Skip to main content
OncoTargets and Therapy logoLink to OncoTargets and Therapy
. 2015 Jul 6;8:1651–1670. doi: 10.2147/OTT.S82718

FH535 inhibited metastasis and growth of pancreatic cancer cells

Meng-Yao Wu 1,*, Rong-Rui Liang 1,*, Kai Chen 1, Meng Shen 1, Ya-Li Tian 1,2, Dao-Ming Li 1, Wei-Ming Duan 1, Qi Gui 1, Fei-Ran Gong 3, Lian Lian 1,2, Wei Li 1,6,, Min Tao 1,4,5,6,
PMCID: PMC4500609  PMID: 26185454

Abstract

FH535 is a small-molecule inhibitor of the Wnt/β-catenin signaling pathway, which a substantial body of evidence has proven is activated in various cancers, including pancreatic cancer. Activation of the Wnt/β-catenin pathway plays an important role in tumor progression and metastasis. We investigated the inhibitory effect of FH535 on the metastasis and growth of pancreatic cancer cells. Western blotting and luciferase reporter gene assay indicated that FH535 markedly inhibited Wnt/β-catenin pathway viability in pancreatic cancer cells. In vitro wound healing, invasion, and adhesion assays revealed that FH535 significantly inhibited pancreatic cancer cell metastasis. We also observed the inhibitory effect of FH535 on pancreatic cancer cell growth via the tetrazolium and plate clone formation assays. Microarray analyses suggested that changes in the expression of multiple genes could be involved in the anti-cancer effect of FH535 on pancreatic cancer cells. Our results indicate for the first time that FH535 inhibits pancreatic cancer cell metastasis and growth, providing new insight into therapy of pancreatic cancer.

Keywords: pancreatic cancer, FH535, β-catenin, metastasis, growth

Introduction

Pancreatic cancer is one of the most aggressive human malignancies worldwide. Despite improvements in surgical and chemotherapeutic approaches over the past decades, the prognosis of pancreatic cancer remains dismal; the average overall 5-year survival rate is <5%.1 The reasons for this are the challenges associated with diagnosis, which tends to be late and uncertain; more importantly, therapeutic options are limited. Even with early diagnosis and surgical resection with curative intention, nearly all patients develop local recurrence or distant metastases following surgery and eventually succumb to the debilitating effects of metastatic growth.2,3 Conventional chemotherapy is rarely curative for metastatic pancreatic cancer. In recent years, there have been important advances in the organization of care for patients with pancreatic cancer; these advances have also resulted in more focused studies on surgical, oncological, and immunological treatment.

The Wnt/β-catenin pathway is a genetically conserved signaling pathway associated with a variety of human conditions such as birth defects and tumors. Abnormal Wnt/β-catenin pathway activation is closely related to the development of many cancers.4,5 An increasing amount of evidence demonstrates that both the β-catenin-dependent (canonical) and β-catenin-independent (non-canonical) Wnt signaling pathways play a key role in regulating pathological processes by facilitating tumor growth, migration, and invasion. In canonical Wnt signaling, glycogen synthase kinase-3β (GSK-3β) phosphorylates β-catenin at certain key residues, leading to its ubiquitination and subsequent degradation.5,6 Non-phosphorylated β-catenin accumulates in the cytoplasm, and pathway activation leads to nuclear accumulation of β-catenin and interaction with T-cell factor (TCF) transcription factors, subsequently stimulating the downstream target genes, which include the genes participating in cell metastasis and proliferation.7,8

Abnormal Wnt/β-catenin pathway activation plays an important role in human pancreatic cancer, where it causes extracellular matrix degradation and uncontrolled cell proliferation and differentiation.9 Recent studies have demonstrated that FH535 is a synthetic inhibitor of the canonical Wnt signaling pathway; it inhibits the growth of colon, lung, breast, and hepatocellular carcinoma lines,10,11 suggesting that small-molecule targeting of the Wnt/β-catenin pathway could be a promising therapeutic approach for cancers in which this pathway is activated.

In this study, we investigated the anti-cancer effect of FH535 on pancreatic cancer and explored the mechanisms underlying the effect, providing a rationale for further development of FH535 as a promising therapeutic agent for treating pancreatic cancer.

Materials and methods

Cell cultures and reagents

The human pancreatic cancer cell lines PANC-1 and BxPC-3 were purchased from American Type Culture Collection (ATCC) (Manassas, VA, USA). The cells were maintained in Dulbecco’s Modified Eagle’s Medium (DMEM; Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal calf serum (FCS), 100 U/mL penicillin, and 100 μg/mL streptomycin (Thermo Fisher Scientific) at 37°C in a 5% CO2 incubator under a humidified atmosphere; the cells were passaged every 2–3 days for exponential growth. FH535 was purchased from EMD Millipore (Billerica, MA, USA).

Western blotting

Total protein was extracted using a lysis buffer (50 mM Tris-HCl [pH 7.4], 150 mM NaCl, 1% Triton X-100, 0.1% sodium dodecyl sulfate [SDS], 1 mM EDTA) supplemented with a protease inhibitor cocktail kit and a phosphatase inhibitor cocktail kit (Hoffman-La Roche Ltd., Basel, Switzerland). The protein extracts were loaded, size-fractionated by SDS-polyacrylamide gel electrophoresis, and transferred to polyvinylidene difluoride membranes (Bio-Rad Laboratories Inc., Hercules, CA, USA). After blocking, the membranes were incubated with the primary antibodies mouse anti-β-catenin (Santa Cruz Biotechnology Inc., Dallas, TX, USA) and rabbit anti-β-actin (Proteintech Group Inc., Chicago, IL, USA) at 4°C overnight. Protein expression was determined using horseradish peroxidase-conjugated anti-mouse or anti-rabbit secondary antibodies, followed by detection using enhanced chemiluminescence (EMD Millipore). Band intensity was visualized using a JS-1035 image analysis scanning system (Shanghai Peiqing Science & Technology, Co., Ltd., Shanghai, People’s Republic of China).

Luciferase reporter assay

β-catenin is a dominant factor in the Wnt/β-catenin/TCF signaling pathway, which regulates gene transcription by binding β-catenin and TCF. The activity of this final step in the pathway can be precisely measured using a luciferase reporter construct. The reporter plasmid pTOPFLASH (TCF reporter plasmid; EMD Millipore) contains two sets (the second set is in the reverse orientation) of three copies of the TCF binding site (wild-type) upstream of the thymidine kinase minimal promoter and luciferase open reading frame. The internal control plasmid pRL-SV40 (Promega Corporation, Fitchburg, WI, USA) contains the Renilla luciferase gene. Cells were transiently cotransfected with pTOPFLASH plasmid (500 ng/well) and pRL-SV40 plasmid (100 ng/well) for 6 hours using Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer’s protocol. Then, the medium was renewed and FH535 was added. After 24 hours of treatment, cell lysates were subjected to the dual luciferase reporter assay according to the manufacturer’s recommendations; luciferase activity was measured using a luminometer (Turner Designs, Sunnyvale, CA, USA). The results are expressed as relative luciferase activity, ie, the ratio of firefly luciferase activity over Renilla luciferase activity.

Wound healing assay

Cells (1×104/well) were seeded in 96-well plates and grown to confluence. The monolayer culture was artificially scrape wounded with a sterile micropipette tip to create a denuded zone of constant width. Each well was washed with phosphate-buffered saline twice to remove the detached cells before FH535 treatment. Cell migration to the wounded region was observed using an XDS-1B inverted microscope (MIC Optical and Electrical Instrument, Chongqing, People’s Republic of China) and photographed (×40 magnification). Images were captured at 0, 8, and 12 hours to monitor the wound healing process. The wound areas were measured using ImageJ (NIH, Bethesda, MA, USA).

Transwell invasion assay

We used a 24-well Transwell plate with an 8 μm pore size polycarbonate filter membrane (Corning Incorporated, Corning, NY, USA). Cells (1×105) in 100 μL serum-free DMEM were added to the Matrigel-coated top chamber (BD Biosciences, San Jose, CA, USA); the bottom chamber contained DMEM with 10% FCS. The cells were incubated for 24 hours; cells that had invaded through the Matrigel-coated membrane were fixed and stained with crystal violet and counted under a light microscope in five random fields in a blinded fashion.

Adhesion assay

Cells were resuspended in complete medium and seeded in 24-well plates at 1×104 cells/mL. After 5-hour incubation, the unattached cells were removed to another well. The attached and unattached cells were evaluated using the 3-[4,5-dimethylthiazol-2-yl] 2,5-diphenyltetrazolium bromide (MTT) assay. The adhesion rate was calculated as follows: (absorbance of attached cells/[absorbance of attached cells + absorbance of unattached cells]) ×100%.

MTT assay

Cell growth was evaluated using the MTT assay. Cells (5×104/well) were seeded in 24-well tissue culture plates. Blank control was treated with DMSO. After FH535 treatment, MTT (Sigma-Aldrich Co., St Louis, MO, USA) was added to each well (final concentration, 0.5 mg/mL), followed by 4-hour incubation at 37°C. The medium was removed, and 800 μL of dimethyl sulfoxide was added to each well. The absorbance of the mixture was measured at 490 nm using a microplate enzyme-linked immunosorbent assay reader (Bio-Rad Laboratories Inc.). The relative cell viability was calculated as follows: relative cell viability = (mean experimental absorbance/mean control absorbance) ×100%.

Plate clone formation assay

Cells (200/well) were seeded in 24-well plates and treated after 12 hours. After 15 days, the cells were stained with 1% methylrosanilinium chloride, and the number of visible colonies was counted. The relative clone formation ability was calculated as follows: (mean experimental clone number/mean control clone number) ×100%.

