autophagy. MDA-MB-231 and BT-549 cells were 290 pretreated with NOK-1 for 2 h in the absence or presence of PTE. NOK-1 repressed 291 PTE-induced the conversion of LC3 from LC3-Ⅰ to LC3-Ⅱ(Figure 6E and 6F). 292 Moreover, we investigated the role of autophagy in PTE caused growth inhibition. 293 Pretreated with 3-MA for 2 h in the absence or presence of PTE, 3-MA alone had no 294 significant effect on growth inhibition but the cell proliferation was significantly 295 enhanced after combined treatment with 3-MA and PTE (Figure 6G). These results 296 showed that autophagy induction by PTE played a cyto-protective and an EMT 297 regulative role in TNBC cells. Additionally, NOK-1 decreased the induction of a 298 protective form of autophagy by PTE. 299 300 4 Discussion 301 PTE is considered a chemopreventive agent and exhibits antiproliferative effects on 302 several types of cancer cells through various pathways including PI3K, MAPK, 16 303 adenosine monophosphate activated protein kinase (AMPK), apoptosis, autophagy, 304 metastatic activity inhibition, ROS, and cytosolic Ca2+ overload.34,35 PTE regulated 305 NO accumulation which related to intracellular adhesion molecules expression,36 306 leading to the inhibition of the potential metastasis in mouse B16 melanoma F10 307 cells.37,38 PTE significantly inhibited MMP-9 gene expression through suppression of 308 the MAPK, PI3K/NF-κB and AP-1-signaling pathways and resulted in blocking 309 invasion and metastasis of human hepatocellular carcinoma cells.26 310 EMT was related to cancer stem cells (CSCs) which contributed to poor 311 prognosis, drug-resistance, and metastasis of cancers. CD44 and CD24 are two of 312 widely identified and isolated breast cancer stem cell markers.39 PTE effectively 313 inhibited tumor-associated macrophages (TAMs)-enriched CD44+/CD24- percentage 314 via modulating NF-B/microRNA 448 circuit.40 Our data showed that PTE transiently 315 inhibited EMT during 6 and 12 h (Figure 2). In addition to apoptosis induction, Fas 316 signaling activated multiple signaling pathways including NF-κB, MAPK and 317 PI3K/AKT to induce non-apoptotic events such as growth regulation, EMT, migration 318 and invasion in cancer cells.41-45 PTE has been demonstrated to induce apoptosis 319 through mitochondrial and Fas/FasL pathway in human gastric adenocarcinoma 320 cells.29 We question whether PTE regulated EMT by modulating Fas signaling. Our 321 results showed that PTE activated Fas/FasL pathway (Figure 3A and 3B) and induced 17 322 cell death by apoptosis and autophagy in TNBC cells (Figure 1B, Figure 6A and 6B). 323 Furthermore, preventing Fas signaling held the ability of PTE-decreased EMT (Figure 324 3E and 3F), suggesting that Fas signaling contributed to the promotion of EMT. 325 ERK1/2 inhibition and loss of Snail or twist expression suppressed Fas 326 signaling-induced EMT and motility, suggesting ERK1/2 activation, Snail and twist 327 were essential for Fas signaling-induced EMT and motility promotion.8 Similarly, our 328 data showed that ERK1/2 inhibition inhibited Fas-induced EMT during PTE treatment 329 (Figure 4). 