1 Chemical constituents of Rhododendron formosanum show pronounced growth inhibitory effect 2 on non-small cell lung carcinoma cells 3 Tzong-Der Way1,2, Shang-Jie Tsai3, Chao-Min Wang3, Chi-Tang Ho4, Chang-Hung Chou1,3,5* 4 1 5 University, Taichung 40402, Taiwan 6 2Department 7 Taichung 41354, Taiwan 8 3Research 9 4Department of Food Science, Rutgers University, New Brunswick, New Jersey, USA 10 5Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan Department of Biological Science and Technology, College of Life Sciences, China Medical of Health and Nutrition Biotechnology, College of Health Science, Asia University, Center for Biodiversity, China Medical University, Taichung 40402, Taiwan 11 12 13 14 15 16 17 18 19 20 1 21 22 *Author for correspondence: 23 Chang-Hung Chou, Ph.D 24 Academician, Academia Sinica 25 Chair Professor and Director 26 Research Center for Biodiversity and Graduate Institute of Ecology and Evolutionary Biology 27 China Medical University, Taichung, Taiwan 28 Address: Room 720, 7F, Lifu Hall, 91, Hsueh-Shih Road, Taichung, 40402, Taiwan. 29 E-mail: choumasa@mail.cmu.edu.tw 30 Tel: +886-4-2205-3366 ext. 1633 31 Fax: +886-4-2207-1500 32 33 34 ABBREVIATION 35 ACC, acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; BuOH, n-butanol; DAPI, 4’,6- 36 diamidino-2-phenylindole; DCM, dichloromethane; DR, death receptor; EA, ethyl acetate; FASN, fatty 37 acid synthase; FBS, fetal bovine serum; LC, liquid chromatography; MeOH, methanol; mTOR, 38 mammalian target of rapamycin; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; 39 NSCLC, non-small cell lung carcinoma; PI, propidium-iodide; PS, phosphatidylserine 40 2 41 ABSTRACT 42 The aim of the present study was to investigate whether Rhododendron formosanum Hemsl. 43 (Ericaceae), an endemic species in Taiwan, exhibit anti-neoplastic potential against non-small cell lung 44 carcinoma (NSCLC). We successively extracted the R. formosanum with methanol, and then separated 45 into dichloromethane (RFL-DCM), ethyl acetate (RFL-EA), n-butanol (RFL-BuOH), and water (RFL- 46 H2O) fractions. Among these extracts, RFL-EA exhibited the most effective anti-neoplastic effect. Our 47 study also demonstrated that fractions 2 and 3 from the RFL-EA extract (RFL-EA-2, RFL-EA-3) 48 possessed the strongest anti-neoplastic potential against NSCLC cells. The major phytochemical 49 constituents of RFL-EA-2 and RFL-EA-3 were ursolic acid, oleanolic acid, and betulinic acid. Our 50 studied indicated that ursolic acid demonstrated the most efficient anti-neoplastic effects on NSCLC 51 cells. Ursolic acid inhibited growth of NSCLC cells in a dose- and time-dependent manner, and 52 stimulated apoptosis. Apoptosis was substantiated by activation of caspase-3 and -9, and decrease in 53 Bcl-2 and elevation of the Bax was also observed following ursolic acid treatment. Ursolic acid 54 activated AMP-activated protein kinase (AMPK) and then inhibited the mammalian target of rapamycin 55 (mTOR) which controls protein synthesis and cell growth. Moreover, ursolic acid decreased the 56 expression and/or activity of lipogenic enzymes, such as acetyl-CoA carboxylase (ACC) and fatty acid 57 synthase (FASN) via AMPK activation. Collectively, these data provide insight into the chemical 58 constituents and anticancer activity of R. formosanum against NSCLC cells and worthy of continued 59 study. 60 61 KEYWORDS: Rhododendron formosanum; Non-small cell lung carcinoma; Ursolic acid, AMP- 62 activated protein kinase 63 64 3 65 66 INTRODUCTION 67 Lung cancer is the second frequent types of cancer, being considered one of the most common 68 causes of death by cancer worldwide.1 Despite the improvements in surgical techniques and other 69 therapies, most patients may present with advanced disease and with an estimated 5-year relative 70 survival rate of 17%.2 Non-small cell lung cancer (NSCLC) accounts for ~80% of primary lung cancer. 71 Surgery, radiation, and chemotherapy are useful treatments for patients with NSCLC. However, patients 72 considered favorable for therapeutic treatment will still keep a high rate of recurrence. Recent studies 73 showed that numerous alterations in oncogenic pathways possess a critical role in NSCLC tumorigenesis 74 and progression. Develop selective drugs to specifically target the NSCLC oncogenic pathways is very 75 critical. 