Cell cycle analysis

Before treatment, the cells were serum starved for 24 hours to synchronize the cell cycle. Then, FCS was added to the cells, followed by various concentrations of FH535. Following 24 hours of FH535 treatment, the cells were fixed in 80% cooled ethanol and incubated with 0.5% Triton X-100 solution containing 1 mg/mL RNase A at 37°C for 30 minutes. Next, propidium iodide (Sigma-Aldrich Co.) was added to the wells (final concentration, 50 μg/mL), followed by 30-minute incubation in the dark. Cellular DNA content was analyzed using a fluorescence-activated cell sorter (Becton Dickinson, Franklin Lakes, NJ, USA). Data were processed using ModFit LT software (Verity Software House, Topsham, ME, USA).

Microarray assay

Sample preparation and processing were performed as described in the GeneChip Expression Analysis Manual (Agilent Technologies, Santa Clara, CA, USA). Differentially expressed genes were screened using Agilent 44K human whole-genome oligonucleotide microarrays. The selection criterion was greater than twofold difference in expression (difference in upregulated expression was greater than twofold; difference in downregulated expression was less than 0.5-fold). Hierarchical clustering of samples was performed using an average linkage algorithm using TIGR MultiExperiment Viewer (The Institute for Genomic Research, Rockville, MD, USA).

Statistical analysis

Each experiment was performed in at least triplicate. Results are expressed as the mean ± standard deviation. Statistical analysis was performed using an unpaired Student’s t-test. P<0.05 was considered significant.

Results

FH535 inhibited the β-catenin pathway in pancreatic cancer cells

Treatment with 20 μM FH53512 did not affect nuclear or total β-catenin expression in the BxPC-3 cells, but downregulated nuclear and total β-catenin in the PANC-1 cells (Figure 1A). The luciferase reporter assay confirmed that FH535 suppressed TCF-dependent transcription, which may have led to dysregulation of the genes downstream of the β-catenin pathway (Figure 1B). To verify this, we performed microarray analyses to determine the mRNA expression changes in 138 genes downstream of the β-catenin pathway using Agilent 44K human whole-genome oligonucleotide microarrays (http://www.stanford.edu/group/nusselab/cgi-bin/wnt/target_genes); 20 μM FH535 upregulated or downregulated multiple genes (Figure 1C, Table 1).

Figure 1.

Figure 1

Figure 1

FH535 suppressed the Wnt/β-catenin pathway in pancreatic cancer cells.

Notes: (A) Time-dependent decrease by FH535 of nuclear and total β-catenin protein levels in PANC-1 cells; FH535 did not affect nuclear or total β-catenin expression in BxPC-3 cells. (B) Dose-dependent decrease by FH535 of TCF-dependent transcription. **P<0.01, significant differences vs the respective control groups. (C) Microarray analysis of expression regulation of genes downstream of the Wnt/β-catenin pathway upon 20 μM FH535 treatment. Up and down arrows indicate gene expression significantly upregulated or downregulated, respectively, by twofold.

Abbreviations: TCF, T-cell factor; h, hours.

Table 1.

Microarray analysis of expression regulation of genes downstream of the Wnt/β-catenin pathway upon 20 μM FH535 treatment

Gene ID Normalized intensity
Control FH535
MYC 4609 16.268158 15.204586
KRT18 3875 15.975001 16.022995
PTTG1 9232 15.680945 15.73604
ANGPTL4 51129 15.190848 15.278334
KRT81 3887 15.0413 14.423697
CD44 960 15.006962 16.199093
PFDN5 5204 14.879261 14.964103
KRT10 3858 14.799751 13.889791
PTTG2 10744 14.772796 14.547727
BIRC5 332 14.757564 14.219355
PTTG1IP 754 14.684395 14.533192
VEGFB 7423 14.498004 13.671163
CYR61 3491 14.279853 14.790296
UBXN1 51035 14.231482 14.049252
KRT7 3855 14.184294 13.285099
WISP2 8839 13.732449 12.493675
SOX9 6662 13.574989 13.415171
EN2 2020 13.393019 12.721889
JAG1 182 12.427784 12.687155
FOSL1 8061 12.344017 11.102832
MYCBP 26292 12.284651 11.974781
SNAI1 6615 12.28132 10.385736
KRT73 319101 12.23975 11.053284
GJA1 2697 12.226766 13.521647
IRX3 79191 12.224495 12.16053
TBX1 6899 12.181493 12.043698
DKK3 27122 12.076692 10.961267
JUN 3725 12.038464 12.673436
MSL1 339287 11.920114 11.438548
KRT80 144501 11.87818 12.039767
CCND3 896 11.576098 10.075832
CDKN2A 1029 11.343829 11.097562
EFNB1 1947 11.337793 10.368351
PTTG3P 26255 11.33311 10.750982
KRT83 3889 11.319811 9.89329
KRT19 3880 11.289505 11.101922
CEBPD 1052 11.196305 10.068165
PPARD 5467 11.19087 10.731722
ANTXR1 84168 11.149265 12.122571
EGFR 1956 11.122326 12.333595
CDX4 1046 10.933424 9.588729
ISLR 3671 10.854443 9.725897
TWIST2 117581 10.853075 10.129753
VEGFA 7422 10.833399 10.087871
CTGF 1490 10.809845 11.98555
FZD7 8324 10.711324 9.901575
KRT85 3891 10.621079 9.606193
CCND1 595 10.543621 10.14267
TLE1 7088 10.343489 9.271956
CDX1 1044 10.329419 9.136879
KRT8 3856 10.267347 8.319682
NRP1 8829 10.246916 9.627893
DKK1 22943 10.211538 10.979951
IRS1 3667 10.175792 10.3609915
MMP2 4313 10.153262 9.412593
IKBKG 8517 10.132635 9.295626
TIAM1 7074 10.117085 11.627998
EGR1 1958 10.007974 8.180263
MET 4233 9.986441 12.74971
BGLAP 632 9.971276 9.414629
VEGFC 7424 9.923567 10.21814
AHR 196 9.886938 11.936481
CACNA1G 8913 9.812038 8.560494
KRT15 3866 9.7214575 8.709181
PPAP2B 8613 9.718731 9.956581
KRT86 3892 9.707824 9.012362
KRT23 25984 9.573925 7.977122
GBX2 2637 9.409858 9.626151
KRT39 390792 9.284486 7.929165
WNT3A 89780 9.275467 8.68014
PLAUR 5329 9.265003 8.37788
ID2 3398 9.226584 8.999163
MAEA 10296 9.087043 8.91366
DAB2 1601 9.034534 10.517419
ETS2 2114 8.999426 10.445461
KRT31 3881 8.998071 7.96933
TNFRSF11A 8792 8.943393 9.029845
RET 5979 8.9224615 7.809108
UBXN6 80700 8.850218 7.6966906
STRA6 64220 8.746183 7.1663184
KLF5 688 8.6543455 8.714795
KRT4 3851 8.640165 7.6698284
UBXN4 23190 8.607909 10.505396
LEF1 51176 8.601926 9.380911
KRT76 51350 8.571253 7.397269
UBXN2B 137886 8.2286415 9.982969
UBXN11 91544 8.179481 7.272692
LRP1 4035 8.175423 6.988433
UBXN2A 165324 8.133479 7.9562063
KRT9 3857 8.110517 7.0731263
EDA 1896 8.09645 7.4072337
KRT32 3882 8.087603 7.7537346
FGF4 2249 7.9492774 6.5977035
KRT3 3850 7.8908534 8.469248
UBXN8 7993 7.86574 9.013798
SIX1 6495 7.818405 7.9264607
FOXN1 8456 7.7998743 6.8640747
ETV6 2120 7.7085342 7.0067773
KRT1 3848 7.5221066 6.7497764
IL8 3576 7.501872 6.6113296
NTRK2 4915 7.497469 7.1365094
RUNX2 860 7.4688272 8.628798
MMP11 4320 7.460847 7.2920337
CDH1 999 7.3595057 7.319695
TCF7L2 6934 7.3556123 8.6040535
KRT78 196374 7.349466 6.8676143
TCF7 6932 7.270456 7.664296
SMO 6608 7.222788 7.0400887
EFNB2 1948 7.1960526 7.26771
CLDN1 9076 7.1643777 8.943991
KRT33A 3883 7.121948 6.808277
VCAN 1462 7.045421 6.763195
MMP9 4318 7.0101504 6.7540355
DLL1 28514 6.969655 6.5782347
KRT13 3860 6.949356 5.971072
IGF2 3481 6.933426 6.170534
KRT26 353288 6.869997 6.697632
TNFRSF9 3604 6.862919 6.6031585
KRT74 121391 6.778076 6.538765
TWIST1 7291 6.765423 6.105777
NRCAM 4897 6.677019 6.7818675
FGF9 2254 6.6647215 5.7855196
TNFRSF11B 4982 6.6092443 6.618697
CHL1 10752 6.6082654 6.3569694
KRT34 3885 6.601664 6.199431
KRT6A 3853 6.536037 5.9656916
EDN1 1906 6.476451 6.7537594
NOS2 4843 6.425461 6.333558
GDF5 8200 6.3569694 6.3291264
CCND2 894 6.3239446 5.996339
DLK1 8788 6.2332454 6.884508
KRT37 8688 5.971611 5.881136
IL6 3569 5.7313643 5.9466343
SOX2 6657 5.6166873 6.7975965
TGFB3 7043 5.5891886 6.0552535
KRT35 3886 5.5883365 6.3580856
PTGS2 5743 5.5262737 7.601541
BTRC 8945 5.3152456 7.7473273

FH535 inhibited pancreatic cancer cell migration

In all, 20 μM FH535 inhibited pancreatic cancer cell migration in a time-dependent manner (Figure 2A). To investigate the mechanisms involved, we analyzed the microarray data to illustrate the expression of genes participating in focal adhesion (Figure 2B, Table 2),13,14 adhesion junctions (Figure 2C, Table 3),1517 tight junctions (Figure 2D, Table 4),1823 and cell motility (Figure 2E, Table 5).2427

Figure 2.

Figure 2

Figure 2

FH535 inhibited pancreatic cancer cell migration.

