Skip to main content
Scientific Reports logoLink to Scientific Reports
. 2016 Mar 2;6:22388. doi: 10.1038/srep22388

Pharmacoproteomic analysis reveals that metapristone (RU486 metabolite) intervenes E-cadherin and vimentin to realize cancer metastasis chemoprevention

Suhong Yu 1,a,*, Cuicui Yan 1,*, Xingtian Yang 1,*, Sudang He 1, Jian Liu 1, Chongtao Qin 2, Chuanzhong Huang 3, Yusheng Lu 1, Zhongping Tian 1, Lee Jia 1,b
PMCID: PMC4773818  PMID: 26932781

Abstract

Metapristone is the most predominant biological active metabolite of mifepristone, and being developed as a novel cancer metastasis chemopreventive agent by us. Despite its prominent metastasis chemopreventive effect, the underlying mechanism remains elusive. Our study, for the first time, demonstrated that metapristone had the ability to prevent breast cancer cells from migration, invasion, and interfere with their adhesion to endothelial cells. To explore the underlying mechanism of metapristone, we employed the iTRAQ technique to assess the effect of metapristone on MDA-MB-231 cells. In total, 5,145 proteins were identified, of which, 311 proteins showed significant differences in metapristone-treated cells compared to the control group (P-value < 0.05). Bioinformatic analysis showed many differentially expressed proteins (DEPs) functionally associated with post-translational modification, chaperones, translation, transcription, replication, signal transduction, etc. Importantly, many of the DEPs, such as E-cadherin, vimentin, TGF-β receptor I/II, smad2/3, β-catenin, caveolin, and dystroglycan were associated with TGF-β and Wnt signaling pathways, which were also linked to epithelial-to-mesenchymal transition (EMT) process. Further validation of the epithelial marker “E-caderin” and mesenchymal marker “vimetin” were carried out using immunoblot and immunofluorescence. These results have revealed a novel mechanism that metapristone-mediated metastasis chemoprevention is through intervening the EMT-related signaling pathways.


Breast cancer is the first leading cause of cancer mortality in women worldwide. Every year, above 1.3 million women are diagnosed with breast cancer and nearly 450,000 women die from it1. Metastasis, a process that cancer cells invade surrounding tissues and migrate to distal organs including lung, liver, brain, bone, and lymph nodes, is a major cause of mortality in breast cancer patients2. Therefore, development of safe and effective cancer metastasis chemopreventive agents is becoming important and badly needed. Metapristone, the most predominant biological active metabolite of mifepristone (RU486), is being developed as a novel cancer metastasis chemopreventive by us3,4.

Metapristone has received considerable attention to its anticancer activity recently. In our previous studies, we showed that metapristone produced comparable antitumor effect on several cancer cell lines. For example, metapristone induced HT-29 cells to be arrested at the G0/G1 stage, induced dose-dependent apoptosis, and interfered with adhesion of HT-29 cells to human umbilical vein endothelial cells (HUVECs) in vitro3,4,5.

Although the anticancer activity of metapristone has been exploited, its exact molecular mechanisms of actions and related pathways and targets towards cancer remain poorly understood. To get a more comprehensive understanding of metapristone functions on cancer, we employed the pharmacoproteomic analysis in the present study as we pioneered ten years ago6. Isobaric tags for relative and absolute quantitation (iTRAQ) technique is considered one of the most robust techniques for differential quantitative proteomic analysis7, which yields very small coefficients of variation in quantitative measurements8. Unlike gel-based proteomic method, iTRAQ exhibits much better sensitivity and allows the identification and accurate quantification of proteins from multiple samples9.

The epithelial–mesenchymal transition (EMT) is an important cellular process during which epithelial polarized cells become motile mesenchymal-appeared cells, which, in turn, promotes cancer cell invasion and metastasis10,11. The EMT process is very complex and controlled by various families of transcriptional regulators through different signaling pathways, including TGF-β, Wnt, MAPK, EGFR, PI3K and others12,13,14. Therefore, preventing cancer cells from epithelial-mesenchymal transition as well as intervening with the key proteins in EMT-related pathways is the main research objective for us to identify safe and effective cancer metastasis chemopreventives.

In the current study, we investigated the cancer metastasis chemopreventive effect of metapristone on the cell growth, migration, invasion and adhesion of MDA-MB-231 cells in vitro, and further explored the underlying molecular mechanism of metapristone by using an isobaric tag for relative and absolute quantitation iTRAQ combined with the tandem mass spectrometry (LC-ESI-MS/MS). We further identified differentially expressed proteins and potential signaling pathways in MDA-MB-231 cells after metapristone treatment. The findings reported in this study support our hypothesis and reveal, for the first time, a novel function for metapristone in the prevention of metastasis of breast cancer by intervening EMT-related signaling pathways.

Results

Effect of metapristone on cell viability

To explore the metastasis chempreventives function of metapristone, the cytostatic effect was examined first on human breast cancer cells MDA-MB-231 after treatment with various concentrations of metapristone for 24 h. As showed in Fig. 1A, the cytotoxicity of metapristone was low. The IC50 value for metapristone to suppress MDA-MB-231 cell proliferation is 91 μM.

Figure 1. Cellular pharmacology analysis of metapristone.

Figure 1

(A) in vitro activity of metapristone against MDA-MB-231 cell line. (B) dose-dependent inhibition by metapristone on cell migration. (C) inhibition by metapristone of MDA-MB-231 cells adhesion to HUVECs. Representative microscopic observation of the inhibition by metapristone at 0, 10, 50, and 75 μM. DMSO (0.1%) was used as vehicle control (average of 10 independent microscope fields for each of 3 independent experiments). (D) a Corning transwell system was used to assay cell invasion as described in methods. The amount of MDA-MB-231 cells invading through polycarbonate membranes was counted by microscopic observation (10×). Each experiment was carried out at least three times. **P < 0.01.

Metapristone inhibits cell migration, adhesion, and invasion

Would healing assay was conducted with MDA-MB-231 cells to examine the effect of metapristone on cell motility. As shown in Fig. 1B, cellular migration was controlled in a concentration-dependent manner by metapristone, being inhibited by up to 15%, 23% and 43% at 10, 50 and 75 μM, respectively (P < 0.01). Metapristone inhibited cell motility and wound closure at concentrations lower than its IC50, suggesting its specific inhibition on cell migration.

Tumor cells adhesion to the ECM is a fundamental step in cancer metastasis, the adherence of MDA-MB-231 cells to HUVECs was assessed to determine whether metapristone can regulate cell adhesion at a non-cytotoxic concentration. Ten fields of each well were randomly selected, and the adhered spots were counted. Compared with the control, the adhesion rate of MDA-MB-231 cells was 84, 68 and 39%, respectively, at 10, 50 and 75 μM of metapristone (Fig. 1C). Metapristone markedly and in a concentration-dependent manner inhibited the adherence of MDA-MB-231 cells to endothelial monolayers, indicating that it may fit into a new class of therapy for the reduction of risk factors of cancer metastasis.

It is well known that MDA-MB-231 cells have strong invasion properties in matrigel. In this study, we investigate the inhibitory effect of metapristone on cell invasion using a transwell system coated with matrigel. We found that treatment with metapristone for 24 h significantly inhibited MDA-MB-231 cells invasion through the transwell membrane. When metapristone was added at 10, 50 and 75 μM, the inhibitory effects were much more obvious compared to that of untreated group, with the inhibition rate of 48.52%, 60.06% and 82.88%, respectively (Fig. 1D).

Overview of quantitative proteomics

The iTRAQ analysis was performed on the purified protein extracts from MDA-MB-231 cells with or without metapristone treatment to understand the mechanism of metapristone-mediated anti-metastasis mechanism on the cellular and molecular level (Fig. 2). In total, 440,119 spectra were obtained from the iTRAQ-LC-MS/MS proteomic analysis. After data filtering to eliminate low-scoring spectra, a total of 93,114 unique spectra that met the strict confidence criteria for identification were matched to 5,145 unique proteins, of which, 311 proteins showed significant differences in metapristone-treated cells (P-value < 0.05). The detailed information including protein accession number, identified peptide number, protein score, sequence coverage, and regulation (fold change) for these identified proteins is shown in Table 1 and 2. Among these differentially expressed proteins (DEPs), 163 proteins were up-regulated (Table 1) and 148 proteins were down-regulated (Table 2). Then, GO analysis was conducted with the GSEABase package from R (http://www.r-project.org/) statistical platform15. Genes were classified in three major groups: the biological process, cellular component, and molecular function (Fig. 3A–C). Approximately 50.94% of the altered proteins were binding proteins, 27.52% were catalytic and 3.91% were enzyme regulators. In addition, we performed COG function prediction and classified these 311 positive proteins into 18 functional categories (Fig. 4).

Figure 2. Workflow used to study differential expressed proteins in MDA-MB-231 cells after metapristone treatment using iTRAQ technology.

Figure 2

Table 1. Annotation of up-regulated proteins after metapristone treatment in MDA-MB-231 cells.