76 Cell death plays an important role in the efficacy of cancer chemotherapy. The process of apoptosis, 77 a major form of cell death, is regulated by programmed cellular signaling pathways. The main 78 mechanism by which anti-cancer drugs kill cells is to induce apoptosis in cancer cells. Apoptosis is 79 associated with characteristic morphological changes including the formation of apoptotic bodies, 80 chromatin and nuclear condensation, and DNA fragmentation. The caspases, a family of cysteine 81 proteases, play a critical role during apoptosis. In the intrinsic pathway, pro-apoptotic factors are 82 released from the mitochondria, leading to caspase 9 and caspase 3 cleavages and then induce 83 apoptosis.3,4 84 The tumor suppressor LKB1 is mutated in at least 15%–30% of NSCLC and play a critical role in 85 NSCLC metastasis.5 The canonical target of LKB1 is AMP-activated protein kinase (AMPK), a crucial 86 cellular energy sensor, that is activated during metabolic stress.6 Phosphorylation of Thr172 in the T-loop 87 of AMPK catalytic α subunit by LKB1 is necessary for AMPK catalytic activity. Extensive evidences 88 have demonstrated that AMPK inhibits anabolic pathways that promote cell growth, such as synthesis of 4 89 cholesterol, fatty acid, glycogen, triglyceride, protein, and ribosomal RNA synthesis. Cancer cells 90 possess mutation or deletion of LKB1 that inactivate the AMPK pathway are highly malignant form of 91 cancer.7,8 Since AMPK activation inhibits anabolic pathways, such as cell growth and proliferation– 92 thereby antagonizing carcinogenesis. Many studies have verified the anti-cancer effects of AMPK in 93 vitro and in vivo, including breast, lung, colorectum, skin, and hematological malignancies.9 94 The mammalian target of rapamycin (mTOR) is one of the major growth regulatory pathways 95 controlled by AMPK. The mTOR pathway plays a major role in proliferation, angiogenesis, and 96 metastasis of NSCLC and other cancers. The target on mTOR signaling pathways is extensively 97 investigated for cancer chemotherapy including NSCLC.10 Moreover, AMPK controlled lipid 98 metabolism at transcriptional and post-translational levels. AMPK phosphorylates and inactivates 99 metabolic enzyme acetyl-CoA carboxylase (ACC) that involves in regulating de novo biosynthesis of 100 fatty acid and cholesterol. Phosphorylated ACC leaded to the decrease of malonyl-CoA levels, thus 101 stimulating mitochondrial carnitine palmitoyltransferase 1 (CPT1) and promoting fatty acid oxidation. 102 Moreover, AMPK inhibits the transcription factor SREBP1c, which controls the entire fatty acid 103 synthetic pathway, or by directly inhibiting the expression of fatty acid synthase (FASN).11 104 Rhododendron formosanum Hemsl. (Ericaceae), is an evergreen, broad leafed tree native to Taiwan 105 and ubiquitously distributed from 800 m to 2,000 m. The vegetation exhibits a unique pattern and forms 106 pure dominant vegetation.12,13 We studied whether R. formosanum exhibited pharmacological activities 107 for NSCLC and investigated its bioactive phytochemical constituents. Our results indicated that 108 treatment of NSCLC cells with R. formosanum had a very potent inhibitory effect on cellular growth. 109 110 111 112 5 113 114 115 116 MATERIALS AND METHODS 117 Materials. The leaves of R. formosanum were collected after flowering in April 2010 and July 118 2010 at the Dasyueshan site (24°14'6.49"N, 120°57'7.29"E at 1911 m asl.). Ursolic acid, oleanolic acid, 119 betulinic 120 diphenyltetrazolium (MTT), and propidium iodide (PI) were purchased from Sigma Chemical Co. (St. 121 Louis, MO, USA). The antibody for LKB1 was purchased from Santa Cruz Biotechnology (Santa Cruz, 122 CA, USA). Primary antibody against phospho-AMPK (Thr 172), AMPK, Bcl-2, Bax, caspase 9, caspase 123 3, FASN, phospho-ACC (Ser 79), mTOR, phospho-mTOR (Ser 2448) and phospho-p70S6k (Thr 389) 124 were purchased from Cell Signaling Technology (Beverly, MA, USA). β-actin antibody was purchased 125 from sigma Chemical Co. (St. Louis, MO, USA). HRP-conjugated Goat anti-Rabbit IgG and Goat anti- 126 Mouse IgG were obtained from Millipore (Billerica, MA, USA). Methanol (MeOH), n-hexane, 127 dichloromethane (DCM), n-butanol (BuOH), and ethyl acetate (EA) were purchased form Seedchem Co. 128 (Melbourne, Australia). Silica gel 60 was purchased from Merck KGaA (Darmstadt, Germany). acid, 4’,6-diamidino-2-phenylindole (DAPI), 3-[4,5-dimethylthylthiazol-2-yl]-2,5- 129 Plant collection, chemical extraction and isolation. The leaves of R. formosanum were air-dried 130 for chemical analysis. The air-dried and powdered leaves of R. formosanum (5.5 kg) were extracted with 131 MeOH for three days at room temperature (three times), and the combined extracts were concentrated in 132 vacuo (under 35 °C) to obtain a dark green gum (1,540 g), which was suspended in H2O and partitioned 133 sequentially with DCM, EA, and BuOH. The EA extract (3.5 g) was subjected to column 134 chromatography on silica gel using n-hexane, n-hexane-EA and EA-MeOH mixtures of increasing 135 polarity for elution to furnish 10 fractions. 