Notes: (A) Time-dependent inhibition by FH535 of PANC-1 and BxPC-3 cell migration. **P<0.01, significant differences vs the respective control groups. Microarray analysis of (B) focal adhesion–related, (C) adhesion junction–related, (D) tight junction–related, and (E) cell motility–related gene expression regulation upon FH535 treatment. Up and down arrows indicate gene expression significantly upregulated or downregulated, respectively, by twofold. Asterisks indicate genes downstream of the Wnt/β-catenin pathway.

Abbreviation: h, hours.

Table 2.

Microarray analysis of focal adhesion–related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
RPSA 3921 16.551584 16.069508
RHOA 387 15.761177 14.786651
ACTN4 81 15.032014 14.01403
CAPN2 824 14.947017 15.841314
RAC1 5879 14.251518 15.113209
FLNA 2316 14.083586 13.488903
FLNB 2317 13.958575 13.296808
ITGB5 3693 13.888797 13.484464
DNM1 1759 13.640091 12.518821
TMEM132A 54972 13.622586 13.150153
RAP1A 5906 13.5597315 14.039375
HGS 9146 13.533683 13.846248
VCL 7414 13.376745 13.681126
DIAPH1 1729 13.16062 13.659487
GNG11 2791 13.022779 13.403848
AKT1 207 12.957863 12.259176
RAC2 5880 12.955015 11.604415
ITGA3 3675 12.797894 12.391577
ITGB1 3688 12.738785 13.636554
CAV1 857 12.61244 13.617725
ITGB2 3689 12.546266 11.473748
ACTN1 87 12.409878 11.967234
ITGAV 3685 12.278682 14.3424
ITGA6 3655 12.273888 14.392418
ITGA5 3678 12.169847 10.866323
ILK 3611 12.11682 11.583433
TLN1 7094 12.096641 10.645829
SGCE 8910 12.047686 12.282321
ITGB4 3691 11.982763 11.56935
PRKCA 5578 11.918201 13.803304
CTNNB1 1499 11.900537 11.841962
FXYD5 53827 11.859393 10.980669
AKT2 208 11.791592 10.995004
CAV2 858 11.534644 11.731664
VAV2 7410 11.322939 10.17948
CDC42 998 11.250544 11.791042
PARVB 29780 11.224628 9.830263
ZYX 7791 10.997072 9.663998
VASP 7408 10.877319 10.418066
RAF1 5894 10.594473 10.865986
SHC1 6464 10.287678 8.595637
DSP 1832 10.226259 11.500326
PARVA 55742 10.0804615 10.301352
ITGAM 3684 10.003317 8.889115
AKT3 10000 9.999831 12.071189
HRAS 3265 9.972342 9.272375
PDPK1 5170 9.005413 9.698432
HPSE 10855 8.978405 8.067395
PTK2 5747 8.939062 10.820772
DNM2 1785 8.613899 7.43392
SH3PXD2A 9644 8.566784 9.949804
BCAR1 9564 8.529809 7.7692404
ACTN2 88 8.437073 7.474538
PARD6B 84612 8.172608 9.636746
SOS1 6654 7.9648976 7.5355105
DST 667 7.7908773 11.245214
ITGA11 22801 7.7725782 7.250522
ITGA9 3680 7.725706 7.2797456
PIK3CA 5290 7.546133 9.475706
CRK 1398 7.5114365 8.404298
ITGA2B 3674 7.474538 6.8249826
PXN 5829 7.426979 6.493304
SPTB 6710 7.143782 6.598218
PTEN 5728 7.0376005 9.120998
CASK 8573 6.554297 9.147331
ITGA2 3673 6.538141 10.171594
SORBS1 10580 6.5160394 7.080136
SELE 6401 5.8820415 7.629673
ARHGAP5 394 5.628543 7.9602804
ITGA1 3672 5.3547735 7.6031985
ZEB2 9839 5.2203803 6.942315

Table 3.

Microarray analysis of adhesion junction–related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
PFN1 5216 16.843973 16.144138
RHOA 387 15.761177 14.786651
ACTN4 81 15.032014 14.01403
CD44 960 15.006962 16.199093
RAC1 5879 14.251518 15.113209
CTNNA1 1495 14.209974 14.654735
FLNA 2316 14.083586 13.488903
MAPRE1 22919 13.413141 13.8757925
DIAPH1 1729 13.16062 13.659487
RAC2 5880 12.955015 11.604415
CD99 4267 12.8705635 12.11682
JUP 3728 12.776809 12.098349
ACTN1 87 12.409878 11.967234
CDH2 1000 12.27524 13.657263
TLN1 7094 12.096641 10.645829
IQGAP1 8826 11.903805 15.008826
CTNNB1 1499 11.900537 11.841962
PKP3 11187 11.572304 11.483009
CAV2 858 11.534644 11.731664
MGAT5 4249 11.399225 12.44798
CSNK2A1 1457 11.389523 10.994029
PLEK2 26499 11.380254 12.049034
ANAPC1 64682 11.330902 11.025982
NOTCH1 4851 11.311136 10.345143
CDC42 998 11.250544 11.791042
NOTCH2 4853 11.125797 10.202223
WASL 8976 10.930079 12.483249
DLG5 9231 10.567565 11.019769
SRC 6714 10.48147 9.648777
PAK4 10298 10.446864 8.676079
VEGFA 7422 10.250756 7.901348
ZEB1 6935 10.177025 13.283847
JAM3 83700 10.084784 9.556893
PVRL2 5819 10.018614 8.147698
MET 4233 9.986441 12.74971
CSNK2A2 1459 9.960693 9.169151
PTPN1 5770 9.82763 9.271097
PTK2B 2185 9.636893 10.68203
DOCK1 1793 9.48097 11.021774
MAPK1 5594 9.240688 9.218932
ARHGEF7 8874 9.037083 9.147919
CBLL1 79872 9.0251875 9.311203
PKP2 5318 9.022291 10.025781
BAIAP2 10458 9.018734 8.004229
JAM2 58494 8.789471 8.162707
CAV3 859 8.535716 8.229119
ACTN2 88 8.437073 7.474538
PKP1 5317 8.423988 7.3570046
CDH3 1001 8.389479 7.50349
CSF1 1435 8.360545 7.4550886
TJP3 27134 8.295799 7.207067
WASF1 8936 8.178464 7.4462004
PVRL1 5818 8.152037 7.283169
ESR1 2099 8.048168 7.405365
OCLN 100506658 8.036663 9.494983
PIP5K1C 23396 7.9606485 7.254657
CTNND1 1500 7.8659673 7.09317
MAP1B 4131 7.7052383 10.589897
DSG2 1829 7.513804 8.2605915
CDH1 999 7.3595057 7.319695
ACTN3 89 7.355976 6.6988516
VCAN 1462 7.045421 6.763195
DLL1 28514 6.969655 6.5782347
VPS13A 23230 6.859817 10.562696
DSG4 147409 6.608555 6.1116643
DSC2 1824 6.3962626 7.24841
INADL 10207 6.08029 8.808925
PNN 5411 5.9342465 7.5790677
APC 324 5.3153567 6.5241365
ITGA2 3673 6.538141 10.171594
SORBS1 10580 6.5160394 7.080136
SELE 6401 5.8820415 7.629673
ARHGAP5 394 5.628543 7.9602804
ITGA1 3672 5.3547735 7.6031985
ZEB2 9839 5.2203803 6.942315

Table 4.

Microarray analysis of tight junction–related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
RHOA 387 15.761177 14.786651
CLDN4 1364 15.11957 14.539507
ACTN4 81 15.032014 14.01403
CD44 960 15.006962 16.199093
CAPN2 824 14.947017 15.841314
TIMP2 7077 14.796619 14.858342
CFL1 1072 14.710272 13.114106
CSNK2B 1460 14.4039135 14.575101
RAC1 5879 14.251518 15.113209
CTNNA1 1495 14.209974 14.654735
MAPRE1 22919 13.413141 13.8757925
ARHGDIA 396 13.207352 11.634186
EZR 7430 13.144885 12.566931
JAG1 182 12.427784 12.687155
ACTN1 87 12.409878 11.967234
TSPAN13 27075 12.246902 11.379381
ILK 3611 12.11682 11.583433
ICAM1 3383 12.0056095 11.118488
CLDN7 1366 11.972866 11.032687
MMP1 4312 11.905035 12.887484
CTNNB1 1499 11.900537 11.841962
COL16A1 1307 11.647751 10.777789
CSNK2A1 1457 11.389523 10.994029
ENAH 55740 11.354481 13.398125
MLLT4 4301 11.299263 13.275789
CDC42 998 11.250544 11.791042
IGF1R 3480 11.2369585 12.140446
CLDN19 149461 11.222952 10.278006
CTGF 1490 10.809845 11.98555
FZD7 8324 10.711324 9.901575
MAPRE2 10982 10.535324 11.375724
SVIL 6840 10.304885 11.463148
CLDN2 9075 10.221999 7.959734
THBS3 7059 10.1687765 9.736564
LIMK1 3984 10.151468 9.52237
MPP5 64398 10.149654 12.064446
TIAM1 7074 10.117085 11.627998
CGN 57530 10.004088 9.987757
CSNK2A2 1459 9.960693 9.169151
PRKCI 5584 9.934886 11.633633
CRKL 1399 9.737389 9.574368
TJAP1 93643 9.66933 9.609078
CLDN12 9069 9.506469 10.83943
TJP1 7082 9.28694 12.134832
PARD6A 50855 9.12321 8.362814
ARHGEF7 8874 9.037083 9.147919
PDPK1 5170 9.005413 9.698432
CDH5 1003 8.708324 7.5856657
LMO7 4008 8.558113 9.277104
SPTAN1 6709 8.494044 7.7864056
ACTN2 88 8.437073 7.474538
CLDN9 9080 8.4181795 7.5940213
CSF1 1435 8.360545 7.4550886
TJP2 9414 8.343918 7.3491254
HAS1 3036 8.124433 7.6573296
CLDN16 10686 7.9999046 7.022292
AMOTL1 154810 7.8963585 7.8100796
CRK 1398 7.5114365 8.404298
ACTN3 89 7.355976 6.6988516
PRKCG 5582 7.321149 6.9112835
CLDN6 9074 7.220466 6.6578355
CLDN1 9076 7.1643777 8.943991
CLDN15 24146 7.0927997 6.5795236
CLDN10 9071 7.0557775 6.613464
PTEN 5728 7.0376005 9.120998
SMAD2 4087 6.9688606 9.496367
PARD3 56288 6.94016 7.33421
CLEC3B 7123 6.6491346 6.5587797
SPP1 6696 6.37645 6.842924
MAGI1 9223 6.3656254 7.168139
CTTN 2017 6.2022476 6.6959023
ERBB3 2065 6.178696 5.9926624

Table 5.