No. Score % Cov Accession number Name Peptides regulation (fold change)a
1 226 17.3 D9HTE9 Plasma membrane citrate carrier 5 1.707*
2 503 14.5 P31040 Succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial 8 3.644*
3 308 39.9 P62280 40S ribosomal protein S11 7 1.709*
4 115 19.2 Q0QEY7 Succinate dehydrogenase complex subunit B 4 3.776*
5 474 27.8 P13073 Cytochrome c oxidase subunit 4 isoform 1, mitochondrial 5 1.671*
6 267 30 Q53EW8 Thiosulfate sulfurtransferase variant 7 3.181*
7 488 30.3 E9PH29 Thioredoxin-dependent peroxide reductase, mitochondrial 6 1.538*
8 446 20.7 A2A274 Aconitate hydratase, mitochondrial 13 1.528*
9 266 24.1 Q5QNZ2 ATP synthase F(0) complex subunit B1, mitochondrial 5 1.681*
10 388 26.7 Q59FZ8 Nebulette non-muscle isoform variant 9 1.591*
11 302 14.5 A6NN80 Annexin 10 2.861*
12 410 50.3 O75947 ATP synthase subunit d, mitochondrial 7 3.498*
13 1130 34.2 Q59GB4 Dihydropyrimidinase-like 2 variant 15 1.781*
14 947 30.7 Q06210-2 Isoform 2 of Glutamine—fructose-6-phosphate aminotransferase [isomerizing] 1 16 4.479**
15 212 16.1 G3V325 Pentatricopeptide repeat-containing protein 1, mitochondrial 4 2.441*
16 2168 59.3 P00338 L-lactate dehydrogenase A chain 17 2.417*
17 135 15.3 B7Z792 cDNA FLJ53932 5 1.713*
18 518 16.9 D3DUJ0 AFG3 ATPase family gene 3-like 2, isoform CRA_a 11 1.981*
19 388 63.2 E9PN17 ATP synthase subunit g, mitochondrial 4 1.709*
20 202 25.1 Q5HYK3 2-methoxy-6-polyprenyl-1,4-benzoquinol methylase, mitochondrial 4 2.408*
21 671 45.4 P15559-2 Isoform 2 of NAD(P)H dehydrogenase [quinone] 1 9 3.405*
22 146 17.5 Q8N4T8 Carbonyl reductase family member 4 4 5.404**
23 474 29.8 P62277 40S ribosomal protein S13 5 2.403*
24 189 23.7 B2RDE0 cDNA, FLJ96567 8 6.402**
25 1329 48.2 Q53FB6 Mitochondrial aldehyde dehydrogenase 2 variant 19 5.389**
26 530 26.7 Q53FC3 Programmed cell death 6 variant 5 2.312*
27 161 20.9 Q9UK22 F-box only protein 2 5 1.975*
28 148 22.6 P18827 Syndecan-1 5 4.562*
29 458 35.2 Q5T9B7 Adenylate kinase isoenzyme 1 6 2.334*
30 237 17.5 B3KMV8 cDNA FLJ12766 fis, clone NT2RP2001520 6 3.366*
31 252 28.7 I3L1P8 Mitochondrial 2-oxoglutarate/malate carrier protein (Fragment) 6 2.364*
32 167 52.2 P56385 ATP synthase subunit e, mitochondrial 4 4.361*
33 216 24.1 B3KTJ1 cDNA FLJ38349 fis, clone FEBRA1000057 6 5.353*
34 259 22.4 Q5M7Z1 RAD23 homolog A (S. cerevisiae) 4 3.351*
35 177 30.2 Q9H479 Fructosamine-3-kinase 4 2.342*
36 160 19.5 B4DNW0 Aminoacylase-1 9 2.921*
37 775 23 O60832 H/ACA ribonucleoprotein complex subunit 10 3.339**
38 104 36.5 Q9NRV9 Heme-binding protein 1 5 1.759*
39 194 43.4 P35754 Glutaredoxin-1 4 4.368*
40 362 22 Q96I99 Succinyl-CoA ligase [GDP-forming] subunit beta, mitochondrial 9 1.837*
41 156 24.6 F5GZW3 Rho GTPase-activating protein 4 4 1.536*
42 144 33.8 A8KA74 cDNA FLJ76065 4 1.625*
43 434 24.4 B4E2Z8 cDNA FLJ61206 8 1.734*
44 904 37 P11177-2 Isoform 2 of Pyruvate dehydrogenase E1 component subunit beta, mitochondrial 10 2.333*
45 699 31.6 P00966 Argininosuccinate synthase 13 2.302*
46 109 22.6 Q5SRD1 Putative mitochondrial import inner membrane translocase subunit Tim23B 4 2.133*
47 969 25.4 Q9NSE4 Isoleucine—tRNA ligase, mitochondrial 22 6.329**
48 254 15 Q8N0 × 4 Citrate lyase subunit beta-like protein, mitochondrial 4 3.327*
49 222 15.2 Q14376 UDP-glucose 4-epimerase 4 7.325**
50 225 36.7 Q86WA8 Lon protease homolog 2, peroxisomal 4 5.324*
51 128 42.1 P43155-2 Isoform 2 of Carnitine O-acetyltransferase 5 1.724*
52 414 19.6 D7PBN3 ESRP1/RAF1 fusion protein 13 2.324*
53 2112 50.5 Q59EI9 ADP,ATP carrier protein, liver isoform T2 variant 16 7.32**
54 381 29.3 D3XNU5 E-cadherin 1 6 7.319**
55 398 22.8 D3DVA5 Rho/rac guanine nucleotide exchange factor (GEF) 2, isoform CRA_a 12 1.619*
56 346 20.8 B4E290 cDNA FLJ50039 10 8.318**
57 1299 50.5 P12429 Annexin A3 15 2.322*
58 379 22.4 B7Z3K9 Fructose-bisphosphate aldolase 8 2.315*
59 151 19.9 Q9H974-2 Isoform 2 of Queuine tRNA-ribosyltransferase subunit QTRTD1 4 1.751*
60 556 23 B4DFP1 cDNA FLJ51818 12 1.815*
61 679 45 P84085 ADP-ribosylation factor 5 8 1.663*
62 317 18.8 P80303-2 Isoform 2 of Nucleobindin-2 7 2.306*
63 402 16.5 Q8NE62 Choline dehydrogenase, mitochondrial 9 1.996*
64 1016 32.6 Q13011 Delta(3, 5)-Delta(2, 4)-dienoyl-CoA isomerase, mitochondrial 10 1.703*
65 149 36.8 Q7Z4G4-2 Isoform 2 of tRNA (guanine(10)-N2)-methyltransferase homolog 5 2.302*
66 227 21.2 B4DG80 LIM and cysteine-rich domains protein 1 5 3.271*
67 122 26.3 O00178 GTP-binding protein 1 4 2.111*
68 196 24.8 H3BNX8 Cytochrome c oxidase subunit 5A, mitochondrial 5 3.022*
69 264 31.6 O95865 N(G), N(G)-dimethylarginine dimethylaminohydrolase 2 7 2.395*
70 286 41.8 P36969-2 Isoform Cytoplasmic of Phospholipid hydroperoxide glutathione peroxidase, mitochondrial 7 1.795*
71 238 21.9 B3KTM6 Ribosomal protein L5, isoform CRA_b 5 1.694*
72 891 48.9 P18085 ADP-ribosylation factor 4 8 1.742*
73 105 18 Q96CF2 Charged multivesicular body protein 4c 4 1.562*
74 714 20.5 Q6XQN6-3 Isoform 3 of Nicotinate phosphoribosyltransferase 4 2.291*
75 419 72.1 Q9HCY8 Protein S100-A14 6 1.287*
76 416 22.7 Q8IYS1 Peptidase M20 domain-containing protein 2 9 2.286*
77 1692 35.4 Q59EK6 TNF receptor-associated protein 1 variant 21 1.986*
78 192 16.7 B3KQQ0 cDNA PSEC0007 fis, clone NT2RM1000634 8 1.784*
79 322 27.7 B4DP80 cDNA FLJ56357 6 2.283*
80 270 14.4 B4DUF1 cDNA FLJ59760 8 2.252*