6 136 The quantification of ursolic acid, oleanolic acid and betulinic acid in RFL-EA-2 and RFL- 137 EA-3 fractions. The EA fractions of R. formosanum were prepared at concentration of 1 mg/mL in 138 MeOH. The samples were passed through 0.22-µm filters (Millipore) and placed in sample vials for 139 liquid chromatography (LC) analysis. LC mass analysis for the quantification of betulinic acid, oleanolic 140 acid and ursolic acid was carried out with an Atlantis T3 RP-18 column (150 × 2.1 mm; 3 µm; Waters, 141 Milford, MA, USA). In each case the injection volume was 5 mL. The column was eluted with buffer A 142 (distilled water/acetonitrile/formic acid; 95/5/0.1, v/v/v) and buffer B (acetonitrile/formic acid; 100/0.1, 143 v/v) at a flow rate of 0.25 mL/min at 25 °C. The column was eluted initially with 100% buffer A, 144 followed by a linear increase in buffer B to 30% from 0 to 5 min, and maintained in 30% buffer B for 145 another 5 min. From 10 to 20 min, a linear increase in buffer B to 80% was carried out and the column 146 was maintained in 80% buffer B for another 10 min. The column was further eluted with a linear 147 increase in buffer B to 95% from 30 to 40 min. The column was finally equilibrated with buffer A for 10 148 min. Quantification of the triterpenoids in EA fraction was performed in the ion monitoring mode 149 selected. Both positive and negative ionization mode MS analyses were undertaken. The molecular ion 150 peaks and mass spectra recorded were compared to those of reference substances. Analysis was carried 151 out using data-dependent MS/MS scanning from m/z 100 to 1000. The temperature of the ion source 152 was maintained at 100 °C, the dry temperature was 365 °C and the desolvation gas, N2, had a flow rate 153 of 12 L/min. Product ion scans for mass were performed by low-energy collision (20 eV) using argon as 154 the collision gas. All liquid chromatography-electrospray ionization/tandem mass spectrometry (LC- 155 ESI-MS/MS) data were processed by Bruker Daltonics Data analysis software (version 4.0). All 156 chemicals were prepared at a concentration range of 0.01–1000 µg/mL. Quantification of triterpenoids 157 was performed with the selected ion monitoring (SIM) mode. The separated [M-H]– ion chromatogram 158 was selected at m/z 455 for the specific parent ion of triterpenoids. The linearity of the calibration 159 curves was demonstrated by the good determination of coefficients (r2) obtained for the regression line. 160 Good linearity was achieved over the calibration range, with all coefficients of correlation greater than 161 0.995. All samples were freshly prepared. The mean values for the regression equation were y=6E+07x 7 162 + 1E+07 (r2=0.9997) for betulinic acid, y=552577x+ 5E+07 (r2=0.9983) for oleanolic acid and 163 y=353895x + 3E+07 (r2=0.9985) for ursolic acid. The limits of quantification (LOQ) and determination 164 (LOD), defined as signal to noise ratios of 3:1 and 10:1, were in the range 0.01–0.1 mg/mL and 0.1 165 mg/mL, receptively. 166 Cell lines and cell culture. Both A549 cells (human lung adenocarcinoma cell line) and H460 cells 167 (human non-small cell lung cancer cell line) were used to study and acquired from American Type 168 Culture Collection. A549 cells were cultured in DMEM (Invitrogen Carlsbad, CA, USA) and H460 cells 169 were cultured in RPMI-1640 (Invitrogen Carlsbad, CA, USA) supplemented with 10% fetal bovine 170 serum (FBS, 10%) (Invitrogen Carlsbad, CA, USA), streptomycin (100 g/mL), and penicillin (100 171 IU/mL) (Invitrogen Carlsbad, CA, USA) in a humidified incubator at 37 oC with 5% CO2. 172 Cell viability assay. To measure the cell viability, A549 cells or H460 cells were incubated on the 173 96-well cell culture cluster (1 × 104 cells/well). MTT stock solution was prepared at con. 5 mg/mL in 174 PBS, and working solution (500 μg/mL) was diluted form stock solution. Remove the medium from 175 each well, add 100 μL MTT working solution for 1 h at 37 °C. When the crystals were formed, remove 176 the solution and add 80 μL DMSO to dissolve the crystals. Finally, use OD 570 nm to detect the 177 absorbance by the ELISA reader. MTT assay was done as described previously.14 178 Western blot analysis. Seed cells (2 × 106) onto the 10 cm cell culture dish overnight, and treat 179 with ursolic acid (30 μM) for 0, 12, 24, and 48 h. After treatment, cells were collected in 1.5 mL 180 eppendorf, and lysed in the lysis buffer (0.1% SDS, 1% NP-40, 10 g/mL leupeptin, 1% sodium 181 deoxycholate, 1 mM PMSF, 10 mM Tris-HCl pH7.5, 150 mM NaCl and 10 g/mL aprotinin). Extract 182 each protein sample and quantify by Bio-Rad protein assay kit, each group was took 50 μg total proteins 183 to run SDS-PAGE, followed transfer to PVDF membrane. Western Blot was done as described 184 previously.15 The results were analyzed and quantified by using Image J software. 8 185 Cell cycle and apoptosis analysis. A549 cells (5×105) were cultured in 6 cm cell culture dish for 186 24 h, and then treated with ursolic acid (10-30 μM) for 24 h and 48 h. After treatment, cells were 187 harvested in a 15 mL tube, washed with PBS, resuspended in PBS, and fixed in 2 mL of 70 % ethanol at 188 20 oC overnight. The cell cycle state and apoptosis was determined by using PI staining as reported 189 previously.15 190 Fluorescence microscopy. After treatment with ursolic acid (30 μM) for 48 h, A549 cells fixed by 191 70 % ethanol at -4 °C for 6 h or overnight. Cells stained with DAPI (1 μg/mL DAPI, 0.1% Triton X- 192 100) at room temperature for 15 min. The morphology of cell nuclei was observed by Nikon TE2000-U 193 fluorescence microscope at 400× magnification. Fluorescence microscopy was done as described 194 previously.16 195 RNA interference suppression of LKB1. A549 cells were transfected with LKB1 small 196 interfering RNA (siRNA) using siRNA transfection reagent (Santa Cruz Biotechnology; Santa Cruz, CA, 197 USA) and incubated for 6 h. The LKB1 siRNA was obtained from Santa Cruz Biotechnology (sc- 198 25816). 