Microarray analysis of cell motility–related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
VIM 7431 18.111416 18.417988
PERP 64065 17.034954 17.530819
MYH9 4627 16.01196 15.906586
RHOA 387 15.761177 14.786651
ACTN4 81 15.032014 14.01403
TIMP2 7077 14.796619 14.858342
MSN 4478 14.751841 15.357616
CFL1 1072 14.710272 13.114106
RAC1 5879 14.251518 15.113209
RAP1B 5908 14.023661 15.037672
TIMP1 7076 13.919523 13.2338505
CDK4 1019 13.87332 13.635977
RHOC 389 13.521647 12.094296
LAMC1 3915 13.492421 14.51922
VCL 7414 13.376745 13.681126
ACTR3 10096 13.228158 14.45616
DIAPH1 1729 13.16062 13.659487
EZR 7430 13.144885 12.566931
VAPA 9218 13.089962 13.857084
AKT1 207 12.957863 12.259176
RAC2 5880 12.955015 11.604415
ACTR2 10097 12.94824 13.651513
ITGB1 3688 12.738785 13.636554
PRKCZ 5590 12.597843 11.836956
ITGB2 3689 12.546266 11.473748
ACTN1 87 12.409878 11.967234
ILK 3611 12.11682 11.583433
SGCE 8910 12.047686 12.282321
ICAM1 3383 12.0056095 11.118488
PPL 5493 11.998627 11.51075
PRKCA 5578 11.918201 13.803304
PPPDE2 27351 11.624274 11.774211
ENAH 55740 11.354481 13.398125
CDC42 998 11.250544 11.791042
EGFR 1956 11.122326 12.333595
WASL 8976 10.930079 12.483249
CALD1 800 10.921519 12.294691
STEAP1 26872 10.895491 11.820029
TGFB1 7040 10.7152 9.03388
CAMK2N1 55450 10.587699 10.065469
RDX 5962 10.522251 12.191257
MCAM 4162 10.452353 9.462444
ARF6 382 10.415711 11.52632
SVIL 6840 10.304885 11.463148
RGS2 5997 10.257294 9.80196
VEGFA 7422 10.250756 7.901348
CAPN1 823 10.239203 8.216266
F11R 50848 10.234683 9.044022
RND3 390 10.199277 12.088578
MMP2 4313 10.153262 9.412593
WASF2 10163 10.085579 8.826545
FAT1 2195 9.970972 12.042841
RHOB 388 9.965946 8.545685
RAPGEF1 2889 9.903289 8.354535
RASA1 5921 9.702747 11.502001
PTK2B 2185 9.636893 10.68203
ROCK1 6093 9.603488 11.484902
MYH10 4628 9.5496025 11.096066
MMP15 4324 9.533566 8.395943
DOCK1 1793 9.48097 11.021774
PAK2 5062 9.287464 10.61245
PLAUR 5329 9.265003 8.37788
CDC27 996 9.129515 11.077047
MST1R 4486 9.085802 9.12538
BAIAP2 10458 9.018734 8.004229
PTK2 5747 8.939062 10.820772
STAT3 6774 8.847785 7.9192953
ARHGEF2 9181 8.798216 8.383045
PKP4 8502 8.663319 9.734335
MARK2 2011 8.612933 8.00314
PVRL3 25945 8.582907 9.851074
BCAR1 9564 8.529809 7.7692404
ARVCF 421 8.524339 8.364944
SPTAN1 6709 8.494044 7.7864056
TJP2 9414 8.343918 7.3491254
HCLS1 3059 8.263556 7.7210197
WASF1 8936 8.178464 7.4462004
HAS1 3036 8.124433 7.6573296
ADAMTS13 11093 8.074093 8.375932
ESR1 2099 8.048168 7.405365
OCLN 100506658 8.036663 9.494983
WAS 7454 7.9598556 7.412658
CTNND1 1500 7.8659673 7.09317
DOCK4 9732 7.829811 10.702755
CDSN 1041 7.738298 7.3062844
MAP1B 4131 7.7052383 10.589897
MMP11 4320 7.460847 7.2920337
PXN 5829 7.426979 6.493304
ACTN3 89 7.355976 6.6988516
MTSS1 9788 7.3144355 8.826939
VCAN 1462 7.045421 6.763195
MMP9 4318 7.0101504 6.7540355
VTN 7448 6.8925853 6.3992944
EXOC2 55770 6.8692775 8.335709
ECM1 1893 6.8224096 7.0186477
TWIST1 7291 6.765423 6.105777
ADAMTS1 9510 6.670437 7.5566187
CASK 8573 6.554297 9.147331
PLCG1 5335 6.326862 6.175169
CTTN 2017 6.2022476 6.6959023
FARP2 9855 5.4352922 5.775334
CTNND2 1501 5.4170265 5.9111185

FH535 inhibited pancreatic cancer cell invasion

The Matrigel invasion assay revealed that FH535-treated cells had significantly decreased invasive capacity as compared with the control cells (Figure 3A), supporting the premise that FH535 inhibits pancreatic cancer cell invasion. Moreover, FH535 inhibited the adhesion ability of pancreatic cancer cells dose-dependently (Figure 3C). We also analyzed the microarray data to explore the changes in the expression of genes involved in the in vitro invasion process, including extracellular matrix degradation (Figure 3B, Table 6), cell adhesion (Figure 3D, Table 7),28,29 and epithelial–mesenchymal transition (EMT) (Figure 3E, Table 8).3033

Figure 3.

Figure 3

Figure 3

FH535 inhibited pancreatic cancer cell invasion.

Notes: (A) Dose-dependent inhibition by FH535 of PANC-1 and BxPC-3 cell invasion. (B) Microarray analysis of extracellular matrix degradation–related gene expression regulation upon FH535 treatment. (C) Dose-dependent inhibition by FH535 of PANC-1 and BxPC-3 cell adhesion. *P<0.05, **P<0.01, significant differences vs the respective control groups. (D) Microarray analysis of adhesion molecule–related gene expression regulation upon FH535 treatment. (E) Microarray analysis of EMT-related gene expression regulation upon FH535 treatment. Up and down arrows indicate gene expression significantly upregulated or downregulated, respectively, by twofold. Asterisks indicate genes downstream of the Wnt/β-catenin pathway.

Abbreviation: EMT, epithelial–mesenchymal transition.

Table 6.

Microarray analysis of extracellular matrix degradation–related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
TGFBI 7045 16.09069 15.894443
TIMP2 7077 14.796619 14.858342
TIMP1 7076 13.919523 13.2338505
LAMC1 3915 13.492421 14.51922
MMP1 4312 11.905035 12.887484
COL16A1 1307 11.647751 10.777789
COL5A1 1289 11.607744 10.272581
COL6A1 1291 11.396863 10.174638
LAMB1 3912 10.813978 9.924841
CTGF 1490 10.809845 11.98555
THBS3 7059 10.1687765 9.736564
MMP2 4313 10.153262 9.412593
COL4A2 1284 9.866227 8.850218
THBS1 7057 9.663341 9.193558
MMP15 4324 9.533566 8.395943
SPARC 6678 9.32407 8.289816
COL7A1 1294 8.711706 7.6560946
LAMB3 3914 8.550647 8.319239
HAS1 3036 8.124433 7.6573296
ADAMTS13 11093 8.074093 8.375932
SPG7 6687 7.799603 7.3888316
MMP11 4320 7.460847 7.2920337
COL12A1 1303 7.404812 8.422412
COL14A1 7373 7.3424816 6.805993
TNC 3371 7.329479 6.564947
VCAN 1462 7.045421 6.763195
MMP9 4318 7.0101504 6.7540355
VTN 7448 6.8925853 6.3992944
ECM1 1893 6.8224096 7.0186477
ADAMTS1 9510 6.670437 7.5566187
CLEC3B 7123 6.6491346 6.5587797
SPP1 6696 6.37645 6.842924
LAMA3 3909 6.1783895 5.889333

Table 7.

Microarray analysis of adhesion molecule–related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
CD44 960 15.006962 16.199093
ITGB5 3693 13.888797 13.484464
LAMC1 3915 13.492421 14.51922
LAMB1 3912 12.817556 13.73472
ITGA3 3675 12.797894 12.391577
ITGB1 3688 12.738785 13.636554
ITGAV 3685 12.278682 14.3424
ITGA6 3655 12.273888 14.392418
ITGA5 3678 12.169847 10.866323
SGCE 8910 12.047686 12.282321
ICAM1 3383 12.0056095 11.118488
ITGB4 3691 11.982763 11.56935
CTNNB1 1499 11.900537 11.841962
CTNNA1 1495 11.841962 11.517105
COL16A1 1307 11.647751 10.777789
COL5A1 1289 11.607744 10.272581
COL6A1 1291 11.396863 10.174638
CTGF 1490 10.809845 11.98555
CTNND1 1500 10.622252 11.350482
THBS3 7059 10.1687765 9.736564
ITGAM 3684 10.003317 8.889115
THBS1 7057 9.663341 9.193558
ITGA7 3679 9.627002 8.878363
MMP15 4324 9.533566 8.395943
COL7A1 1294 8.711706 7.6560946
LAMB3 3914 8.550647 8.319239
HAS1 3036 8.124433 7.6573296
ADAMTS13 11093 8.074093 8.375932
SPG7 6687 7.990712 6.850328
COL12A1 1303 7.404812 8.422412
CDH1 999 7.3595057 7.319695
COL14A1 7373 7.3424816 6.805993
TNC 3371 7.329479 6.564947
VCAN 1462 7.045421 6.763195
VTN 7448 6.8925853 6.3992944
CLEC3B 7123 6.6491346 6.5587797
ITGB2 3689 6.6435785 7.713477
ITGA2 3673 6.538141 10.171594
SPP1 6696 6.37645 6.842924
LAMA3 3909 6.1783895 5.889333
SELE 6401 5.8820415 7.629673
CTNND2 1501 5.4170265 5.9111185
ITGA1 3672 5.3547735 7.6031985

Table 8.