330 The serine/threonine kinase, GSK3β is inactivated through various signal 331 mechanisms such as PI3K/AKT and ERK1/2 pathways and regulates cell cycle 332 progression, anti-apoptosis, invasion and various oncogenic transcriptional factors 333 such as vimentin, uPAR and metalloproteinases. Inactivation of GSK3β results in 334 stabilization of β-catenin which translocates from cytosol into nucleus and then binds 335 to the T cell-specific transcription factor (TCF)/ lymphoid enhancer-binding factor 336 (LEF) to alter the target genes and then to induce EMT and cancer metastasis.46-48 337 FasL treatment inactivated GSK3β which leaded to accumulated Snail and β-catenin 338 in nuclear and, thus, E-cadherin was downregulated and vimentin and MMP9 were 339 upregulated.49 Similarly, our data showed that PTE regulated EMT by Fas signaling 340 through modulating GSK3β and β-catenin (Figure 5). 18 341 Autophagy induces cyto-protective role in breast cancer cells50 and promotes invasion 342 by inducing EMT through TGF-β/Smad3 signaling modulation in HCC cells, 343 indicating that autophagy accelerates the invasion of cancer cells.33,51 Autophagy 344 induction is known to relate with T-cell-mediated immune surveillance and EMT.52 345 Our data showed that autophagy induction by PTE played a cyto-protective role 346 (Figure 6G) and autophagy inhibition by 3-MA maintained PTE-inhibited EMT 347 (Figure 6C and 6D), suggesting the regulation of EMT by PTE related with autophagy. 348 Similarly, NOK-1 increased the effect of PTE-inhibited anti-proliferation may relate 349 with protective autophagy inhibition (Figure 3G). It may suggest that Fas signaling 350 inhibition increased PTE-triggered growth inhibition via switching autophagy to 351 apoptosis. Our results suggest that PTE-induced a protective form of autophagy 352 against cell death and the inhibition of autophagy could enhance PTE-caused cell 353 death through Fas signaling inhibition. 354 In summary, we demonstrated that PTE transiently inhibited EMT due to Fas 355 signaling stimulation and autophagy induction in TNBC cells. The broader 356 implication of this study supports that Fas signaling and autophagy enhance 357 aggressiveness in TNBC and could lead to the development of target therapy. Further 358 studies will focus on whether EMT regulation by PTE was similar to other cancer cell 359 types. 19 360 361 Abbreviations 362 TNBC, triple negative breast cancer; EMT, epithelial-mesenchymal transition; PTE, 363 pterostilbene; FBS, fetal bovine serum; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5- 364 diphenyl tetrazolium bromide; SDS-PAGE, sodium dodecyl sulfate- polyacrylamide; 365 ECL; enhanced chemiluminescence; ER, estrogen receptor; HER2, epidermal growth 366 factor receptor-2; PR, progesterone receptor; 3-MA, 3-Methyladenine; DMSO, 367 dimethyl sulfoxide; DMEM/F12, Dulbecco’s Modified Eagle’s Medium/Nutrient 368 Mixture F12; RPMI 1640, Roswell Park Memorial Institute (RPMI) 1640. 