199 Statistical analysis. Data were presented as the mean ± SD of at least three independent 200 experiments. For statistical analysis, the independent Student’s t-test was used to compare the 201 continuous variables between two groups, and the chi-squared test was applied for compare the 202 dichotomous variables. Asterisk indicates that the values were significantly different from the control (*, 203 P < 0.05). 204 205 206 207 9 208 209 210 211 212 213 214 215 216 RESULTS 217 R. formosanum induced NSCLC A549 cells growth inhibition. To evaluate the bioactive 218 phytochemical constituents of R. formosanum, we extracted with methanol and separated the methanol 219 extract into dichloromethane (RFL-DCM), ethyl acetate (RFL-EA), n-butanol (RFL-BuOH), and water 220 (RFL-H2O) fractions (Figure 1A). We next examined anti-proliferative activity of R. formosanum 221 extracts on NSCLC A549 cells. Among these R. formosanum extracts, RFL-EA was the most effective 222 one in our assay (Figure 1B). Further fractionation of the RFL-EA by column chromatography was used 223 to analyze the detailed bioactive phytochemical constituents (Figure 2A). Our study demonstrated that 224 fractions 2 and 3 from the RFL-EA extract (RFL-EA-2, RFL-EA-3) possessed the strongest anti- 225 proliferative activity against A549 cells (Figure 2B). The morphology variations of R. formosanum- 226 treated A549 cells were investigated by microscopic inspection. After treatment with different 227 concentrations of RFL-EA-2 or RFL-EA-3 for 48 h, apoptotic bodies were observed in A549 cells 228 (Figure 2C). These results may provide a rationale for the potential use of R. formosanum against 229 NSCLC. 10 230 Comparative study of the phytochemical constituents of of RFL-EA-2 and RFL-EA-3. The 231 HPLC fingerprint chromatogram was established to analyze the detailed phytochemical constituents of 232 RFL-EA-2 and RFL-EA-3. Figure 3A showed the HPLC fingerprint chromatogram for the mixture of 233 ursolic acid, oleanolic acid, and betulinic acid. As shown in Figure 3B, RFL-EA-3 contains ursolic acid, 234 oleanolic acid, and betulinic acid. Interestingly, our results revealed that ursolic acid was the most 235 abundant constituents in RFL-EA-2 and RFL-EA-3 (Table 1). 236 Effect of oleanolic acid, ursolic acid, and betulinic acid on NSCLC cells proliferation. The 237 potential anti-proliferative activities of oleanolic acid, ursolic acid, and betulinic acid were evaluated 238 using MTT assay. A549 and H460 cells were treated with various concentrations of ursolic acid (Figure 239 4A), oleanolic acid (Figure 4B), and betulinic acid (Figure 4C) for 24 and 48 h. Among these 240 constituents, ursolic acid and betulinic acid exhibited the potent cytotoxic effects on NSCLC cells 241 (Figure 4). Because ursolic acid was the most abundant constituents and exhibited the potent cytotoxic 242 effects, we chose ursolic acid for the future experiments. 243 Ursolic acid induces NSCLC cells apoptosis. To test whether ursolic acid could induce apoptosis, 244 apoptosis and morphology variations were investigated by microscopic inspection. Treatment with 245 different concentrations of ursolic acid for 48 h, apoptotic bodies were observed in A549 cells (Figure 246 5A). We next elucidated whether ursolic acid induce chromatin condensation in A549 cells. After 30 247 M ursolic acid treatment, chromatin condensation was seen in A549 cells, as evidenced by staining 248 with DAPI. (Figure 5B). To further confirm ursolic acid-induced A549 cells apoptosis, flow cytometric 249 analysis was performed. After treatment with various concentrations of ursolic acid for 24 and 48 h, 250 A549 cells underwent apoptosis (Figure 5C). Apoptosis-related modulators were studied by western 251 blot analysis. Treatment with 30 μM ursolic acid increased the cleavages of Caspase-3 and Caspase-9 252 (Figure 5D). Moreover, there was a marked increase of proapoptotic protein Bax and marked decrease 253 of antiapoptotic protein Bcl-2 in ursolic acid-treated A549 cells (Figure 5E). These results indicated 254 that ursolic acid induced apoptosis in NSCLC cells. 11 255 Ursolic acid decreases protein synthesis via the up-regulation of AMPK activity in NSCLC 256 cells. Abnormalities in the AMPK function has emerged as an important pathway implicated in cancer 257 development.9 We examined whether ursolic acid was involved in the regulation of AMPK. Figure 6A 258 indicated that ursolic acid stimulated AMPK phosphorylation in a time-dependent manner. While 259 AMPK activation resulted in marked inhibition of mammalian target of rapamycin (mTOR) and 260 p70S6K in a time-dependent manner (Figure 6B). Our studies suggest that the phosphorylation of 261 AMPK is required for ursolic acid to decrease protein synthesis in NSCLC cells. 