Microarray analysis of EMT-related gene expression regulation upon FH535 treatment

Gene ID Normalized intensity
Control FH535
VIM 7431 18.111416 18.417988
TGFBI 7045 16.09069 15.894443
NME1 4830 15.692858 15.573043
IGFBP4 3487 14.852157 13.246835
MSN 4478 14.751841 15.357616
RAC1 5879 14.251518 15.113209
KRT7 3855 14.184294 13.285099
TIMP1 7076 13.919523 13.2338505
COL5A2 1290 13.175857 12.833253
TCF3 6929 13.00084 12.29279
AKT1 207 12.957863 12.259176
ITGB1 3688 12.738785 13.636554
CAV1 857 12.61244 13.617725
SNAI1 6615 12.28132 10.385736
ITGA5 3678 12.169847 10.866323
TLN1 7094 12.096641 10.645829
SYMPK 8189 11.942529 12.312602
CTNNB1 1499 11.900537 11.841962
FXYD5 53827 11.859393 10.980669
BMP7 655 11.688513 10.938241
COL5A1 1289 11.607744 10.272581
AHNAK 79026 11.605762 10.785921
CAV2 858 11.534644 11.731664
NOTCH3 4854 11.523583 10.370972
TLN2 83660 11.404076 10.886059
COL6A1 1291 11.396863 10.174638
KRT19 3880 11.289505 11.101922
IGF1R 3480 11.2369585 12.140446
EGFR 1956 11.122326 12.333595
FYN 2534 10.966112 11.325876
WASL 8976 10.930079 12.483249
CALD1 800 10.921519 12.294691
LAMB1 3912 10.813978 9.924841
TGFB1 7040 10.7152 9.03388
FZD7 8324 10.711324 9.901575
SERPINE1 5054 10.639182 10.452353
GRB2 2885 10.605613 9.416897
RDX 5962 10.522251 12.191257
SVIL 6840 10.304885 11.463148
PLEC 5339 10.301559 9.204668
SHC1 6464 10.287678 8.595637
RGS2 5997 10.257294 9.80196
MMP2 4313 10.153262 9.412593
SSX2IP 117178 10.144432 11.167568
COL4A2 1284 9.866227 8.850218
PPAP2B 8613 9.718731 9.956581
THBS1 7057 9.663341 9.193558
ESAM 90952 9.472261 8.959825
NOTCH4 4855 9.433491 9.707824
SPARC 6678 9.32407 8.289816
VEZT 55591 9.128493 11.195451
MST1R 4486 9.085802 9.12538
STAT3 6774 8.847785 7.9192953
ZAK 51776 8.719432 11.337164
COL7A1 1294 8.711706 7.6560946
SMURF1 57154 8.629299 9.522539
LAMB3 3914 8.550647 8.319239
TNS1 7145 8.064375 7.5303655
SPG7 6687 7.990712 6.850328
SOS1 6654 7.9648976 7.5355105
WIPF1 7456 7.8996034 7.0742846
BMP1 649 7.73736 6.776738
FOXC2 2303 7.5557323 6.690961
COL12A1 1303 7.404812 8.422412
CDH1 999 7.3595057 7.319695
COL14A1 7373 7.3424816 6.805993
TNC 3371 7.329479 6.564947
IL1RN 3557 7.2758436 6.734858
SOX10 6663 7.0939784 6.8492174
VCAN 1462 7.045421 6.763195
PTEN 5728 7.0376005 9.120998
MMP9 4318 7.0101504 6.7540355
MPP6 51678 6.9906545 8.912582
SYK 6850 6.4246235 6.223468
SPP1 6696 6.37645 6.842924
ERBB3 2065 6.178696 5.9926624
LAMA3 3909 6.1783895 5.889333
BMP2 650 6.0141077 7.1390386
SOS2 6655 5.6132765 8.797646
TGFB3 7043 5.5891886 6.0552535

Abbreviation: EMT, epithelial–mesenchymal transition.

FH535 inhibited pancreatic cancer cell growth

Using MTT assay, we evaluated the inhibitory effect of FH535 on pancreatic cancer cell line growth. The proliferation of PANC-1 and BxPC-3 cells cultured for up to 48 hours with FH535 was significantly inhibited time-dependently and dose-dependently as compared to the control cells (Figure 4A). The clone formation assays confirmed the dose-dependent inhibitory effect of FH535 on pancreatic cancer cell growth (Figure 4B). We performed cell cycle analysis to confirm the antimitogenic effect of FH535. FH535 induced G2/M accumulation and decreased the cell population in the G0/G1 and S phases dose-dependently (Figure 4C). The expression profile of the cell cycle–related genes obtained from microarray analyses was analyzed (Figure 4D, Table 9).34

Figure 4.

Figure 4

Figure 4

Inhibitory effect of FH535 on pancreatic cancer cell growth.

Notes: (A) Dose- and time-dependent inhibition by FH535 of PANC-1 and BxPC-3 cell growth. (B) Dose-dependent inhibition by FH535 of the clone formation ability of BxPC-3 cells. *P<0.05, **P<0.01, significant differences vs the respective control groups. (C) Significant dose-dependent G2/M arrest following FH535 treatment in BxPC-3 cells. (D) Microarray analysis of cell cycle–related gene expression regulation upon 20 μM FH535 treatment. Up and down arrows indicate gene expression significantly upregulated or downregulated, respectively, by twofold. Asterisks indicate genes downstream of the Wnt/β-catenin pathway.

Abbreviation: h, hours.

Table 9.

Microarray analysis of cell cycle–related gene expression regulation upon 20 μM FH535 treatment

Gene ID Normalized intensity
Control FH535
MYC 4609 16.268158 15.204586
CKS2 1164 15.878164 15.394571
RHOA 387 15.761177 14.786651
BIRC5 332 14.757564 14.219355
CCNB1 891 14.478785 14.022737
CCNB2 9133 14.019871 14.271269
KPNA2 3838 13.950185 14.971469
CDK4 1019 13.87332 13.635977
CDKN2D 1032 13.839806 12.513427
GADD45A 1647 13.765451 13.130958
CDKN1C 1028 13.694702 12.537176
PCNA 5111 13.611973 13.2414665
CKS1B 1163 13.609195 12.845673
MCM3 4172 13.580797 13.911951
PRC1 9055 13.4114275 14.32471
MAD2L1 4085 13.297964 12.867024
CDK1 983 13.286596 13.600226
DDIT3 1649 13.135856 11.871853
SERTAD1 29950 13.060848 11.212
IGF2 3481 13.0203 13.799717
AKT1 207 12.957863 12.259176
GNL3 26354 12.891848 14.223899
ITGB1 3688 12.738785 13.636554
RASSF1 11186 12.559567 11.458946
CDK7 1022 12.553116 13.288865
CDC34 997 12.53036 11.302476
TFDP2 7029 12.527303 12.6644745
CDC20 991 12.526335 11.232994
CDKN3 1033 12.486179 12.966997
MAP2K1 5604 12.483637 11.714967
CUL3 8452 12.389215 13.715795
FOSL1 8061 12.344017 11.102832
ILK 3611 12.11682 11.583433
FOXO3 2309 12.095011 13.865094
MNAT1 4331 12.074865 12.456777
JUN 3725 12.038464 12.673436
PDK2 5164 11.944916 10.781894
CTNNB1 1499 11.900537 11.841962
WEE1 7465 11.894105 13.381722
MCM2 4171 11.862871 11.269842
AURKB 9212 11.800928 9.697033
PPP1R15A 23645 11.676006 10.124018
MDC1 9656 11.649198 10.940614
CDKN2C 1031 11.6150875 11.096327
CCND3 896 11.576098 10.075832
CCNE1 898 11.407219 11.045229
CDKN2A 1029 11.343829 11.097562
AURKA 6790 11.181647 10.961699
RAD1 5810 11.162613 9.890079
NOTCH2 4853 11.125797 10.202223
EGFR 1956 11.122326 12.333595
CDK5 1020 11.117936 10.542482
RAD51 5888 11.1082325 10.784544
CCNA2 890 10.911861 10.9871645
MCM4 4173 10.849212 11.05679
CDC16 8881 10.808938 13.084003
BCCIP 56647 10.778181 12.021907
CDC25C 995 10.672214 10.564099
RB1 5925 10.640414 12.741512
CCNC 892 10.610059 10.50804
CDK9 1025 10.606858 9.6394415
GRB2 2885 10.605613 9.416897
ATM 472 10.5960865 9.790577
PPM1D 8493 10.580978 9.338833
CCND1 595 10.543621 10.14267
PKD1 5310 10.471296 9.29677
E2F3 1871 10.412796 12.061844
CCNG1 900 10.409301 9.063653
CDK5R1 8851 10.322197 10.292204
PDK1 5163 10.321034 10.933424
MAD2L2 10459 10.306548 9.040705
SHC1 6464 10.287678 8.595637
CCNT1 904 10.17827 11.456285
CDK2 1017 10.10528 9.357359
RUVBL1 8607 10.000026 8.814938
SKP2 6502 9.937107 8.872935
CASP3 836 9.893856 10.658698
AHR 196 9.886938 11.936481
RAD9A 5883 9.855825 8.961951
PA2G4 5036 9.737894 8.966842
CHEK2 11200 9.726725 10.255492
INS 3630 9.673779 8.813154
BBS4 585 9.597437 9.65494
SIAH1 6477 9.530121 9.295226
CCNG2 901 9.4692955 8.337656
RBBP8 5932 9.454372 12.631638
MAPK3 5595 9.447224 7.818405
TFDP1 7027 9.4434185 9.030092
E2F1 1869 9.397123 8.754342
CDK5RAP2 55755 9.390803 10.79269
MAPK1 5594 9.240688 9.218932
MKI67 4288 9.233824 8.092867
ID2 3398 9.226584 8.999163
CUL1 8454 9.221058 10.794849
RBL2 5934 9.160267 10.348637
JAG2 3714 9.075178 8.140677
GTSE1 51512 9.04477 8.488665
CHEK1 1111 9.04231 9.651725
E2F4 1874 9.035055 8.016477
G0S2 50486 9.032754 8.410861
SESN2 83667 8.951754 8.138044
PTK2 5747 8.939062 10.820772
CDC25A 993 8.677163 8.042739
MDM2 4193 8.622698 7.73673
CDK5RAP1 51654 8.508385 6.9220624
AXIN1 8312 8.351635 7.6164603
ATR 545 8.152882 11.634625
MCM5 4174 8.055058 7.4200873
CDKN1A 1026 8.007444 6.7135
SOS1 6654 7.9648976 7.5355105
CDKN1B 1027 7.8906517 6.7354736
GML 2765 7.8596773 6.9595275
TSC1 7248 7.6562896 9.700143
BRCA1 672 7.620017 10.24544
CUL2 8453 7.566332 9.5496025
STMN1 3925 7.5155845 6.8102884
CDK8 1024 7.5133963 6.9160185
TERT 7015 7.4070444 7.4882307
ABL1 25 7.3099413 6.6226487
PTEN 5728 7.0376005 9.120998
MDM4 4194 6.996299 8.63818
HUS1 3364 6.976554 7.3689637
RBL1 5933 6.7265186 8.035306
CCND2 894 6.3239446 5.996339
BMP2 650 6.0141077 7.1390386
ATRIP 84126 5.912352 6.682503
CDC6 990 5.8019896 6.064807
SOX2 6657 5.6166873 6.7975965
PTGS2 5743 5.5262737 7.601541
APC 324 5.3153567 6.5241365
BTRC 8945 5.3152456 7.7473273