81 519 28.8 P31930 Cytochrome b-c1 complex subunit 1, mitochondrial 10 3.281*
82 278 33.5 B7Z4B7 cDNA FLJ52561 7 2.218*
83 1591 37.3 K7EKE6 Lon protease homolog, mitochondrial 25 3.278*
84 345 25 P04040 Catalase 11 3.215*
85 1420 58.6 P30044-2 Isoform Cytoplasmic + peroxisomal of Peroxiredoxin-5, mitochondrial 7 2.275*
86 571 44.9 B4DNR3 cDNA FLJ52710 6 8.214**
87 213 46.8 B4DRT2 28S ribosomal protein S27, mitochondrial 4 7.212**
88 412 24.4 Q6NVY1 3-hydroxyisobutyryl-CoA hydrolase, mitochondrial 9 6.274**
89 636 47 M0R0F0 40S ribosomal protein S5 (Fragment) 10 11.271**
90 254 45.4 P61081 NEDD8-conjugating enzyme Ubc12 8 6.269**
91 524 23.1 P17858 6-phosphofructokinase, liver type 14 7.368**
92 146 11.2 Q53H22 Amidophosphoribosyltransferase 5 2.068*
93 152 19.1 P46781 40S ribosomal protein S9 4 1.968*
94 227 25.6 Q9NWV4 UPF0587 protein C1orf123 5 1.266*
95 159 19.4 B3KRI2 NADH dehydrogenase [ubiquinone] iron-sulfur protein 7, mitochondrial 4 3.214*
96 646 21.6 B2R9S4 cDNA, FLJ94534 6 1.963*
97 296 18.4 B2R6S5 Cytidylate kinase, isoform CRA_a 4 10.262**
98 576 16.1 Q9UJS0-2 Isoform 2 of Calcium-binding mitochondrial carrier protein Aralar2 8 8.261**
99 269 39.5 O75368 SH3 domain-binding glutamic acid-rich-like protein 4 7.259**
100 522 45.2 O75223 Gamma-glutamylcyclotransferase 8 6.229*
101 168 24.7 B2R9 × 3 cDNA, FLJ94599 10 5.257*
102 2592 45.5 Q6NVC0 SLC25A5 protein (Fragment) OS = Homo sapiens 16 3.255*
103 319 22.5 Q9BQ69 O-acetyl-ADP-ribose deacetylase MACROD1 6 3.154*
104 179 14.8 Q6V9R7 Solute carrier family 25 member 19 4 1.752*
105 108 23.6 Q8IW45 ATP-dependent (S)-NAD(P)H-hydrate dehydratase 5 1.922*
106 878 32.4 P35908 Keratin, type II cytoskeletal 2 epidermal 18 3.251*
107 953 37.6 P49419-2 Isoform 2 of Alpha-aminoadipic semialdehyde dehydrogenase 16 5.338**
108 751 30.1 P07384 Calpain-1 catalytic subunit 19 7.247*
109 564 62 O95336 6-phosphogluconolactonase 11 11.246**
110 176 14.1 Q9H9T3-2 Isoform 2 of Elongator complex protein 3 6 5.245*
111 2382 35.4 Q53F91 Villin 1 variant 27 3.245**
112 247 31.4 B4DP27 cDNA FLJ52153 5 2.242*
113 365 25.9 Q9NUQ9 Protein FAM49B 7 3.551*
114 268 35.6 E7EW20 Unconventional myosin-VI 9 7.241**
115 4889 43.5 B7Z2 × 9 Gamma-enolase 13 5.244*
116 494 15.2 P10253 Lysosomal alpha-glucosidase 10 3.379*
117 167 20.2 P15328 Folate receptor alphas 4 2.238*
118 166 28.4 Q13315 Serine-protein kinase ATM 6 5.238*
119 232 20.2 B3KM98 cDNA FLJ10556 fis, clone NT2RP2002479 6 6.238*
120 267 29.7 Q02338 D-beta-hydroxybutyrate dehydrogenase, mitochondrial 7 4.235*
121 673 44.2 B7Z6B8 2,4-dienoyl-CoA reductase, mitochondrial 11 9.215**
122 114 20.9 B4DQ51 Short/branched chain-specific acyl-CoA dehydrogenase, mitochondrial 4 8.234**
123 2746 43.8 P40939 Trifunctional enzyme subunit alpha, mitochondrial 31 7.212**
124 437 28.9 P31937 3-hydroxyisobutyrate dehydrogenase, mitochondrial 6 5.212*
125 214 33.5 B3KTS4 cDNA FLJ38665 fis, clone HLUNG2003378 8 4.222*
126 3670 56.3 P06899 Histone H2B type 1-J 8 10.231**
127 5762 59.3 Q13885 Tubulin beta-2A chain 21 1.831*
128 647 27.9 B2RAH7 cDNA, FLJ94921 16 1.529*
129 443 17.5 O95202 LETM1 and EF-hand domain-containing protein 1, mitochondrial 11 2.229*
130 766 24.7 Q0VGA5 SARS protein s 10 1.729*
131 407 33.7 Q9Y305-2 Isoform 2 of Acyl-coenzyme A thioesterase 9, mitochondrial 12 1.855*
132 317 35.1 P63000-2 Isoform B of Ras-related C3 botulinum toxin substrate 1 7 2.277*
133 103 21.5 Q9C0C9 Ubiquitin-conjugating enzyme E2 O 5 6.217**
134 114 14.1 P04792 Heat shock protein beta-1 4 5.246*
135 248 15.7 Q9UBF2 Coatomer subunit gamma-2 4 6.213*
136 356 63.9 P07741 Adenine phosphoribosyltransferase 8 7.273**
137 260 14.7 Q8TE67-2 Isoform 2 of Epidermal growth factor receptor kinase substrate 8-like protein 3 8 2.222*
138 2952 49 P09211 Glutathione S-transferase P 9 1.825*
139 343 43.8 Q8TCD5 5~(3~)-deoxyribonucleotidase, cytosolic type 6 2.261*
140 234 26.9 Q8NCF7 cDNA FLJ90278 fis, clone NT2RP1000325 10 2.254*
141 739 45.2 P14550 Alcohol dehydrogenase [NADP(+)] 13 1.722*
142 138 19 B2RCC2 cDNA, FLJ95978 5 1.998*
143 235 16.4 Q9BRQ8 Apoptosis-inducing factor 2 6 9.217**
144 267 39 P30046 D-dopachrome decarboxylase 4 3.217*
145 144 16.4 B4DRN7 C2 domain-containing protein 5 5 8.216**
146 1762 51.4 Q6LES2 Annexin (Fragment) 15 8.113**
147 383 21.8 Q5JNW7 Proteasome subunit beta type-8 4 12.212**
148 126 20.9 Q96GD0 Pyridoxal phosphate phosphatase 4 7.202*
149 516 45.7 Q9NQR4 Omega-amidase NIT2 11 7.254**
150 278 16.6 B3KM97 cDNA FLJ10554 fis, clone NT2RP2002385 5 1.911*
151 427 25.1 J3QQX3 NADPH:adrenodoxin oxidoreductase, mitochondrial 8 1.721*
152 184 19.1 P47929 Galectin-7 4 7.209**
153 125 22.9 Q6PJ77 BTF3L4 protein (Fragment) 4 6.209**
154 228 17.3 R4GMU1 GDH/6PGL endoplasmic bifunctional protein 9 5.248**
155 326 32.7 B2R7T6 cDNA, FLJ93596 12 7.217**
156 175 21.2 Q9BUL8 Programmed cell death protein 10 5 3.252*
157 264 19.6 B2R673 cDNA, FLJ92818 9 1.884*
158 341 34.1 G8JLB3 tRNA pseudouridine synthase (Fragment) 11 1.653*
159 1093 26 F8W930 Insulin-like growth factor 2 mRNA-binding protein 2 13 8.224**
160 169 16 Q15031 Probable leucine—tRNA ligase, mitochondrial 5 5.263*
161 2146 44.7 O43175 D-3-phosphoglycerate dehydrogenase 20 5.211*
162 516 22.2 B4E0B1 cDNA FLJ52100 4 3.202*
163 151 20.3 B4DKL4 Lipolysis-stimulated lipoprotein receptor 6 2.328*