262 Ursolic acid inhibits lipogenesis through activation of AMPK in NSCLC cells. AMPK 263 negatively regulated the activities of lipogenic enzymes FASN and ACC. We next examined whether 264 reduction of FASN expression and ACC activity in ursolic acid-treated A549 cells. Our result indicated 265 that ursolic acid decreased the expression of FASN and increased ACC phosphorylation in a time- 266 dependent manner (Figure 6C). Our results suggested that ursolic acid suppressed lipogenesis through 267 modulation of AMPK activity. 268 Ursolic acid induces AMPK phosphorylation via a LKB1-dependent pathway. The canonical 269 target of LKB1 is AMPK.6 To further investigate whether ursolic acid induced AMPK phosphorylation 270 via a LKB1-dependent pathway, LKB1 siRNA was used to inhibit LKB1 expression in A549 cells. 271 Treatment with ursolic acid, an increasing level of phosphorylated AMPK was observed, however, 272 under LKB1 siRNA transfection, despite the addition of ursolic acid, AMPK phosphorylation was 273 reduced (Figure 6D). Our results showed that the activation of AMPK by ursolic acid treatment might 274 occur via a LKB1-dependent pathway. 275 276 277 278 12 279 280 281 282 283 284 285 286 287 DISCUSSION 288 The present study has clearly evaluated the bioactive constituents of R. formosanum and 289 investigated the anti-proliferative activity on NSCLC. We extracted the R. formosanum with fractions of 290 dichloromethane (RFL-DCM), ethyl acetate (RFL-EA), n-butanol (RFL-BuOH), and water (RFL-H2O). 291 RFL-EA possesses the most effective bioactivity in our cell viability assay. Moreover, our study also 292 demonstrated that fractions 2 and 3 from the RFL-EA extract (RFL-EA-2, RFL-EA-3) showed the most 293 anti-proliferative effect on NSCLC cells. In our previous study, eighteen known compounds were 294 isolated from the six fractions obtained by column chromatography from the methanolic extract of R. 295 formosanum leaves.13 In this study, the major compositions of RFL-EA-2 and RFL-EA-3 were ursolic 296 acid, oleanolic acid, and betulinic acid. Among the three constituents, ursolic acid was the most 297 abundant compound in RFL-EA-2 and RFL-EA-3. Interestingly, our results also revealed that ursolic 298 acid possessed the most efficient anti-proliferative effect on NSCLC cells. 299 Ursolic acid is a natural pentacyclic triterpene acid, which exists in elder flower, apples, lavender, 300 peppermint rosemary, basil, bilberries, cranberries, thyme, oregano, hawthorn, prunes and medicinal 13 301 herbs. Since ursolic acid is relatively non-toxic, there is a growing interest in the elucidation of the 302 pharmacological activities. Several reports described that ursolic acid has many health benefits including 303 hepatoprotective, anti-inflammatory, and anti-cancer effects.17-19 Ursolic acid possesses strong 304 anticancer activity against several cancers of prostate,20 breast,21 lung,22 pancreas,23 ovary,24 colon,25 and 305 bladder26. In the present study, we focused on the anti-neoplastic effect of ursolic acid on NSCLC cells. 306 These findings suggested that ursolic acid is a potent anti-cancer agent. 307 Induction of cell apoptosis is thought to be the principal mechanism by which anti-cancer drugs kill 308 cells. Ursolic acid has been reported to induce apoptosis through calcium-dependent and activation of 309 sphingomyelinase.27,28 To confirm whether the cytotoxic effect of ursolic acid was due to apoptosis in 310 NSCLC cells, phenotypic characteristics, cell cycle analysis, and Western blotting were used in the 311 present study. Our results indicated that ursolic acid induced cleavages of caspase-9 and caspase-3 and 312 also attenuated the expression of Bcl-2, indicating that ursolic acid induced apoptosis via caspase 313 dependent pathway. The Bcl-2 family proteins play a crucial role in the mitochondrial pathway of 314 apoptosis. The ratio between Bcl-2 and Bax proteins has been suggested as a primary event in 315 determining the sensitivity to the mitochondrial pathway of apoptosis. Bax translocation to the 316 mitochondria and insertion into the membrane and induces cytochrome c and other apoptogenic factors 317 release that subsequently activates caspase-3 leading to downstream apoptotic responses.29 From the 318 above results, we suggest that the enhancement of Bax and the attenuation of Bcl-2 a might be the 319 important molecular pathway that is involved in ursolic acid-induced apoptosis in A549 cells. 