Discussion

It is widely acknowledged that the prognosis of pancreatic cancer is very poor. The canonical Wnt/β-catenin signaling pathway plays a key role in tumor development and dissemination. Classical Wnt signaling pathway causes accumulation of β-catenin in cytoplasm in complex with the transcription factor TCF/LEF that regulates target gene expression.9,35 Dysregulation of Wnt/β-catenin signaling and altered transcription of β-catenin/TCF-regulated genes are found in many cancers,36 including pancreatic cancer.37 In this regard, we focused on characterizing the mechanisms of the anti-tumor effect of FH535 on pancreatic cancer cells.

Western blotting revealed that FH535 did not affect β-catenin expression in BxPC-3 cells. Interestingly, FH535 downregulated the protein level of total β-catenin in the PANC-1 cells, which differed from the results of most previous studies.10 This cell type–dependent downregulation of β-catenin could have been due to the stabilization of axin, which suppresses β-catenin.11 Axin is characterized as a tumor-suppressor gene, and it plays a key role in inhibiting the canonical Wnt pathway by forming molecular complexes with other proteins such as GSK-3β and adenomatous polyposis coli (APC).38 Whether or not β-catenin expression was inhibited, the luciferase reporter assay proved that transcriptional activity of β-catenin pathway was decreased, which was consistent with previous study findings.10

Metastasis, the leading cause of cancer-related death, is a complex process comprising several steps, all of which we found were affected by FH535. First, FH535 inhibited pancreatic cancer cell migration. Microarray analyses revealed that FH535 altered the expression of several migration-related genes, which participate in focal adhesion, adhesion junctions, tight junctions, and/or motility regulation. Among these genes, the focal adhesion–related gene PTEN, considered “the most highly mutated tumor-suppressor gene in the post-p53 era”,39 plays a role in controlling cell migration.40 The loss of PTEN protein expression or function has been reported in many human cancers, including ovarian, endometrial, and prostate carcinoma; breast cancer; and primary gastrointestinal stromal tumor.41,42 We also found that FH535 downregulated the adhesion junction–related gene TLN1, which encodes a cytoskeletal protein that is concentrated in areas of cell–substratum and cell–cell contact. The encoded protein plays a significant role in actin filament assembly and in the spread and migration of various cell types.43,44 TLN1 is codistributed with integrins in the cell surface membrane, aiding the attachment of adherent cells to extracellular matrices and lymphocytes to other cells. In our study, tight junction protein 1 (TJP1), which plays a critical role in cell–cell interaction, proliferation, and differentiation, was upregulated. TJP1 is an important marker of tight junction integrity, which is disrupted in many highly invasive cancers; upregulated TJP1 correlates with favorable survival in breast cancer and gastrointestinal stromal tumor.45,46 The motility-related gene VEGFA significantly increases the motility of pancreatic cancer cells. The vascular endothelial growth factor/vascular endothelial growth factor receptor (VEGF/VEGFR) inhibitors bevacizumab and sunitinib significantly decrease pancreatic cancer cell motility.47 In our study, FH535 not only suppressed VEGFA expression but also inhibited cell motility, suggesting the involvement of a similar mechanism.

To establish metastasis, tumor cells must traverse the basement membrane to reach the connective tissues. Accordingly, we investigated the anti-invasive effect of FH535. The Transwell assay proved that FH535 inhibited invasion. In vitro invasion can be divided into several steps, including matrix adhesion, matrix degradation, and EMT. We analyzed the expression of the genes involved in these steps using microarray and found that FH535 significantly downregulated the cell adhesion molecule ITGA5; ITGA5 knockdown results in decreased adhesion in pancreatic cancer cells.48 The ability of matrix metalloproteinases (MMPs) to degrade extracellular matrix proteins has been well characterized; therefore, they have been studied extensively to elucidate their involvement in both tumor development and progression. Different MMPs play different roles in tumorigenesis. MMP15 appears to be upregulated during colorectal tumorigenesis, and past research has shown stromal localization of MMP15 in the early phases of neoplastic transformation in colorectal cancer.49 In our study, FH535 downregulated MMP15. Epithelial cells are characterized by well-developed junctions and apical–basolateral polarization; on the contrary, mesenchymal cells lack polarization due to the loss of an organized junctional layer. Cell metastasis is correlated with EMT. In the present study, FH535 downregulated Snail, which is upregulated during EMT.50 In human colorectal cancer cells, overexpression of Snail induces not only EMT but also a cancer stem cell–like phenotype, which enhances cell migration and invasion in vitro and increases metastasis formation in vivo.51 Snail also plays an essential role in human pancreatic cancer progression and metastasis.52,53 In the clinical setting, overexpression of Snail was previously associated with poorer prognosis and a more invasive phenotype in many malignancies.5456 We also detected the downregulation of TGFB1, a classic EMT stimulator.57 TGFB1 overexpression is associated with early recurrence following resection and decreased survival;58 consistent with our study, the suppression of TGFB1 activity in immune-deficient orthotopic mouse models of pancreatic cancer attenuated tumor growth and metastasis.59,60

Besides metastasis, FH535 also induced G2/M arrest and inhibited pancreatic cancer cell proliferation. FH535 significantly upregulated the G2/M regulator gene BCCIP while downregulating the cell cycle regulatory genes CCNG1 and SERTAD1. Human BCCIP, a protein that interacts with BRCA2 and CDKN1A (Cip1, p21), has been implicated in many cellular processes, including cell cycle regulation, DNA recombination and damage repair, telomere maintenance, embryonic development, and genomic stability.6163 BCCIP knockdown and concomitant p53 deletion causes rapid development of medulloblastomas, which have a wide spectrum of alterations involving the Sonic hedgehog pathway, consistent with the caretaker responsibility of BCCIP in genomic integrity.64 BCCIP expression is downregulated in human ovarian cancer, renal cell carcinoma, and colorectal cancer tissues, suggesting that the gene plays a role in the pathogenesis of these cancers.63 The positive expression rate and intensity of CCNG1 in gastric carcinoma is significantly correlated with tumor differentiation. Elevated amounts of CCNG1 are frequently detected in malignant tissue tumors, including astrocytoma; melanoma; carcinoma of the esophagus, lung, and breast; and cancer of the cervix, uterus, and ovary.65 It plays a pivotal role in hepatocellular carcinoma metastasis and may be a novel prognostic biomarker and therapeutic target.66 SERTAD1 is involved in positive regulation of the cell cycle and proliferation;67,68 accordingly, its expression is upregulated in several tumor types.69,70 Studies indicate that SERTAD1 promotes proliferation by binding to the transcription factor E2F1 and by enhancing its transcriptional activity.71 Experimental overexpression of SERTAD1 provoked hyperproliferation,72 genomic instability,68 and inhibition of apoptosis.73

We demonstrated that FH535 significantly inhibits pancreatic cancer cell metastasis by suppressing migration, invasion, and adhesion and induces the accumulation of cells in the G2/M phase to suppress proliferation. These results suggest that FH535 is a potential candidate for pancreatic cancer treatment. Some of the identified genes that responded to FH535 are well-established direct targets of the Wnt/β-catenin pathway. However, it has not been proven that the other identified genes are located downstream of the pathway. FH535 might affect the expression of these genes through the Wnt/β-catenin pathway indirectly or in a β-catenin independent manner. In fact, FH535 not only antagonizes β-catenin/TCF-mediated transcription but also inhibits recruitment of the coactivators glucocorticoid receptor-interacting protein 1 (GRIP1) and β-catenin to peroxisome proliferator-activated receptor (PPAR)δ and PPARγ,10 suggesting that these mechanisms could also be involved in the anti-cancer effect of FH535.