aRegulations (fold-changes) of differentially expressed proteins in MDA-MB-231 cells (metapristone-treatment versus control). *P < 0.05; **P < 0.01.

Table 2. Annotation of down-regulated proteins after metapristone treatment in MDA-MB-231 cells.

No. Score % Cov Accession number Name Peptides regulation (fold change)a
1 701 35.3 A8K9B9 cDNA FLJ77391 17 0.331*
2 286 31.4 B3KQF5 cDNA FLJ90381 fis, clone NT2RP2005035 8 0.511*
3 451 20.1 Q9H089 Large subunit GTPase 1 homolog 11 0.621*
4 167 22.4 A7UJ17 DnaJ 4 0.431*
5 178 18.3 Q53HF3 Galactosidase, alpha variant 4 0.383*
6 165 14.7 Q9BTM9-2 Isoform 2 of Ubiquitin-related modifier 1 4 0.501*
7 263 22.7 Q8NAF0 Zinc finger protein 579 4 0.528*
8 221 18.1 Q9BPX3 Condensin complex subunit 3 10 0.438*
9 122 21.3 J3QTQ0 Dystonin 8 0.607*
10 175 16.2 B4DQM4 GPN-loop GTPase 1 4 0.527*
11 437 27 J3KQA0 Synaptotagmin I, isoform CRA_b 10 0.694*
12 119 15.4 B2R728 cDNA, FLJ9325 4 0.603*
13 133 23.1 Q13308-2 Isoform 2 of Inactive tyrosine-protein kinase 7 6 0.324**
14 2854 46 A8K2 × 8 cDNA FLJ78433 25 0.523*
15 226 12.8 Q9ULX6 A-kinase anchor protein 8-like 6 0.612*
16 704 30.3 Q6FHK7 PSME3 protein 7 0.521*
17 479 25.4 A8K878 cDNA FLJ77177 4 0.282**
18 612 36 P84022 Smad3 5 0.233**
19 532 20.1 M0QY97 Zinc finger CCCH domain-containing protein 4 13 0.619*
20 316 22.2 Q7Z417 Nuclear fragile X mental retardation-interacting protein 2 7 0.518*
21 117 25.4 Q9UHN6 Transmembrane protein 2 5 0.317**
22 274 14.6 B4DNN4 Ribonucleoside-diphosphate reductase 9 0.516*
23 172 21.9 Q12846 Syntaxin-4 5 0.415*
24 139 25.8 H0Y5K5 Endoplasmic reticulum-Golgi intermediate compartment protein 3 4 0.414**
25 215 40.1 B4DGU4 Catenin beta-1 5 0.446**
26 287 32.7 K7EPB2 cAMP-dependent protein kinase type I-alpha regulatory subunit 9 0.332**
27 131 16.1 B2WTI3 Bifunctional arginine demethylase and lysyl-hydroxylase JMJD6 4 0.632*
28 260 15 B3KN49 cDNA FLJ13562 fis, clone PLACE1008080 5 0.211**
29 269 17.7 B3KSG9 cDNA FLJ36188 fis, clone TESTI2027179 5 0.441**
30 519 27.1 P46013 Antigen KI-67 20 0.281**
31 219 32.9 Q96A35 39S ribosomal protein L24, mitochondrial 6 0.181**
32 445 21.6 B7Z591 Transmembrane and coiled-coil domains 1, isoform CRA_a 4 0.409**
33 457 17.9 Q9NYF8-2 Isoform 2 of Bcl-2-associated transcription factor 1 11 0.338**
34 222 25.8 Q9UNK0 Syntaxin-8 6 0.467*
35 247 21.5 Q9BYK8 Helicase with zinc finger domain 2 4 0.607*
36 156 19.7 Q6LEU0 STX12 protein 4 0.557*
37 164 29.1 B2R6J0 Homo sapiens SRY (sex determining region Y)-box 2 (SOX2) 4 0.204**
38 394 16.7 Q92896-2 Isoform 2 of Golgi apparatus protein 1 19 0.304*
39 226 15.8 B4DRG7 Condensin complex subunit 10 0.514*
40 161 21.4 Q9BXK1 Krueppel-like factor 16 4 0.353*
41 281 17.4 Q8NFC6 Biorientation of chromosomes in cell division protein 1-like 1 4 0.409*
42 118 24.8 P17301 Integrin alpha-2 5 0.322**
43 457 22.9 Q15796-2 Smad2 6 0.201**
44 845 36.8 P61586 Transforming protein RhoA 6 0.374*
45 124 15.7 Q15628 Tumor necrosis factor receptor type 1-associated DEATH domain protein 4 0.26**
46 437 19.5 Q01650 Large neutral amino acids transporter small subunit 1 4 0.201*
47 718 22.9 Q86U75 Dihydropyrimidinase-like 2 10 0.244*
48 895 19.3 H3BUX2 Cytochrome b5 type B 4 0.508*
49 674 43.9 H0YKC5 Deoxyuridine 5~-triphosphate nucleotidohydrolase, mitochondrial 7 0.299*
50 355 27.2 P62906 60S ribosomal protein L10a 5 0.633*
51 287 14.5 B3KM90 cDNA FLJ10529 fis, clone NT2RP2000965 8 0.672*
52 134 17.9 Q8NCC3 Group XV phospholipase A2 4 0.495*
53 148 19.8 Q3LIB1 Putative uncharacterized protein Nbla00445 8 0.612*
54 163 20.4 O43752 Syntaxin-6 5 0.679*
55 1445 53.8 P04083 Annexin A1 15 0.586*
56 507 29.2 P20645 Cation-dependent mannose-6-phosphate receptor 7 0.385**
57 349 41.9 P60520 Gamma-aminobutyric acid receptor-associated protein-like 2 6 0.283**
58 1061 47.1 Q6FI35 Proliferating cell nuclear antigen 11 0.381**
59 156 33.4 B2RMQ4 Cytoskeleton associated protein 2 4 0.607*
60 192 16.5 G3V5T9 Cyclin-dependent kinase 2 5 0.633*
61 263 37.9 Q6IAA8 Ragulator complex protein LAMTOR1 4 0.576*
62 150 18.1 B4DJI2 cDNA FLJ53342 4 0.624*
63 108 22 B2R7M1 cDNA, FLJ93507 4 0.376*
64 140 17.3 H0Y3T6 45 kDa calcium-binding protein 4 0.558*
65 147 27.4 F8VX04 Sodium-coupled neutral amino acid transporter 1 4 0.471*
66 1103 54.2 B4DJP7 Small nuclear ribonucleoprotein Sm D3 5 0.277**
67 731 26.1 O94925-3 Isoform 3 of Glutaminase kidney isoform, mitochondrial 13 0.167**
68 178 35.3 P51151 Ras-related protein Rab-9A 5 0.267**
69 197 20.7 P15529-16 Isoform 3 of Membrane cofactor protein 4 0.566*
70 125 18.9 P46087-4 Isoform 4 of Putative ribosomal RNA methyltransferase NOP2 6 0.162**
71 268 23.6 D6W4Z6 HCG23833, isoform CRA_b 4 0.654*
72 271 27.9 B7ZM24 SLC12A2 protein 9 0.64**
73 583 42.1 P36897.1 TGF-beta receptor type-1 4 0.335**
74 321 20.2 P98172 Ephrin-B1 5 0.539*
75 192 25.3 B7Z5A7 cDNA FLJ57557 4 0.454*
76 186 39.4 B4E324 cDNA FLJ60397 4 0.552*
77 200 15.1 Q9H5V8-2 Isoform 2 of CUB domain-containing protein 1 10 0.648*
78 249 18.6 Q96T88-2 Isoform 2 of E3 ubiquitin-protein ligase UHRF1 6 0.145**
79 241 35.9 B2R7 × 3 cDNA, FLJ93645 4 0.245**
80 191 22.6 P54709 Sodium/potassium-transporting ATPase subunit beta-3 6 0.544*
81 119 18 O14672 Disintegrin and metalloproteinase domain-containing protein 10 6 0.342**
82 257 18 B3KXC3 Ferritin 5 0.557*
83 236 51.1 K7EJT5 60S ribosomal protein L22 6 0.544*
84 278 14.9 B7Z4 × 6 cDNA FLJ51012, highly similar to Plasminogen activator inhibitor 1 5 0.339**
85 1951 38.6 Q9NR30 Nucleolar RNA helicase 2 25 0.445*
86 175 15.4 B2RAK1 cDNA, FLJ94965 11 0.537*
87 209 26.9 B4DMR3 cDNA FLJ51896, highly similar to Glia-derived nexin 8 0.235**
88 332 18.6 Q53G91 Solute carrier family 16, member 3 variant (Fragment) 4 0.334*
89 130 16.3 Q5U8S2 Syntaxin 10 4 0.233**
90 136 18.2 Q9UNE7 E3 ubiquitin-protein ligase CHIP 5 0.402*
91 385 26.5 Q7Z4F3 Caveolin 4 0.471*
92 130 18.2 A8KAQ6 cDNA FLJ76490 4 0.322*
93 2202 38.5 B4DMF5 Glutamate dehydrogenase 16 0.207**
94 143 22.7 B2R6P4 cDNA, FLJ93048 4 0.113**
95 1581 68.6 P51149 Ras-related protein Rab-7a 13 0.246**
96 257 15.9 H3BRB3 Kinesin-like protein KIF22 4 0.517*
97 277 18.2 P81605-2 Isoform 2 of Dermcidin 4 0.606*
98 263 19.1 Q8N353 TMEM106B protein 5 0.399*
99 1189 34.5 Q53G71 Calreticulin variant 11 0.601*
100 395 20.9 Q13217 DnaJ homolog subfamily C member 3 5 0.374*
101 126 17.4 B2RE34 cDNA, FLJ96901 4 0.442*
102 174 26 Q53GY1 BCL2-associated athanogene 3 variant 4 0.501*
103 200 38 Q9NQW6 Actin-binding protein anillin 8 0.695*
104 110 27.9 A8K3S3 cDNA FLJ75664 5 0.326*
105 326 18.1 A8K201 cDNA FLJ75605 4 0.425*
106 115 17.9 A8K274 cDNA FLJ78227 4 0.689*
107 438 25.8 Q9NQ29-2 Isoform 2 of Putative RNA-binding protein Luc7-like 1 8 0.686*
108 20862 51.2 Q15149-4 Isoform 4 of Plectin 20 0.686*
109 164 27.5 O00161 Synaptosomal-associated protein 23 4 0.68*
110 457 18.5 Q59EZ3 Insulin-like growth factor 2 receptor variant 25 0.678*
111 362 18 A8MXZ4 G-protein-coupled receptor family C group 5 member C 5 0.673*
112 682 14.9 B3KRY3 cDNA FLJ35079 fis, clone PLACE6005283 6 0.398**
113 120 26.3 B4DN85 E3 ubiquitin-protein ligase 4 0.595*
114 108 10.8 O15269 Serine palmitoyltransferase 1 5 0.67*
115 382 24.2 B4DL49 cDNA FLJ58073, moderately similar to Cathepsin B 5 0.67*
116 139 16 H0YDJ9 CD81 antigen 4 0.657*
117 522 32.4 B4DKJ4 cDNA FLJ57738 4 0.232**
118 149 25.2 B5BU32 Thymidine kinase 5 0.652*
119 122 33.8 O75976 Carboxypeptidase D 4 0.332*
120 618 19.3 P29317 Ephrin type-A receptor 2 11 0.651*
121 117 24.1 D6RAR4 Hepatocyte growth factor activator 4 0.451*
122 198 32.6 G3V3D1 Epididymal secretory protein E1 6 0.643*
123 286 30.7 C0JYY2 Apolipoprotein B 4 0.64*
124 114 29 F5H569 V-type proton ATPase 116 kDa subunit a isoform 1 6 0.276**
125 178 31.7 B4E1K0 Kinesin-like protein KIF23 4 0.633*
126 156 51.4 Q53HU8 vimentin 5 0.413*
127 125 34.5 Q14118 Dystroglycan 4 0.614*
128 128 16.2 C1K3N4 Tumor necrosis factor receptor superfamily member 10a 4 0.592*
129 165 32.6 Q13501-2 Isoform 2 of Sequestosome-1 4 0.58*
130 290 26.8 F5GZY0 Amyloid-like protein 2 4 0.576*
131 410 26.2 B4DJQ8 cDNA FLJ5569 8 0.174**
132 1790 32.8 P11387 DNA topoisomerase 1 26 0.044**
133 186 17.3 B2R686 Trans-golgi network protein 2, isoform CRA_a 4 0.542*
134 191 30.7 H0Y8A7 NEDD4 family-interacting protein 2 4 0.331**
135 112 27.7 P62266 40S ribosomal protein S23 5 0.53*
136 159 33.4 B3KMB6 cDNA FLJ10642 fis, clone NT2RP2005752 7 0.53*
137 175 25 B4DSG5 cDNA FLJ56149 5 0.525*
138 893 24.8 Q71UA6 Neutral amino acid transporter 10 0.499*
139 179 15.8 A8K6H9 cDNA FLJ75876 5 0.486*
140 198 28.6 Q9NRX5 Serine incorporator 1 4 0.442*
141 125 18.8 B4DIB1 cDNA FLJ55065 5 0.427*
142 137 17.3 P37173-2 Isoform 2 of TGF-beta receptor type-2 4 0.349*
143 753 25.1 P55010 Eukaryotic translation initiation factor 5 9 0.315*
144 121 35.7 P14174 Macrophage migration inhibitory factor 4 0.299**
145 325 24.1 E7EQY1 Protein FAM136A 5 0.432*
146 124 19.6 Q9NY27 Serine/threonine-protein phosphatase 4 regulatory subunit 2 4 0.411*
147 900 24.4 P62306 Small nuclear ribonucleoprotein F 4 0.558*
148 256 19.8 C9JEH3 Angio-associated migratory cell protein 7 0.512**