320 Recent studies have focused on the potential of targeting cellular metabolic pathways that may be 321 altered during NSCLC tumorigenesis and progression. AMPK has recently been considered as a critical 322 energy-sensing serine/threonine kinase in the regulation of cellular metabolism. The activation of 323 AMPK switches on catabolic pathways that generate ATP, while switching off ATP-consuming 324 processes. Recently, our and other groups have reported that sustained AMPK activation inhibits cancer 325 cell growth and proliferation, and induces cancer cell apoptosis.15,30 These findings are given that cancer 14 326 cell DNA replication, mitosis, cell growth, and proliferation are all ATP-consuming processes. In this 327 study, ursolic acid induced growth inhibition and apoptosis in NSCLC A549 cells, and activation of 328 AMPK may contribute to the process. Similarly, Zheng et al. suggested that activation of AMPK by 329 ursolic acid contributes to growth inhibition and apoptosis in human bladder cancer T24 cells.31 Son et 330 al. reported that ursolic acid induced apoptosis in HepG2 cells via AMPK activation and GSK3β 331 phosphorylation.32 Lee et al. found that ursolic acid potentiated cell cycle arrest and UVR-induced 332 apoptosis in skin melanoma cells via AMPK activation.33 These studies are consistent with our findings 333 here that AMPK activation by ursolic acid inducing A549 cells apoptosis. However, how AMPK 334 activation induces A549 cells apoptosis needs further study. Our study provides evidence that ursolic 335 acid may as a potential cancer chemopreventive agent for NSCLC and AMPK might be the key 336 mechanism for its action. 337 Targeting mTOR pathway may lead to the development of novel drug for cancers. Many laboratories 338 and pharmaceutical companies have focused intensely on developing approaches to block the mTOR 339 pathway. Particularly due to the fact that mTOR pathway plays such a crucial role in cancer biology. The 340 intervention of some target proteins will lower mTOR activity, for example, activation of AMPK results 341 in decreased mTOR signaling and in turn inhibition of protein synthesis and cellular growth.14,15,34 The 342 present data shows that ursolic acid inhibits protein synthesis via AMPK-mTOR pathway. 343 In conclusion, R. formosanum is an endemic species distributed widely in Taiwan, the 344 pharmacological activities of R. formosanum have not yet been fully explained. To the best of our 345 knowledge, the anti-neoplastic activity of R. formosanum has no been explored in the previous studies. 346 In this study, we have investigated the chemical constituents and anti-neoplastic activity of R. 347 formosanum against NSCLC cells. Based on these studies, it is tempting to propose that R. formosanum 348 could be developed as an anti-neoplastic agent for NSCLC. 349 15 350 351 352 353 354 355 356 357 358 359 360 361 ACKNOWLEDGMENT 362 This study was financially supported by grants from the National Science Council of Taiwan 363 (NSC101-2811-B-039-013, NSC101-2621-B-039-001, NSC102-2313-B-039-001 and NSC102-2811-B- 364 039-005) to C. H. Chou. Additionally, technical assistance with chemical data analyses from Instrument 365 Analysis Centers at the China Medical University and National Chung Hsing University is greatly 366 appreciated. 367 368 369 16 370 371 372 373 374 375 376 377 378 379 380 381 382 References 383 (1) Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics 2012. CA Cancer J. Clin. 2012, 62, 10–29. 384 (2) Askoxylakis, V.; Thieke, C.; Pleger, S. T.; Most, P.; Tanner, J.; Lindel, K.; Katus, H. A.; Debus, J.; 385 Bischof, M. Long-term survival of cancer patients compared to heart failure and stroke: a systematic 386 review. BMC Cancer 2010, 10, 105. 387 (3) Green, D. R.; Reed, J. C. Mitochondria and apoptosis. Science 1998, 281, 1309-1312. 388 (4) Riedl, S. J.; Salvesen, G. S. The apoptosome: signalling platform of cell death. Nat. Rev. Mol. Cell 389 Biol. 2007, 8, 405-413. 17 390 (5) Carretero, J.; Medina, P. P.; Pio, R.; Montuenga, L. M.; Sanchez-Cespedes, M. Novel and natural 391 knockout lung cancer cell lines for the LKB1/STK11 tumor suppressor gene. Oncogene 2004, 23, 392 4037–4040. 393 (6) Mu, J.; Brozinick, J. T. Jr.; Valladares, O.; Bucan, M.; Birnbaum, M. J. A role for AMP-activated 394 protein kinase in contractionand hypoxia-regulated glucose transport in skeletal muscle. Mol. Cell 395 2001, 7, 1085–1094. 396 397 (7) Hardie, D. G. AMP-activated protein kinase: a cellular energy sensor with a key role in metabolic disorders and in cancer. Biochem. Soc. Trans. 2011, 39, 1-13. 398 (8) Hadad, S. M.; Baker, L.; Quinlan, P. R.; Robertson, K. E.; Bray, S. E.; Thomson, G.; Kellock, D.; 399 Jordan, L. B.; Purdie, C. A.; Hardie, D. G.; Fleming, S.; Thompson, A. M. Histological evaluation of 400 AMPK signalling in primary breast cancer. BMC Cancer 2009, 9, 307. 401 402 403 404 405 406 (9) Kim, I.; He, Y. Y. Targeting the AMP-activated protein kinase for cancer prevention and therapy. Front. Oncol. 2013, 3, 175. (10) Alayev, A.; Holz, M. K. mTOR signaling for biological control and cancer. J. Cell. Physiol. 2013, 228, 1658-1664. (11) Russo, G. L.; Russo, M.; Ungaro, P. AMP-activated protein kinase: A target for old drugs against diabetes and cancer. Biochem. Pharmacol. 2013, 86, 339-350. 407 (12) Krishna, V.; Chang, C. I.; Chou, C. H. Two isomeric epoxysitosterols from Rhododendron 408 formosanum: 1H and 13C NMR chemical shift assignments. Magn. Reson. Chem. 2006, 44, 817- 409 819. 