369 370 371 Acknowledgement 372 This study is supported in part by Taiwan Ministry of Health and Welfare Clinical 373 Trial and Research Center of Excellence (DOH102-TD-B-111-004) 374 375 376 377 378 20 379 References 380 1 W. D. Foulkes, I. E. Smith and J. S. Reis-Filho, N. Engl. J. Med. 2010, 363, 1938– 381 1948. 382 2 L. C. Liua, C. H. Sua, H. C. Wang, W. S. Chang, C. W. Tsai, M. C. Ma, C. H. Tsai, F. 383 J. Tsai and D. T. Baua. BioMedicine, 2014, 4, 16-19 384 3 M. C. Yin. BioMedicine, 2012, 3, 83 385 4 S. T. Lee, M. Feng, Y. Wei, Z. Li, Y. Qiao, P. Guan, X. Jiang, C. H. Wong, K. Huynh, 386 J. Wang, J. Li, K. Murthy Karuturi, E. Y. Tan, D. S. Hoon, Y. Kang and Q. Yu, PNAS. 387 2013, 110, 11121–11126. 388 5 R. Derynck and R. J. Akhurst, Nat. Cell Biol. 2007, 9, 1000–1004. 389 6 V. Byles, L. Zhu, L. K. Chemilewski, J. Wang, D. V. Faller and Y. Dai, Oncogene. 390 2012, 43, 4619–4629. 391 392 7 S. C. Hsu, J. H. Lin, S. W. Weng, F. S. Chueh, C. C. Yu, K. W. Lu, W. Gibson Wood and J. G. Chung. BioMedicine, 2013, 3, 120-129 393 8 H. X. Zheng, Y. D. Cai, Y. D. Wang, X. B. Cui, T. T. Xie, W. Li, W. J. Peng, L. 394 Zhang, Z. Q. Wang, J. Wang and B. Jiang. Oncogene. 2012, 32, 1183–1192. 395 9 J. M. Lee, S. Dedhar, R. Kalluri and W. W. Thompson. J. Cell. Biol. 2006, 172, 396 973–981. 397 10 J. Zavadil and E. P. Bottinger, Oncogene. 2005, 24, 5764–5774. 398 11 A. Staeli and S. Li, Cell Mol. Life Sce. 2011, 68, 3033–3046. 21 399 12 N. Yamashita, E. Tokunaga, H. Kitao, Y. Hisamatsu, K. Taketani, S. Akiyoshi, S. 400 Okada, S. Aishima, M. Morita and Y. Maehara, J. Cancer Res. Clin. Oncol. 2013, 401 139, 739–746. 402 403 404 405 406 407 13 H. Zheng, W. Li, Y. Wang, T. Xie, Y. Cai, Z. Wang and B. Jiang, Carcinogenesis, 2013, 35, 173–183. 14 O. Barca, M. Seoane, R. M. Senaris and V. M. Arce, Cell. Physiol. Biochem. 2013, 32, 111–120. 15 F. Liu, K. Bardhan, D. Yang, M. Thangaraju, V. Ganapathy, J. L. Waller, G. B. Liles, J. R. Lee and K. Liu, J. Boil. Chem. 2012, 30, 25530–25540. 408 16 S. K. Bhutia, R. Dash, S. K. Das, B. Azab, Z. Z. Su, S. G. Lee, S. Grant, A. 409 Yacoub, P. Dent, D. T. Curiel, D. Sarkar and P. B. Fisher. Cancer Res. 2010, 70, 410 3667–3676. 411 412 413 414 17 J. S. Carew, S. T. Nawrocki, C. N. Kahue, H. Zhang, C. Yang, L. Chung, J. A. Houghton, P. Huang, F. J. Giles and J. L. Cleveland, Blood. 2007, 110, 313–322. 18 M. J. Abedin, D. Wang, M. A. McDonnell, U. Lehmann and A. Kelekar, Cell Death Differ. 2007, 14, 500–510. 415 19 Q. Tang, G. Li, X. Wei, J. Zhang and J. F. Chiu, Cancer Lett. 2013, 336, 325–337. 416 20 D. McCormack and D. McFadden, J. Surg. Res. 2012, 173, e53–e61. 417 21 C. Stockley, P. L. Teissedre, M. Boban, C. Di Lorenzo and P. Restani, Food Funct. 22 418 2012, 10, 995-1007 419 22 C. L. Hsu, Y. J. Lin, C. T. Ho and G. C. Yen. Food Funct. 2012, 3, 49-57. 420 23 M. J. Ruiz, M. Fernadez, Y. Pico, J. Manes, M. Asensi, C. Carda, G. Asensio and J. 421 422 423 424 425 426 427 428 429 M. Estrela. J. Agric. Food Chem. 2009, 57, 3180–3186. D. M. Rich, L. C. McEwen, K. D. Riche, J. J. Sherman, M. R. Wofford, D. Deschamp and M. Griswold. J Toxicol. 