aRegulations (fold-changes) of differentially expressed proteins in MDA-MB-231 cells (metapristone-treatment versus control). *P < 0.05; **P < 0.01.

Figure 3. Categorization of all differential expressed proteins by GO analysis.

Figure 3

(A) cellular component. (B) biological process. C,molecular function (P < 0.05).

Figure 4. Functional category coverage of the proteins identified.

Figure 4

KEGG pathway analysis was also performed based on the 311 DEPs. A total of 249 metapristone-related pathways were identified, which were assigned into 33 statistically remarkable categories (P value < 0.01) (Table 3), including metabolic (such as “NADH dehydrogenase”, P56181-2), Oxidative phosphorylation (such as “ATP synthase”, O75947), MAPK signaling pathway (such as “Rac GTPase activating protein 1”, B2RE34), Wnt signaling pathway (such as “RhoA”, P61586), Focal adhesion (such as “Integrin alpha-2”, P17301), ECM-receptor interaction (such as “Dystroglycan”, Q14118), VEGF signaling pathway (such as “Protein kinase C”, Q2TSD3), and TGF-beta signaling pathway (such as “TGF-beta receptor type-2”, P37173-2).

Table 3. Pathway analysis of the DEPs obtained from the iTRAQ analysis.

Pathway description Count P-value
Metabolic pathways 81 5.21E-11
RNA transport 74 1.13E-10
Endocytosis 58 1.49E-10
Oxidative phosphorylation 56 2.83E-09
Apoptosis 53 5.78E-09
Focal adhesion 48 1.18E-08
MAPK signaling pathway 36 5.62E-08
Regulation of actin cytoskeleton 35 8.21E-08
GnRH signaling pathway 33 1.91E-07
B cell receptor signaling pathway 31 2.05E-07
Calcium signaling pathway 30 1.13E-06
Chemokine signaling pathway 28 3.61E-06
NF-kappa B signaling pathwy 27 4.13E-06
Peroxisome 27 1.70E-05
T cell receptor signaling pathway 25 2.18E-05
ErbB signaling pathway 23 3.62E-05
Neurotrophin signaling pathway 23 4.21E-05
Toll-like receptor signaling pathway 23 4.36E-05
Jak-STAT signaling pathway 23 6.99E-05
Insulin signaling pathway 20 0.000134
Notch signaling pathway 21 0.000313
ECM-receptor interaction 19 0.000397
mTOR signaling pathway 19 0.000724
p53 signaling pathway 17 0.000797
TGF-beta signaling pathway 17 0.000913
VEGF signaling pathway 16 0.001033
PPAR signaling pathway 15 0.001334
Adherens junction 13 0.001427
Wnt signaling pathway 12 0.003628
Cell adhesion molecules (CAMs) 11 0.003316
Drug metabolism-cytochrome P450 8 0.004733
ABC transporters 7 0.007124
Regulation of autophagy 6 0.008114

There were 249 pathways revealed. Among them, the following 33 signaling pathways were significant (P < 0.01).

Western blot validation of the proteomics analysis

Following the database search and classification of proteins, many differentially expressed proteins were reported to be involved in epithelial-to-mesenchymal transition (EMT), such as E-cadherin, vimentin, syndecan-1, β-catenin, dystroglycan, Smad2/3, glutaredoxin, TGF-β receptor, and so on. Western blots were performed on some selected proteins (E-cadherin, vimentin, β-catenin, and Smad 2) to further verify the iTRAQ results (Fig. 5B,C). While vimentin, one mesenchymal cell marker, was down-regulated by metapristone treatment, E-cadherin, one epithelial cell marker, strengthened with the increasing concentration of metapristone. Moreover, the expression of phosphorylation of Smad 2 was also found to be decreased by metapristone treatment. Notably, the western blot images correlated very well and thus confirmed the iTRAQ data obtained.

Figure 5. The effect of metapristone on cell morphology and EMT markers in MDA-MB-231 cells.

Figure 5

(A) morphological changes were observed by phase-contrast microscopy. (B,C) the expression of vimentin E-cadherin, β-catenin, Smad2, and pSmad2 in MDA-MB-231 cells treated with or without metapristone (50 μM) was assessed by immunoblotting analysis. (D) confocal microscope images of vimentin immunostained with goat anti-rabbit IgG-CY3 antibody (green) and E-cadherin immunostained with goat anti-mouse IgG-FITC antibody (red) in MDA-MB-231 cells untreated or treated metapristone (50 μM).

Metapristone impedes EMT in MDA-MB-231cells in vitro

Epithelial to mesenchymal transition (EMT) and extracellular matrix degradation are critical for the initiation and progression of tumor invasion. As shown in Fig. 5A, MDA-MB-231 cells initially exhibited a typical mesenchymal-like morphology with long and narrow stretch, while cells under the treatment of metapristone showed epithelial-like morphology with relatively round extension on the plastic surface. Furthermore, we sought to determine whether metapristone could inhibit Epithelial-mesenchymal transition by regulating EMT-related markers, such as vimentin (mesenchymal-specific marker) and E-cadherin (epithelial-specific marker). As shown in Fig. 5D, up-regulated E-cadherin accumulated in the cell to cell junctions after metapristone treatment. Accordingly, the significantly reduced expression of the mesenchymal-specific marker vimentin was observed in the presence of metapristone.

Discussion

Breast cancer metastasis accounts for the lethality of the disease and therefore there is an urgent need to develop new chemopreventives to inhibit cancer cell metastasis16,17. Experimental, epidemiological, and clinical data from the last three decades have each supported the hypothesis that oral contraceptive, such as mifepristone, possesses anticancer properties18,19. Then metapristone, the most predominant biological active metabolite of mifepristone, is being developed as a novel cancer metastasis chemopreventive agent by us.

Metastasis is a hallmark of cancer and the leading cause of mortality among cancer patients. The first step in metastasis is the migration of cancer cells away from the primary tumor, a process called tumor invasion20. Therefore, much research effortin recent years has been directed toward disruption of this step of the metastatic process21,22. In this study, we chose MDA-MB-231 cells with high metastatic potential to explore the effects of metapristone on the metastatic activity of human breast cancer cells. We showed that metapristone markedly inhibited their migratory (Fig. 1B) and invasive (Fig. 1D) abilities of MDA-MB-231 cells at low concentrations. Adhesion of cancer cells to ECM or vascular endothelium is also a crucial starting point of metastasis23. Here, we also found that metapristone markedly and in a concentration-dependent manner inhibited the adherence of MDA-MB-231 cells to endothelial monolayers. Collectively, these results suggested that metapristone had the ability to inhibit breast cancer cells metastasis. However, the underlying mechanism remains elusive.

Pharmacoproteomic, especially quantitative pharmacoproteomics, has been emerging as a powerful tool in cancer research, providing a unique avenue to investigate direct drug targets at a functional level24,25. Here, we have demonstrated the ability of the isobaric tags to detect and quantify differences in expression levels of proteins between metapristone-treated and untreated MDA-MB-231 cells that reflect functions associated with cancer cells metastasis. Temporal iTRAQ analysis identified 311 proteins as differentially expressed, with 163 as up-regulated (Table 1) and 148 as down-regulated (Table 2). Followed by GO analysis and KEGG pathway analysis, we established their potentially functional classification for the first time: there are 249 pathways, including metabolic, oxidative phosphorylation, p53, MAPK, Wnt, focal adhesion, VEGF, TGF-beta signaling pathways and so on (Table 3). Importantly, some of these pathways were reported to be linked to epithelial-to-mesenchymal transition (EMT) process, which was related with cancer carcinogenesis, prognosis and especially metastasis14,26.