410 411 (13) Chou, S. C.; Krishna, V.; Chou, C. H. Hydrophobic metabolites from Rhododendron formosanum and their allelopathic activities. Nat. Prod. Commun. 2009, 4, 1189-1192. 18 412 (14) Shieh, J. M.; Chen, Y. C.; Lin, Y. C.; Lin, J. N.; Chen, W. C.; Chen, Y. Y.; Ho, C. T.; Way, T. D. 413 Demethoxycurcumin inhibits energy metabolic and oncogenic signaling pathways through AMPK 414 activation in triple-negative breast cancer cells. J. Agric. Food Chem. 2013, 61, 6366-6375. 415 (15) Lin, V. C.; Tsai, Y. C.; Lin, J. N.; Fan, L. L.; Pan, M. H.; Ho, C. T.; Wu, J. Y.; Way, T. D. 416 Activation of AMPK by pterostilbene suppresses lipogenesis and cell-cycle progression in p53 417 positive and negative human prostate cancer cells. J. Agric. Food Chem. 2012, 60, 6399-6407. 418 (16) Liu, L. C.; Tsao, T. C.; Hsu, S. R.; Wang, H. C.; Tsai, T. C.; Kao, J. Y.; Way, T. D. EGCG inhibits 419 transforming growth factor-β-mediated epithelial-to-mesenchymal transition via the inhibition of 420 Smad2 and Erk1/2 signaling pathways in nonsmall cell lung cancer cells. J. Agric. Food Chem. 421 2012, 60, 9863-9873. 422 423 (17) Takeoka, G.; Dao, L.; Teranishi, R.; Wong, R.; Flessa, S.; Harden, L.; Edwards, R. Identification of three triterpenoids in almond hulls. J. Agric. Food Chem. 2000, 48, 3437-3439. 424 (18) Tang, X.; Gao, J.; Chen, J.; Fang, F.; Wang, Y.; Dou, H.; Xu, Q. Qian, Z. Inhibition of ursolic acid 425 on calcium-induced mitochondrial permeability transition and release of two proapoptotic proteins. 426 Biochem. Biophys. Res. Commun. 2005, 337, 320-324. 427 (19) Andersson, D.; Liu, J. J.; Nilsson, A.; Duan, R. D. Ursolic acid inhibits proliferation and stimulates 428 apoptosis in HT29 cells following activation of alkaline sphingomyelinase. Anticancer Res. 2003, 429 23, 3317-3322. 430 (20) Kassi, E.; Papoutsi, Z.; Pratsinis, H.; Aligiannis, N.; Manoussakis, M.; Moutsatsou, P. Ursolic acid, 431 a naturally occurring triterpenoid, demonstrates anticancer activity on human prostate cancer cells. 432 J. Cancer Res. Clin. Oncol. 2007, 133, 493–500. 433 (21) Yeh, C. T. ; Wu, C. H.; Yen, G. C. Ursolic acid, a naturally occurring triterpenoid, suppresses 434 migration and invasion of human breast cancer cells by modulating c-Jun N-terminal kinase, Akt 435 and mammalian target of rapamycin signaling. Mol. Nutr. Food Res. 2010, 54, 1285–1295. 436 (22) Hsu, Y. L.; Kuo, P. L.; Lin, C. C. Proliferative inhibition, cell-cycle dysregulation, and induction of 19 437 apoptosis by ursolic acid in human non-small cell lung cancer A549 cells. Life Sci. 2004, 75, 2303– 438 2316. 439 (23) Li, J.; Liang, X.; Yang, X. Ursolic acid inhibits growth and induces apoptosis in gemcitabine- 440 resistant human pancreatic cancer via the JNK and PI3K/Akt/NF-kappaB pathways. Oncol. Rep. 441 2012, 28, 501–510. 442 (24) Song, Y. H.; Jeong, S. J.; Kwon, H. Y.; Kim, B.; Kim, S. H.; Yoo, D. Y. Ursolic acid from 443 Oldenlandia diffusa induces apoptosis via activation of caspases and phosphorylation of glycogen 444 synthase kinase 3 beta in SK-OV-3 ovarian cancer cells. Biol. Pharm. Bull. 2012, 35, 1022–1028. 445 (25) Wang, J.; Liu, L.; Qiu, H.; Zhang, X.; Guo, W.; Chen, W.; Tian, Y.; Fu, L.; Shi, D.; Cheng, J.; 446 Huang, W.; Deng, W. Ursolic acid simultaneously targets multiple signaling pathways to suppress 447 proliferation and induce apoptosis in colon cancer cells. PLoS One 2013, 8, e63872. 448 (26) Zheng, Q. Y.; Li, P. P.; Jin, F. S.; Yao, C.; Zhang, G. H.; Zang, T.; Ai, X. Ursolic acid induces ER 449 stress response to activate ASK1-JNK signaling and induce apoptosis in human bladder cancer T24 450 cells. Cell. Signal. 2013, 25, 206–213. 451 (27) Andersson, D.; Liu, J. J.; Nilsson, A.; Duan, R. D. Ursolic acid inhibits proliferation and stimulates 452 apoptosis in HT29 cells following activation of alkaline sphingomyelinase. Anticancer Res. 2003, 453 23, 3317–3322. 454 455 456 457 458 459 (28) Lauthier, F.; Taillet, L.; Trouillas, P.; Delage, C.; Simon, A. Ursolic acid triggers calciumdependent apoptosis in human Daudi cells. Anticancer Drugs 2000, 11, 737–745. (29) Fesik, S. W. Promoting apoptosis as a strategy for cancer drug discovery. Nat. Rev. Cancer 2005, 5, 876–885. (30) Russo, G. L.; Russo, M.; Ungaro, P. AMP-activated protein kinase: A target for old drugs against diabetes and cancer. Biochem. Pharmacol. 2013, 86, 339-350. 20 460 (31) Zheng, Q. Y.; Jin, F. S.; Yao, C.; Zhang, T. ; Zhang, G. H.; Ai, X. Ursolic acid-induced AMP 461 activated protein kinase (AMPK) activation contributes to growth inhibition and apoptosis in 462 human bladder cancer T24 cells. Biochem. Biophys. Res. Commun. 2012, 419, 741–747. 463 (32) Son, H. S.; Kwon, H. Y.; Sohn, E. J.; Lee, J. H.; Woo, H. J.; Yun, M.; Kim, S. H.; Kim, Y. C. 464 Activation of AMP-activated protein kinase and phosphorylation of glycogen synthase kinase3 β 465 mediate ursolic acid induced apoptosis in HepG2 liver cancer cells. Phytother. Res. 2013, 27, 1714- 466 1722. 467 (33) Lee, Y. H.; Wang, E.; Kumar, N.; Glickman, R. D. Ursolic acid differentially modulates apoptosis 468 in skin melanoma and retinal pigment epithelial cells exposed to UV-VIS broadband radiation. 