2013, Article ID 463595 24 D. Moon, D. McCormack, D. McDonald and D. McFadder, J. Surg. Res. 2012, 180, 208–215. 25 M. Cichocki, J. Paluszczak, H. Szaefer, A. Piechowiak, A. M. Rimando and W. Baser-Dubowska, Mol Nutr Food Res, 2008, 52, S62–S70. 26 M. H. Pan, Y. S. Chiou, W. J. Chen, J. M. Wang, V. Badmaev and C. T. Ho, Carcinogenesis. 2009, 30, 1234–1242. 430 27 27 W. E. Pierceall, A. S. Woodard, J. S. Morrow, D. Rimm and E. R, Fearon. 431 Oncogene, 1995, 11, 1319-1326. 432 28 V. P. Tryndyak, F. A. Beland and I. P. Pogribny. Int. J. Cancer, 2010, 126, 433 2575-2583. 434 29 M. H. Pan, Y. H. Chang, V. Badmaev, K. Nagabhushanam and C. T. Ho, J. Agric. 435 Food Chem. 2007, 55, 7777–7785. 436 30 Q. Ding, W. Xia, J. C. Liu, et al, Mol. Cell. 2005, 19, 159–70. 23 437 31 R. J. Chen, C. T. Ho and Y. J. Wang, Mol. Nutr. Food Res. 2010, 54, 1819–1832. 438 32 R. L. Macinosh, P. Timpson, J. Thorburn, K. I. Anderson, A. Thorburn and K. M. 439 Ryan, Cell Cycle. 2012, 11, 2022–2029. 440 33 J. Li, B. Yang, Q. Zhou, Y. Wu, D. Shang, Y. Guo, Z. Song, Q. Zheng and J. Xiong, 441 442 443 Carcinogenesis. 2013, 34, 1343–1351. 34 R. J. Chen, S. J. Tsai, C. T. Ho, M. H. Pan, Y. S. Ho, C. H. Wu and Y. J. Wang, J. Agric. Food Chem. 2012, 60, 11533–11541. 444 35 M. L. Tsai, C. S. Lai, Y. H. Chang, W. J. Chen, C. T. Ho and M. H. Pan. Food 445 Funct. 2012, 11, 1185-1194. 446 36 Y. S. Chiou, M. L. Tsai, K. Nagabhushanam, Y. J. Wang, C. H. Wu, C. T. Ho and M. 447 H. Pan, J. Agric. Food Chem. 2011, 59, 2725–2733. 448 37 P. Ferrer, M. Asensi, S. Priego, M. Benlloch, S. Mena, A. Ortega, E. Obrador, J. M. 449 450 451 452 453 Esteve and J. M. Estrela, J. Biol. Chem. 2007, 282, 2880–2890. 38 P. Ferrer, M. Asensi, R. Segarra, A. Ortega, M. Benlloch, E. Obrador, M. T. Varea, G. Asensio, L. Jorda and J. M. Estrela, Neoplasia. 2005, 7, 37–47. 39 P. Mallini, T. Lennard, J. Kirby and A. Meeson, Cancer Treat. Rev. 2014, 40, 341–348. 24 454 40 K. K. Mak, A. T. H. Wu, W. H. Lee, T. C. Chang, J. F. Chiou, L. S. Wang, C. H. 455 Wu, C. Y. F. Hugng, Y. S. Shieh, T. Y. Chao, C. T. Ho, G. C. Yen and C. T. Yeh, 456 Mol. Nutr. Food Res. 2013, 57, 1123–1134. 457 458 459 460 461 462 463 464 41 H. Shinohara, H. Yagita, Y. Ikawa and N. Oyaizu, Cancer Res. 2000, 15, 1766–1772. 42 C. Choi, X. Xu, J. W. Oh, S. J. Lee, G. Y. Gillespie, H. Park, H. Jo and E. N. Benveniste, Cancer Res. 2001, 61, 3084–3091. 43 B. C. Barnhart, P. Legembre, E. Pietras, C. Bubici, G. Franzoso and M. E. Peter, EMBO J. 2004, 23, 3175–3185. 44 L. Chen, S. M. Park, A. V. Tumanov, A. Hau, K. Sawada, C. Feig, J. R. Turner, X. X. Fu, I. L. Romero, E. Lengyel and M. E. Peter, Nature. 2010, 465, 492–496. 465 45 M. W. Nijkamp, F. J. Hoogwater, E. J. Steller, B. F. Westendorp, T. A. van der 466 Meulen, M. W. Leenders, I. H. Borel Rinkes and O. Kranenburg, J. Hepatol. 2010, 467 53, 1069–1077. 468 46 S. H. Kao, W. L. Wang, C. Y. Chen, Y. L. Chang, Y. Y. Wu, Y. T. Wang, S. P. 469 Wang, A. I. Nesvizhskii, Y. J. Chen, T. M. Hong and P. C. Yang, Oncogene. 