Acknowledgments

This study was supported by the National Natural Science Foundation of China (grant nos 81472296, 81101867, 81272542, 81200369, and 81372443), the CSPAC-Celgene Foundation, the China International Medical Foundation (grant no CIMF-F-H001-057), the Special Foundation of Clinical Medicine of Jiangsu Provincial Bureau of Science and Technology (grant no BL2014039), the Scientific Research Project of Jiangsu Provincial Bureau of Traditional Chinese Medicine (grant no L213236), the Medical Scientific Research Project of Jiangsu Provincial Bureau of Health (grant no Z201206), the Special Foundation of Wu Jieping Medical Foundation for Clinical Scientific Research (grant nos 320.6753.1225 and 320.6750.12242), the Science and Education for Health Foundation of Suzhou for Youth (grant nos SWKQ1003 and SWKQ1011), the Science and Technology Project Foundation of Suzhou (grant nos SYS201112, SYSD2012137, and SYS201335), the Science and Technology Foundation of Suzhou Xiangcheng (grant nos SZXC2012-70 and XJ201451), and a project founded by the priority academic program development of Jiangsu higher education institutions.

Footnotes

Disclosure

The authors report no conflicts of interest in this work.

References

  • 1.Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2006. CA Cancer J Clin. 2006;56(2):106–130. doi: 10.3322/canjclin.56.2.106. [DOI] [PubMed] [Google Scholar]
  • 2.Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363(9414):1049–1057. doi: 10.1016/S0140-6736(04)15841-8. [DOI] [PubMed] [Google Scholar]
  • 3.Bosetti C, Bertuccio P, Malvezzi M, et al. Cancer mortality in Europe, 2005–2009, and an overview of trends since 1980. Ann Oncol. 2013;24(10):2657–2671. doi: 10.1093/annonc/mdt301. [DOI] [PubMed] [Google Scholar]
  • 4.Cai HH, Sun YM, Miao Y, et al. Aberrant methylation frequency of TNFRSF10C promoter in pancreatic cancer cell lines. Hepatobiliary Pancreat Dis Int. 2011;10(1):95–100. doi: 10.1016/s1499-3872(11)60014-3. [DOI] [PubMed] [Google Scholar]
  • 5.Iida J, Wilhelmson KL, Price MA, et al. Membrane type-1 matrix metalloproteinase promotes human melanoma invasion and growth. J Invest Dermatol. 2004;122(1):167–176. doi: 10.1046/j.0022-202X.2003.22114.x. [DOI] [PubMed] [Google Scholar]
  • 6.Eisenmann KM, McCarthy JB, Simpson MA, et al. Melanoma chondroitin sulphate proteoglycan regulates cell spreading through Cdc42, Ack-1 and p130cas. Nat Cell Biol. 1999;1(8):507–513. doi: 10.1038/70302. [DOI] [PubMed] [Google Scholar]
  • 7.Vaid M, Prasad R, Sun Q, Katiyar SK. Silymarin targets beta-catenin signaling in blocking migration/invasion of human melanoma cells. PLoS One. 2011;6(7):e23000. doi: 10.1371/journal.pone.0023000. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 8.Iida J, Pei D, Kang T, et al. Melanoma chondroitin sulfate proteoglycan regulates matrix metalloproteinase-dependent human melanoma invasion into type I collagen. J Biol Chem. 2001;276(22):18786–18794. doi: 10.1074/jbc.M010053200. [DOI] [PubMed] [Google Scholar]
  • 9.Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127(3):469–480. doi: 10.1016/j.cell.2006.10.018. [DOI] [PubMed] [Google Scholar]
  • 10.Handeli S, Simon JA. A small-molecule inhibitor of Tcf/beta-catenin signaling down-regulates PPARgamma and PPARdelta activities. Mol Cancer Ther. 2008;7(3):521–529. doi: 10.1158/1535-7163.MCT-07-2063. [DOI] [PubMed] [Google Scholar]
  • 11.Iida J, Dorchak J, Lehman JR, et al. FH535 inhibited migration and growth of breast cancer cells. PLoS One. 2012;7(9):e44418. doi: 10.1371/journal.pone.0044418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ren J, Wang R, Song H, Huang G, Chen L. Secreted frizzled related protein 1 modulates taxane resistance of human lung adenocarcinoma. Mol Med. 2014;20:164–178. doi: 10.2119/molmed.2013.00149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hannigan G, Troussard AA, Dedhar S. Integrin-linked kinase: a cancer therapeutic target unique among its ILK. Nat Rev Cancer. 2005;5(1):51–63. doi: 10.1038/nrc1524. [DOI] [PubMed] [Google Scholar]
  • 14.Legate KR, Montanez E, Kudlacek O, Fassler R. ILK, PINCH and parvin: the tIPP of integrin signalling. Nat Rev Mol Cell Biol. 2006;7(1):20–31. doi: 10.1038/nrm1789. [DOI] [PubMed] [Google Scholar]
  • 15.Palacios F, Price L, Schweitzer J, Collard JG, D’Souza-Schorey C. An essential role for ARF6-regulated membrane traffic in adherens junction turnover and epithelial cell migration. EMBO J. 2001;20(17):4973–4986. doi: 10.1093/emboj/20.17.4973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Pece S, Gutkind JS. E-cadherin and Hakai: signalling, remodeling or destruction? Nat Cell Biol. 2002;4(4):E72–E74. doi: 10.1038/ncb0402-e72. [DOI] [PubMed] [Google Scholar]
  • 17.D’Souza-Schorey C. Disassembling adherens junctions: breaking up is hard to do. Trends Cell Biol. 2005;15(1):19–26. doi: 10.1016/j.tcb.2004.11.002. [DOI] [PubMed] [Google Scholar]
  • 18.Tsukita S, Furuse M, Itoh M. Multifunctional strands in tight junctions. Nat Rev Mol Cell Biol. 2001;2(4):285–293. doi: 10.1038/35067088. [DOI] [PubMed] [Google Scholar]
  • 19.Cheng CY, Mruk DD. Cell junction dynamics in the testis: sertoli-germ cell interactions and male contraceptive development. Physiol Rev. 2002;82(4):825–874. doi: 10.1152/physrev.00009.2002. [DOI] [PubMed] [Google Scholar]
  • 20.Matter K, Balda MS. Signalling to and from tight junctions. Nat Rev Mol Cell Biol. 2003;4(3):225–236. doi: 10.1038/nrm1055. [DOI] [PubMed] [Google Scholar]
  • 21.Balda MS, Matter K. Epithelial cell adhesion and the regulation of gene expression. Trends Cell Biol. 2003;13(6):310–318. doi: 10.1016/s0962-8924(03)00105-3. [DOI] [PubMed] [Google Scholar]
  • 22.Bazzoni G, Dejana E. Endothelial cell-to-cell junctions: molecular organization and role in vascular homeostasis. Physiol Rev. 2004;84(3):869–901. doi: 10.1152/physrev.00035.2003. [DOI] [PubMed] [Google Scholar]
  • 23.Furuse M, Tsukita S. Claudins in occluding junctions of humans and flies. Trends Cell Biol. 2006;16(4):181–188. doi: 10.1016/j.tcb.2006.02.006. [DOI] [PubMed] [Google Scholar]
  • 24.Linder S. The matrix corroded: podosomes and invadopodia in extracellular matrix degradation. Trends Cell Biol. 2007;17(3):107–117. doi: 10.1016/j.tcb.2007.01.002. [DOI] [PubMed] [Google Scholar]
  • 25.Chhabra ES, Higgs HN. The many faces of actin: matching assembly factors with cellular structures. Nat Cell Biol. 2007;9(10):1110–1121. doi: 10.1038/ncb1007-1110. [DOI] [PubMed] [Google Scholar]
  • 26.McEver RP, Zhu C. Rolling cell adhesion. Annu Rev Cell Dev Biol. 2010;26:363–396. doi: 10.1146/annurev.cellbio.042308.113238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Parsons JT, Horwitz AR, Schwartz MA. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nat Rev Mol Cell Biol. 2010;11(9):633–643. doi: 10.1038/nrm2957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Mullins RF, Skeie JM, Folk JC, et al. Evaluation of variants in the selectin genes in age-related macular degeneration. BMC Med Genet. 2011;12:58. doi: 10.1186/1471-2350-12-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ciriza J, Garcia-Ojeda ME. Expression of migration-related genes is progressively upregulated in murine lineage-Sca-1+c-Kit+ population from the fetal to adult stages of development. Stem Cell Res Ther. 2010;1(2):14. doi: 10.1186/scrt14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kaartinen V, Voncken JW, Shuler C, et al. Abnormal lung development and cleft palate in mice lacking TGF-beta 3 indicates defects of epithelial-mesenchymal interaction. Nat Genet. 1995;11(4):415–421. doi: 10.1038/ng1295-415. [DOI] [PubMed] [Google Scholar]
  • 31.Timmerman LA, Grego-Bessa J, Raya A, et al. Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004;18(1):99–115. doi: 10.1101/gad.276304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Moreno-Bueno G, Cubillo E, Sarrió D, et al. Genetic profiling of epithelial cells expressing E-cadherin repressors reveals a distinct role for snail, slug, and E47 factors in epithelial-mesenchymal transition. Cancer Res. 2006;66(19):9543–9556. doi: 10.1158/0008-5472.CAN-06-0479. [DOI] [PubMed] [Google Scholar]
  • 33.Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14(6):818–829. doi: 10.1016/j.devcel.2008.05.009. [DOI] [PubMed] [Google Scholar]
  • 34.Hoffman AE, Zheng T, Ba Y, et al. Phenotypic effects of the circadian gene cryptochrome 2 on cancer-related pathways. BMC Cancer. 2010;10:110. doi: 10.1186/1471-2407-10-110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999;398(6726):422–426. doi: 10.1038/18884. [DOI] [PubMed] [Google Scholar]
  • 36.Fearon ER. PARsing the phrase “all in for axin” – Wnt pathway targets in cancer. Cancer Cell. 2009;16(5):366–368. doi: 10.1016/j.ccr.2009.10.007. [DOI] [PubMed] [Google Scholar]