469 Apoptosis 2013, [Epub ahead of print]. 470 (34) Lin, J. N.; Lin, V. C.; Rau, K. M.; Shieh, P. C.; Kuo, D. H.; Shieh, J. C.; Chen, W. J.; Tsai, S. C.; 471 Way, T. D. Resveratrol modulates tumor cell proliferation and protein translation via SIRT1- 472 dependent AMPK activation. J. Agric. Food Chem. 2010, 58, 1584-1592. 473 474 475 476 477 478 Figure legends 479 Figure 1. The anti-proliferation activities of different fractions from R. formosanum. (A) Leaf 480 powder of R. formosanum was extracted by methanol and partitioned into four fractions including 481 dichloromethane, ethyl acetate, n-butanol and water to get RFL-DCM, RFL-EA, RFL-BuOH, and RFL- 482 H2O fractions. (B) A549 cells were treated with four fractions (40-160 μg/mL) for 24 h. The cell 21 483 viability was then determined using MTT assay. This experiment was repeated three times. The data 484 represented the mean ± S.D. Values significantly were different from the control group. *, P < 0.05. 485 Figure 2. The anti-proliferation activities of different RFL-EA fractions against NSCLC A549 486 cells. (A) RFL-EA was subjected into silica column and eluted with n-hexane, ethyl actate and methanol 487 at different combination rates to get ten fractions. (B) A549 cells were treated with ten fractions (10-80 488 μg/mL) for 24 h. The cell viability was then determined using MTT assay. This experiment was repeated 489 three times. The data represented the mean ± S.D. Values significantly were different from the control 490 group. *, P < 0.05. (C) A549 cells were treated with RFL-EA-2 (40-80 μg/mL) and RFL-EA-3 (40-80 491 μg/mL) for 24 h and the cell morphology was observed by photomicroscope. 492 Figure 3. Quantification of triterpenoids by LC-ESI-MS/MS analysis. (A) Individual standard 493 triterpenoids including betulinic acid (B), oleanolic acid (O) and ursolic acid (U) were subjected to LC- 494 ESI-MS/MS analysis for chemical identification and quantification. (B) Quantification of the betulinic 495 acid, oleanolic acid and ursolic acid from RFL-EA-2 and RFL-EA-3 fractions. 496 Figure 4. The anti-proliferation effect of ursolic acid, oleanolic acid and betulinic acid against 497 NSCLC cells. A549 cells and H460 cells were treated with (A) ursolic acid, (B) oleanolic acid and (C) 498 betulinic acid at concentration of 10-160 μM for 24 h or 48 h. The cell viability was then determined 499 using MTT assay. This experiment was repeated three times. The data represented the mean ± S.D. 500 Values significantly were different from the control group. *, P < 0.05. 501 Figure 5. Ursolic acid induced A549 cells apoptosis. (A) A549 cells were incubated with ursolic acid 502 (10-40 μM) for 24 h, and morphology was observed by photomicroscope, and (B) the morphology of 503 cell nuclei was observed by fluorescence microscope. (C) To observe cell cycle statements and apoptosis 504 levels, cell were stained with PI and measured by flow cytometry. (D) A549 cells were treated with 505 ursolic acid (30 μM) for the indicated time. Cells were then harvested and lysed for the detection of 506 caspase-3, cleaved caspase-3, caspase-9, cleaved caspase-9 and β-actin. (E) A549 cells were treated with 22 507 ursolic acid (30 μM) for the indicated time. Cells were then harvested and lysed for the detection of Bcl- 508 2, Bax and β-actin. Western blot data presented are representative of those obtained in at least three 509 separate experiments. The values below the figures represent change in protein expression of the bands 510 normalized to β-actin. 511 Figure 6. Ursolic acid decreases general mRNA translation and the activity of fatty acid synthesis 512 via activating of AMPK. (A) A549 cells were treated with ursolic acid (30 μM) for the indicated time. 513 Cells were then harvested and lysed for the detection of phosphorylated AMPK (Thr 172), AMPK and 514 β-actin. (B) A549 cells were treated with ursolic acid (30 μM) for the indicated time. Cells were then 515 harvested and lysed for the detection of phosphorylated mTOR (Ser 2448), mTOR, phosphorylated 516 p70S6K and β-actin. (C) A549 cells were treated with ursolic acid (30 μM) for the indicated time. Cells 517 were then harvested and lysed for the detection of FASN, phospho-ACC (Ser79) and β-actin. (D) A549 518 cells were transfected with 50 nmol/L LKB1-siRNA using Oligofectamine. A total of 24 h after 519 transfection, cells were treated with ursolic acid (30 μM) for 24 h. After harvesting, cells were lysed and 520 prepared for Western blotting analysis using antibodies against LKB-1, phosphorylated AMPK (Thr 521 172) and β-actin. Western blot data presented are representative of those obtained in at least three 522 separate experiments. The values below the figures represent change in protein expression of the bands 523 normalized to β-actin. 524 525 526 527 23 528 24 529 530 531 532 25 533 534 535 26 536 537 27 538 28 539 29 540 541 542 543 544 545 546 547 30 548 549 550 551 552 553 31