2013, 470 DOI: 10.1038/onc.2013.279. 25 471 47 J. C. Lim, K. D. Kania, H. Wijesuriya, S. Chawla, J. K. Sethi, L. Pulaski, I. A. 472 Romero, P. O. Couraud, B. B. Weksler, S. B. Hladky and M. A. Barrand, 473 Neurochem. 2008, 106, 1855–1865. 474 48 D. A. Cross, D. R. Alessi and P. Cohen, et al, Nature. 1995, 378, 785–789. 475 49 H. Zheng, W. Li, Y. Wang, Z. Liu, Y. Cai, T. Xie, M. Shi, Z. Wang and B. Jiang, 476 477 478 479 480 Euro. J. Cancer. 2013, 49, 2734–2746. 50 Y. Wang, L. Ding, X. Wang, J. Zhang, W. Han, L. Feng, J. Sun, H. Jin and X. J. Wang, Ann. J. Transl. Res. 2012, 4, 44–51. 51 H. Y. Zheng, X. Y. Zhang, X. F. Wang and B. C. Sun, Cancer Biol. Med. 2012, 9, 105–110. 481 52 I. Akalay, B. Janji, M. Hasmim, M. Z. Noman, F. André, P. De Cremoux, P. 482 Bertheau, C. Badoual, P. Vielh, A. K. Larsen, M. Sabbah, T. Z. Tan, J. H. Keira, N. T. 483 Hung, J. P. Thiery, F. Mami-Chouaib and S. Chouaib, Cancer Res. 2013, 73, 484 2418–2427. 485 486 Figure legends 487 Figure 1. The anti-proliferation activities of PTE against TNBC cells. (A) 488 Structure of PTE. (B) MDA-MB-231 and BT-549 cells were treated with various 26 489 concentrations (5, 10, 20, 40 and 80 M) of PTE for 48 h. Cell proliferation was 490 assessed using the MTT assay. The percentage of cell growth inhibition was 491 calculated by the absorption of control cells as 100%. *p 0.05 compared with 492 control group. a and c, MDA-MB-231; b and d, BT-549. 493 Figure 2. PTE transiently regulated EMT in TNBC cells. MDA-MB-231 and 494 BT-549 cells were treated with 80 M PTE for 6, 12 and 24 h. (A), (B) Phase-contrast 495 images of MDA-MB-231 and BT-549 cells. The sub-confluent cultures were shown 496 the morphological differences. (C), (D) Cells were then harvested and lysed for the 497 detection of E-cadherin and vimentin and β-actin. (E), (F) Cells were then harvested 498 and RNA was isolated. Real-time PCR was performed using primers directed against 499 E-cadherin, vimentin and GAPDH. (G), (H) Cells were then harvested and lysed for 500 the detection of Zeb1, twist, MMP2, MMP9 and β-actin. Western blot data presented 501 are representative of those obtained in at least three separate experiments. The values 502 below the figures represent change in protein expression of the bands normalized to 503 β-actin. 504 Figure 3. PTE transiently regulated EMT through Fas-signaling in TNBC cells. 505 (A) MDA-MB-231 and (B) BT-549 cells were incubated with PTE (80 M) for 6, 12 506 and 24 h. Cells were then harvested and lysed for the detection of Fas and FasL and 27 507 β-actin. MDA-MB-231 and BT-549 cells were incubated PTE (80 M) for 24 h or 508 pretreated with NOK-1 (10 g/mL) before PTE treatment. Phase-contrast images of 509 (C) MDA-MB-231 and (D) BT-549 cells. The sub-confluent cultures were shown the 510 morphological differences. (E), (F) Cells were then harvested and lysed for the 511 detection of vimentin and β-actin. Western blot data presented are representative of 512 those obtained in at least three separate experiments. The values below the figures 513 represent change in protein expression of the bands normalized to β-actin. (G) Cell 514 proliferation was assessed using the MTT assay. The percentage of cell growth 515 inhibition was calculated by the absorption of control cells as 100%. *p 0.05 516 compared with control group. a, MDA-MB-231; b, BT-549. 