The epithelial-to-mesenchymal transition (EMT) has been considered as the initiation process of cancer metastasis, when non-invasive and non-metastatic tumor cells lose their epithelial phenotype, acquire invasive properties, infiltrate surrounding tissues and metastasize to secondary sites27,28.Turning an epithelial cell into a mesenchymal cell requires loss of epithelial polarity, alteration in cellular architecture and acquisition of migrationcapacity29. It has also been described that during EMT, the epithelial cells acquire mesenchymal morphology, hence the expression of epithelial markers decreases and the expression of mesenchymal markers increases30,31. Here, we found that MDA-MB-231 cells initially exhibited a typical mesenchymal-like morphology with long and narrow stretch, while under the treatment of metapristone, cells showed epithelial-like morphology with relatively round extension on the plastic surface (Fig. 5A). We also found that metapristone-treatment resulted in decreased expression of mesenchymal marker “vimentin” and increased expression of epithelial marker “E-cadherin” in MDA-MB-231. Vimentin is a well-known metastasis marker and therapeutic target, as inhibiting vimentin function reduces the ability of cells to migrate32. Some anti-cancer drugs that are currently used in the clinic directly target vimentin such as “silibinin”33 and “withaferin A”34. One of the hallmarks of EMT is the functional loss of E-cadherin, which is thought to be a metastatic suppressor during tumor progression35. E-cadherin, encoded by the gene CDH1, is a transmembrane glycoprotein responsible for calcium-dependent cell-to-cell adhesion. E-cadherin plays a pivotal role in cadherin-catenin-cytoskeleton complexes, and it grants anti-invasive and anti-migratory properties to epithelial cells36,37. Our results suggest that metapristone inhibits cell migration, adhesion and invasion in highly metastatic human breast cancer cells, maybe in part, through the regulation of significant EMT-related markers which then leads to reversal of EMT.

Epithelial-to-mesenchymal transition, the process closely related to tumor development, is often regulated by a variety of signaling pathways and cytokines12,13,14,26. In this work, we performed KEGG pathway analysis based on the differential expressed proteins in MDA-MB-231 cells under metapristone-treated and untreated. We found some DEPs, including TGF β receptor I/II, Smad 2/3, RhoA, and Glutaredoxin, were related with Transforming Growth Factor β (TGF β) signaling pathway. TGF β signaling pathway has been characterized as an important inducer of EMT via several downstream signaling moleculars13. TGF β signals via formation of a heterotetrameric complex of TGF β receptor I/II (TGF β RI/RII), in which the active TGF β RII phosphorylates and activates the TGF β RI at the plasma membrane38,39. This conformational switch allows activated TGF β RI to interact with Smad2/3 through their MH2 domain. The activated type I receptor then propagates the signal to the nucleus by phosphorylating Smad 2 and Smad 3. Then, Smad2/3 can directly or indirectly regulate gene expression by controlling epigenetic processes, such as chromatin remodeling or by maintaining promoter DNA methylation, which is critical in silencing epithelial gene expression in cells that have undergone EMT40. Meanwhile, there exists a non-Smad pathway induced by TGF β41. In this non-Smad pathway, TGF β RII phosphorylates PAR6 (partitioning-defective protein 6), then inactivates the epithelial polarity complex, as well as activating of the small GTPase RhoA, which is contribute to cell invasion leading to breast cancer metastasis42. Furthermore, glutaredoxin (Grx), an anti-oxidant enzyme, was reported to play an important role in intervening TGF β-induced EMT process by reducing ROS generation in intracellular and suppressing the expression of mesenchymal markers43. Our results demonstrated that metapristone significantly inhibited the protein expression levels of TGF β RI/RII, RhoA, Smad 2/3, and up-regulated the expression level of glutaredoxin, implying that metapristone maybe in part, reverse EMT through attenuating TGFβ signaling pathway in MDA-MB-231 cells.

In addition to the TGF β signaling pathway, the Wnt signaling pathway also plays an important role in EMT44,45. Wnt pathway contributes to EMT by activating β-catenin, and then activating Snail, which in turn suppresses epithelial markers expression like E-cadherin45,46. Meanwhile, Caveolin-1 (CAV1), the principal structural protein of the cholesterol-rich plasma membrane invaginations, could induce EMT process through Wnt/β-catenin pathway to promote cancer metastasis47,48. Caveolin-1 is also an important regulator of cell polarity and directional movement49. The decreases in caveolin-1 expression follows classically described cellular changes associated with MET (including changes in cell morphology and expression of the E-cadherins and fibronectin)50. Our studies show that metapristone inhibits cell growth, and reverses EMT in conjunction with the activation of E-cadherin, and the inactivation of β-catenin and Caveolin-1 in MDA-MB-231 cells, implying that the MET potential of metapristone maybe related with Wnt signaling pathway.

In conclusion, our data show that metapristone inhibits migration, adhesion, and invasion abilities of the breast cancer cells. The pharmacoproteomic study reveals that metapristone intervenes EMT-related signaling pathways, such as TGF-β and Wnt signaling pathways, in conjunction with the activation of E-cadherin and glutaredoxin and inactivation of vimentin, TGFβ RI/RII, Smad2/3, RhoA, β-catenin and Caveolin-1 (Fig. 6). These findings imply that the application of metapristone is a possible new method to control EMT, which contributes to metastatic processes in breast cancer. Our results suggest that knowledge of the putative pharmacoproteomic mechanisms will promote better use of existing drugs and facilitate the conception of new therapies and new drug development.

Figure 6. The schematic representation represents the MET potential of metapristone in MDA-MB-231 cells.

Figure 6

Metapristone inhibits EMT by regulating TGF-β or Wnt signaling pathways. Metapristone inhibits EMT through Smad or non-Smad pathways involved in TGF-β signaling pathway, which results in suppression of mesenchymal and up-regulation of epithelial marker expression. Metapristone depressed EMT through regulating Wnt signaling pathway mediated by β-catenin and caveolin.

Materials and Methods

Cell culture, antibodies and reagents

MDA-MB-231 human breast cancer cells were purchased from American Type Culture Collection (ATCC, Manassas, VA) and maintained in ATCC-formulated Leibovitz’s L-15 Medium (Catalog No. 30-2008). Cells were supplemented with heat inactivated fetal bovine serum to a final concentration of 10%, and incubated at 37 °C in a free gas exchange with atmospheric air. Mouse monoclonal anti-vimentin (ab8978), -E-cadherin (ab1416), -β-actin antibodies (ab6276), goat anti-rabbit (ab150077) and goat anti-mouse (ab150115) antibodies were all obtained from Abcam Corporation.

In vitro cytotoxicity studies

The cytotoxicity of metapristone was investigated by the MTT assayas described previously by this lab51,52. Briefly, MDA-MB-231 cells were seeded into 96-well plates at a density of 1 × 104 cells/well, and then incubated at 37 °C in a humidified atmosphere with 100% air. After overnight incubation, the cells were treated with different concentrations of metapristone for 24 h. Culture medium was used as a blank control. Then, cells were incubation with the MTT solution (5 mg/ml) in the medium without phenol red and serum for another 4 h. The MTT-formazan formed by metabolically viable cells was dissolved in 150 μl of dimethyl sulfoxide (DMSO). Cell viability was determined by detecting the absorbance at 565 nm using an infinite M200 Pro microplate reader (Tecan, Switzerland). The absorbance of untreated cells was considered as 100%. Each sample was assayed in triplicate in three independent experiments.

Wound healing assay

Migration of MDA-MB-231 cells was investigated in the in vitro wound-healing assay as described previously by this lab3,51. The MDA-MB-231 cells were seeded in 6-well plate; once confluent, 10 μg/ml mitomycin C was added. The scratch wound was generated in the surface of the plate using a pipette tip, followed by extensive washing with serum-free medium to remove cell debris. DMSO (final concentration: 0.1%) as vehicle control was added after wounding. Cells were then cultured and allowed to migrate into the wound area for up to 24 h at 37 °C. At indicated time points, motility was quantified by measuring the average extent of wound closure. Each well was counted under a light microscope (Zeiss, Germany) at a magnification of 10 × and then photographed.

Cell invasion assay

Cell invasion assay was performed using 24-well transwells (Costar, Coring Incorporated, USA), which allows cells to migrate through a polycarbonate membrane with 8-μm pore size as we described previously52,53. Briefly, in transwell cell culture chambers, filters of 8 mm pore size were coated with Matrigel on the upper surface. MDA-MB-231 cells were resuspended with reduced serum L-15 medium and seeded 5 × 104 per well on the upper chamber of the transwell apparatus. Invasion assay was performed in the presence of 0, 10, 50, 75 μM of metapristone. DMSO (final concentration: 0.1%) was used as vehicle control. After 24 h incubation, the cells on the inner layer were softly removed with a cotton swab. Then, the adherent cells on undersurface of the insert were fixed in methanol and stained with 0.1% crystal violet for 20 min. The filters were washed with PBS and images were taken by a light microscope (Zeiss, Germany) at × 200 magnification. Five fields were counted per filter in each group and the experiment was conducted in triplicate.

Cell adhesion assay

The adhesion assay of MDA-MB-231 cells to the HUVECs was assessed according to the method described previously by this lab with minor modifications3,52. Briefly, Human umbilical vein endothelial cells (HUVECs) were isolated and utilized between passages 2 and 5, and grown to confluence in 24-well culture plates. Then, TNF-α (final concentration: 10 ng/ml) was used to activate HUVECs for 4 hours. Rhodamine 123-labled MDA-MB-231 cells were co-cultured with the HUVEC monlayers in each well, followed by treatment with metapristone for 1 hour. DMSO (0.1%) was used as the vehicle control. The nonadherent cells were removed from the plate by careful three-time washings with PBS, and the MDA-MB-231 cells bound to the HUVECs were measured by a fluorescence microscope (Zeiss, Germany). Then, ten visual fields for each well were selected randomly and taken pictures. Mean inhibition of adhesion for 10 visual fields was calculated by using the equation: % of control adhesion = [the number of adhered cells in treated group/the number of adhered cells in the control group] × 100%.