  • 37.White BD, Chien AJ, Dawson DW. Dysregulation of Wnt/beta-catenin signaling in gastrointestinal cancers. Gastroenterology. 2012;142(2):219–232. doi: 10.1053/j.gastro.2011.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17(1):9–26. doi: 10.1016/j.devcel.2009.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Di Cristofano A, Pandolfi PP. The multiple roles of PTEN in tumor suppression. Cell. 2000;100(4):387–390. doi: 10.1016/s0092-8674(00)80674-1. [DOI] [PubMed] [Google Scholar]
  • 40.Waite KA, Eng C. Protean PTEN: form and function. Am J Hum Genet. 2002;70(4):829–844. doi: 10.1086/340026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chalhoub N, Baker SJ. PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol. 2009;4:127–150. doi: 10.1146/annurev.pathol.4.110807.092311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wang H, Chen P, Liu XX, et al. Prognostic impact of gastrointestinal bleeding and expression of PTEN and Ki-67 on primary gastrointestinal stromal tumors. World J Surg Oncol. 2014;12:89. doi: 10.1186/1477-7819-12-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tang H, Yao L, Tao X, et al. miR-9 functions as a tumor suppressor in ovarian serous carcinoma by targeting TLN1. Int J Mol Med. 2013;32(2):381–388. doi: 10.3892/ijmm.2013.1400. [DOI] [PubMed] [Google Scholar]
  • 44.Sakamoto S, McCann RO, Dhir R, Kyprianou N. Talin1 promotes tumor invasion and metastasis via focal adhesion signaling and anoikis resistance. Cancer Res. 2010;70(5):1885–1895. doi: 10.1158/0008-5472.CAN-09-2833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sommers CL, Byers SW, Thompson EW, Torri JA, Gelmann EP. Differentiation state and invasiveness of human breast cancer cell lines. Breast Cancer Res Treat. 1994;31(2–3):325–335. doi: 10.1007/BF00666165. [DOI] [PubMed] [Google Scholar]
  • 46.Zhu H, Lu J, Wang X, et al. Upregulated ZO-1 correlates with favorable survival of gastrointestinal stromal tumor. Med Oncol. 2013;30(3):631. doi: 10.1007/s12032-013-0631-7. [DOI] [PubMed] [Google Scholar]
  • 47.Doi Y, Yashiro M, Yamada N, Amano R, Noda S, Hirakawa K. VEGF-A/VEGFR-2 signaling plays an important role for the motility of pancreas cancer cells. Ann Surg Oncol. 2012;19(8):2733–2743. doi: 10.1245/s10434-011-2181-6. [DOI] [PubMed] [Google Scholar]
  • 48.Walsh N, Clynes M, Crown J, O’Donovan N. Alterations in integrin expression modulates invasion of pancreatic cancer cells. J Exp Clin Cancer Res. 2009;28:140. doi: 10.1186/1756-9966-28-140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sena P, Mariani F, Marzona L, et al. Matrix metalloproteinases 15 and 19 are stromal regulators of colorectal cancer development from the early stages. Int J Oncol. 2012;41(1):260–266. doi: 10.3892/ijo.2012.1441. [DOI] [PubMed] [Google Scholar]
  • 50.Wu Y, Zhou BP. Snail: more than EMT. Cell Adh Migr. 2010;4(2):199–203. doi: 10.4161/cam.4.2.10943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Fan F, Samuel S, Evans KW, et al. Overexpression of snail induces epithelial-mesenchymal transition and a cancer stem cell-like phenotype in human colorectal cancer cells. Cancer Med. 2012;1(1):5–16. doi: 10.1002/cam4.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hotz B, Arndt M, Dullat S, Bhargava S, Buhr HJ, Hotz HG. Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin Cancer Res. 2007;13(16):4769–4776. doi: 10.1158/1078-0432.CCR-06-2926. [DOI] [PubMed] [Google Scholar]
  • 53.von Burstin J, Eser S, Paul MC, et al. E-cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex. Gastroenterology. 2009;137(1):e361–e365. doi: 10.1053/j.gastro.2009.04.004. [DOI] [PubMed] [Google Scholar]
  • 54.Shin NR, Jeong EH, Choi CI, et al. Overexpression of snail is associated with lymph node metastasis and poor prognosis in patients with gastric cancer. BMC Cancer. 2012;12:521. doi: 10.1186/1471-2407-12-521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kuo KT, Chou TY, Hsu HS, Chen WL, Wang LS. Prognostic significance of NBS1 and snail expression in esophageal squamous cell carcinoma. Ann Surg Oncol. 2012;19(suppl 3):S549–S557. doi: 10.1245/s10434-011-2043-2. [DOI] [PubMed] [Google Scholar]
  • 56.van Nes JG, de Kruijf EM, Putter H, et al. Co-expression of SNAIL and TWIST determines prognosis in estrogen receptor-positive early breast cancer patients. Breast Cancer Res Treat. 2012;133(1):49–59. doi: 10.1007/s10549-011-1684-y. [DOI] [PubMed] [Google Scholar]
  • 57.Wu Y, Zhou BP. New insights of epithelial-mesenchymal transition in cancer metastasis. Chin J Biochem Biophys. 2008;40(7):643–650. doi: 10.1111/j.1745-7270.2008.00443.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Friess H, Yamanaka Y, Büchler M, et al. Enhanced expression of transforming growth factor beta isoforms in pancreatic cancer correlates with decreased survival. Gastroenterology. 1993;105(6):1846–1856. doi: 10.1016/0016-5085(93)91084-u. [DOI] [PubMed] [Google Scholar]
  • 59.Rowland-Goldsmith MA, Maruyama H, Kusama T, Ralli S, Korc M. Soluble type II transforming growth factor-beta (TGF-beta) receptor inhibits TGF-beta signaling in COLO-357 pancreatic cancer cells in vitro and attenuates tumor formation. Clin Cancer Res. 2001;7(9):2931–2940. [PubMed] [Google Scholar]
  • 60.Melisi D, Ishiyama S, Sclabas GM, 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. 2008;7(4):829–840. doi: 10.1158/1535-7163.MCT-07-0337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Misra S, Sharma S, Agarwal A, et al. Cell cycle-dependent regulation of the bi-directional overlapping promoter of human BRCA2/ZAR2 genes in breast cancer cells. Mol Cancer. 2010;9:50. doi: 10.1186/1476-4598-9-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lu H, Yue J, Meng X, Nickoloff JA, Shen Z. BCCIP regulates homologous recombination by distinct domains and suppresses spontaneous DNA damage. Nucleic Acids Res. 2007;35(21):7160–7170. doi: 10.1093/nar/gkm732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Liu X, Cao L, Ni J, et al. Differential BCCIP gene expression in primary human ovarian cancer, renal cell carcinoma and colorectal cancer tissues. Int J Oncol. 2013;43(6):1925–1934. doi: 10.3892/ijo.2013.2124. [DOI] [PubMed] [Google Scholar]
  • 64.Huang YY, Dai L, Gaines D, et al. BCCIP suppresses tumor initiation but is required for tumor progression. Cancer Res. 2013;73(23):7122–7133. doi: 10.1158/0008-5472.CAN-13-1766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Cui X, Yu L, Wang Y, et al. The relationship between cyclin G1 and survival in patients treated surgically for HCC. Hepatogastroenterology. 2013;60(121):153–159. doi: 10.5754/hge12549. [DOI] [PubMed] [Google Scholar]
  • 66.Wen W, Ding J, Sun W, et al. Cyclin G1-mediated epithelial-mesenchymal transition via phosphoinositide 3-kinase/Akt signaling facilitates liver cancer progression. Hepatology. 2012;55(6):1787–1798. doi: 10.1002/hep.25596. [DOI] [PubMed] [Google Scholar]
  • 67.Li J, Muscarella P, Joo SH, et al. Dissection of CDK4-binding and transactivation activities of p34(SEI-1) and comparison between functions of p34(SEI-1) and p16(INK4A) Biochemistry. 2005;44(40):13246–13256. doi: 10.1021/bi0504658. [DOI] [PubMed] [Google Scholar]
  • 68.Tang DJ, Hu L, Xie D, et al. Oncogenic transformation by SEI-1 is associated with chromosomal instability. Cancer Res. 2005;65(15):6504–6508. doi: 10.1158/0008-5472.CAN-05-0351. [DOI] [PubMed] [Google Scholar]
  • 69.van Dekken H, Alers JC, Riegman PH, Rosenberg C, Tilanus HW, Vissers K. Molecular cytogenetic evaluation of gastric cardia adenocarcinoma and precursor lesions. Am J Pathol. 2001;158(6):1961–1967. doi: 10.1016/S0002-9440(10)64666-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Tang TC, Sham JS, Xie D, et al. Identification of a candidate oncogene SEI-1 within a minimal amplified region at 19q13.1 in ovarian cancer cell lines. Cancer Res. 2002;62(24):7157–7161. [PubMed] [Google Scholar]
  • 71.Hsu SI, Yang CM, Sim KG, Hentschel DM, O’Leary E, Bonventre JV. TRIP-Br: a novel family of PHD zinc finger- and bromodomain-interacting proteins that regulate the transcriptional activity of E2F-1/DP-1. EMBO J. 2001;20(9):2273–2285. doi: 10.1093/emboj/20.9.2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sugimoto M, Nakamura T, Ohtani N, et al. Regulation of CDK4 activity by a novel CDK4-binding protein, p34(SEI-1) Genes Dev. 1999;13(22):3027–3033. doi: 10.1101/gad.13.22.3027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hong SW, Kim CJ, Park WS, et al. p34SEI-1 inhibits apoptosis through the stabilization of the X-linked inhibitor of apoptosis protein: p34SEI-1 as a novel target for anti-breast cancer strategies. Cancer Res. 2009;69(3):741–746. doi: 10.1158/0008-5472.CAN-08-1189. [DOI] [PubMed] [Google Scholar]

Articles from OncoTargets and therapy are provided here courtesy of Dove Press

RESOURCES