517 518 Figure 4. Fas-mediated activation of ERK1/2 corrected with PTE transiently 519 regulated EMT. (A) MDA-MB-231 and (B) BT-549 cells were incubated with PTE 520 (80 M) for 15, 30, 60 and 90 min. Cells were then harvested and lysed for the 521 detection of ERK1/2, p-ERK1/2 and β-actin. (C) MDA-MB-231 and (D) BT-549 cells 522 were incubated with PTE (80 M) for 24 h or pretreated with PD98059 (10 M). 523 Cells were then harvested and lysed for the detection of vimentin and β-actin. Western 524 blot data presented are representative of those obtained in at least three separate 525 experiments. The values below the figures represent change in protein expression of 28 526 the bands normalized to β-actin. 527 Figure 5. Fas-induced glycogen synthase kinase-3 beta (GSK3) inhibition 528 involved in PTE transiently regulated EMT. (A) MDA-MB-231 and (B) BT-549 529 cells were incubated with PTE (80 M) for 6, 12 and 24 h. Cells were then harvested 530 and lysed for the detection of p-GSK3, -catenin and β-actin. (C) MDA-MB-231 531 and (D) BT-549 cells were incubated with PTE (80 M) for 24 h or pretreated with 532 NOK-1 (10 g/mL). Cells were then harvested and lysed for the detection of 533 p-GSK3, -catenin and β-actin. Western blot data presented are representative of 534 those obtained in at least three separate experiments. The values below the figures 535 represent change in protein expression of the bands normalized to β-actin. 536 Figure 6. PTE-induced autophagy participated in PTE transiently regulated 537 EMT. (A) MDA-MB-231 and (B) BT-549 cells were incubated with PTE (80 M) for 538 6, 12 and 24 h. Cells were then harvested and lysed for the detection of LC3Ⅰ/Ⅱ, 539 Beclin-1 and β-actin. (C) MDA-MB-231 and (D) BT-549 cells were incubated with 540 PTE (80 M) for 24 h or pretreated with 3-MA (5 mM). Cells were then harvested 541 and lysed for the detection of vimentin and β-actin. MDA-MB-231(E) and BT-549 (F) 542 cells were incubated PTE (80 M) for 24 h or pretreated with NOK-1 (10 g/mL) 543 before PTE treatment. Cells were then harvested and lysed for the detection of LC3Ⅰ 544 /Ⅱ and β-actin. Western blot data presented are representative of those obtained in at 29 545 least three separate experiments. The values below the figures represent change in 546 protein expression of the bands normalized to β-actin. (G) MDA-MB-231 and BT-549 547 cells were incubated with PTE (80 M) for 24 h or pretreated with 3-MA (5 mM). 548 Cell proliferation was assessed using the MTT assay. The percentage of cell growth 549 inhibition was calculated by the absorption of control cells as 100%. *p 0.05 550 compared with control group. a, MDA-MB-231; b, BT-549. 30 551 552 31 553 32 554 33 555 34 556 35 557 36