Protein preparation and iTRAQ labeling

MDA-MB-231 cells were cultured and treated with 50 μM metapristone. Treated and untreated cells were suspended in the Lysis buffer (7 M Urea, 2 M Thiourea, 4% CHAPS, 40 mM Tris-HCl, pH8.5, 1 mM PMSF, 2 mM EDTA) and sonicated in ice. The proteins were reduced with 10 mM DTT (final concentration) at 56°C for 1 h and then alkylated by 55 mM IAM (final concentration) in the darkroom for 1 h. The reduced and alkylated protein mixtures were precipitated by adding 4 × volume of chilled acetone at −20 °C overnight. After centrifugation at 4 °C, 30000 g, the pellet was dissolved in 0.5 M TEAB (Applied Biosystems, Milan, Italy) and sonicated in ice. After centrifuging at 30000 g at 4°C, an aliquot of the supernatant was taken and protein concentration was determined using the Bradford method. Then, total protein (100 μg) of each sample was digested with Trypsin Gold (Promega, Madison, WI, USA) with the ratio of protein: trypsin = 30:1 at 37°C for 16 hours. After trypsin digestion, peptides were dried by vacuum centrifugation, reconstituted in 0.5 M TEAB, and processed according to the manufacture’s protocol for 8-plex iTRAQ reagent (Applied Biosystems, Foster City,CA). The labeled peptide mixtures were pooled and dried by vacuum centrifugation, and then fractionated using Poly SULFOETHYL ATM SCX column (200 × 4.6 mm, 5 μm particle size, 200 A° pore size) by HPLC system (Shimadzu, Japan) at flow rate 1.0 ml min-1. The eluted peptides were pooled into 20 fractions, desalted with a Strata × C 18 column, concentrated to dryness using vacuum centrifuge and then reconstituted in 0.1% formic acid for LC-MS/MS analysis.

LC-ESI-MS/MS analysis based on Q EXACTIVE

The mass spectroscopy analysis was performed using a tandem mass spectrometry (MS/MS) in an Q EXACTIVE (Thermo Fisher Scientific, San Jose, CA) coupled online to the HPLC as described before54,55. Peptides were selected for MS/MS using high-energy collision dissociation (HCD) operating mode with a normalized collision energy setting of 27.0; ion fragments were detected in the Orbitrap at a resolution of 17500. A data-dependent procedure that alternated between one MS scan followed by 15 MS/MS scan with a following Dynamic Exclusion duration of 15s. Proteins identification was performed by using Mascot search engine (Matrix Science, London, UK; version 2.3.02). For protein quantitation, it was required that a protein contains at least two unique peptides. The quantitative protein ratios were weighted and normalized by the median ratio in Mascot. We only used ratios with p-values < 0.05, and only fold changes of >1.5 were considered as significant.

Proteomic data analysis

Functional annotations of the proteins were conducted using Blast2GO program against the non-redundant protein database (NR; NCBI). The keg database (http:www.genome.jp/keg/), the COG database (http://www.ncbi.nlm.nih.gov/COG/), and GO (Gene Ontology) analyses (http://www.geneontology.org) were used to classify and group these identified proteins according to the methods reported in early literature15,56.

Western blot analysis

Cell lysates were collected using radio immunoprecipitation (RIPA) lysis buffer, supplemented with HALT protease and phosphatase inhibitor cocktail (Thermo Scientific), and immunodetection of electrophoresis-resolved proteins was performed using standard protocols. The E-Cadherin, vimentin, Smad2, pSmad2, β-catenin, and β-actin antibodies were from Abcam. Immunodetection was accomplished using enhanced chemiluminescence, and data were acquired with a quantitative digital imaging system (Quantity One, Bio-Rad) allowing it to check for saturation. Overall emitted photons were quantified for each band, particularly for homogeneously the loading controls.

Immunofluorescence staining and high-content confocal imaging

MDA-MB-231 cells were cultured on a 35 mm cell culture dish (NEST, GBD-35-20) to 50% confluence at least 2 days before carrying out the immunofluorescence assay. Cells (with or without metapristone-treatment) were first washed by phosphate buffer 3 times and then fixed by 4% paraformaldehyde for 30 minutes. One milliliter of 0.1% Triton-X-100 was subsequently added to culture cells for ten minutes to increase cell permeability. Cells were blocked for 30 minutes at room temperature with 10% goat serum followed by culturing with primary antibodies, mouse monoclonal anti-vimentin antibody and E-cadherin for 1 h at room temperature. Then cells were added with secondary antibodies, Goat Anti-Mouse IgG-FITC antibody (Boster, BA1101) and Goat Anti-Rabbit IgG-CY3 antibody (Boster, BA1032) respectively, and cultured in the dark for 1 h at room temperature. Phosphate buffer was used to wash cells for at least three times between every two contiguous steps. Confocal analysis was performed on a Leica-TCS-SP8 confocal microscope and the images were taken under the same parameter configuration.

Statistical analysis

All data were analyzed using SASS software and expressed as the mean ± SD or SE. Statistical comparisons between different groups were performed using Student t-test. A P value of <0.05 was considered to be statistically significant.

Additional Information

How to cite this article: Yu, S. et al. Pharmacoproteomic analysis reveals that metapristone (RU486 metabolite) intervenes E-cadherin and vimentin to realize cancer metastasis chemoprevention. Sci. Rep. 6, 22388; doi: 10.1038/srep22388 (2016).

Acknowledgments

This work was supported by the National Natural Science Foundation of China (NSFC) (No. 81502617, 81273548), Fujian Development and Reform Commission (2014/168), and Fujian Science and Technology plan project (2015Y0071).

Footnotes

Author Contributions S.Y. and L.J. conceived and designed the experiments. C.Y. and X.Y. performed cell culture experiments and carried out the iTRAQ analysis. S.H. and Z.T. carried out the cell migration and adhesion experiments, J.L. performed the western blot experiments, C.Q., C.H. and Y.L. acquired and analyzed the experimental data. L.J. and S.Y. wrote the manuscript. All authors reviewed the manuscript.

References

  1. Siegel R., Ma J., Zou Z. & Jemal A. Cancer statistics, 2014. CA Cancer J Clin 64, 9–29 (2014). [DOI] [PubMed] [Google Scholar]
  2. Jemal A. et al. Global cancer statistics. CA Cancer J Clin 61, 69–90 (2011). [DOI] [PubMed] [Google Scholar]
  3. Wang J. et al. Synthesis, spectral characterization, and in vitro cellular activities of metapristone, a potential cancer metastatic chemopreventive agent derived from mifepristone (RU486). AAPS J 16, 289–298 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Chen J. Z. et al. A novel UPLC/MS/MS method for rapid determination of metapristone in rat plasma, a new cancer metastasis chemopreventive agent derived from mifepristone (RU486). J Pharm Biomed Anal 95, 158–163 (2014). [DOI] [PubMed] [Google Scholar]
  5. Chen J. et al. The unique pharmacological characteristics of mifepristone (RU486): from terminating pregnancy to preventing cancer metastasis. Med Res Rev 34, 979–1000 (2014). [DOI] [PubMed] [Google Scholar]
  6. Jia L., Coward L., Gorman G. S., Noker P. E. & Tomaszewski J. E. Pharmacoproteomic effects of isoniazid, ethambutol, and N-geranyl-N’-(2-adamantyl)ethane-1,2-diamine (SQ109) on Mycobacterium tuberculosis H37Rv. J Pharmacol Exp Ther 315, 905–911 (2005). [DOI] [PubMed] [Google Scholar]
  7. Chahrour O., Cobice D. & Malone J. Stable isotope labelling methods in mass spectrometry-based quantitative proteomics. J Pharm Biomed Anal 113, 2–20 (2015). [DOI] [PubMed] [Google Scholar]
  8. Gan C. S., Chong P. K., Pham T. K. & Wright P. C. Technical, experimental, and biological variations in isobaric tags for relative and absolute quantitation (iTRAQ). J Proteome Res 6, 821–827 (2007). [DOI] [PubMed] [Google Scholar]
  9. Martinez-Esteso M. J., Casado-Vela J., Selles-Marchart S., Pedreno M. A. & Bru-Martinez R. Differential plant proteome analysis by isobaric tags for relative and absolute quantitation (iTRAQ). Methods Mol Biol 1072, 155–169 (2014). [DOI] [PubMed] [Google Scholar]
  10. Li L. & Li W. Epithelial-mesenchymal transition in human cancer: comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol Ther 150, 33–46 (2015). [DOI] [PubMed] [Google Scholar]
  11. Mitra A., Mishra L. & Li S. EMT, CTCs and CSCs in tumor relapse and drug-resistance. Oncotarget 6, 10697–10711 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chen Z., Shao Y. & Li X. The roles of signaling pathways in epithelial-to-mesenchymal transition of PVR. Mol Vis 21, 706–710 (2015). [PMC free article] [PubMed] [Google Scholar]
  13. Papageorgis P. TGFbeta Signaling in Tumor Initiation, Epithelial-to-Mesenchymal Transition, and Metastasis. J Oncol 2015, 587193 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Gonzalez D. M. & Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal 7, re8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bauer S., Robinson P. N. & Gagneur J. Model-based gene set analysis for Bioconductor. Bioinformatics 27, 1882–1883 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Yin W. et al. Efficacy and safety of capecitabine-based first-line chemotherapy in advanced or metastatic breast cancer: a meta-analysis of randomised controlled trials. Oncotarget 6, 39365–39372 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Mustacchi G. & De Laurentiis M. The role of taxanes in triple-negative breast cancer: literature review. Drug Des Devel Ther 9, 4303–4318 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Masters G. A. et al. Clinical cancer advances 2015: Annual report on progress against cancer from the American Society of Clinical Oncology. J Clin Oncol 33, 786–809 (2015). [DOI] [PubMed] [Google Scholar]
  19. Wargon V. et al. Progestin and antiprogestin responsiveness in breast cancer is driven by the PRA/PRB ratio via AIB1 or SMRT recruitment to the CCND1 and MYC promoters. Int J Cancer 136, 2680–2692 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Clark A. G. & Vignjevic D. M. Modes of cancer cell invasion and the role of the microenvironment. Curr Opin Cell Biol 36, 13–22 (2015). [DOI] [PubMed] [Google Scholar]
  21. Herbertz S. et al. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 9, 4479–4499 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Lai Y. H. et al. Rhodomycin A, a novel Src-targeted compound, can suppress lung cancer cell progression via modulating Src-related pathways. Oncotarget 6, 26252–26265, (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Yates C. M., McGettrick H. M., Nash G. B. & Rainger G. E. Adhesion of tumor cells to matrices and endothelium. Methods Mol Biol 1070, 57–75, (2014). [DOI] [PubMed] [Google Scholar]
  24. Roumeliotis T. I. et al. Pharmacoproteomic study of the natural product Ebenfuran III in DU-145 prostate cancer cells: the quantitative and temporal interrogation of chemically induced cell death at the protein level. J Proteome Res 12, 1591–1603 (2013). [DOI] [PubMed] [Google Scholar]
  25. Bang J. Y. et al. Pharmacoproteomic analysis of a novel cell-permeable peptide inhibitor of tumor-induced angiogenesis. Mol Cell Proteomics : MCP 10, M110 005264 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Saitoh M. Epithelial-mesenchymal transition is regulated at post-transcriptional levels by transforming growth factor-beta signaling during tumor progression. Cancer Sci 106, 481–488 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Pasquier J., Abu-Kaoud N., Al Thani H. & Rafii A. Epithelial to Mesenchymal Transition in a Clinical Perspective. J Oncol 2015, 792182 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Zhou L., Shi L., Guo H. & Yao X. MYSM-1 suppresses migration and invasion in renal carcinoma through inhibiting epithelial-mesenchymal transition. Tumour Biol, doi: 10.1007/s13277-015-4138-z (2015). [DOI] [PubMed] [Google Scholar]
  29. Lee J. M., Dedhar S., Kalluri R. & Thompson E. W. The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol 172, 973–981 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Barriere G. et al. Circulating tumor cells and epithelial, mesenchymal and stemness markers: characterization of cell subpopulations. Ann Transl Med 2, 109 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Mimeault M. & Batra S. K. Molecular biomarkers of cancer stem/progenitor cells associated with progression, metastases, and treatment resistance of aggressive cancers. Cancer Epidemiol Biomarkers Prev 23, 234–254 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Vuoriluoto K. et al. Vimentin regulates EMT induction by Slug and oncogenic H-Ras and migration by governing Axl expression in breast cancer. Oncogene 30, 1436–1448 (2011). [DOI] [PubMed] [Google Scholar]
  33. Wu K. J. et al. Silibinin inhibits prostate cancer invasion, motility and migration by suppressing vimentin and MMP-2 expression. Acta Pharmacol Sin 30, 1162–1168 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Challa A. A., Vukmirovic M., Blackmon J. & Stefanovic B. Withaferin-A reduces type I collagen expression in vitro and inhibits development of myocardial fibrosis in vivo. PloS one 7, e42989 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Pecina-Slaus N. Tumor suppressor gene E-cadherin and its role in normal and malignant cells. Cancer Cell Int 3, 17 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Buckley C. D. et al. Cell adhesion. The minimal cadherin-catenin complex binds to actin filaments under force. Science 346, 1254211 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Semina E. V. et al. Novel mechanism regulating endothelial permeability via T-cadherin-dependent VE-cadherin phosphorylation and clathrin-mediated endocytosis. Mol Cell Biochem 387, 39–53 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Forbes K., Souquet B., Garside R., Aplin J. D. & Westwood M. Transforming growth factor-{beta} (TGF{beta}) receptors I/II differentially regulate TGF{beta}1 and IGF-binding protein-3 mitogenic effects in the human placenta. Endocrinology 151, 1723–1731 (2010). [DOI] [PubMed] [Google Scholar]
  39. Melisi D. et al. LY2109761, a novel transforming growth factor beta receptor type I and type II dual inhibitor, as a therapeutic approach to suppressing pancreatic cancer metastasis. Mol Cancer Ther 7, 829–840 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Qiu X., Cheng J. C., Zhao J., Chang H. M. & Leung P. C. Transforming growth factor-beta stimulates human ovarian cancer cell migration by up-regulating connexin43 expression via Smad2/3 signaling. Cell Signal 27, 1956–1962 (2015). [DOI] [PubMed] [Google Scholar]
  41. Fazilaty H., Gardaneh M., Bahrami T., Salmaninejad A. & Behnam B. Crosstalk between breast cancer stem cells and metastatic niche: emerging molecular metastasis pathway? Tumour Biol 34, 2019–2030 (2013). [DOI] [PubMed] [Google Scholar]
  42. Fleming Y. M. et al. TGF-beta-mediated activation of RhoA signalling is required for efficient (V12)HaRas and (V600E)BRAF transformation. Oncogene 28, 983–993 (2009). [DOI] [PubMed] [Google Scholar]
  43. Lee E. K. et al. Decreased expression of glutaredoxin 1 is required for transforming growth factor-beta1-mediated epithelial-mesenchymal transition of EpRas mammary epithelial cells. Biochem Biophys Res Commun 391, 1021–1027 (2010). [DOI] [PubMed] [Google Scholar]
  44. Qi J. et al. New Wnt/beta-catenin target genes promote experimental metastasis and migration of colorectal cancer cells through different signals. Gut, doi: 10.1136/gutjnl-2014-307900 (2015). [DOI] [PubMed] [Google Scholar]
  45. Ghahhari N. M. & Babashah S. Interplay between microRNAs and WNT/beta-catenin signalling pathway regulates epithelial-mesenchymal transition in cancer. Eur J Cancer 51, 1638–1649 (2015). [DOI] [PubMed] [Google Scholar]
  46. Gnemmi V. et al. MUC1 drives epithelial-mesenchymal transition in renal carcinoma through Wnt/beta-catenin pathway and interaction with SNAIL promoter. Cancer Lett 346, 225–236 (2014). [DOI] [PubMed] [Google Scholar]
  47. Hwangbo C. et al. Syntenin regulates TGF-beta1-induced Smad activation and the epithelial-to-mesenchymal transition by inhibiting caveolin-mediated TGF-beta type I receptor internalization. Oncogene, doi: 10.1038/onc.2015.100 (2015). [DOI] [PubMed] [Google Scholar]
  48. Kannan A. et al. Caveolin-1 promotes gastric cancer progression by up-regulating epithelial to mesenchymal transition by crosstalk of signalling mechanisms under hypoxic condition. Eur J Cancer 50, 204–215 (2014). [DOI] [PubMed] [Google Scholar]
  49. Gai X., Lu Z., Tu K., Liang Z. & Zheng X. Caveolin-1 is up-regulated by GLI1 and contributes to GLI1-driven EMT in hepatocellular carcinoma. PloS one 9, e84551 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Chanvorachote P., Pongrakhananon V. & Halim H. Caveolin-1 regulates metastatic behaviors of anoikis resistant lung cancer cells. Mol Cell Biochem 399, 291–302 (2015). [DOI] [PubMed] [Google Scholar]
  51. Lu Y. et al. Nitric oxide inhibits hetero-adhesion of cancer cells to endothelial cells: restraining circulating tumor cells from initiating metastatic cascade. Sci Rep 4, 4344 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Yu S. et al. Systems pharmacology of mifepristone (RU486) reveals its 47 hub targets and network: comprehensive analysis and pharmacological focus on FAK-Src-Paxillin complex. Sci Rep 5, 7830 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Jiang Z. et al. Bioactivity-guided fast screen and identification of cancer metastasis chemopreventive components from raw extracts of Murraya exotica. J Pharm Biomed Anal 107, 341–345 (2015). [DOI] [PubMed] [Google Scholar]
  54. Nie J. et al. Comparative analysis of dynamic proteomic profiles between in vivo and in vitro produced mouse embryos during postimplantation period. J Proteome Res 12, 3843–3856 (2013). [DOI] [PubMed] [Google Scholar]
  55. Rubert J., Lacina O., Fauhl-Hassek C. & Hajslova J. Metabolic fingerprinting based on high-resolution tandem mass spectrometry: a reliable tool for wine authentication? Anal Bioanal Chem 406, 6791–6803 (2014). [DOI] [PubMed] [Google Scholar]
  56. Dahlquist K. D., Salomonis N., Vranizan K., Lawlor S. C. & Conklin B. R. GenMAPP, a new tool for viewing and analyzing microarray data on biological pathways. Nat Genet 31, 19–20 (2002). [DOI] [PubMed] [Google Scholar]

Articles from Scientific Reports are provided here courtesy of Nature Publishing Group

RESOURCES